Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Vet Microbiol ; 264: 109304, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34922148

RESUMEN

DEAD (Asp-Glu-Ala-Asp)-box RNA helicases (DDX) play important roles in viral infection, either as cytosolic viral nucleic acids sensors or as essential host factors for viral replication. In this study, we identified DDX56 as a positive regulator for encephalomyocarditis virus (EMCV) replication. EMCV infection promotes DDX56 expression via its viral proteins, VP3 and 3C. We showed that DDX56 overexpression promotes EMCV replication whereas its loss dampened EMCV replication. Consequently, knockdown of DDX56 increases type I interferon (IFN) expression during EMCV infection. We also showed that DDX56 interrupts IFN regulatory factor 3 (IRF3) phosphorylation and its nucleus translocation by directly targeting KPNA3 and KPNA4 in an EMCV-triggered MDA5 signaling activation cascade leading to the blockade of IFN-ß production. Overall, we showed that DDX56 is a novel negative regulator of EMCV-mediated IFN-ß responses and that DDX56 plays a critical role in EMCV replication. These findings reveal a novel strategy for EMCV to utilize a host factor to evade the host innate immune response and provide us new insight into the function of DDX56.


Asunto(s)
ARN Helicasas DEAD-box , Virus de la Encefalomiocarditis , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón , Interferón beta , Transporte de Proteínas , Replicación Viral , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Línea Celular , ARN Helicasas DEAD-box/metabolismo , Virus de la Encefalomiocarditis/fisiología , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón beta/genética , Interferón beta/metabolismo , Replicación Viral/genética
2.
PLoS One ; 12(4): e0176406, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28445497

RESUMEN

Infection of various cells with Theiler's murine encephalomyelitis virus (TMEV) activates the TLR- and melanoma differentiation-associated gene 5 (MDA5)-dependent pathways, resulting in the production of IL-1ß via the activation of caspase-1 upon assembly of the node-like receptor protein 3 (NLRP3) inflammasome. The role of IL-1ß in the pathogenesis of TMEV-induced demyelinating disease was previously investigated. However, the signaling effects of prostaglandin E2 (PGE2) downstream of the NLRP3 inflammasome on the immune responses to viral determinants and the pathogenesis of demyelinating disease are unknown. In this study, we investigated the levels of intermediate molecules leading to PGE2 signaling and the effects of blocking PGE2 signaling on the immune response to TMEV infection, viral persistence and the development of demyelinating disease. We demonstrate here that TMEV infection activates the NLRP3 inflammasome and PGE2 signaling much more vigorously in dendritic cells (DCs) and CD11b+ cells from susceptible SJL mice than in cells from resistant B6 mice. Inhibition of virus-induced PGE2 signaling using AH23848 resulted in decreased pathogenesis of demyelinating disease and viral loads in the central nervous system (CNS). In addition, AH23848 treatment caused the elevation of protective early IFN-γ-producing CD4+ and CD8+ T cell responses. Because the levels of IFN-ß were lower in AH23848-treated mice but the level of IL-6 was similar, over-production of pathogenic IFN-ß was modulated and the generation of IFN-γ-producing T cell responses was enhanced by the inhibition of PGE2 signaling. These results strongly suggest that excessive activation of the NLRP3 inflammasome and downstream PGE2 signaling contribute to the pathogenesis of TMEV-induced demyelinating disease.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Enfermedades Desmielinizantes/fisiopatología , Dinoprostona/metabolismo , Theilovirus/fisiología , Animales , Compuestos de Bifenilo/farmacología , Células de la Médula Ósea/citología , Encéfalo/metabolismo , Encéfalo/patología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Infecciones por Cardiovirus/fisiopatología , Células Cultivadas , Enfermedades Desmielinizantes/virología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Neuroglía/citología , Neuroglía/metabolismo , Subtipo EP4 de Receptores de Prostaglandina E/antagonistas & inhibidores , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/patología , Carga Viral
3.
Neurosci Lett ; 609: 210-5, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26477780

RESUMEN

Viral infections of the central nervous system are often associated with seizures, and while patients usually recover from the infection and the seizures cease, there is an increased lifetime incidence of epilepsy. These viral infections can result in mesial temporal sclerosis, and, subsequently, a type of epilepsy that is difficult to treat. In previous work, we have shown that Theiler's murine encephalomyelitis virus (TMEV) infections in C57B/6 mice, an animal model of virus-induced epilepsy, results in changes in excitatory currents of CA3 neurons both during the acute infection and two months later, at a time when seizure thresholds are reduced and when spontaneous seizures can occur. The changes in the excitatory system differ at these two time points, suggesting different mechanisms for seizure generation. In the present paper, we examine GABAergic mediated inhibition in CA3 pyramidal cells at these two time points following TMEV infection. We found that amplitudes of sIPSCs and mIPSCs were reduced during the acute infection, but recovered at the two-month time point. These observations are consistent with previous measurements of excitatory currents suggesting different mechanisms of seizure generation during the acute infection and during chronic epilepsy.


Asunto(s)
Región CA3 Hipocampal/fisiopatología , Infecciones por Cardiovirus/fisiopatología , Epilepsia/fisiopatología , Theilovirus , Animales , Infecciones por Cardiovirus/virología , Epilepsia/virología , Potenciales Postsinápticos Inhibidores , Masculino , Ratones Endogámicos C57BL , Células Piramidales/fisiología , Factores de Tiempo , Ácido gamma-Aminobutírico/fisiología
4.
Biomed Res Int ; 2015: 645153, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25821814

RESUMEN

Viral-myocarditis is an important cause of heart failure for which no specific treatment is available. We previously showed the neuropeptide substance P (SP) is associated with the pathogenesis of murine myocarditis caused by encephalomyocarditis virus (EMCV). The current studies determined if pharmacological inhibition of SP-signaling via its high affinity receptor, NK1R and downstream G-protein, Ras homolog gene family, member-A (RhoA), will be beneficial in viral-myocarditis. Aprepitant (1.2 mg/kg), a SP-receptor antagonist, or fasudil (10 mg/kg), a RhoA inhibitor, or saline control was administered daily to mice orally for 3 days, prior to, or 5 days following, intraperitoneal infection with and without 50 PFU of EMCV, following which disease assessment studies, including echocardiogram and cardiac Doppler were performed in day 14 after infection. Pretreatment and posttreatment with aprepitant significantly reduced mortality, heart and cardiomyocyte size, and cardiac viral RNA levels (P < 0.05 all, ANOVA). Only aprepitant pretreatment improved heart functions; it significantly decreased end systolic diameter, improved fractional shortening, and increased peak aortic flow velocity (P < 0.05 all, ANOVA). Pre- or posttreatment with fasudil did not significantly impact disease manifestations. These findings indicate that SP contributes to cardiac-remodeling and dysfunction following ECMV infection via its high affinity receptor, but not through the Rho-A pathway. These studies suggest that SP-receptor antagonism may be a novel therapeutic-option for patients with viral-myocarditis.


Asunto(s)
Infecciones por Cardiovirus/tratamiento farmacológico , Infecciones por Cardiovirus/fisiopatología , Miocarditis/tratamiento farmacológico , Miocarditis/fisiopatología , Antagonistas del Receptor de Neuroquinina-1/uso terapéutico , Receptores de Neuroquinina-1/metabolismo , Animales , Infecciones por Cardiovirus/virología , Estudios de Factibilidad , Masculino , Ratones , Ratones Endogámicos C57BL , Miocarditis/virología , Resultado del Tratamiento , Carga Viral/efectos de los fármacos
5.
Virus Res ; 177(2): 222-5, 2013 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-24036175

RESUMEN

The Theiler's murine encephalomyelitis virus (TMEV) leader (L) protein zinc-finger domain was mutated to study its role in cell death in infection of the murine macrophage cell line M1-D, revealing that an intact zinc-finger domain is required for full apoptotic activity. A functional L zinc-finger domain was also required for activation of p38 MAPK that results in phosphorylation and activation of p53, and in turn, alteration of the conformation of the anti-apoptotic proteins Puma and Mcl-1, leading to the release of pro-apoptotic Bax and apoptosis through the intrinsic pathway. TMEV infection also inhibits host protein synthesis, a stress shown by others to induce apoptosis. Since inhibition of host protein synthesis follows rather than precedes activation of MKK3/6 and p38, it seems less likely that it triggers apoptosis in infected cells. Finally, we showed that the levels of reactive oxygen species following infection were consistent with apoptotic rather than necrotic cell death. Thus, these experiments support an important role for the TMEV L protein zinc-finger domain in apoptosis in an infected murine macrophage line.


Asunto(s)
Apoptosis , Infecciones por Cardiovirus/veterinaria , Macrófagos/citología , Enfermedades de los Roedores/fisiopatología , Enfermedades de los Roedores/virología , Theilovirus/genética , Proteínas Virales/química , Proteínas Virales/genética , Animales , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Mutación , Estructura Terciaria de Proteína , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/metabolismo , Theilovirus/química , Theilovirus/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Virales/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
6.
Circ Heart Fail ; 6(2): 326-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23271791

RESUMEN

BACKGROUND: Viral myocarditis is among the most common causes of heart failure in children and young adults. The RNA helicase melanoma differentiation-associated gene-5 (MDA5) is critical for host antiviral responses against members of the Picornaviridae family, such as encephalomyocarditis virus (EMCV). MDA5-knockout mice are highly susceptible to EMCV infection and develop significant myocardial injury and left ventricular dysfunction. However, the mechanisms by which MDA5 signaling within cardiac myocytes contributes to the host response against viral infection have not been defined. METHODS AND RESULTS: We generated cardiac-specific MDA5 transgenic (alpha-myosin heavy chain [αMHC]-MDA5) mice. These mice showed increased baseline cardiac expression of antiviral cytokines and increased cellular infiltration but no alterations in cardiac function and structure. αMHC-MDA5 mice were less susceptible to EMCV infection and had a significantly lower cardiac viral load compared with littermate control mice. The severity of myocarditis, prevalence of cardiac myocyte apoptosis, and cleavage of caspase 3 after EMCV infection were attenuated in αMHC-MDA5 mice. Furthermore, αMHC-MDA5 mice were protected against EMCV-induced myocardial dysfunction. CONCLUSIONS: Our data suggest that myocardial MDA5 may be a key molecule in protecting the heart from direct viral injury and myocardial dysfunction.


Asunto(s)
Infecciones por Cardiovirus/metabolismo , ARN Helicasas DEAD-box/metabolismo , Virus Maus Elberfeld/patogenicidad , Miocarditis/prevención & control , Miocardio/metabolismo , Animales , Apoptosis , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Caspasa 3/metabolismo , ARN Helicasas DEAD-box/deficiencia , ARN Helicasas DEAD-box/genética , Modelos Animales de Enfermedad , Femenino , Genotipo , Helicasa Inducida por Interferón IFIH1 , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocarditis/genética , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/fisiopatología , Miocarditis/virología , Miocardio/patología , Fenotipo , Volumen Sistólico , Factores de Tiempo , Disfunción Ventricular Izquierda/metabolismo , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Izquierda/prevención & control , Disfunción Ventricular Izquierda/virología , Función Ventricular Izquierda , Replicación Viral
7.
J Neuroimmunol ; 254(1-2): 19-27, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23021485

RESUMEN

Prior exposure to social disruption (SDR) stress exacerbates Theiler's murine encephalomyelitis virus (TMEV) infection, a model of multiple sclerosis. Here we examined the impact of SDR on T cell responses to TMEV infection in SJL mice. SDR impaired viral clearance and exacerbated acute disease. Moreover, TMEV infection alone increased CD4 and CD8 mRNA expression in brain and spleen while SDR impaired this response. SDR decreased both CD4(+) and CD8(+) virus-specific T cells in CNS, but not spleen. These findings suggest that SDR-induced suppression of virus-specific T cell responses contributes to impairments in viral clearance and exacerbation of acute disease.


Asunto(s)
Inmunidad Adaptativa/fisiología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/fisiopatología , Poliomielitis/inmunología , Poliomielitis/fisiopatología , Estrés Psicológico/fisiopatología , Enfermedad Aguda , Animales , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/virología , Antígenos CD4/genética , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/patología , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos , ARN Mensajero/metabolismo , Bazo/inmunología , Bazo/metabolismo , Bazo/virología , Estrés Psicológico/inmunología , Theilovirus/patogenicidad
8.
J Virol ; 86(4): 1922-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22130544

RESUMEN

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.


Asunto(s)
Apoptosis , Infecciones por Cardiovirus/veterinaria , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Enfermedades de los Roedores/metabolismo , Enfermedades de los Roedores/fisiopatología , Theilovirus/fisiología , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/fisiopatología , Muerte Celular , Cricetinae , Ratones , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Necrosis , Proteínas Proto-Oncogénicas c-bcl-2/genética , Enfermedades de los Roedores/genética , Enfermedades de los Roedores/virología , Theilovirus/genética
9.
PLoS One ; 6(10): e26001, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022490

RESUMEN

Our laboratory demonstrated that a natural human serum antibody, sHIgM12, binds to neurons in vitro and promotes neurite outgrowth. We generated a recombinant form, rHIgM12, with identical properties. Intracerebral infection with Theiler's Murine Encephalomyelitis Virus (TMEV) of susceptible mouse strains results in chronic demyelinating disease with progressive axonal loss and neurologic dysfunction similar to progressive forms of multiple sclerosis. To study the effects of rHIgM12 on the motor function of TMEV-infected mice, we monitored spontaneous nocturnal activity over many weeks. Nocturnal behavior is a sensitive measure of rodent neurologic function because maximal activity changes are expected to occur during the normally active night time monitoring period. Mice were placed in activity boxes eight days prior to treatment to collect baseline spontaneous activity. After treatment, activity in each group was continuously recorded over 8 weeks. We chose a long 8-week monitoring period for two reasons: (1) we previously demonstrated that IgM induced remyelination is present by 5 weeks post treatment, and (2) TMEV-induced demyelinating disease in this strain progresses very slowly. Due to the long observation periods and large data sets, differences among treatment groups may be difficult to appreciate studying the original unfiltered recordings. To clearly delineate changes in the highly fluctuating original data we applied three different methods: (1) binning, (2) application of Gaussian low-pass filters (GF) and (3) polynomial fitting. Using each of the three methods we showed that compared to control IgM and saline, early treatment with rHIgM12 induced improvement in both horizontal and vertical motor function, whereas later treatment improved only horizontal activity. rHIgM12 did not alter activity of normal, uninfected mice. This study supports the hypothesis that treatment with a neuron-binding IgM not only protects neurons in vitro, but also influences functional motor improvement.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Enfermedades Desmielinizantes/fisiopatología , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Oscuridad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inyecciones Intraperitoneales , Ratones , Unión Proteica/efectos de los fármacos , Theilovirus/efectos de los fármacos
10.
Immunology ; 133(3): 360-9, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21564094

RESUMEN

This paper describes an experimental model of neuroinflammation based on the production of interleukin-6 (IL-6) by neural glial cells infected with Theiler's murine encephalomyelitis virus (TMEV). Production of IL-6 mRNA in mock-infected and TMEV-infected SJL/J murine astrocytes was examined using the Affymetrix murine genome U74v2 DNA microarray. The IL-6 mRNA from infected cells showed an eightfold increase in hybridization to a sequence encoding IL-6 located on chromosome number 5. Quantitative real-time reverse transcription PCR (qPCR) was used to study the regulation of IL-6 expression. The presence of IL-6 in the supernatants of TMEV-infected astrocyte cultures was quantified by ELISA and found to be weaker than in cultures of infected macrophages. The IL-6 was induced by whole TMEV virions, but not by Ad.ßGal adenovirus, purified TMEV capsid proteins, or UV-inactivated virus. Two recombinant inflammatory cytokines, IL-1α and tumour necrosis factor-α were also found to be potent inducers of IL-6. The secreted IL-6 was biologically active because it fully supported B9 hybridoma proliferation in a [(3) H]thymidine incorporation bioassay. The cerebrospinal fluid of infected mice contained IL-6 during the acute encephalitis phase, peaking at days 2-4 post-infection. Finally, this in vitro neuroinflammation model was fully inhibited, as demonstrated by ELISA and qPCR, by five selective oestrogen receptor modulators.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitos/virología , Infecciones por Cardiovirus/inmunología , Moduladores de los Receptores de Estrógeno/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-6/inmunología , Theilovirus , Animales , Infecciones por Cardiovirus/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Inflamación/tratamiento farmacológico , Interleucina-18/metabolismo , Interleucina-6/líquido cefalorraquídeo , Interleucina-6/metabolismo , Macrófagos/virología , Ratones , Modelos Inmunológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Theilovirus/inmunología
11.
Int Immunol ; 22(9): 729-38, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20685674

RESUMEN

We examined the role of Notch ligand Delta-like 4 (Dll4) in the development of Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD). Treatment with mAb to Dll4, especially during the effector phase, resulted in significant suppression of the disease development both clinically and histologically. The number of infiltrating mononuclear inflammatory cells in the spinal cords was also decreased in mice treated with anti-Dll4 mAb. Semi-quantitative analysis of mRNA by using real-time PCR revealed that mRNAs of T(h)1-derived cytokines such as IFN-gamma and T(h)17-derived cytokines such as IL-17 were decreased in mice treated with anti-Dll4 mAb, whereas those of T(h)2-derived cytokines such as IL-4 and IL-10 were not. Flow cytometric analysis of cytokines indicated that there were no significant differences between mAb-treated mice and control mice in the relative frequency of splenic T(h)1 and T(h)2. However, absolute cell numbers of T(h)1-derived cytokine-producing cells in spinal cord were markedly decreased in mice treated with anti-Dll4 mAb in effector phase compared with control mice treated with non-specific IgG. These data suggest that Dll4 is critically involved in the pathogenesis of TMEV-IDD and that antibodies to Dll4 could be used as a novel therapeutic treatment of demyelinating diseases such as human multiple sclerosis.


Asunto(s)
Anticuerpos Bloqueadores/administración & dosificación , Infecciones por Cardiovirus/inmunología , Enfermedades Desmielinizantes/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Médula Espinal/efectos de los fármacos , Theilovirus/inmunología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Infecciones por Cardiovirus/tratamiento farmacológico , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología , Balance Th1 - Th2/efectos de los fármacos , Theilovirus/patogenicidad
12.
Can J Physiol Pharmacol ; 88(5): 568-75, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20555426

RESUMEN

Exposure to tobacco smoke is known to have deleterious cardiovascular effects. In this study, we tested whether exposure to tobacco smoke exacerbates the severity of viral myocarditis in mice. Viral myocarditis was generated in 4-week-old male BALB/c mice by injection of Encephalomyocarditis virus (EMCV). Four groups were studied: (1) control (C, no smoke and no virus); (2) smoke only (S, exposure to cigarette smoke for 90 min/day for 15 days); (3) virus only (V); and (4) exposure to smoke for 5 days before plus 10 days following virus injection (S+V). We found that viral inoculation preceded by smoke exposure increased mortality more than twofold compared with virus inoculation alone. In addition, the mRNA level of atrial natriuretic factor was significantly higher in S+V than among any of the other 3 groups. Virus injection significantly decreased cardiac function compared with controls, with further deterioration observed in the S+V group. We also observed a significantly increased rate of apoptosis, with an increased activation of apoptosis-inducing factor in hearts exposed to S+V compared with those exposed to V alone. Our results suggest that preexposure to smoke significantly exacerbates the severity of viral myocarditis, likely through increased viral load and increased cardiomyocyte cell death.


Asunto(s)
Infecciones por Cardiovirus/virología , Corazón/virología , Miocarditis/virología , Contaminación por Humo de Tabaco/efectos adversos , Carga Viral , Animales , Apoptosis , Factor Inductor de la Apoptosis/biosíntesis , Factor Natriurético Atrial/biosíntesis , Western Blotting , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Virus de la Encefalomiocarditis , Hemodinámica/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/metabolismo , Miocarditis/patología , Miocarditis/fisiopatología , Miocardio/metabolismo , Miocardio/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Neuroimmunomodulation ; 17(3): 169-72, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20134194

RESUMEN

Psychological stress is an important factor in susceptibility to many diseases. Our laboratory has been investigating the impact of stress on the susceptibility to Theiler's virus-induced demyelination (TVID), a mouse model of multiple sclerosis. Using immunodominant viral peptides specific for identification of either CD4(+) or CD8(+) T cells, stress reduced IFN-gamma-producing virus-specific CD4(+) and CD8(+) T cells in the spleen and CD8(+) T cells in the CNS. Expression of mRNA for the Th1 transcription factor T-bet and the Th2 transcription factor GATA-3 were decreased in spleen cells isolated from stressed mice. Cytokine production by cells isolated from the CNS or spleens following stimulation with virus indicated that stress decreased both type 1 and type 2 responses. The adverse effects of stress were partially reversed by concurrent RU486 administration but mimicked by dexamethasone, indicating a major role for glucocorticoids. Global stress-induced immunosuppression resulted in higher levels of virus replication and dissemination. The higher viral load subsequently led to an earlier disease onset and more severe clinical and histological signs of demyelinating disease. Our results have important implications for understanding the pathogenesis of MS, and suggest that stressful events during early infection with an agent capable of inducing demyelination may result in immunosuppression and failure to eliminate the pathogen, which in turn may lead to the development of MS.


Asunto(s)
Autoinmunidad/fisiología , Infecciones por Cardiovirus/inmunología , Esclerosis Múltiple/inmunología , Estrés Psicológico/inmunología , Theilovirus/inmunología , Animales , Infecciones por Cardiovirus/fisiopatología , Citocinas/inmunología , Citocinas/metabolismo , Glucocorticoides/metabolismo , Humanos , Tolerancia Inmunológica/inmunología , Ratones , Esclerosis Múltiple/fisiopatología , Estrés Psicológico/complicaciones , Estrés Psicológico/fisiopatología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/metabolismo
14.
J Neuroimmunol ; 220(1-2): 79-89, 2010 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-20167380

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) infection is a well-characterized model of multiple sclerosis (MS). Previous research has shown that chronic restraint stress (RS) during early TMEV infection exacerbates behavioral signs of the disease. The present data suggest that RS-induced increases in CNS inflammation, demyelination, and axonal degeneration may underlie this exacerbation. In addition, we report that males exhibit greater CNS inflammation and higher numbers of demyelinating lesions while females show greater susceptibility to RS-induced exacerbation. These findings indicate that RS during early TMEV infection increases CNS lesion formation during the late phase and suggest that the effects of RS are sex-dependent.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Sistema Nervioso Central/inmunología , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Encefalomielitis/inmunología , Estrés Psicológico/inmunología , Theilovirus/inmunología , Animales , Axones/inmunología , Axones/patología , Axones/virología , Infecciones por Cardiovirus/fisiopatología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Enfermedad Crónica , Enfermedades Autoinmunes Desmielinizantes SNC/fisiopatología , Enfermedades Autoinmunes Desmielinizantes SNC/virología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Encefalomielitis/fisiopatología , Encefalomielitis/virología , Femenino , Masculino , Ratones , Fibras Nerviosas Mielínicas/inmunología , Fibras Nerviosas Mielínicas/patología , Fibras Nerviosas Mielínicas/virología , Restricción Física/efectos adversos , Restricción Física/psicología , Índice de Severidad de la Enfermedad , Caracteres Sexuales , Estrés Psicológico/fisiopatología , Degeneración Walleriana/inmunología , Degeneración Walleriana/patología , Degeneración Walleriana/virología
15.
Rev Neurol (Paris) ; 165(10): 789-95, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19656540

RESUMEN

The involvement of a viral infection in the physiopathology of multiple sclerosis has been said to cause certain viruses to target the central nervous system and induce neuroinflammation leading to cell dysfunction, as seen, for example, by demyelination or neuronal death. The most recent results of the literature have focused on the Herpes family viruses (HHV-6 and HHV-4/Epstein-Barr virus) and their possible role in the development of multiple sclerosis. Even if no virus has been identified so far as the multiple sclerosis etiological agent, our aim here is to show that some viruses may be responsible for triggering or sustaining neurological diseases. This is particularly the case for Paramyxoviruses, in the late appearance of functional alterations, Picornaviruses, in inducing a breakdown of immune tolerance, epitope spreading and demyelination, and Herpes viruses in inducing T and B lymphocyte activation, T lymphocytes dysregulation and autoimmunity after their reactivation. Therefore, "common" viruses can play a role as potential modulators of the immune and nervous systems which, in the specific context of dysimmunity and genetic susceptibility, stimulate a favorable background to the development of multiple sclerosis. Tracing and studying viruses in multiple sclerosis patients may improve our understanding of their actual involvement in multiple sclerosis physiopathology.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/fisiopatología , Esclerosis Múltiple/fisiopatología , Animales , Autoinmunidad/inmunología , Linfocitos B/virología , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/psicología , Enfermedades Virales del Sistema Nervioso Central/complicaciones , Enfermedades Virales del Sistema Nervioso Central/patología , Enfermedades Virales del Sistema Nervioso Central/virología , Enfermedad Crónica , Epítopos , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/fisiopatología , Humanos , Inflamación/etiología , Inflamación/patología , Sarampión/complicaciones , Sarampión/patología , Sarampión/fisiopatología , Sarampión/virología , Proteína Cofactora de Membrana/genética , Ratones , Esclerosis Múltiple/etiología , Esclerosis Múltiple/patología , Esclerosis Múltiple/virología , Theilovirus
16.
J Virol ; 83(16): 8004-11, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19535454

RESUMEN

The D variant of encephalomyocarditis virus (EMC-D virus) causes diabetes in mice by destroying pancreatic beta cells. In mice infected with a low dose of EMC-D virus, macrophages play an important role in beta-cell destruction by producing soluble mediators such as interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNF-alpha), and nitric oxide (NO). To investigate the role of NO and inducible NO synthase (iNOS) in the development of diabetes in EMC-D virus-infected mice, we infected iNOS-deficient DBA/2 mice with EMC-D virus (2 x 10(2) PFU/mouse). Mean blood glucose levels in EMC-D virus-infected iNOS-deficient mice and wild-type mice were 205.5 and 466.7 mg/dl, respectively. Insulitis and macrophage infiltration were reduced in islets of iNOS-deficient mice compared with wild-type mice at 3 days after EMC-D virus infection. Apoptosis of beta cells was decreased in iNOS-deficient mice, as evidenced by reduced numbers of terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling-positive cells. There were no differences in mRNA expression of antiapoptotic molecules Bcl-2, Bcl-xL, Bcl-w, Mcl-1, cIAP-1, and cIAP-2 between wild-type and iNOS-deficient mice, whereas expression of proapoptotic Bax and Bak mRNAs was significantly decreased in iNOS-deficient mice. Expression of IL-1beta and TNF-alpha mRNAs was significantly decreased in both islets and macrophages of iNOS-deficient mice compared with wild-type mice after EMC-D virus infection. Nuclear factor kappaB was less activated in macrophages of iNOS-deficient mice after virus infection. We conclude that NO plays an important role in the activation of macrophages and apoptosis of pancreatic beta cells in EMC-D virus-infected mice and that deficient iNOS gene expression inhibits macrophage activation and beta-cell apoptosis, contributing to prevention of EMC-D virus-induced diabetes.


Asunto(s)
Infecciones por Cardiovirus/virología , Diabetes Mellitus/virología , Virus de la Encefalomiocarditis/patogenicidad , Óxido Nítrico/inmunología , Animales , Apoptosis , Glucemia , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/fisiopatología , Células Cultivadas , Diabetes Mellitus/inmunología , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis/inmunología , Virus de la Encefalomiocarditis/fisiología , Femenino , Humanos , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/inmunología , Macrófagos/inmunología , Macrófagos/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , FN-kappa B/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología
17.
J Virol ; 83(14): 7273-84, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19420082

RESUMEN

Apoptosis is a common antiviral defensive mechanism that potentially limits viral reproduction and spread. Many viruses possess apoptosis-suppressing tools. Here, we show that the productive infection of HeLa cells with encephalomyocarditis virus (a cardiovirus) was not accompanied by full-fledged apoptosis (although the activation of caspases was detected late in infection) but rather elicited a strong antiapoptotic state, as evidenced by the resistance of infected cells to viral and nonviral apoptosis inducers. The development of the antiapoptotic state appeared to depend on a function(s) of the viral leader (L) protein, since its mutational inactivation resulted in the efflux of cytochrome c from mitochondria, the early activation of caspases, and the appearance of morphological and biochemical signs of apoptosis in a significant proportion of infected cells. Infection with both wild-type and L-deficient viruses induced the fragmentation of mitochondria, which in the former case was not accompanied with cytochrome c efflux. Although the exact nature of the antiapoptotic function(s) of cardioviruses remains obscure, our results suggested that it includes previously undescribed mechanisms operating upstream and possibly downstream of the mitochondrial level, and that L is involved in the control of these mechanisms. We propose that cardiovirus L belongs to a class of viral proteins, dubbed here security proteins, whose roles consist solely, or largely, in counteracting host antidefenses. Unrelated L proteins of other picornaviruses as well as their highly variable 2A proteins also may be security proteins. These proteins appear to be independent acquisitions in the evolution of picornaviruses, implying multiple cases of functional (though not structural) convergence.


Asunto(s)
Apoptosis , Infecciones por Cardiovirus/fisiopatología , Virus de la Encefalomiocarditis/metabolismo , Proteínas Virales/metabolismo , Animales , Cardiovirus/genética , Cardiovirus/metabolismo , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/virología , Línea Celular , Cricetinae , Citocromos c/metabolismo , Virus de la Encefalomiocarditis/genética , Células HeLa , Humanos , Mitocondrias/metabolismo , Proteínas Virales/genética
18.
Mol Cell Neurosci ; 40(2): 258-66, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19059482

RESUMEN

Adhesion molecules are critical players in the regulation of transmigration of blood leukocytes across the blood-brain barrier in multiple sclerosis (MS). Cannabinoids (CBs) are potential therapeutic agents in the treatment of MS, but the mechanisms involved are only partially known. Using a viral model of MS we observed that the cannabinoid agonist WIN55,212-2 administered at the time of virus infection suppresses intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) in brain endothelium, together with a reduction in perivascular CD4+ T lymphocytes infiltrates and microglial responses. WIN55,212-2 also interferes with later progression of the disease by reducing symptomatology and neuroinflammation. In vitro data from brain endothelial cell cultures, provide the first evidence of a role of peroxisome proliferator-activated receptors gamma (PPARgamma) in WIN55,212-2-induced downregulation of VCAM-1. This study highlights that inhibition of brain adhesion molecules by WIN55,212-2 might underline its therapeutic effects in MS models by targeting PPAR-gamma receptors.


Asunto(s)
Cannabinoides/agonistas , Cannabinoides/farmacología , Endotelio/efectos de los fármacos , Molécula 1 de Adhesión Intercelular/metabolismo , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/patología , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Conducta Animal/fisiología , Benzoxazinas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Cannabinoides/uso terapéutico , Infecciones por Cardiovirus/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Endotelio/citología , Endotelio/metabolismo , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/genética , Ratones , Morfolinas/farmacología , Actividad Motora/fisiología , Esclerosis Múltiple/fisiopatología , Naftalenos/farmacología , PPAR gamma/metabolismo , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Theilovirus/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética
19.
J Neuroinflammation ; 5: 44, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18922161

RESUMEN

BACKGROUND: The periventricular subventricular zone (SVZ) contains stem cells and is an area of active neurogenesis and migration. Since inflammation can reduce neurogenesis, we tested whether Theiler's murine encephalomyelitis virus (TMEV) induces inflammation and reduces neurogenesis in the SVZ. METHODS: We performed immmunohistochemistry for the hematopoietic cell marker CD45 throughout the central nervous system and then examined neuroblasts in the SVZ. RESULTS: CD45+ activation (inflammation) occurred early in the forebrain and preceded cerebellar and spinal cord inflammation. Inflammation in the brain was regionally stochastic except for the SVZ and surrounding periventricular regions where it was remarkably pronounced and consistent. In preclinical mice, SVZ neuroblasts emigrated into inflamed periventricular regions. The number of proliferating phoshpohistone3+ cells and Doublecortin+ (Dcx) SVZ neuroblasts was overall unaffected during the periods of greatest inflammation. However the number of Dcx+ and polysialylated neural cell adhesion molecule (PSA-NCAM+) SVZ neuroblasts decreased only after periventricular inflammation abated. CONCLUSION: Our results suggest that after TMEV infection, the SVZ may mount an attempt at neuronal repair via emigration, a process dampened by decreases in neuroblast numbers.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Encefalitis/inmunología , Células Madre Hematopoyéticas/inmunología , Antígenos Comunes de Leucocito/inmunología , Neurogénesis/inmunología , Theilovirus/inmunología , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Infecciones por Cardiovirus/fisiopatología , Linaje de la Célula/inmunología , Movimiento Celular/inmunología , Proliferación Celular , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Encefalitis/fisiopatología , Femenino , Células Madre Hematopoyéticas/virología , Histonas/metabolismo , Ventrículos Laterales , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Regeneración Nerviosa/inmunología , Plasticidad Neuronal/inmunología , Neuronas/citología , Neuronas/inmunología , Neuropéptidos/metabolismo , Prosencéfalo/inmunología , Prosencéfalo/patología , Prosencéfalo/fisiopatología
20.
Physiol Behav ; 95(1-2): 63-71, 2008 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-18538803

RESUMEN

Previous research has shown that chronic restraint stress exacerbates Theiler's virus infection, a murine model for CNS inflammation and multiple sclerosis. The current set of experiments was designed to evaluate the potential role of glucocorticoids in the deleterious effects of restraint stress on acute CNS inflammatory disease. Exposure to chronic restraint stress resulted in elevated levels of corticosterone as well as increased clinical scores and weight loss (Experiment 1). In addition, corticosterone administration alone exacerbated behavioral signs of TMEV-induced sickness (i.e. decreased body weight, increased symptoms of encephalitis, and increased mortality) and reduced inflammation in the CNS (Experiment 2). Infected subjects receiving exogenous corticosterone showed exacerbation of acute phase measures of sickness and severe mortality as well as decreased viral clearance from CNS (Experiment 3). These findings indicate that corticosterone exposure alone is sufficient to exacerbate acute CNS inflammatory disease.


Asunto(s)
Infecciones por Cardiovirus/etiología , Infecciones por Cardiovirus/fisiopatología , Glucocorticoides/administración & dosificación , Theilovirus/patogenicidad , Animales , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/mortalidad , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Glucocorticoides/metabolismo , Masculino , Meningitis/etiología , Meningitis/patología , Meningitis/virología , Ratones , Ratones Endogámicos CBA , Mortalidad , Índice de Severidad de la Enfermedad , Estrés Psicológico/fisiopatología , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...