Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
Nat Med ; 4(12): 1383-91, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9846575

RESUMEN

In viral myocarditis, inflammation and destruction of cardiac myocytes leads to fibrosis, causing progressive impairment in cardiac function. Here we show the etiologic importance of serine elastase activity in the pathophysiology of acute viral myocarditis and the therapeutic efficacy of an elastase inhibitor. In DBA/2 mice inoculated with the encephalomyocarditis virus, a more than 150% increase in myocardial serine elastase activity is observed. This is suppressed by a selective serine elastase inhibitor, ZD0892, which is biologically effective after oral administration. Mice treated with this compound had little evidence of microvascular constriction and obstruction associated with myocarditis-induced ischemia reperfusion injury, much less inflammation and necrosis, only mild fibrosis and myocardial collagen deposition, and normal ventricular function, compared with the infected nontreated group.


Asunto(s)
Infecciones por Cardiovirus/tratamiento farmacológico , Corazón/fisiología , Inflamación/tratamiento farmacológico , Miocarditis/tratamiento farmacológico , Pirroles/uso terapéutico , Inhibidores de Serina Proteinasa/uso terapéutico , Sulfonamidas/uso terapéutico , Administración Oral , Animales , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Modelos Animales de Enfermedad , Virus de la Encefalomiocarditis , Fibrosis/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos DBA , Microcirculación/efectos de los fármacos , Miocarditis/fisiopatología , Miocarditis/virología , Miocardio/patología , Perfusión , Peroxidasa/metabolismo , Pirroles/administración & dosificación , Inhibidores de Serina Proteinasa/administración & dosificación , Sulfonamidas/administración & dosificación
6.
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
7.
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
8.
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
9.
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
10.
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
11.
Curr Top Microbiol Immunol ; 318: 177-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18219818

RESUMEN

Myelin repair (remyelination) following the demyelination of central nervous system (CNS) axons in diseases such as multiple sclerosis plays a critical role in determining the level of accompanying neurologic disability. While remyelination can be quite robust, in multiple sclerosis it often fails. Understanding and stimulating the remyelination process are therefore important goals in MS research. Remyelination is a complex cellular process that involves an intimate interplay between the myelin-producing cells of the CNS (oligodendrocytes), the axons to be myelinated, as well as CNS-infiltrating immune cells. Genetic analysis can be a powerful tool for the functional analysis of complex cellular processes and has recently been applied to the problem of remyelination failure during disease. This chapter reviews the recent use of genetic approaches for the study of CNS remyelination in mouse models of demyelinating disease.


Asunto(s)
Enfermedades del Sistema Nervioso Central/genética , Enfermedades del Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología , Animales , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/fisiopatología , Enfermedades del Sistema Nervioso Central/fisiopatología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Humanos , Ratones , Vaina de Mielina/patología , Theilovirus
12.
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
13.
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
14.
Neuropathol Appl Neurobiol ; 34(6): 603-20, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18466224

RESUMEN

AIMS: Limited remyelination is a key feature of demyelinating Theiler's murine encephalomyelitis (TME). It is hypothesized that a dysregulation of differentiation of oligodendroglial progenitor cells (OPCs) represents the main cause of insufficient regeneration in this model of multiple sclerosis. METHODS: TME virus (TMEV)-infected SJL/J mice were evaluated by footprint analysis, light and electron microscopy, immunohistology, confocal immunofluorescence and RT-qPCR at multiple time points ranging from 1 h to 196 days post infection (dpi). RESULTS: Footprint analysis revealed a significantly decreased stride length at 147 and 196 dpi. Demyelination progressively increased from 14 towards 196 dpi. A mild amount of remyelination was detected at 147 and 196 dpi. Early onset axonal injury was detected from 14 dpi on. TMEV RNA was detectable throughout the observation period and markedly increased between 7 and 28 dpi. Intralesional nerve/glial antigen 2 (NG2)-positive OPCs were temporarily increased between 28 and 98 dpi. Similarly, a transient upregulation of NG2 and platelet-derived growth factor alpha-receptor mRNA was noticed. In contrast, intralesional 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase)-positive oligodendrocytes were decreased between 56 and 196 dpi. Although CNPase mRNA remained unchanged, myelin basic protein mRNA and especially its exon 2 containing splice variants were decreased. Glial fibrillary acidic protein (GFAP)-positive astrocytes and GFAP mRNA were increased in the late phase of TME. A mildly increased colocalization of both NG2/CNPase and NG2/GFAP was revealed at 196 dpi. CONCLUSIONS: Summarized, the present results indicated a dysregulation of OPC maturation as the main cause for the delayed and limited remyelination in TME. A shift of OPC differentiation from oligodendroglial towards astrocytic differentiation is postulated.


Asunto(s)
Antígenos/metabolismo , Infecciones por Cardiovirus/fisiopatología , Encefalomielitis/fisiopatología , Esclerosis Múltiple/fisiopatología , Vaina de Mielina/fisiología , Proteoglicanos/metabolismo , Células Madre/citología , Theilovirus , 2',3'-Nucleótido Cíclico Fosfodiesterasas/genética , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Antígenos/genética , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/virología , Diferenciación Celular , Encefalomielitis/patología , Encefalomielitis/virología , Femenino , Expresión Génica , Proteína Ácida Fibrilar de la Glía , Inmunohistoquímica , Ratones , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Esclerosis Múltiple/patología , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Vaina de Mielina/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Oligodendroglía/citología , Oligodendroglía/fisiología , Proteoglicanos/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Médula Espinal/patología , Médula Espinal/fisiopatología , Médula Espinal/virología , Células Madre/fisiología , Theilovirus/aislamiento & purificación
15.
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
16.
J Neuroimmunol ; 190(1-2): 112-20, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17919740

RESUMEN

Little is known about antibody production within the central nervous system, called intrathecal antibody production (ITAbP). Mice infected with Theiler's murine encephalomyocarditis virus (TMEV) develop an immune-mediated demyelinating disease(TMEV-IDD), characterized by progressive weakness associated with robust ITAbP, CNS inflammation and demyelination. TMEV-IDD represents an excellent model of human multiple sclerosis (MS), especially its progressive disability. The mechanism of the weakness in this disease is unknown but there is considerable evidence that it is immune-mediated. Our hypothesis was that the disability in the model is induced by robust ITAbP by antibody-secreting cells(ASCs) in the CNS. We further hypothesized that these ASCs are predominantly CD138+ plasma cells, driven by the persistence of virus at high levels in the central nervous system (CNS) as well as high levels of B-cell activating factor (BAFF). In order to test these hypotheses we infected mice with TMEV, and correlated measures of ITAbP with disability. Measures of ITAbP were ELISpot and IgG gene expression, while disability was tested by Rotarod. We found that disability was highly correlated with ITAbP, assayed by number of CNS ASCs as well as amount of IgG mRNA. CNS ASCs were primarily CD138+, and produced high levels of IgG enriched for reactivity to TMEV. There were high levels of BAFF production in the CNS, but these levels were only minimally higher in infected mice than in uninfected controls. TMEV and IgG RNA were distributed throughout the spinal cord. These data are the first demonstrating that ITAbP is highly correlated with disability in TMEV infection, an excellent model of human MS. Our data raise the possibility that ITAbP contributes significantly to disability, both in this model and in MS.


Asunto(s)
Anticuerpos/inmunología , Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Esclerosis Múltiple/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Animales , Factor Activador de Células B/inmunología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Sistema Nervioso Central/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Inmunoglobulina G/inmunología , Ratones , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/virología , Fenotipo , Células Plasmáticas/virología , ARN Mensajero/metabolismo , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/fisiopatología , Sindecano-1/inmunología , Theilovirus/inmunología
17.
Ann N Y Acad Sci ; 1103: 152-6, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17376832

RESUMEN

The ability to study the immediate, early events in the demyelinating process has been difficult on account of the lack of model systems that address this phase of lesion development. The vast majority of animal models used to study multiple sclerosis (MS) focuses on the later events of myelin destruction. To address this deficiency, we have modified the currently used Theiler's murine encephalomyelitis virus (TMEV)-induced model of demyelination to precisely identify the area where virus-induced demyelination first occurs. Following surgical exposure of the spinal cord, we directly injected TMEV into the spinal cord of female SJL/J mice. Characterization of the events in the spinal cord in the days following injection of virus support the use of this model to dissect the pathways triggered in the host in the early phases of demyelination. A complete understanding of the genesis of the sclerotic plaque may provide insights into enhanced treatment for patients with central nervous system (CNS) demyelination.


Asunto(s)
Infecciones por Cardiovirus/fisiopatología , Theilovirus , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos , Proteína Proteolipídica de la Mielina/genética , ARN Mensajero/genética , Médula Espinal/fisiopatología , Médula Espinal/virología
18.
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
19.
J Neuroimmunol ; 177(1-2): 46-62, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16762424

RESUMEN

Chronic restraint stress, administered during early infection with Theiler's virus, was found to exacerbate the acute CNS viral infection in male and female mice. During the subsequent demyelinating phase of disease (a model of multiple sclerosis), the effect of stress on disease progression was sex-dependent. Previously stressed male mice had less severe behavioral signs of the chronic phase, better rotarod performance and decreased inflammatory lesions of the spinal cord, while the opposite pattern was observed in females. In addition, mice in all groups developed autoantibodies to MBP, PLP139-151 and MOG33-55.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Enfermedades Autoinmunes Desmielinizantes SNC/inmunología , Tolerancia Inmunológica/inmunología , Estrés Psicológico/complicaciones , Estrés Psicológico/inmunología , Enfermedad Aguda , Animales , Autoanticuerpos/inmunología , Conducta Animal/fisiología , Infecciones por Cardiovirus/fisiopatología , Enfermedad Crónica , Enfermedades Autoinmunes Desmielinizantes SNC/fisiopatología , Enfermedades Autoinmunes Desmielinizantes SNC/virología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos CBA , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/virología , Proteínas de la Mielina/inmunología , Fibras Nerviosas Mielínicas/inmunología , Fibras Nerviosas Mielínicas/patología , Restricción Física , Factores Sexuales , Médula Espinal/inmunología , Médula Espinal/patología , Médula Espinal/fisiopatología , Estrés Psicológico/fisiopatología , Theilovirus/inmunología , Factores de Tiempo
20.
Virus Res ; 111(2): 214-23, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15893838

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) infection of mice, in which persistent central nervous system (CNS) infection induces Th1 CD4+ T cell responses to both virus and myelin proteins, provides a relevant experimental animal model for MS. During persistence, >10(9) TMEV genome equivalents per spinal cord are detectable by real-time reverse transcription-polymerase chain reaction (RT-PCR). Because of the short half-life of TMEV (<1 day), continual viral replication is needed to sustain these very high TMEV copy numbers. An essential role for macrophages in TMEV persistence has been documented and, although limited by host anti-viral immune responses, TMEV nonetheless spreads during persistence to infect other cells, particularly oligodendrocytes, in which the infection is productive and lytic. Virus factors influencing persistence of TMEV are expression of the out-of-frame L* protein and use of sialic acid co-receptors.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central/virología , Macrófagos/virología , Oligodendroglía/virología , Theilovirus/fisiología , Theilovirus/patogenicidad , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/fisiopatología , Infecciones por Cardiovirus/virología , Enfermedades Virales del Sistema Nervioso Central/inmunología , Enfermedades Virales del Sistema Nervioso Central/fisiopatología , Modelos Animales de Enfermedad , Humanos , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Esclerosis Múltiple/virología , Theilovirus/genética , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA