Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(4): e1012133, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38662794

RESUMO

The discovery that extracellular vesicles (EVs) serve as carriers of virus particles calls for a reevaluation of the release strategies of non-enveloped viruses. Little is currently known about the molecular mechanisms that determine the release and composition of EVs produced by virus-infected cells, as well as conservation of these mechanisms among viruses. We previously described an important role for the Leader protein of the picornavirus encephalomyocarditis virus (EMCV) in the induction of virus-carrying EV subsets with distinct molecular and physical properties. EMCV L acts as a 'viral security protein' by suppressing host antiviral stress and type-I interferon (IFN) responses. Here, we tested the ability of functionally related picornavirus proteins of Theilers murine encephalitis virus (TMEV L), Saffold virus (SAFV L), and coxsackievirus B3 (CVB3 2Apro), to rescue EV and EV-enclosed virus release when introduced in Leader-deficient EMCV. We show that all viral security proteins tested were able to promote virus packaging in EVs, but that only the expression of EMCV L and CVB3 2Apro increased overall EV production. We provide evidence that one of the main antiviral pathways counteracted by this class of picornaviral proteins, i.e. the inhibition of PKR-mediated stress responses, affected EV and EV-enclosed virus release during infection. Moreover, we show that the enhanced capacity of the viral proteins EMCV L and CVB3 2Apro to promote EV-enclosed virus release is linked to their ability to simultaneously promote the activation of the stress kinase P38 MAPK. Taken together, we demonstrate that cellular stress pathways involving the kinases PKR and P38 are modulated by the activity of non-structural viral proteins to increase the release EV-enclosed viruses during picornavirus infections. These data shed new light on the molecular regulation of EV production in response to virus infection.


Assuntos
Vesículas Extracelulares , Picornaviridae , Proteínas Virais , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/virologia , Humanos , Picornaviridae/metabolismo , Picornaviridae/fisiologia , Proteínas Virais/metabolismo , Proteínas Virais/genética , Animais , eIF-2 Quinase/metabolismo , Liberação de Vírus/fisiologia , Camundongos , Theilovirus/metabolismo , Infecções por Cardiovirus/virologia , Infecções por Cardiovirus/metabolismo , Vírus da Encefalomiocardite/metabolismo , Vírus da Encefalomiocardite/fisiologia
2.
J Immunol ; 206(1): 154-163, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33219146

RESUMO

Viral RNA in the cytoplasm of mammalian host cells is recognized by retinoic acid-inducible protein-I-like receptors (RLRs), which localize to cytoplasmic stress granules (SGs). Activated RLRs associate with the mitochondrial adaptor protein IPS-1, which activates antiviral host defense mechanisms, including type I IFN induction. It has remained unclear, however, how RLRs in SGs and IPS-1 in the mitochondrial outer membrane associate physically and engage in information transfer. In this study, we show that NUDT21, an RNA-binding protein that regulates alternative transcript polyadenylation, physically associates with IPS-1 and mediates its localization to SGs in response to transfection with polyinosinic-polycytidylic acid [poly(I:C)], a mimic of viral dsRNA. We found that despite its well-established function in the nucleus, a fraction of NUDT21 localizes to mitochondria in resting cells and becomes localized to SGs in response to poly(I:C) transfection. NUDT21 was also found to be required for efficient type I IFN induction in response to viral infection in both human HeLa cells and mouse macrophage cell line RAW264.7 cells. Our results together indicate that NUDT21 links RLRs in SGs to mitochondrial IPS-1 and thereby activates host defense responses to viral infection.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Infecções por Cardiovirus/metabolismo , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Proteína DEAD-box 58/metabolismo , Vírus da Encefalomiocardite/fisiologia , Mitocôndrias/metabolismo , Doença de Newcastle/metabolismo , Vírus da Doença de Newcastle/fisiologia , Receptores Imunológicos/metabolismo , Vesículas Secretórias/metabolismo , Animais , Fator de Especificidade de Clivagem e Poliadenilação/genética , Regulação da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Camundongos , Poli I-C/imunologia , Transporte Proteico , Células RAW 264.7 , RNA Interferente Pequeno/genética , RNA Viral/imunologia , Estresse Fisiológico
3.
J Biol Chem ; 295(52): 18189-18198, 2020 12 25.
Artigo em Inglês | MEDLINE | ID: mdl-33100269

RESUMO

Environmental factors, such as viral infection, are proposed to play a role in the initiation of autoimmune diabetes. In response to encephalomyocarditis virus (EMCV) infection, resident islet macrophages release the pro-inflammatory cytokine IL-1ß, to levels that are sufficient to stimulate inducible nitric oxide synthase (iNOS) expression and production of micromolar levels of the free radical nitric oxide in neighboring ß-cells. We have recently shown that nitric oxide inhibits EMCV replication and EMCV-mediated ß-cell lysis and that this protection is associated with an inhibition of mitochondrial oxidative metabolism. Here we show that the protective actions of nitric oxide against EMCV infection are selective for ß-cells and associated with the metabolic coupling of glycolysis and mitochondrial oxidation that is necessary for insulin secretion. Inhibitors of mitochondrial respiration attenuate EMCV replication in ß-cells, and this inhibition is associated with a decrease in ATP levels. In mouse embryonic fibroblasts (MEFs), inhibition of mitochondrial metabolism does not modify EMCV replication or decrease ATP levels. Like most cell types, MEFs have the capacity to uncouple the glycolytic utilization of glucose from mitochondrial respiration, allowing for the maintenance of ATP levels under conditions of impaired mitochondrial respiration. It is only when MEFs are forced to use mitochondrial oxidative metabolism for ATP generation that mitochondrial inhibitors attenuate viral replication. In a ß-cell selective manner, these findings indicate that nitric oxide targets the same metabolic pathways necessary for glucose stimulated insulin secretion for protection from viral lysis.


Assuntos
Infecções por Cardiovirus/tratamento farmacológico , Vírus da Encefalomiocardite/fisiologia , Sequestradores de Radicais Livres/farmacologia , Galactose/metabolismo , Glicólise , Ilhotas Pancreáticas/efeitos dos fármacos , Óxido Nítrico/farmacologia , Animais , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/virologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/virologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Estresse Oxidativo
4.
Int J Mol Sci ; 22(10)2021 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-34067536

RESUMO

Several virus-induced models were used to study the underlying mechanisms of multiple sclerosis (MS). The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) establishes persistent viral infections and induces chronic inflammatory demyelinating disease. In this review, the innate and adaptive immune responses to TMEV are discussed to better understand the pathogenic mechanisms of viral infections. Professional (dendritic cells (DCs), macrophages, and B cells) and non-professional (microglia, astrocytes, and oligodendrocytes) antigen-presenting cells (APCs) are the major cell populations permissive to viral infection and involved in cytokine production. The levels of viral loads and cytokine production in the APCs correspond to the degrees of susceptibility of the mice to the TMEV-induced demyelinating diseases. TMEV infection leads to the activation of cytokine production via TLRs and MDA-5 coupled with NF-κB activation, which is required for TMEV replication. These activation signals further amplify the cytokine production and viral loads, promote the differentiation of pathogenic Th17 responses, and prevent cellular apoptosis, enabling viral persistence. Among the many chemokines and cytokines induced after viral infection, IFN α/ß plays an essential role in the downstream expression of costimulatory molecules in APCs. The excessive levels of cytokine production after viral infection facilitate the pathogenesis of TMEV-induced demyelinating disease. In particular, IL-6 and IL-1ß play critical roles in the development of pathogenic Th17 responses to viral antigens and autoantigens. These cytokines, together with TLR2, may preferentially generate deficient FoxP3+CD25- regulatory cells converting to Th17. These cytokines also inhibit the apoptosis of TMEV-infected cells and cytolytic function of CD8+ T lymphocytes (CTLs) and prolong the survival of B cells reactive to viral and self-antigens, which preferentially stimulate Th17 responses.


Assuntos
Doenças Desmielinizantes/imunologia , Esclerose Múltipla/imunologia , Theilovirus/fisiologia , Imunidade Adaptativa/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Astrócitos/metabolismo , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/virologia , Citocinas , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Humanos , Imunidade Inata/imunologia , Camundongos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais/imunologia , Theilovirus/patogenicidade
5.
J Neuroinflammation ; 17(1): 307, 2020 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-33069239

RESUMO

BACKGROUND: Experimental autoimmune encephalitis (EAE) and virally induced demyelinating disease are two major experimental model systems used to study human multiple sclerosis. Although endothelin-1 level elevation was previously observed in the CNS of mice with EAE and viral demyelinating disease, the potential role of endothelin-1 in the development of these demyelinating diseases is unknown. METHODS AND RESULTS: In this study, the involvement of endothelin-1 in the development and progression of demyelinating diseases was investigated using these two experimental models. Administration of endothelin-1 significantly promoted the progression of both experimental diseases accompanied with elevated inflammatory T cell responses. In contrast, administration of specific endothelin-1 inhibitors (BQ610 and BQ788) significantly inhibited progression of these diseases accompanied with reduced T cell responses to the respective antigens. CONCLUSIONS: These results strongly suggest that the level of endothelin-1 plays an important role in the pathogenesis of immune-mediated CNS demyelinating diseases by promoting immune responses.


Assuntos
Infecções por Cardiovirus/metabolismo , Doenças Desmielinizantes/metabolismo , Endotelina-1/biossíntese , Theilovirus , Animais , Infecções por Cardiovirus/induzido quimicamente , Infecções por Cardiovirus/imunologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/imunologia , Endotelina-1/antagonistas & inibidores , Endotelina-1/toxicidade , Feminino , Camundongos , Oligopeptídeos/farmacologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
J Neuroinflammation ; 14(1): 238, 2017 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-29202854

RESUMO

BACKGROUND: Viral encephalitis is a dangerous compromise between the need to robustly clear pathogen from the brain and the need to protect neurons from bystander injury. Theiler's murine encephalomyelitis virus (TMEV) infection of C57Bl/6 mice is a model of viral encephalitis in which the compromise results in hippocampal damage and permanent neurological sequelae. We previously identified brain-infiltrating inflammatory monocytes as the primary driver of this hippocampal pathology, but the mechanisms involved in recruiting these cells to the brain were unclear. METHODS: Chemokine expression levels in the hippocampus were assessed by microarray, ELISA, RT-PCR, and immunofluorescence. Monocyte infiltration during acute TMEV infection was measured by flow cytometry. CCL2 levels were manipulated by immunodepletion and by specific removal from neurons in mice generated by crossing a line expressing the Cre recombinase behind the synapsin promoter to animals with floxed CCL2. RESULTS: Inoculation of the brain with TMEV induced hippocampal production of the proinflammatory chemokine CCL2 that peaked at 6 h postinfection, whereas inoculation with UV-inactivated TMEV did not elicit this response. Immunofluorescence revealed that hippocampal neurons expressed high levels of CCL2 at this timepoint. Genetic deletion of CCR2 and systemic immunodepletion of CCL2 abrogated or blunted the infiltration of inflammatory monocytes into the brain during acute infection. Specific genetic deletion of CCL2 from neurons reduced serum and hippocampal CCL2 levels and inhibited inflammatory monocyte infiltration into the brain. CONCLUSIONS: We conclude that intracranial inoculation with infectious TMEV rapidly induces the expression of CCL2 in neurons, and this cellular source is necessary for CCR2-dependent infiltration of inflammatory monocytes into the brain during the most acute stage of encephalitis. These findings highlight a unique role for neuronal production of chemokines in the initiation of leukocytic infiltration into the infected central nervous system.


Assuntos
Quimiocina CCL2/biossíntese , Encefalite Viral/mortalidade , Hipocampo/patologia , Monócitos/imunologia , Neurônios/metabolismo , Animais , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/patologia , Quimiotaxia de Leucócito/imunologia , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Hipocampo/imunologia , Hipocampo/virologia , Camundongos , Camundongos Endogâmicos C57BL , Theilovirus
7.
PLoS Pathog ; 11(9): e1005185, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26406250

RESUMO

Cardioviruses, including encephalomyocarditis virus (EMCV) and the human Saffold virus, are small non-enveloped viruses belonging to the Picornaviridae, a large family of positive-sense RNA [(+)RNA] viruses. All (+)RNA viruses remodel intracellular membranes into unique structures for viral genome replication. Accumulating evidence suggests that picornaviruses from different genera use different strategies to generate viral replication organelles (ROs). For instance, enteroviruses (e.g. poliovirus, coxsackievirus, rhinovirus) rely on the Golgi-localized phosphatidylinositol 4-kinase III beta (PI4KB), while cardioviruses replicate independently of the kinase. By which mechanisms cardioviruses develop their ROs is currently unknown. Here we show that cardioviruses manipulate another PI4K, namely the ER-localized phosphatidylinositol 4-kinase III alpha (PI4KA), to generate PI4P-enriched ROs. By siRNA-mediated knockdown and pharmacological inhibition, we demonstrate that PI4KA is an essential host factor for EMCV genome replication. We reveal that the EMCV nonstructural protein 3A interacts with and is responsible for PI4KA recruitment to viral ROs. The ensuing phosphatidylinositol 4-phosphate (PI4P) proved important for the recruitment of oxysterol-binding protein (OSBP), which delivers cholesterol to EMCV ROs in a PI4P-dependent manner. PI4P lipids and cholesterol are shown to be required for the global organization of the ROs and for viral genome replication. Consistently, inhibition of OSBP expression or function efficiently blocked EMCV RNA replication. In conclusion, we describe for the first time a cellular pathway involved in the biogenesis of cardiovirus ROs. Remarkably, the same pathway was reported to promote formation of the replication sites of hepatitis C virus, a member of the Flaviviridae family, but not other picornaviruses or flaviviruses. Thus, our results highlight the convergent recruitment by distantly related (+)RNA viruses of a host lipid-modifying pathway underlying formation of viral replication sites.


Assuntos
Infecções por Cardiovirus/metabolismo , Vírus da Encefalomiocardite/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Metabolismo dos Lipídeos/fisiologia , Replicação Viral/fisiologia , 1-Fosfatidilinositol 4-Quinase/metabolismo , Animais , Western Blotting , Hepacivirus/fisiologia , Humanos , Imunoprecipitação , Microscopia de Fluorescência , Fosfatos de Fosfatidilinositol/metabolismo , Picornaviridae , Vírus de RNA , RNA Interferente Pequeno , Receptores de Esteroides/metabolismo , Transfecção
8.
J Neurovirol ; 23(5): 696-703, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28741149

RESUMO

Seizure disorders are often associated with infectious etiologies. Infection, via the intracerebral (i.c.) route, of C57BL/6J mice with the Daniels (DA) strain of Theiler's murine encephalomyelitis virus (TMEV) results in approximately 50% of the mice developing acute behavioral seizures. TMEV-DA is the wild-type strain of the virus that replicates within the parenchyma of the brain. A variant of TMEV-DA, TMEV-H101, does not replicate within the parenchyma of the brain. However, infection with TMEV-H101 via the i.c. route still results in approximately 40% of the mice developing acute behavioral seizures. Infiltrating macrophages producing interleukin-6 (IL-6) have been implicated in the induction of acute seizures following TMEV-DA infection. We examined macrophage infiltration and microglial activation within the brain and cytokine levels in the periphery in mice infected with TMEV-DA or TMEV-H101 and assessed the effects of the addition of recombinant IL-6 to the periphery in wild-type and IL-6 knockout mice infected with TMEV-DA. We found that pathologic levels of IL-6 in the periphery may play a role in the development of seizures when viral replication within the brain is limited. Examination of the role played by the peripheral immune system in the development of seizures/epilepsy in the TMEV-induced seizure model, the first viral infection driven model for epilepsy, could lead to the elucidation of novel therapeutics.


Assuntos
Infecções por Cardiovirus/complicações , Infecções por Cardiovirus/imunologia , Interleucina-6/imunologia , Convulsões/metabolismo , Convulsões/virologia , Animais , Infecções por Cardiovirus/metabolismo , Interleucina-6/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/metabolismo , Theilovirus/imunologia
9.
Biochem Biophys Res Commun ; 480(2): 187-193, 2016 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-27743889

RESUMO

LGP2 and MDA5 cooperate to detect viral RNA in the cytoplasm of Picornavirus-infected cells and activate innate immune responses. To further define regulatory components of RNA recognition by LGP2/MDA5, a yeast two-hybrid screen was used to identify LGP2-interacting proteins. The screening has identified the TAR-RNA binding protein (TRBP), which is known to be an essential factor for RNA interference (RNAi). Immuno-precipitation experiments demonstrated that TRBP interacted specifically with LGP2 but not with related RIG-I-like receptors, RIG-I or MDA5. siRNA knockdown experiments indicate that TRBP is important for Cardiovirus-triggered interferon responses, but TRBP is not involved in Sendai virus-triggered interferon response that is mediated mainly by RIG-I. To support functional interaction with LGP2, overexpressed TRBP increased Cardiovirus-triggered interferon promoter activity only when LGP2 and MDA5 are co-expressed but not MDA5 alone. Together, our findings illustrate a possible connection between an RNAi-regulatory factor and antiviral RNA recognition that is specifically required for a branch of the virus induced innate immune response.


Assuntos
Infecções por Cardiovirus/metabolismo , Interações Hospedeiro-Patógeno , Proteínas de Ligação a RNA/metabolismo , Animais , Cardiovirus/patogenicidade , Infecções por Cardiovirus/imunologia , Chlorocebus aethiops , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Células HEK293 , Humanos , Helicase IFIH1 Induzida por Interferon/genética , Interferon beta/genética , Camundongos , Regiões Promotoras Genéticas , RNA Helicases/genética , RNA Helicases/metabolismo , RNA Interferente Pequeno , Proteínas de Ligação a RNA/genética , Receptores Imunológicos , Vírus Sendai/patogenicidade , Células Vero
10.
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
11.
Mol Cell Proteomics ; 13(10): 2604-17, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24997996

RESUMO

Viral double-stranded RNA (dsRNA) is the most important viral structure recognized by cytosolic pattern-recognition receptors of the innate immune system, and its recognition results in the activation of signaling cascades that stimulate the production of antiviral cytokines and apoptosis of infected cells. 14-3-3 proteins are ubiquitously expressed regulatory molecules that participate in a variety of cellular processes, and 14-3-3 protein-mediated signaling pathways are activated by cytoplasmic dsRNA in human keratinocytes. However, the functional role of 14-3-3 protein-mediated interactions during viral dsRNA stimulation has remained uncharacterized. Here, we used functional proteomics to identify proteins whose phosphorylation and interaction with 14-3-3 is modulated by dsRNA and to characterize the signaling pathways activated during cytosolic dsRNA-induced innate immune response in human HaCaT keratinocytes. Phosphoproteome analysis showed that several MAPK- and immune-response-related signaling pathways were activated after dsRNA stimulation. Interactome analysis identified RelA-associated inhibitor, high-mobility group proteins, and several proteins associated with host responses to viral infection as novel 14-3-3 target proteins. Functional studies showed that RelA-associated inhibitor regulated dsRNA-induced apoptosis and TNF production. Integrated network analyses of proteomic data revealed that sirtuin1 was a central molecule regulated by 14-3-3s during dsRNA stimulation. Further experiments showed that sirtuin 1 negatively regulated dsRNA-induced NFκB transcriptional activity, suppressed expression of antiviral cytokines, and protected cells from apoptosis in dsRNA-stimulated and encephalomyocarditis-virus-infected keratinocytes. In conclusion, our data highlight the importance of 14-3-3 proteins in antiviral responses and identify RelA-associated inhibitor and sirtuin 1 as novel regulators of antiviral innate immune responses.


Assuntos
Proteínas 14-3-3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Queratinócitos/metabolismo , Proteômica/métodos , RNA de Cadeia Dupla/metabolismo , Proteínas Repressoras/metabolismo , Sirtuína 1/metabolismo , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Linhagem Celular , Citosol/metabolismo , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Humanos , Imunidade Inata , Queratinócitos/citologia , Queratinócitos/imunologia , Queratinócitos/virologia , Fosforilação , RNA de Cadeia Dupla/imunologia , RNA Viral/imunologia , RNA Viral/metabolismo , Transdução de Sinais
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
J Virol ; 86(4): 1922-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22130544

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) results in a persistent central nervous system infection (CNS) and immune-mediated demyelination in mice. TMEV largely persists in macrophages (Ms) in the CNS, and infected Ms in vitro undergo apoptosis, whereas the infection of other rodent cells produces necrosis. We have found that necrosis is the dominant form of cell death in BeAn virus-infected BHK-21 cells but that ~20% of cells undergo apoptosis. Mcl-1 was highly expressed in BHK-21 cells, and protein levels decreased upon infection, consistent with onset of apoptosis. In infected BHK-21 cells in which Mcl-1 expression was knocked down using silencing RNAs there was a 3-fold increase in apoptotic cell death compared to parental cells. The apoptotic program switched on by BeAn virus is similar to that in mouse Ms, with hallmarks of activation of the intrinsic apoptotic pathway in a tumor suppressor protein p53-dependent manner. Infection of stable Mcl-1-knockdown cells led to restricted virus titers and increased physical to infectious particle (PFU) ratios, with additional data suggesting that a late step in the viral life cycle after viral RNA replication, protein synthesis, and polyprotein processing is affected by apoptosis. Together, these results indicate that Mcl-1 acts as a critical prosurvival factor that protects against apoptosis and allows high yields of infectious virus in BHK-21 cells.


Assuntos
Apoptose , Infecções por Cardiovirus/veterinária , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Doenças dos Roedores/metabolismo , Doenças dos Roedores/fisiopatologia , Theilovirus/fisiologia , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/fisiopatologia , Morte Celular , Cricetinae , Camundongos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Necrose , Proteínas Proto-Oncogênicas c-bcl-2/genética , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Theilovirus/genética
19.
J Neurovirol ; 18(5): 354-63, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22638909

RESUMO

This paper reports the upregulation of the gene coding for the apoptosis regulator family member "surviving" in SJL/J mouse brain astrocyte cultures infected with the BeAn strain of TMEV. cRNA from mock- and TMEV-infected SJL/J astrocytes was hybridised to an Affymetrix whole murine genome DNA microarray. Analysis revealed the upregulation of two sequences coding for the survivin protein in infected cells; this was confirmed by RT-PCR and qPCR. Western blotting showed an increase in the synthesis of survivin and caspase-3 after infection. Unexpectedly, no enzymatic activity was detected in BeAn-infected cell lysates in caspase-3-specific colorimetric assays. Cross-linking experiments showed survivin and caspase-3 to exist as a complex containing one molecule of caspase-3 (17 kDa) and one of either 16 kDa or 14 kDa survivin. The neutralization of caspase-3 by survivin-containing lysates was demonstrated using recombinant caspase-3. Brains from TMEV-infected mice, but not from naïve mice, contained survivin mRNA during the acute phase of encephalitis. The present results suggest that astrocytes infected by the BeAn strain do not undergo apoptosis due to the production of survivin.


Assuntos
Astrócitos/metabolismo , Encéfalo/metabolismo , Infecções por Cardiovirus/metabolismo , Caspase 3/genética , Proteínas Inibidoras de Apoptose/genética , RNA Viral/metabolismo , Proteínas Repressoras/genética , Theilovirus/fisiologia , Animais , Apoptose , Astrócitos/virologia , Encéfalo/virologia , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , Caspase 3/metabolismo , Expressão Gênica , Proteínas Inibidoras de Apoptose/metabolismo , Masculino , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Cultura Primária de Células , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , RNA Viral/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/metabolismo , Survivina , Regulação para Cima
20.
Int J Mol Sci ; 13(6): 7365-7374, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22837699

RESUMO

The single-stranded RNA encephalomyocarditis virus (EMCV) can replicate in the central nervous system (CNS) and lead to prominent brain lesions in the stratum pyramidale hippocampus and the stratum granulosum cerebelli. Activated microglia cells infected by EMCV produce a massive burst of reactive oxygen species (ROS) via NADPH oxidase 2 (NOX2) activation, leading to neuronal death. Balancing this effect is mechanisms by which ROS are eliminated from the CNS. Cellular prion protein (PrP(C)) plays an important antioxidant role and contributes to cellular defense against EMCV infection. This review introduces recent knowledge on brain injury induced by EMCV infection via ROS generation as well as the involvement of various mediators and regulators in the pathogenesis.


Assuntos
Infecções por Cardiovirus/metabolismo , Vírus da Encefalomiocardite/metabolismo , Hipocampo/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , NADPH Oxidases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Infecções por Cardiovirus/patologia , Hipocampo/patologia , Hipocampo/virologia , Humanos , Microglia/patologia , Microglia/virologia , NADPH Oxidase 2 , Proteínas PrPC/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA