Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
Nat Immunol ; 18(7): 744-752, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28553952

RESUMO

The single-nucleotide polymorphism rs1990760 in the gene encoding the cytosolic viral sensor IFIH1 results in an amino-acid change (A946T; IFIH1T946) that is associated with multiple autoimmune diseases. The effect of this polymorphism on both viral sensing and autoimmune pathogenesis remains poorly understood. Here we found that human peripheral blood mononuclear cells (PBMCs) and cell lines expressing the risk variant IFIH1T946 exhibited heightened basal and ligand-triggered production of type I interferons. Consistent with those findings, mice with a knock-in mutation encoding IFIH1T946 displayed enhanced basal expression of type I interferons, survived a lethal viral challenge and exhibited increased penetrance in autoimmune models, including a combinatorial effect with other risk variants. Furthermore, IFIH1T946 mice manifested an embryonic survival defect consistent with enhanced responsiveness to RNA self ligands. Together our data support a model wherein the production of type I interferons driven by an autoimmune risk variant and triggered by ligand functions to protect against viral challenge, which probably accounts for its selection within human populations but provides this advantage at the cost of modestly promoting the risk of autoimmunity.


Assuntos
Autoimunidade/genética , Infecções por Cardiovirus/genética , Interferon Tipo I/imunologia , Helicase IFIH1 Induzida por Interferon/genética , Adolescente , Adulto , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Southern Blotting , Infecções por Cardiovirus/imunologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Vírus da Encefalomiocardite/imunologia , Feminino , Predisposição Genética para Doença , Células HEK293 , Humanos , Immunoblotting , Helicase IFIH1 Induzida por Interferon/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Viroses/genética , Viroses/imunologia , Adulto Jovem
2.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28916264

RESUMO

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Assuntos
Imunidade Inata , Proteínas de Membrana/metabolismo , Transporte de RNA , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Linhagem Celular , Citoplasma , Proteína DEAD-box 58/metabolismo , Modelos Animais de Doenças , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Endossomos/metabolismo , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Herpes Simples/genética , Herpes Simples/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Lisossomos/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Transporte de Nucleotídeos , Ligação Proteica , Transporte Proteico , RNA Viral/genética , RNA Viral/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo
3.
Nat Immunol ; 13(11): 1055-62, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23042151

RESUMO

Toll-like receptors (TLRs) sense pathogen-associated molecules and respond by inducing cytokines and type I interferon. Here we show that genetic ablation of the E3 ubiquitin ligase Pellino3 augmented the expression of type I interferon but not of proinflammatory cytokines in response to TLR3 activation. Pellino3-deficient mice had greater resistance against the pathogenic and lethal effects of encephalomyocarditis virus (EMCV). TLR3 signaling induced Pellino3, which in turn interacted with and ubiquitinated TRAF6. This modification suppressed the ability of TRAF6 to interact with and activate IRF7, resulting in downregulation of type I interferon expression. Our findings highlight a new physiological role for Pellino3 and define a new autoregulatory network for controlling type I interferon expression.


Assuntos
Infecções por Cardiovirus/imunologia , Regulação da Expressão Gênica , Interferon Tipo I/imunologia , Receptor 3 Toll-Like/imunologia , Ubiquitina-Proteína Ligases/imunologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/mortalidade , Infecções por Cardiovirus/virologia , Vírus da Encefalomiocardite/imunologia , Homeostase , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/genética , Camundongos , Camundongos Knockout , Transdução de Sinais , Taxa de Sobrevida , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/imunologia , Receptor 3 Toll-Like/genética , Ubiquitina/genética , Ubiquitina/imunologia , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Ubiquitinação
4.
PLoS Pathog ; 16(4): e1008457, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32251420

RESUMO

The retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), including RIG-I and melanoma differentiation-associated gene 5 (MDA5), sense cytoplasmic viral RNA and initiate innate antiviral responses. How RIG-I and MDA5 are differentially regulated remains enigmatic. In this study, we identified the guanylate-binding protein (GBP) and zinc-finger FYVE domain-containing protein ZFYVE1 as a negative regulator of MDA5- but not RIG-I-mediated innate antiviral responses. ZFYVE1-deficiency promoted MDA5- but not RIG-I-mediated transcription of downstream antiviral genes. Comparing to wild-type mice, Zfyve1-/- mice were significantly protected from lethality induced by encephalomyocarditis virus (EMCV) that is sensed by MDA5, whereas Zfyve1-/- and Zfyve1+/+ mice were comparable to death induced by vesicular stomatitis virus (VSV) that is sensed by RIG-I. Mechanistically, ZFYVE1 interacted with MDA5 but not RIG-I. ZFYVE1 bound to viral RNA and decreased the ligand binding and oligomerization of MDA5. These findings suggest that ZFYVE1 acts as a specific negative regulator of MDA5-mediated innate immune responses by inhibiting its ligand binding and oligomerization.


Assuntos
Infecções por Cardiovirus/imunologia , Proteína DEAD-box 58/imunologia , Vírus da Encefalomiocardite/fisiologia , Helicase IFIH1 Induzida por Interferon/imunologia , Proteínas de Membrana/imunologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Proteína DEAD-box 58/genética , Vírus da Encefalomiocardite/genética , Humanos , Imunidade Inata , Helicase IFIH1 Induzida por Interferon/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
5.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34768809

RESUMO

Virus-induced neurological sequelae resulting from infection by Theiler's murine encephalomyelitis virus (TMEV) are used for studying human conditions ranging from epileptic seizures to demyelinating disease. Mouse strains are typically considered susceptible or resistant to TMEV infection based on viral persistence and extreme phenotypes, such as demyelination. We have identified a broader spectrum of phenotypic outcomes by infecting strains of the genetically diverse Collaborative Cross (CC) mouse resource. We evaluated the chronic-infection gene expression profiles of hippocampi and thoracic spinal cords for 19 CC strains in relation to phenotypic severity and TMEV persistence. Strains were clustered based on similar phenotypic profiles and TMEV levels at 90 days post-infection, and we categorized distinct TMEV response profiles. The three most common profiles included "resistant" and "susceptible," as before, as well as a "resilient" TMEV response group which experienced both TMEV persistence and mild neurological phenotypes even at 90 days post-infection. Each profile had a distinct gene expression signature, allowing the identification of pathways and networks specific to each TMEV response group. CC founder haplotypes for genes involved in these pathways/networks revealed candidate response-specific alleles. These alleles demonstrated pleiotropy and epigenetic (miRNA) regulation in long-term TMEV infection, with particular relevance for resilient mouse strains.


Assuntos
Infecções por Cardiovirus/genética , Regulação da Expressão Gênica , Hipocampo/metabolismo , Medula Espinal/metabolismo , Theilovirus , Animais , Doenças Desmielinizantes , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Masculino , Camundongos , Análise de Sequência de RNA
6.
PLoS Pathog ; 14(2): e1006886, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29462185

RESUMO

Melanoma differentiation-associated gene-5 (MDA5) recognizes distinct subsets of viruses including Encephalomyocarditis virus (EMCV) of picornavirus family, but the molecular mechanisms underlying the specificity of the viral recognition of MDA5 in immune cells remain obscure. DHX29 is an RNA helicase required for the translation of 5' structured mRNA of host and many picornaviruses (such as EMCV). We identify that DXH29 as a key RNA co-sensor, plays a significant role for specific recognition and triggering anti-EMCV immunity. We have observed that DHX29 regulates MDA5-, but not RIG-I-, mediated type I interferon signaling by preferentially interacting with structured RNAs and specifically with MDA5 for enhancing MDA5-dsRNA binding affinity. Overall, our results identify a critical role for DHX29 in innate immune response and provide molecular insights into the mechanisms by which DHX29 recognizes 5' structured EMCV RNA and interacts with MDA5 for potent type I interferon signaling and antiviral immunity.


Assuntos
Infecções por Cardiovirus/imunologia , Vírus da Encefalomiocardite/imunologia , Imunidade Inata/genética , Helicase IFIH1 Induzida por Interferon/fisiologia , RNA Helicases/fisiologia , RNA Viral/imunologia , Animais , Infecções por Cardiovirus/genética , Células Cultivadas , Chlorocebus aethiops , Vírus da Encefalomiocardite/genética , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , RNA Helicases/genética , RNA Viral/genética , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Células Vero
7.
PLoS Pathog ; 14(10): e1007307, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30308076

RESUMO

As antimicrobial signalling molecules, type III or lambda interferons (IFNλs) are critical for defence against infection by diverse pathogens, including bacteria, fungi and viruses. Counter-intuitively, expression of one member of the family, IFNλ4, is associated with decreased clearance of hepatitis C virus (HCV) in the human population; by contrast, a natural frameshift mutation that abrogates IFNλ4 production improves HCV clearance. To further understand how genetic variation between and within species affects IFNλ4 function, we screened a panel of all known extant coding variants of human IFNλ4 for their antiviral potential and identify three that substantially affect activity: P70S, L79F and K154E. The most notable variant was K154E, which was found in African Congo rainforest 'Pygmy' hunter-gatherers. K154E greatly enhanced in vitro activity in a range of antiviral (HCV, Zika virus, influenza virus and encephalomyocarditis virus) and gene expression assays. Remarkably, E154 is the ancestral residue in mammalian IFNλ4s and is extremely well conserved, yet K154 has been fixed throughout evolution of the hominid genus Homo, including Neanderthals. Compared to chimpanzee IFNλ4, the human orthologue had reduced activity due to amino acid K154. Comparison of published gene expression data from humans and chimpanzees showed that this difference in activity between K154 and E154 in IFNλ4 correlates with differences in antiviral gene expression in vivo during HCV infection. Mechanistically, our data show that the human-specific K154 negatively affects IFNλ4 activity through a novel means by reducing its secretion and potency. We thus demonstrate that attenuated activity of IFNλ4 is conserved among humans and postulate that differences in IFNλ4 activity between species contribute to distinct host-specific responses to-and outcomes of-infection, such as HCV infection. The driver of reduced IFNλ4 antiviral activity in humans remains unknown but likely arose between 6 million and 360,000 years ago in Africa.


Assuntos
Antivirais/uso terapêutico , Infecções por Cardiovirus/tratamento farmacológico , Hepatite C/tratamento farmacológico , Interleucinas/genética , Polimorfismo de Nucleotídeo Único , Infecção por Zika virus/tratamento farmacológico , Animais , Evolução Biológica , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Células Cultivadas , Vírus da Encefalomiocardite/efeitos dos fármacos , Vírus da Encefalomiocardite/isolamento & purificação , Regulação da Expressão Gênica , Hepacivirus/efeitos dos fármacos , Hepacivirus/isolamento & purificação , Hepatite C/genética , Hepatite C/virologia , Humanos , Pan troglodytes , Especificidade da Espécie , Zika virus/efeitos dos fármacos , Zika virus/isolamento & purificação , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
8.
Proc Natl Acad Sci U S A ; 113(37): 10388-93, 2016 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-27582466

RESUMO

Despite the breadth of knowledge that exists regarding the function of long noncoding RNAs (lncRNAs) in biological phenomena, the role of lncRNAs in host antiviral responses is poorly understood. Here, we report that lncRNA#32 is associated with type I IFN signaling. The silencing of lncRNA#32 dramatically reduced the level of IFN-stimulated gene (ISG) expression, resulting in sensitivity to encephalomyocarditis virus (EMCV) infection. In contrast, the ectopic expression of lncRNA#32 significantly suppressed EMCV replication, suggesting that lncRNA#32 positively regulates the host antiviral response. We further demonstrated the suppressive function of lncRNA#32 in hepatitis B virus and hepatitis C virus infection. lncRNA#32 bound to activating transcription factor 2 (ATF2) and regulated ISG expression. Our results reveal a role for lncRNA#32 in host antiviral responses.


Assuntos
Fator 2 Ativador da Transcrição/genética , Interações Hospedeiro-Patógeno/genética , Interferon Tipo I/genética , RNA Longo não Codificante/genética , Fator 2 Ativador da Transcrição/metabolismo , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Linhagem Celular Tumoral , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/patogenicidade , Regulação da Expressão Gênica , Inativação Gênica , Hepacivirus/genética , Hepacivirus/patogenicidade , Hepatite C/genética , Hepatite C/virologia , Hepatócitos/metabolismo , Hepatócitos/virologia , Humanos , Interferon Tipo I/metabolismo , RNA Longo não Codificante/metabolismo , Transdução de Sinais/genética , Replicação Viral/genética
9.
Biochem Biophys Res Commun ; 494(1-2): 227-233, 2017 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-29032202

RESUMO

Laboratory of genetics and physiology 2 (LGP2) and melanoma differentiation-associated gene 5 (MDA5) cooperatively detect viral RNA in the cytoplasm of Cardiovirus-infected cells and activate innate immune responses. Here, we evaluated whether the double-stranded RNA-binding protein PACT plays a role in this anti-viral response to further elucidate the mechanism. Immunoprecipitation experiments demonstrated that PACT interacts with LGP2 and that this interaction is enhanced by encephalomyocarditis virus (EMCV) infection. In vitro interaction analyses using purified recombinant proteins confirmed that the single-stranded Theiler's murine encephalitis virus genome enhanced the interaction between LGP2 and PACT. Small interfering RNA knockdown experiments further indicated that PACT is required for Cardiovirus-triggered interferon responses. To support this functional interaction with LGP2, overexpressed PACT was shown to enhance EMCV-triggered interferon promoter activity only when LGP2 and MDA5 were co-expressed but not when MDA5 is expressed alone. Together, our findings indicate a possible role of PACT in regulating the Cardiovirus-triggered immune responses mediated by MDA5 and LGP2, which opens the door to novel therapeutic strategies in interferon-related autoimmune diseases and cancer.


Assuntos
Infecções por Cardiovirus/imunologia , Vírus da Encefalomiocardite , Helicase IFIH1 Induzida por Interferon/imunologia , RNA Helicases/imunologia , Proteínas de Ligação a RNA/imunologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Linhagem Celular , Chlorocebus aethiops , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/imunologia , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Técnicas de Silenciamento de Genes , Células HEK293 , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Imunidade Inata/genética , Helicase IFIH1 Induzida por Interferon/genética , Interferon beta/genética , Camundongos , Regiões Promotoras Genéticas , RNA Helicases/genética , RNA Interferente Pequeno/genética , RNA Viral/genética , RNA Viral/imunologia , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Ribonuclease III/antagonistas & inibidores , Ribonuclease III/genética , Ribonuclease III/imunologia , Células Vero
10.
J Immunol ; 194(6): 2796-809, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25681345

RESUMO

Virus-induced myositis is an emerging global affliction that remains poorly characterized with few treatment options. Moreover, muscle-tropic viruses often spread to the CNS, causing dramatically increased morbidity. Therefore, there is an urgent need to explore genetic factors involved in this class of human disease. This report investigates critical innate immune pathways affecting murine virus-induced myositis. Of particular importance, the key immune regulator src homology region 2 domain-containing phosphatase 1 (SHP-1), which normally suppresses macrophage-mediated inflammation, is a major factor in promoting clinical disease in muscle. We show that Theiler's murine encephalomyelitis virus (TMEV) infection of skeletal myofibers induces inflammation and subsequent dystrophic calcification, with loss of ambulation in wild-type (WT) mice. Surprisingly, although similar extensive myofiber infection and inflammation are observed in SHP-1(-/-) mice, these mice neither accumulate dead calcified myofibers nor lose ambulation. Macrophages were the predominant effector cells infiltrating WT and SHP-1(-/-) muscle, and an increased infiltration of immature monocytes/macrophages correlated with an absence of clinical disease in SHP-1(-/-) mice, whereas mature M1-like macrophages corresponded with increased myofiber degeneration in WT mice. Furthermore, blocking SHP-1 activation in WT macrophages blocked virus-induced myofiber degeneration, and pharmacologic ablation of macrophages inhibited muscle calcification in TMEV-infected WT animals. These data suggest that, following TMEV infection of muscle, SHP-1 promotes M1 differentiation of infiltrating macrophages, and these inflammatory macrophages are likely involved in damaging muscle fibers. These findings reveal a pathological role for SHP-1 in promoting inflammatory macrophage differentiation and myofiber damage in virus-infected skeletal muscle, thus identifying SHP-1 and M1 macrophages as essential mediators of virus-induced myopathy.


Assuntos
Infecções por Cardiovirus/imunologia , Diferenciação Celular/imunologia , Macrófagos/imunologia , Miosite/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Theilovirus/imunologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/farmacologia , Antígeno CD47/imunologia , Antígeno CD47/metabolismo , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Diferenciação Celular/genética , Citometria de Fluxo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/virologia , Miosite/genética , Miosite/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Proteína Tirosina Fosfatase não Receptora Tipo 6/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Theilovirus/fisiologia , Transcriptoma/imunologia , Replicação Viral/imunologia
11.
J Neurovirol ; 22(1): 66-79, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26260496

RESUMO

Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) is an important model of the progressive disability caused by irreversible CNS tissue injury, and provides an example of how a CNS pathogen can cause inflammation, demyelination, and neuronal damage. We were interested in which molecules, especially inflammatory mediators, might be upregulated in the CNS throughout TMEV-IDD. We quantitated by a real-time RT-PCR multi-gene system the expression of a pathway-focused panel of genes at 30 and 165 days post infection, characterizing both the early inflammatory and the late neurodegenerative stages of TMEV-IDD. Also, we measured 32 cytokines/chemokines by multiplex Luminex analysis in CSF specimens from early and late TMEV-IDD as well as sham-treated mice. Results indicate that, in the later stage of TMEV-IDD, activation of the innate immune response is most prominent: TLRs, type I IFN response genes, and innate immunity-associated cytokines were highly expressed in late TMEV-IDD compared to sham (p ≤ 0.0001) and early TMEV-IDD (p < 0.05). Conversely, several molecular mediators of adaptive immune response were highly expressed in early TMEV-IDD (all p ≤ 0.001). Protein detection in the CSF was broadly concordant with mRNA abundance of the corresponding gene measured by real-time RT-PCR in the spinal cord, since several cytokines/chemokines were increased in the CSF of TMEV-IDD mice. Results show a clear shift from adaptive to innate immunity from early to late TMEV-IDD, indicating that adaptive and innate immune pathways are likely involved in the development and progression of the disease to different extents. CSF provides an optimal source of biomarkers of CNS neuroinflammation.


Assuntos
Imunidade Adaptativa , Infecções por Cardiovirus/imunologia , Doenças Desmielinizantes/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Animais , Infecções por Cardiovirus/líquido cefalorraquidiano , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Citocinas/líquido cefalorraquidiano , Citocinas/genética , Citocinas/imunologia , Doenças Desmielinizantes/líquido cefalorraquidiano , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/virologia , Progressão da Doença , Regulação da Expressão Gênica , Inflamação , Camundongos , Anotação de Sequência Molecular , RNA Mensageiro/líquido cefalorraquidiano , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Theilovirus/crescimento & desenvolvimento , Theilovirus/imunologia , Theilovirus/patogenicidade , Fatores de Tempo , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
12.
Exp Mol Pathol ; 100(3): 434-40, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27079771

RESUMO

The present study was carried out to clarify the mechanisms of EMC virus-induced sialodacryoadenitis in mice during the acute phase infection focusing on the activation of type I interferon (IFN) signaling in the parotid and exorbital lachrymal glands. In the parotid gland, a few apoptotic acinar cells were detected at 2days post inoculation (DPI). The ratio of apoptotic acinar cells increased at 3 and 4DPI. On the other hand, in the exorbital lachrymal gland, apoptosis of acinar cells and infiltration of inflammatory cells mainly composed of mononuclear cells started at 3DPI, and prominent acinar cell damage developed at 4DPI. Viral RNA was detected at 3 and 4DPI in both glands and the expression level was higher in the exorbital lachrymal gland than in the parotid gland. The up-regulation of IFN-stimulated genes (ISGs), such as Irf7, Pkr and Oas, was quickly induced at 2DPI in the parotid gland, and this probably contributed to suppress viral replication and to eliminate affected cells by apoptosis. In the exorbital lachrymal gland, the expression levels of ISGs mRNAs were not elevated at 2DPI, suggesting no induction of an effective anti-viral response such as apoptosis at this time point. In the exorbital lachrymal gland, the mRNA expression of IFN beta and IFN alpha (type I IFNs) was weak- to strong-positive at 1DPI, and became negative at 2DPI. The weak- to strong-positive expression of IFNs at 1DPI is likely related to the abrupt viral replication and pathological changes in the exorbital lachrymal gland through activating the negative feedback regulation that depressed the IFN signaling cascade at 2DPI. In conclusion, the present study showed the changes in factors involved in the activation of type I IFN signaling cascade in the parotid and exorbital lachrymal glands and their differences between the two glands during the acute phase of EMC virus infection in mice.


Assuntos
Infecções por Cardiovirus/metabolismo , Interferon Tipo I/genética , Aparelho Lacrimal/metabolismo , Glândula Parótida/metabolismo , Transdução de Sinais/genética , Doença Aguda , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/fisiologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Fator Regulador 7 de Interferon/genética , Interferon-alfa/genética , Aparelho Lacrimal/virologia , Masculino , Camundongos Endogâmicos DBA , Glândula Parótida/virologia , RNA Viral/genética , RNA Viral/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
13.
J Virol ; 88(18): 10748-57, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25008915

RESUMO

UNLABELLED: Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are essential intracellular detectors of viral RNA. They contribute to the type I interferon (IFN) response that is crucial for host defense against viral infections. Given the potent antiviral and proinflammatory activities elicited by the type I IFNs, induction of the type I IFN response is tightly regulated. Members of the tripartite motif (TRIM) family of proteins have recently emerged as key regulators of antiviral immunity. We show that TRIM13, an E3 ubiquitin ligase, is expressed in immune cells and is upregulated in bone marrow-derived macrophages upon stimulation with inducers of type I IFN. TRIM13 interacts with MDA5 and negatively regulates MDA5-mediated type I IFN production in vitro, acting upstream of IFN regulatory factor 3. We generated Trim13(-/-) mice and show that upon lethal challenge with encephalomyocarditis virus (EMCV), which is sensed by MDA5, Trim13(-/-) mice produce increased amounts of type I IFNs and survive longer than wild-type mice. Trim13(-/-) murine embryonic fibroblasts (MEFs) challenged with EMCV or poly(I · C) also show a significant increase in beta IFN (IFN-ß) levels, but, in contrast, IFN-ß responses to the RIG-I-detected Sendai virus were diminished, suggesting that TRIM13 may play a role in positively regulating RIG-I function. Together, these results demonstrate that TRIM13 regulates the type I IFN response through inhibition of MDA5 activity and that it functions nonredundantly to modulate MDA5 during EMCV infection. IMPORTANCE: The type I interferon (IFN) response is crucial for host defense against viral infections, and proper regulation of this pathway contributes to maintaining immune homeostasis. Retinoic acid-inducible gene I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5) are intracellular detectors of viral RNA that induce the type I IFN response. In this study, we show that expression of the gene tripartite motif 13 (Trim13) is upregulated in response to inducers of type I IFN and that TRIM13 interacts with both MDA5 and RIG-I in vitro. Through the use of multiple in vitro and in vivo model systems, we show that TRIM13 is a negative regulator of MDA5-mediated type I IFN production and may also impact RIG-I-mediated type I IFN production by enhancing RIG-I activity. This places TRIM13 at a key junction within the viral response pathway and identifies it as one of the few known modulators of MDA5 activity.


Assuntos
Infecções por Cardiovirus/enzimologia , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Vírus da Encefalomiocardite/fisiologia , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/virologia , RNA Helicases DEAD-box/genética , Proteínas de Ligação a DNA/genética , Feminino , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Helicase IFIH1 Induzida por Interferon , Interferon-alfa/genética , Interferon beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/genética
14.
PLoS Pathog ; 9(6): e1003474, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23825954

RESUMO

Theiler's virus is a neurotropic picornavirus responsible for chronic infections of the central nervous system. The establishment of a persistent infection and the subsequent demyelinating disease triggered by the virus depend on the expression of L*, a viral accessory protein encoded by an alternative open reading frame of the virus. We discovered that L* potently inhibits the interferon-inducible OAS/RNase L pathway. The antagonism of RNase L by L* was particularly prominent in macrophages where baseline oligoadenylate synthetase (OAS) and RNase L expression levels are elevated, but was detectable in fibroblasts after IFN pretreatment. L* mutations significantly affected Theiler's virus replication in primary macrophages derived from wild-type but not from RNase L-deficient mice. L* counteracted the OAS/RNase L pathway through direct interaction with the ankyrin domain of RNase L, resulting in the inhibition of this enzyme. Interestingly, RNase L inhibition was species-specific as Theiler's virus L* protein blocked murine RNase L but not human RNase L or RNase L of other mammals or birds. Direct RNase L inhibition by L* and species specificity were confirmed in an in vitro assay performed with purified proteins. These results demonstrate a novel viral mechanism to elude the antiviral OAS/RNase L pathway. By targeting the effector enzyme of this antiviral pathway, L* potently inhibits RNase L, underscoring the importance of this enzyme in innate immunity against Theiler's virus.


Assuntos
Infecções por Cardiovirus/metabolismo , Endorribonucleases/antagonistas & inibidores , Evasão da Resposta Imune/fisiologia , Imunidade Inata , Theilovirus/metabolismo , Proteínas Virais/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/patologia , Linhagem Celular , Cricetinae , Endorribonucleases/genética , Endorribonucleases/imunologia , Endorribonucleases/metabolismo , Humanos , Camundongos , Camundongos Mutantes , Estrutura Terciária de Proteína , Especificidade da Espécie , Theilovirus/genética , Theilovirus/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
15.
J Virol ; 87(11): 6314-25, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536668

RESUMO

Virus infection can initiate a type I interferon (IFN-α/ß) response via activation of the cytosolic RNA sensors retinoic acid-inducible gene-I (RIG-I) and melanoma differentiation-associated gene 5 (MDA5). Furthermore, it can activate kinases that phosphorylate eukaryotic translation initiation factor 2α (eIF2α), which leads to inhibition of (viral) protein translation and formation of stress granules (SG). Most viruses have evolved mechanisms to suppress these cellular responses. Here, we show that a mutant mengovirus expressing an inactive leader (L) protein, which we have previously shown to be unable to suppress IFN-α/ß, triggered SG formation in a protein kinase R (PKR)-dependent manner. Furthermore, we show that infection of cells that are defective in SG formation yielded higher viral RNA levels, suggesting that SG formation acts as an antiviral defense mechanism. Since the induction of both IFN-α/ß and SG is suppressed by mengovirus L, we set out to investigate a potential link between these pathways. We observed that MDA5, the intracellular RNA sensor that recognizes picornaviruses, localized to SG. However, activation of the MDA5 signaling pathway did not trigger and was not required for SG formation. Moreover, cells that were unable to form SG-by protein kinase R (PKR) depletion, using cells expressing a nonphosphorylatable eIF2α protein, or by drug treatment that inhibits SG formation-displayed a normal IFN-α/ß response. Thus, although MDA5 localizes to SG, this localization seems to be dispensable for induction of the IFN-α/ß pathway.


Assuntos
Infecções por Cardiovirus/enzimologia , Grânulos Citoplasmáticos/enzimologia , RNA Helicases DEAD-box/metabolismo , Interferon-alfa/genética , Interferon beta/genética , Mengovirus/fisiologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Grânulos Citoplasmáticos/genética , RNA Helicases DEAD-box/genética , Humanos , Helicase IFIH1 Induzida por Interferon , Interferon-alfa/metabolismo , Interferon beta/metabolismo , Mengovirus/genética , Camundongos , Camundongos Knockout , Transporte Proteico
16.
J Virol ; 87(11): 6517-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536659

RESUMO

The leader (L) protein of encephalomyocarditis virus (EMCV) shuts off host cell nucleocytoplasmic trafficking (NCT) by inducing hyperphosphorylation of nuclear pore proteins. This dramatic effect by a nonenzymatic protein of 6 kDa is not well understood but clearly involves L binding to cellular Ran GTPase, a critical factor of active NCT. Exogenous GDP and GTP are inhibitory to L-Ran binding, but the guanine-nucleotide exchange factor RCC1 can relieve this inhibition. In the presence of RCC1, L binds Ran with a KD (equilibrium dissociation constant) of ≈ 3 nM and reaches saturation within 20 min. The results of fluorescently tagged nucleotide experiments suggest that L-Ran interactions affect the nucleotide-binding pocket of Ran.


Assuntos
Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Proteínas de Ciclo Celular/metabolismo , Vírus da Encefalomiocardite/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Virais/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Infecções por Cardiovirus/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Vírus da Encefalomiocardite/genética , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/genética , Interações Hospedeiro-Patógeno , Humanos , Cinética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Virais/química , Proteínas Virais/genética , Proteína ran de Ligação ao GTP/química , Proteína ran de Ligação ao GTP/genética
17.
PLoS Pathog ; 8(8): e1002857, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22916014

RESUMO

Nod-like receptors (NLRs) comprise a large family of intracellular pattern- recognition receptors. Members of the NLR family assemble into large multiprotein complexes, termed the inflammasomes. The NLR family, pyrin domain-containing 3 (NLRP3) is triggered by a diverse set of molecules and signals, and forms the NLRP3 inflammasome. Recent studies have indicated that both DNA and RNA viruses stimulate the NLRP3 inflammasome, leading to the secretion of interleukin 1 beta (IL-1ß) and IL-18 following the activation of caspase-1. We previously demonstrated that the proton-selective ion channel M2 protein of influenza virus activates the NLRP3 inflammasome. However, the precise mechanism by which NLRP3 recognizes viral infections remains to be defined. Here, we demonstrate that encephalomyocarditis virus (EMCV), a positive strand RNA virus of the family Picornaviridae, activates the NLRP3 inflammasome in mouse dendritic cells and macrophages. Although transfection with RNA from EMCV virions or EMCV-infected cells induced robust expression of type I interferons in macrophages, it failed to stimulate secretion of IL-1ß. Instead, the EMCV viroporin 2B was sufficient to cause inflammasome activation in lipopolysaccharide-primed macrophages. While cells untransfected or transfected with the gene encoding the EMCV non-structural protein 2A or 2C expressed NLRP3 uniformly throughout the cytoplasm, NLRP3 was redistributed to the perinuclear space in cells transfected with the gene encoding the EMCV 2B or influenza virus M2 protein. 2B proteins of other picornaviruses, poliovirus and enterovirus 71, also caused the NLRP3 redistribution. Elevation of the intracellular Ca(2+) level, but not mitochondrial reactive oxygen species and lysosomal cathepsin B, was important in EMCV-induced NLRP3 inflammasome activation. Chelation of extracellular Ca(2+) did not reduce virus-induced IL-1ß secretion. These results indicate that EMCV activates the NLRP3 inflammasome by stimulating Ca(2+) flux from intracellular storages to the cytosol, and highlight the importance of viroporins, transmembrane pore-forming viral proteins, in virus-induced NLRP3 inflammasome activation.


Assuntos
Cálcio/metabolismo , Infecções por Cardiovirus/metabolismo , Proteínas de Transporte/metabolismo , Vírus da Encefalomiocardite/metabolismo , Inflamassomos/metabolismo , Proteínas Virais/metabolismo , Animais , Infecções por Cardiovirus/genética , Proteínas de Transporte/genética , Vírus da Encefalomiocardite/genética , Células HEK293 , Células HeLa , Humanos , Inflamassomos/genética , Interleucina-18/genética , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR , RNA Viral/genética , RNA Viral/metabolismo , Proteínas Virais/genética , Vírion/genética , Vírion/metabolismo
18.
J Neurovirol ; 20(5): 485-95, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25052192

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) induces demyelination in susceptible strains of mice (SJL/J) through an immunopathological process that is mediated by CD4(+) Th1 T cell. These T cells are chemoattracted to the central nervous system by chemokines. Hence, in this study, we focused on the production of the chemokine "interferon-gamma-inducible protein 10 kDa," or IP-10/CXCL10, by cultured SJL/J mouse astrocytes infected with the BeAn strain of TMEV and its capacity to attract activated T cells. The analysis of the whole murine genome by DNA hybridization with cRNAs from mock- and TMEV-infected cultures revealed the upregulation of six sequences that potentially encode for CXCL10. This increased CXCL10 expression was validated by PCR and qPCR. The presence of this chemokine was further demonstrated by enzyme-linked immunoassay (ELISA). Significantly, astrocytes from BALB/c mice, a strain resistant to demyelination, did not produce CXCL10. The secreted CXCL10 was biologically active, inducing chemoattraction of activated lymphocytes. The inflammatory cytokines, IL-1α, IFN-γ, and TNF-α, were strong inducers of CXCL10 in astrocytes. Serum from TMEV-infected SJL/J but not BALB/c mice contains CXCL10, the levels of which peak at the onset of the clinical disease. Finally, this in vitro inflammation model was fully inhibited by 17ß-estradiol and four selective estrogen receptor modulators, as demonstrated by ELISA and qPCR.


Assuntos
Astrócitos/virologia , Infecções por Cardiovirus/imunologia , Quimiocina CXCL10/biossíntese , Linfócitos T/imunologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Western Blotting , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/metabolismo , Células Cultivadas , Quimiocina CXCL10/genética , Ensaio de Imunoadsorção Enzimática , Hibridização In Situ , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Theilovirus/imunologia , Regulação para Cima
19.
Microbiol Immunol ; 58(7): 361-74, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24866463

RESUMO

Deletion of cellular isoform of prion protein (PrP(C)) increases neuronal predisposition to damage by modulating apoptosis and the negative consequences of oxidative stress. In vivo studies have demonstrated that PrP(C)-deficient mice are more prone to seizure, depression, and induction of epilepsy and experience extensive cerebral damage following ischemic challenge or viral infection. In addition, adenovirus-mediated overexpression of PrP(C) reduces brain damage in rat models of cerebral ischemia. In experimental autoimmune encephalomyelitis, PrP(C)-deficient mice reportedly have a more aggressive disease onset and less clinical improvement during the chronic phase than wild-type mice mice. In mice given oral dextran sulfate, PrP(C) has a potential protective role against inflammatory bowel disease. PrP(C)-deficient mice demonstrate significantly greater increases in blood glucose concentrations after intraperitoneal injection of glucose than wild-type mice. Further in vivo challenges to PrP gene-deficient models and conditional knockout models with siRNA and in vivo administration of PrP-ligating agents may assist in refining knowledge of the lymphoid function of PrP(C) and predicting the effects of anti-PrP treatment on the immune system. Together, these findings indicate that PrP(C) may have multiple neuroprotective and anti-inflammatory roles, which explains why this protein is so widely expressed.


Assuntos
Suscetibilidade a Doenças , Príons/genética , Estresse Fisiológico , Animais , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Isquemia Encefálica/genética , Isquemia Encefálica/imunologia , Isquemia Encefálica/metabolismo , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Colite/genética , Colite/imunologia , Colite/metabolismo , Depressão/genética , Depressão/imunologia , Depressão/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Epilepsia/genética , Epilepsia/imunologia , Epilepsia/metabolismo , Técnicas de Inativação de Genes , Humanos , Imunidade/genética , Camundongos , Camundongos Knockout , Príons/metabolismo , Interferência de RNA
20.
J Biol Chem ; 287(52): 43651-64, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23109342

RESUMO

Autophagy is a tightly regulated mechanism that mediates sequestration, degradation, and recycling of cellular proteins, organelles, and pathogens. Several proteins associated with autophagy regulate host responses to viral infections. Ribonuclease L (RNase L) is activated during viral infections and cleaves cellular and viral single-stranded RNAs, including rRNAs in ribosomes. Here we demonstrate that direct activation of RNase L coordinates the activation of c-Jun N-terminal kinase (JNK) and double-stranded RNA-dependent protein kinase (PKR) to induce autophagy with hallmarks as accumulation of autophagic vacuoles, p62(SQSTM1) degradation and conversion of Microtubule-associated Protein Light Chain 3-I (LC3-I) to LC3-II. Accordingly, treatment of cells with pharmacological inhibitors of JNK or PKR and mouse embryonic fibroblasts (MEFs) lacking JNK1/2 or PKR showed reduced autophagy levels. Furthermore, RNase L-induced JNK activity promoted Bcl-2 phosphorylation, disrupted the Beclin1-Bcl-2 complex and stimulated autophagy. Viral infection with Encephalomyocarditis virus (EMCV) or Sendai virus led to higher levels of autophagy in wild-type (WT) MEFs compared with RNase L knock out (KO) MEFs. Inhibition of RNase L-induced autophagy using Bafilomycin A1 or 3-methyladenine suppressed viral growth in initial stages; in later stages autophagy promoted viral replication dampening the antiviral effect. Induction of autophagy by activated RNase L is independent of the paracrine effects of interferon (IFN). Our findings suggest a novel role of RNase L in inducing autophagy affecting the outcomes of viral pathogenesis.


Assuntos
Autofagia , Endorribonucleases/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , eIF-2 Quinase/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/patologia , Linhagem Celular Tumoral , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Vírus da Encefalomiocardite/fisiologia , Endorribonucleases/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Quinase 8 Ativada por Mitógeno/genética , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA de Cadeia Dupla/genética , RNA de Cadeia Dupla/metabolismo , Infecções por Respirovirus/genética , Infecções por Respirovirus/metabolismo , Infecções por Respirovirus/patologia , Vírus Sendai/fisiologia , Proteína Sequestossoma-1 , Replicação Viral/fisiologia , eIF-2 Quinase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA