Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-30959793

RESUMO

Theiler's murine encephalomyelitis (TME) represents a versatile animal model for studying the pathogenesis of demyelinating diseases such as multiple sclerosis. Hallmarks of TME are demyelination, astrogliosis, as well as inflammation. Previous studies showed that matrix metalloproteinase 12 knockout (Mmp12-/-) mice display an ameliorated clinical course associated with reduced demyelination. The present study aims to elucidate the impact of MMP12 deficiency in TME with special emphasis on astrogliosis, macrophages infiltrating the central nervous system (CNS), and the phenotype of microglia/macrophages (M1 or M2). SJL wild-type and Mmp12-/- mice were infected with TME virus (TMEV) or vehicle (mock) and euthanized at 28 and 98 days post infection (dpi). Immunohistochemistry or immunofluorescence of cervical and thoracic spinal cord for detecting glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba1), chemokine receptor 2 (CCR2), CD107b, CD16/32, and arginase I was performed and quantitatively evaluated. Statistical analyses included the Kruskal⁻Wallis test followed by Mann⁻Whitney U post hoc tests. TMEV-infected Mmp12-/- mice showed transiently reduced astrogliosis in association with a strong trend (p = 0.051) for a reduced density of activated/reactive microglia/macrophages compared with wild-type mice at 28 dpi. As astrocytes are an important source of cytokine production, including proinflammatory cytokines triggering or activating phagocytes, the origin of intralesional microglia/macrophages as well as their phenotype were determined. Only few phagocytes in wild-type and Mmp12-/- mice expressed CCR2, indicating that the majority of phagocytes are represented by microglia. In parallel to the reduced density of activated/reactive microglia at 98 dpi, TMEV-infected Mmp12-/- showed a trend (p = 0.073) for a reduced density of M1 (CD16/32- and CD107b-positive) microglia, while no difference regarding the density of M2 (arginase I- and CD107b-positive) cells was observed. However, a dominance of M1 cells was detected in the spinal cord of TMEV-infected mice at all time points. Reduced astrogliosis in Mmp12-/- mice was associated with a reduced density of activated/reactive microglia and a trend for a reduced density of M1 cells. This indicates that MMP12 plays an important role in microglia activation, polarization, and migration as well as astrogliosis and microglia/astrocyte interaction.


Assuntos
Astrócitos/patologia , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Gliose/enzimologia , Gliose/virologia , Metaloproteinase 12 da Matriz/deficiência , Microglia/patologia , Medula Espinal/patologia , Animais , Movimento Celular , Proteína Glial Fibrilar Ácida/metabolismo , Imunofenotipagem , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Metaloproteinase 12 da Matriz/metabolismo , Camundongos Knockout , Fenótipo
2.
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
3.
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
4.
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
5.
J Biol Chem ; 286(30): 26319-26, 2011 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-21636578

RESUMO

RNase L and RNA-dependent protein kinase (PKR) are effectors of the interferon antiviral response that share homology in their pseudokinase and protein kinase domains, respectively. Sunitinib is an orally available, ATP-competitive inhibitor of VEGF and PDGF receptors used clinically to suppress angiogenesis and tumor growth. Sunitinib also impacts IRE1, an endoplasmic reticulum protein involved in the unfolded protein response that is closely related to RNase L. Here, we report that sunitinib is a potent inhibitor of both RNase L and PKR with IC(50) values of 1.4 and 0.3 µM, respectively. In addition, flavonol activators of IRE1 inhibited RNase L. Sunitinib treatment of wild type (WT) mouse embryonic fibroblasts resulted in about a 12-fold increase in encephalomyocarditis virus titers. However, sunitinib had no effect on encephalomyocarditis virus growth in cells lacking both PKR and RNase L. Furthermore, oral delivery of sunitinib in WT mice resulted in 10-fold higher viral titers in heart tissues while suppressing by about 2-fold the IFN-ß levels. In contrast, sunitinib had no effect on viral titers in mice deficient in both RNase L and PKR. Also, sunitinib reduced mean survival times from 12 to 6 days in virus-infected WT mice while having no effect on survival of mice lacking both RNase L and PKR. Results indicate that sunitinib treatments prevent antiviral innate immune responses mediated by RNase L and PKR.


Assuntos
Antineoplásicos/farmacologia , Infecções por Cardiovirus/imunologia , Endorribonucleases/antagonistas & inibidores , Imunidade Inata/efeitos dos fármacos , Indóis/farmacologia , Proteínas de Membrana/imunologia , Proteínas Serina-Treonina Quinases/imunologia , Pirróis/farmacologia , eIF-2 Quinase/antagonistas & inibidores , Animais , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/genética , Vírus da Encefalomiocardite , Endorribonucleases/genética , Endorribonucleases/imunologia , Endorribonucleases/metabolismo , Humanos , Imunidade Inata/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Sunitinibe , eIF-2 Quinase/genética , eIF-2 Quinase/imunologia
6.
J Innate Immun ; 2(1): 77-86, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20375625

RESUMO

Virus infection of macrophages stimulates the expression of proinflammatory and antiviral genes interleukin-1 (IL-1), inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). In this study, we show that phosphatidylinositol 3-kinase (PI3K) is required for the inflammatory response of macrophages to virus infection. When macrophages are infected with encephalomyocarditis virus (EMCV) there is a rapid and transient activation of PI3K and phosphorylation of its downstream target Akt. Inhibitors of PI3K attenuate EMCV- and double-stranded RNA-induced iNOS, COX-2 and IL-1 beta expression in RAW264.7 cells and mouse peritoneal macrophages. The attenuation of inflammatory gene expression in response to PI3K inhibition correlates with the induction of macrophage apoptosis. The morphology of macrophages shifts from activation in response to EMCV infection to apoptosis in the cells treated with PI3K inhibitors and EMCV. These morphological changes are accompanied by the activation of caspase-3. These findings suggest that PI3K plays a central role in the regulation of macrophage responses to EMCV infection. When PI3K is activated, it participates in the regulation of inflammatory gene expression; however, if PI3K is inhibited macrophages are unable to mount an inflammatory antiviral response and die by apoptosis.


Assuntos
Infecções por Cardiovirus/imunologia , Vírus da Encefalomiocardite/imunologia , Ativação de Macrófagos/imunologia , Fosfatidilinositol 3-Quinases/imunologia , RNA de Cadeia Dupla/imunologia , Animais , Infecções por Cardiovirus/enzimologia , Caspase 3/imunologia , Linhagem Celular , Ciclo-Oxigenase 2/imunologia , Vírus da Encefalomiocardite/enzimologia , Interleucina-1beta/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo II/imunologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia
7.
J Neurochem ; 105(6): 2511-23, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18331586

RESUMO

We have previously shown that the protein tyrosine phosphatase SHP-1 is highly expressed in CNS glia and is an important modulator of cytokine signaling. As such, mice genetically lacking SHP-1 display constitutive myelin abnormalities, severe virus-induced demyelinating disease, and defects in innate anti-viral responses in the CNS. In this study, we show the differential distribution of the SHP-1 promoter-specific transcripts and demonstrate that several cytokines significantly induce SHP-1 expression in CNS glia. Consistent with these cytokine effects, infection with a neurotropic virus both in vitro and in vivo up-regulates SHP-1 transcripts and protein in CNS cells. Using CNS glial cultures of gene knockout mice, we show that interferons-beta and interferons-gamma act through STAT-1 and interferon regulatory factor-1 to induce the SHP-1 promoter I transcripts. Conversely, interferons-beta and IL-6 act through STAT-3 to induce SHP-1 promoter II transcripts. This study demonstrates that interferons and other cytokines associated with virus infections in the CNS can significantly induce the expression of SHP-1 through STAT-1/3 activity and provides a better understanding of the molecular mechanisms regulating cytokine-induced expression important for multiple homeostatic functions of SHP-1 in the CNS.


Assuntos
Infecções por Cardiovirus/enzimologia , Citocinas/fisiologia , Encefalite Viral/enzimologia , Neuroglia/enzimologia , Regiões Promotoras Genéticas/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/biossíntese , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/metabolismo , Células Cultivadas , Encefalite Viral/genética , Encefalite Viral/metabolismo , Indução Enzimática/fisiologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Viral da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Knockout , Neuroglia/virologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/deficiência , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Theilovirus
8.
J Immunol ; 178(10): 6444-55, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17475874

RESUMO

The DExD/H box RNA helicase retinoic acid-inducible gene I (RIG-I) and the melanoma differentiation-associated gene 5 (MDA5) are key intracellular receptors that recognize virus infection to produce type I IFN. A third helicase gene, Lgp2, is homologous to Rig-I and Mda5 but lacks a caspase activation and recruitment domain. We generated Lgp2-deficient mice and report that the loss of this gene greatly sensitizes cells to cytosolic polyinosinic/polycytidylic acid-mediated induction of type I IFN. However, negative feedback inhibition of IFN-beta transcription was found to be normal in the absence of LGP2, indicating that LGP2 is not the primary negative regulator of type I IFN production. Our data further indicate that Lgp2-/- mice exhibited resistance to lethal vesicular stomatitis virus infection, a virus whose replicative RNA intermediates are recognized specifically by RIG-I rather than by MDA5 to trigger the production of type I IFN. However, mice lacking LGP2 were observed to exhibit a defect in type I IFN production in response to infection by the encephalomyocarditis virus, the replication of which activates MDA5-dependent innate immune responses. Collectively, our data indicate a disparate regulatory role for LGP2 in the triggering of innate immune signaling pathways following RNA virus infection.


Assuntos
Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/prevenção & controle , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , Infecções por Rhabdoviridae/enzimologia , Infecções por Rhabdoviridae/prevenção & controle , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Células Cultivadas , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , RNA Helicases DEAD-box/fisiologia , Vírus da Encefalomiocardite/imunologia , Feminino , Imunidade Inata/genética , Helicase IFIH1 Induzida por Interferon , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas do Tecido Nervoso/metabolismo , Receptores de Superfície Celular , Infecções por Rhabdoviridae/genética , Infecções por Rhabdoviridae/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia
9.
J Neuropathol Exp Neurol ; 65(8): 783-93, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16896312

RESUMO

Theiler murine encephalomyelitis (TME) represents a highly relevant viral model for multiple sclerosis. Matrix metalloproteinases (MMPs) degrade extracellular matrix molecules and are involved in demyelination processes. To elucidate their impact on demyelination in TME, spinal cords of TME virus (TMEV)-infected SJL/J mice were taken at 9 different time points postinfection (pi) ranging from 1 hour to 196 days pi and investigated for the expression of TMEV, MMP-2, -3, -7, -9, -10, -11, -12, -13, -14, -15, -24, and TIMP-1 to -4 by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). High TMEV RNA levels were detectable throughout the observation period using RT-qPCR. In addition, TMEV RNA was visualized within demyelinated lesions by in situ hybridization. MMP-3 mRNA was significantly upregulated at 1 day pi and again in the late phase of infection. TIMP-1 mRNA was significantly elevated throughout the observation period. MMP-12 mRNA was most prominently upregulated in the late phase of infection and MMP-12 protein was localized in intralesional microglia/macrophages and astrocytes by immunohistochemistry. In summary, in early TMEV infection, MMP-3 and TIMP-1 mRNA upregulation might be directly virus-induced, whereas persistent TMEV infection directly or indirectly stimulated MMP-12 production in microglia/macrophages and astrocytes and might account for ongoing demyelination in TME.


Assuntos
Infecções por Cardiovirus/enzimologia , Doenças Desmielinizantes/enzimologia , Encefalomielite/enzimologia , Metaloproteinases da Matriz/metabolismo , Theilovirus/genética , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/patologia , Sistema Nervoso Central/enzimologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiopatologia , Doença Crônica , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/virologia , Modelos Animais de Doenças , Encefalomielite/genética , Encefalomielite/virologia , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Macrófagos/metabolismo , Metaloproteinase 12 da Matriz , Metaloproteinase 3 da Matriz/metabolismo , Metaloendopeptidases/metabolismo , Camundongos , Microglia/metabolismo , Esclerose Múltipla/enzimologia , Esclerose Múltipla/genética , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia , RNA Mensageiro/análise , RNA Mensageiro/genética , RNA Viral/análise , RNA Viral/genética , Fatores de Tempo , Regulação para Cima , Carga Viral
10.
Life Sci ; 78(2): 195-204, 2005 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-16107267

RESUMO

To determine critical role of cyclooxygenase-2 (COX-2) for development of viral myocarditis, a mouse model of encephalomyocarditis virus-induced myocarditis was used. The virus was intraperitoneally given to COX-2 gene-deficient heterozygote mice (COX-2+/-) and wild-type mice (WT). We examined differences in heart weights, cardiac histological scores, numbers of infiltrating or apoptotic cells in myocardium, cardiac expression levels of COX-2, tumor necrosis factor-alpha (TNF-alpha), and adiponectin mRNA, immunoreactivity of COX-2, TNF-alpha, and adiponectin in myocytes, cardiac concentrations of TNF-alpha and adiponectin, prostaglandin E2 (PGE2) levels in hearts, and viral titers in tissues between COX-2+/- and WT. We observed significantly decreased expression of COX-2 mRNA and reactivity in hearts from COX-2+/- on day 8 after viral inoculation as compared with that from WT, together with elevated cardiac weights and severe inflammatory myocardial damage in COX-2+/-. Cardiac expression of TNF-alpha mRNA, reactivity, and protein on day 8 was significantly higher in COX-2+/- than in WT, together with reciprocal expression of adiponectin mRNA, reactivity, and protein in hearts. Significantly reduced cardiac PGE2 levels on day 8 were found in COX-2+/- compared with those in WT. There was no difference in local viral titers between both groups on day 4. Infected WT treated with a selective COX-2 inhibitor, NS-398, also showed the augmented myocardial damage on day 8. These results suggest that inhibition of COX-2 may enhance myocardial damage through reciprocal cardiac expression of TNF-alpha and adiponectin in a mouse model of viral myocarditis.


Assuntos
Infecções por Cardiovirus/patologia , Inibidores de Ciclo-Oxigenase 2/toxicidade , Ciclo-Oxigenase 2/fisiologia , Vírus da Encefalomiocardite , Miocardite/patologia , Miocárdio/patologia , Adiponectina/biossíntese , Adiponectina/genética , Animais , Peso Corporal/fisiologia , Infecções por Cardiovirus/enzimologia , Ciclo-Oxigenase 2/biossíntese , Ciclo-Oxigenase 2/genética , Dinoprostona/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/enzimologia , Miocardite/virologia , Miocárdio/enzimologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Miócitos Cardíacos/virologia , Nitrobenzenos/farmacologia , Tamanho do Órgão/fisiologia , RNA Mensageiro/biossíntese , Sulfonamidas/farmacologia , Fator de Necrose Tumoral alfa/biossíntese
11.
J Virol ; 76(16): 8335-46, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12134038

RESUMO

We previously characterized the expression and function of the protein tyrosine phosphatase SHP-1 in the glia of the central nervous system (CNS). In the present study, we describe the role of SHP-1 in virus infection of glia and virus-induced demyelination in the CNS. For in vivo studies, SHP-1-deficient mice and their normal littermates received an intracerebral inoculation of an attenuated strain of Theiler's murine encephalomyelitis virus (TMEV). At various times after infection, virus replication, TMEV antigen expression, and demyelination were monitored. It was found that the CNS of SHP-1-deficient mice uniquely displayed demyelination and contained substantially higher levels of virus than did that of normal littermate mice. Many infected astrocytes and oligodendrocytes were detected in both brains and spinal cords of SHP-1-deficient but not normal littermate mice, showing that the virus replicated and spread at a much higher rate in the glia of SHP-1-deficient animals. To ascertain whether the lack of SHP-1 in the glia was primarily responsible for these differences, glial samples from these mice were cultured in vitro and infected with TMEV. As in vivo, infected astrocytes and oligodendrocytes of SHP-1-deficient mice were much more numerous and produced more virus than did those of normal littermate mice. These findings indicate that SHP-1 is a critical factor in controlling virus replication in the CNS glia and virus-induced demyelination.


Assuntos
Infecções por Cardiovirus/etiologia , Infecções do Sistema Nervoso Central/etiologia , Doenças Desmielinizantes/etiologia , Proteínas Tirosina Fosfatases/metabolismo , Theilovirus/patogenicidade , Animais , Astrócitos/enzimologia , Astrócitos/virologia , Encéfalo/patologia , Encéfalo/virologia , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Infecções do Sistema Nervoso Central/enzimologia , Infecções do Sistema Nervoso Central/patologia , Infecções do Sistema Nervoso Central/virologia , Doenças Desmielinizantes/enzimologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Neuroglia/enzimologia , Neuroglia/virologia , Oligodendroglia/enzimologia , Oligodendroglia/virologia , Paralisia/enzimologia , Paralisia/etiologia , Paralisia/virologia , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , Medula Espinal/patologia , Medula Espinal/virologia , Theilovirus/fisiologia , Replicação Viral/fisiologia
12.
Vet Microbiol ; 70(3-4): 171-7, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10596801

RESUMO

Six piglets that had survived experimental infection with encephalomyocarditis virus (EMCV) were treated with dexamethasone for a period of 5 days. The virus had not been detected in excretions of putative carriers for a period of 13-20 days before the treatment. All piglets showed a rise in cardiac isoenzyme (CK-MB) activity, from the first day of treatment, suggesting myocardial damage. Antibody titres against EMCV remained stable or slightly decreased during treatment. EMCV was isolated from blood, nasal and faecal samples from all piglets on days 2 and 3 after initiation of treatment and from various tissues of three piglets. Four contact piglets, that were housed together with the dexamethasone-treated piglets, became infected, indicating that EMCV was shed by treated piglets. It is suggested that recovered pigs may play an important role in the dissemination of EMCV.


Assuntos
Infecções por Cardiovirus/veterinária , Vírus da Encefalomiocardite/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Biomarcadores , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Creatina Quinase/análise , Ensaio de Imunoadsorção Enzimática/veterinária , Isoenzimas , Reação em Cadeia da Polimerase/veterinária , Suínos , Doenças dos Suínos/enzimologia
13.
J Gen Virol ; 80 ( Pt 7): 1701-1705, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10423138

RESUMO

Increasing evidence suggests that macrophages (M(phi)s) are necessary for persistence of Theiler's murine encephalomyelitis virus (TMEV) in the mouse central nervous system. Analysis of BeAn virus infection in the Mphi cell lines P388D1, J774A.1 and PU5-1.8, which are intermediate in their state of differentiation and resemble multifunctional resident M(phi)s, revealed restricted TMEV growth. As a result of the restricted infection, these Mphi cell lines were induced to undergo apoptosis as demonstrated by cellular morphology, DNA fragmentation, caspase protease activity, and in individual cells, by terminal deoxytransferase dUTP nick-end labelling (TUNEL).


Assuntos
Apoptose , Infecções por Cardiovirus/patologia , Macrófagos/patologia , Macrófagos/virologia , Theilovirus , Animais , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Caspases/metabolismo , Fragmentação do DNA , Ativação Enzimática , Marcação In Situ das Extremidades Cortadas , Macrófagos/enzimologia , Camundongos
14.
Cell Immunol ; 194(2): 186-93, 1999 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10383821

RESUMO

Intracerebral inoculation of susceptible strains of mice with Theiler's murine encephalomyelitis virus (TMEV) results in immune-mediated demyelinating disease. We examined the pathogenic roles of nitric oxide (NO) and inducible NO synthase (iNOS) in TMEV-induced demyelinating disease (TMEV-IDD). The presence of iNOS was confirmed in the spinal cords of TMEV-infected mice using immunohistochemical staining with anti-iNOS antibody on day 0 (control) and days 15, 30, 60, and 120. Aminoguanidine (AG), a specific inhibitor of iNOS, was injected intraperitoneally (ip) on 1, 3, 5, 8, 10, and 12 days post-TMEV inoculation as induction phase or 15, 17, 19, 22, 24, and 26 days as effector phase. Control animals in each experiment received phosphate-buffered saline (PBS) ip at similar time intervals. Few iNOS-positive cells were observed in the spinal cords of naive SJL/J mice. In the early phase (day 15) of TMEV-IDD, an increase of iNOS-positive cells was detected in the leptomeninges and perivascular space of the spinal cords. The number of iNOS-positive cells was increased and reached its peak on day 60, when histology of the animals showed peak infiltration with inflammatory cells. The clinical course of TMEV-IDD on each day postintracerebral infection was significantly reduced in mice treated with AG in the effector phase, and there was no significant difference between mice treated with AG in induction phase versus those administered PBS. Thus, NO production via iNOS appears to be a pathogenic factor in the effector phase of TMEV-IDD.


Assuntos
Infecções por Cardiovirus/enzimologia , Sistema Nervoso Central/enzimologia , Doenças Desmielinizantes/enzimologia , Óxido Nítrico Sintase/fisiologia , Theilovirus , Animais , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/patologia , Linhagem Celular , Cricetinae , Citocinas/biossíntese , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Inibidores Enzimáticos/administração & dosagem , Feminino , Guanidinas/administração & dosagem , Camundongos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/biossíntese , Óxido Nítrico Sintase Tipo II , Medula Espinal/enzimologia , Medula Espinal/patologia , Baço/imunologia , Baço/metabolismo
15.
Eur J Biochem ; 254(2): 248-55, 1998 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-9660177

RESUMO

The interferon-(IFN)-inducible 2',5'-oligoadenylate (2-5A)/endoribonuclease L (RNase L) pathway plays a major role in the antiviral and antiproliferative effects of IFN. The 2-5A/RNase L pathway appears to be regulated by the cell-growth status or viral infection. Viruses, and picornaviruses in particular, have evolved strategies to escape the 2-5A/RNase L-pathway-associated antiviral activity. We have recently cloned a cDNA coding for RLI, a RNase-L-specific protein inhibitor. Its regulated expression by viral infection could provide a new strategy to modulate the 2-5A/RNase L pathway. Since RNase L had been shown to be down regulated upon encephalomyocarditis (EMCV) infection, we stably transfected HeLa cells with a RLI antisense cDNA expressing vector. Four independent clones named VAS1, VAS2, VAS3 and VAS4 and one clone transfected with the empty vector (VV) as control, were analyzed. The level of RLI was decreased by 20% for VAS1, 25% for VAS2, 75% for VAS3 and 50% for VAS4. The inactivation of RNase L observed during EMCV infection was decreased in these clones as compared to control HeLa cells. Here again the results vary between the four clones. The maximum inhibition of RNase L (90%) was observed in control cells and in VAS1 while 48% inhibition was observed in VAS4 and 25% in VAS3. The reversal in RNase L inhibition thus reflects closely the resulting RLI level, in keeping with a major role of RLI in EMCV-induced down regulation of 2-5A-binding activity of RNase L. Moreover, cells expressing a low level of RLI (VAS3 and VAS 4) are partially resistant to EMCV infection.


Assuntos
Transportadores de Cassetes de Ligação de ATP , Infecções por Cardiovirus/enzimologia , Chaperoninas , Vírus da Encefalomiocardite , Endorribonucleases/antagonistas & inibidores , Inibidores Enzimáticos/metabolismo , Proteínas/antagonistas & inibidores , Anticorpos , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/virologia , DNA Antissenso/genética , DNA Antissenso/farmacologia , Regulação para Baixo/efeitos dos fármacos , Vírus da Encefalomiocardite/isolamento & purificação , Endorribonucleases/metabolismo , Inibidores Enzimáticos/farmacologia , Células HeLa , Humanos , Proteínas/genética , Proteínas/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA