Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Sci Rep ; 11(1): 23819, 2021 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-34893671

RESUMEN

Neurotropic viruses target the brain and contribute to neurologic diseases. C-type lectin receptors (CLRs) are pattern recognition receptors that recognize carbohydrate structures on endogenous molecules and pathogens. The myeloid CLR dendritic cell immunoreceptor (DCIR) is expressed by antigen presenting cells and mediates inhibitory intracellular signalling. To investigate the effect of DCIR on neurotropic virus infection, mice were infected experimentally with Theiler's murine encephalomyelitis virus (TMEV). Brain tissue of TMEV-infected C57BL/6 mice and DCIR-/- mice were analysed by histology, immunohistochemistry and RT-qPCR, and spleen tissue by flow cytometry. To determine the impact of DCIR deficiency on T cell responses upon TMEV infection in vitro, antigen presentation assays were utilised. Genetic DCIR ablation in C57BL/6 mice was associated with an ameliorated hippocampal integrity together with reduced cerebral cytokine responses and reduced TMEV loads in the brain. Additionally, absence of DCIR favoured increased peripheral cytotoxic CD8+ T cell responses following TMEV infection. Co-culture experiments revealed that DCIR deficiency enhances the activation of antigen-specific CD8+ T cells by virus-exposed dendritic cells (DCs), indicated by increased release of interleukin-2 and interferon-γ. Results suggest that DCIR deficiency has a supportive influence on antiviral immune mechanisms, facilitating virus control in the brain and ameliorates neuropathology during acute neurotropic virus infection.


Asunto(s)
Infecciones por Cardiovirus/virología , Hipocampo/metabolismo , Hipocampo/virología , Lectinas Tipo C/metabolismo , Theilovirus/fisiología , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Biomarcadores , Biopsia , Citocinas/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Hipocampo/patología , Inmunohistoquímica , Inmunomodulación , Lectinas Tipo C/genética , Ratones , Ratones Noqueados , Enfermedades Neuroinflamatorias/inmunología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología , Carga Viral
2.
Biochem Biophys Res Commun ; 585: 162-168, 2021 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-34808499

RESUMEN

tRNase ZS (ELAC1) and TRNT1 function in tRNA recycling. Recently, we have shown that these genes are upregulated in the cells infected with Theiler's mouse encephalitis virus (TMEV), implying that tRNA recycling functions in response to viral infection. To address the molecular mechanism underlying the ELAC1 upregulation in the cells infected with TMEV, we performed luciferase assays using various plasmid constructs harboring the ELAC1 promoter region. The luciferase expression from a construct containing the full-length ELAC1 promoter was augmented by TMEV, poly IC, IFN-ß, or IFN-γ. We identified four IFN-stimulated responsible elements (ISREs) in the proximal promoter region. The luciferase expression from the constructs that lack all the ISREs was strongly reduced compared with that from the constructs with the four ISREs in the presence of IFN-ß or IFN-γ. The observation that the ISREs from the ELAC1 promoter are essential for the gene upregulation by IFN-ß or IFN-γ suggests that the ELAC1 gene is upregulated by IFNs.


Asunto(s)
Interferones/farmacología , Regiones Promotoras Genéticas/genética , ARN de Transferencia/genética , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba/efectos de los fármacos , Antivirales/farmacología , Secuencia de Bases , Células HeLa , Humanos , Interferón beta/farmacología , Interferón gamma/farmacología , ARN de Transferencia/metabolismo , Elementos de Respuesta/genética , Theilovirus/efectos de los fármacos , Theilovirus/fisiología , Regulación hacia Arriba/genética
3.
Artículo en Inglés | MEDLINE | ID: mdl-29410948

RESUMEN

CXCL-1, also called keratinocyte-derived cytokine (KC), is a predominant chemokine produced in glial cells upon infection with Theiler's murine encephalomyelitis virus (TMEV). In this study, we assessed the role of KC in the development of TMEV-induced demyelinating disease by utilizing polyclonal anti-KC antibodies as well as KC-expressing recombinant TMEV. Our results indicate that the level of KC produced after infection with TMEV or stimulation with various TLRs is significantly higher in various cells from susceptible SJL mice compared to those in cells from resistant B6 mice. SJL mice treated with rabbit anti-KC antibodies displayed accelerated development of TMEV-induced demyelinating disease, elevated viral loads in the CNS and decreased antiviral T cell responses. In addition, infection of susceptible SJL mice with recombinant KC-TMEV produced biologically active KC, which resulted in the accelerated pathogenesis of demyelinating disease and elevated T cell responses to viral antigens compared to mice infected with control recombinant HEL-TMEV. These results strongly suggest that both the lack of KC during TMEV infection and the excessive presence of the chemokine promote the pathogenesis of demyelinating disease. Therefore, a balance in the level of KC during TMEV infection appears to be critically important in controlling the pathogenesis of demyelinating disease.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/virología , Quimiocina CXCL1/metabolismo , Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/metabolismo , Theilovirus/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Astrocitos/inmunología , Astrocitos/metabolismo , Astrocitos/virología , Quimiocina CXCL1/antagonistas & inhibidores , Enfermedades Desmielinizantes/diagnóstico , Enfermedades Desmielinizantes/tratamiento farmacológico , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/farmacología , Interferón gamma/biosíntesis , Queratinocitos/metabolismo , Queratinocitos/virología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/virología , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Neutrófilos/virología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Virology ; 512: 21-24, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28898711

RESUMEN

It is well known that SJL mice are susceptible to Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease while C57BL6 (B6) and B10 mice are resistant, and H-2s on a B10 background (B10.S) contributes modestly to susceptibility. A recent study linked two IRF3 non-conservative mutations in SJL compared to B10.S mice to resistance to TMEV infection of SJL peritoneal-derived macrophages, an observation of practical interest in light of the central role of IRF3 transcription factor in the type I interferon (IFN) response. However, we did not find these non-conservative mutations among SJL, B10.S, B6 and B10 mice in the IRF3 amino acid sequence, and show SJL bone marrow-derived macrophages infected with TMEV exhibit increased virus RNA replication and infectious virus yields as well as greater IL-6 production than C57Bl strain (including B10.S) cultures.


Asunto(s)
Infecciones por Cardiovirus/virología , Predisposición Genética a la Enfermedad , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/metabolismo , Theilovirus/fisiología , Animales , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Factor 3 Regulador del Interferón/genética , Interleucina-6/genética , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos , Mutación
5.
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
6.
Genomics ; 107(4): 150-4, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26826341

RESUMEN

Long noncoding (lnc)RNAs comprise a diverse group of transcripts including large intervening noncoding (linc)RNAs, natural antisense transcripts (NATs) and intronic lncRNAs. The functions and mechanisms of more than 200 lncRNAs have been studied in vitro and the results suggest that lncRNAs may be molecular markers of prognosis in cancer patients. Some lncRNAs can promote virus replication and allow escape from cytosolic surveillance to suppress antiviral immunity. For example, lncRNA can cause persistent infection by Theiler's virus, and microRNA (miR)-27a/b is important for efficient murine cytomegalovirus (MCMV) replication. The available evidence suggests that lncRNAs may be potential targets of novel antiviral drugs.


Asunto(s)
ARN Largo no Codificante/genética , Replicación Viral , Virus , Adenovirus Humanos/fisiología , Animales , Humanos , Intrones , Ratones , Muromegalovirus/fisiología , Theilovirus/fisiología
7.
J Virol ; 90(7): 3573-83, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26792734

RESUMEN

UNLABELLED: Infected macrophages in spinal cords of mice persistently infected with Theiler's murine encephalomyelitis virus (TMEV) undergo apoptosis, resulting in restricted virus yields, as do infected macrophages in culture. Apoptosis of murine macrophages in culture occurs via the intrinsic pathway later in infection (>10 h postinfection [p.i.]) after maximal virus titers (150 to 200 PFU/cell) have been reached, with loss of most infectious virus (<5 PFU/cell) by 20 to 24 h p.i. Here, we show that BeAn virus RNA replication, translation, polyprotein processing into final protein products, and assembly of protomers and pentamers in infected M1-D macrophages did not differ from those processes in TMEV-infected BHK-21 cells, which undergo necroptosis. However, the initial difference from BHK-21 cell infection was seen at 10 to 12 h p.i., where virions from the 160S peak in sucrose gradients had incompletely processed VP0 (compared to that in infected BHK-21 cells). Thereafter, there was a gradual loss of the 160S virion peak in sucrose gradients, with replacement by a 216S peak that was observed to contain pentamers among lipid debris in negatively stained grids by electron microscopy. After infection or incubation of purified virions with activated caspase-3 in vitro, 13- and 17-kDa capsid peptide fragments were observed and were predicted by algorithms to contain cleavage sites within proteins by cysteine-dependent aspartate-directed proteases. These findings suggest that caspase cleavage of sites in exposed capsid loops of assembled virions occurs contemporaneously with the onset and progression of apoptosis later in the infection. IMPORTANCE: Theiler's murine encephalomyelitis virus (TMEV) infection in mice results in establishment of virus persistence in the central nervous system and chronic inflammatory demyelinating disease, providing an experimental animal model for multiple sclerosis. Virus persistence takes place primarily in macrophages recruited into the spinal cord that undergo apoptosis and in turn may facilitate viral spread via infected apoptotic blebs. Infection of murine macrophages in culture results in restricted virus yields late in infection. Here it is shown that the early steps of the virus life cycle in infected macrophages in vitro do not differ from these processes in TMEV-infected BHK-21 cells, which undergo necroptosis. However, the findings late in infection suggest that caspases cleave sites in exposed capsid loops and possibly internal sites of assembled virions occurring contemporaneously with onset and progression of apoptosis. Mechanistically, this would explain the dramatic loss in virus yields during TMEV-induced apoptosis and attenuate the virus, enabling persistence.


Asunto(s)
Caspasas/metabolismo , Macrófagos/virología , Theilovirus/fisiología , Virión/metabolismo , Ensamble de Virus , Animales , Apoptosis , Células Cultivadas , Cricetinae , Ratones , Proteolisis , Carga Viral
8.
Neurosci Lett ; 593: 124-8, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25796181

RESUMEN

Changes in the expression of xCT, the specific subunit of system xc(-) or the cystine/glutamate antiporter, have been associated with several neurological disorders and system xc(-) was recently proposed as a potential target for the development of new treatment strategies for multiple sclerosis (MS). In this study we used Theiler's murine encephalomyelitis virus (TMEV) infection, both in vitro and in vivo, as a model to further evaluate the involvement of system xc(-) in MS. Protein levels of xCT, as well as activity of system xc(-) were unaffected in RAW264.7 macrophages after infection with the demyelinating DA strain of TMEV. Also, protein expression of xCT remained stable in spinal cord and brain of FVB mice 1-2 and 6 weeks after intracranial injection of the DA strain of TMEV. These results demonstrate that TMEV infection of macrophages or FVB mice has no effect on system xc(-) and as such cannot be used as a model to study the involvement of system xc(-) in MS.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Infecciones por Cardiovirus/metabolismo , Macrófagos/metabolismo , Theilovirus/fisiología , Animales , Encéfalo/metabolismo , Infecciones por Cardiovirus/virología , Cisteína/metabolismo , Femenino , Macrófagos/virología , Ratones , Médula Espinal/metabolismo
9.
J Immunol ; 194(6): 2796-809, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25681345

RESUMEN

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


Asunto(s)
Infecciones por Cardiovirus/inmunología , Diferenciación Celular/inmunología , Macrófagos/inmunología , Miositis/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/inmunología , Theilovirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Antígeno CD47/inmunología , Antígeno CD47/metabolismo , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Diferenciación Celular/genética , Citometría de Flujo , Interacciones Huésped-Patógeno/efectos de los fármacos , Interacciones Huésped-Patógeno/inmunología , Humanos , Macrófagos/metabolismo , Macrófagos/patología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Monocitos/inmunología , Monocitos/metabolismo , Monocitos/patología , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/virología , Miositis/genética , Miositis/virología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Theilovirus/fisiología , Transcriptoma/inmunología , Replicación Viral/inmunología
10.
Virus Res ; 195: 177-82, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25449910

RESUMEN

In mice Theiler's murine encephalomyelitis virus (TMEV) persists in macrophages that eventually undergo apoptosis. TMEV infection of macrophages in culture induces apoptosis through the intrinsic pathway, restricting virus yields. We show that inhibition of TMEV-induced apoptosis leads to phosphorylation of receptor interacting protein 1 (RIP1), localization of RIP1 and RIP3 to mitochondria, ROS production independent of MAPK activation and programmed necrosis (necroptosis). Blocking both apoptosis and necroptosis restored virus yields.


Asunto(s)
Muerte Celular , Interacciones Huésped-Patógeno , Macrófagos/fisiología , Macrófagos/virología , Theilovirus/fisiología , Animales , Proteínas Activadoras de GTPasa/metabolismo , Ratones , Mitocondrias/química , Fosforilación , Procesamiento Proteico-Postraduccional , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Carga Viral
11.
Virology ; 462-463: 236-40, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24999048

RESUMEN

Cardioviruses of the Encephalomyocarditis virus (EMCV) and Theilovirus species encode small, amino-terminal proteins called Leaders (L). Phosphorylation of the EMCV L (LE) at two distinct sites by CK2 and Syk kinases is important for virus-induced Nup phosphorylation and nucleocytoplasmic trafficking inhibition. Despite similar biological activities, the LE phosphorylation sites are not conserved in the Theiloviruses, Saffold virus (LS, SafV) or Theiler׳s murine encephalitis virus (LT, TMEV) sequences even though these proteins also become phosphorylated in cells and cell-free extracts. Site prediction algorithms, combined with panels of site-specific protein mutations now identify analogous, but not homologous phosphorylation sites in the Ser/Thr and Theilo protein domains of LT and LS, respectively. In both cases, recombinant AMP-activated kinase (AMPK) was reactive with the proteins at these sites, and also with LE, modifying the same residue recognized by CK2.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiovirus/fisiología , Virus de la Encefalomiocarditis/fisiología , Procesamiento Proteico-Postraduccional , Theilovirus/fisiología , Proteínas Estructurales Virales/metabolismo , Animales , Ratones
12.
J Virol ; 88(15): 8479-89, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24829345

RESUMEN

UNLABELLED: Interleukin-6 (IL-6) plays an important role in the development and progression of inflammatory responses, autoimmune diseases, and cancers. Many viral infections, including Theiler's murine encephalomyelitis virus (TMEV), result in the vigorous production of IL-6. However, the role of IL-6 in the development of virus-induced inflammatory responses is unclear. The infection of susceptible mice with TMEV induces the development of chronic demyelinating disease, which is considered a relevant infectious model for multiple sclerosis. In this study, we demonstrate that resistant C57BL/6 mice carrying an IL-6 transgene (IL-6 Tg) develop a TMEV-induced demyelinating disease accompanied by an increase in viral persistence and an elevated Th17 cell response in the central nervous system. Either IL-6 or IL-17 induced the expression of Bcl-2 and Bcl-xL at a high concentration. The upregulated expression of prosurvival molecules in turn inhibited target cell destruction by virus-specific CD8(+) T cells. More interestingly, IL-6 and IL-17 synergistically promoted the expression of these prosurvival molecules, preventing cellular apoptosis at a much lower (<5-fold) concentration. The signals involved in the synergy appear to include the activation of both STAT3 and NF-κB via distinct cytokine-dependent pathways. Thus, the excessive IL-6 promotes the generation of Th17 cells, and the resulting IL-6 and IL-17 synergistically promote viral persistence by protecting virus-infected cells from apoptosis and CD8(+) T cell-mediated target destruction. These results suggest that blocking both IL-6 and IL-17 functions are important considerations for therapies of chronic viral diseases, autoimmune diseases, and cancers. IMPORTANCE: This study indicates that an excessive level of IL-6 cytokine produced following viral infection promotes the development of IL-17-producing pathogenic helper T cells. We demonstrate here for the first time that IL-6 together with IL-17 synergistically enhances the expression of survival molecules to hinder critical host defense mechanisms removing virus-infected cells. This finding has an important implication in controlling not only chronic viral infections but also autoimmune diseases and cancers, which are associated with prolonged cell survival.


Asunto(s)
Apoptosis , Evasión Inmune , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Linfocitos T Citotóxicos/inmunología , Theilovirus/inmunología , Theilovirus/fisiología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
13.
Virus Res ; 178(2): 226-33, 2013 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-24140628

RESUMEN

IRF3 is an innate anti-viral factor whose role in limiting Theiler's murine encephalomyelitis virus (TMEV) infection and preventing TMEV-induced disease is unclear. Acute disease and innate immune responses of macrophages were examined in IRF3 knockout mice compared with C57Bl/6 mice following in vitro or intracranial infection with either TMEV GDVII or DA. IRF3 deficiency augmented viral infection, as well as morbidity and mortality following intracranial infection with neurovirulent TMEV GDVII. In contrast, IRF3 deficiency prevented hippocampal injury following intracranial infection with persistent TMEV DA. The extent of TMEV infection in macrophages from C57Bl/6 mice was significantly less than that in IRF3 deficient macrophages, which was associated with poor IFN-ß and IL-6 expression in response to TMEV. Reestablishing IRF3 expression in IRF3 deficient macrophages increased control of TMEV replication and increased expression of IFN-ß and IL-6. In addition, IRF3 deficient macrophages failed to exhibit IL-6 antiviral effects, which was associated with inability to sustain IL-6-induced STAT1 activation compared with C57BL/6 macrophages. Altogether, IRF3 contributes to early control of TMEV replication through induction of IL-6 and IFN-ß and support of IL-6 antiviral effects, but contributes to TMEV-induced hippocampal injury.


Asunto(s)
Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Interacciones Huésped-Patógeno , Factor 3 Regulador del Interferón/metabolismo , Interleucina-6/inmunología , Theilovirus/inmunología , Animales , Infecciones por Cardiovirus/virología , Hipocampo/inmunología , Hipocampo/patología , Hipocampo/virología , Factor 3 Regulador del Interferón/deficiencia , Interferón beta/biosíntesis , Interferón beta/inmunología , Interleucina-6/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Índice de Severidad de la Enfermedad , Análisis de Supervivencia , Theilovirus/fisiología
14.
Am J Pathol ; 183(5): 1390-1396, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24091251

RESUMEN

The polyphenol compound resveratrol is reported to have multiple functions, including neuroprotection, and no major adverse effects have been reported. Although the neuroprotective effects have been associated with sirtuin 1 activation by resveratrol, the mechanisms by which resveratrol exerts such functions are a matter of controversy. We examined whether resveratrol can be neuroprotective in two models of multiple sclerosis: experimental autoimmune encephalomyelitis (EAE) and Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). EAE was induced in C57BL/6 mice, which were fed a control diet or a diet containing resveratrol during either the induction or effector phase or through the whole course of EAE. SJL/J mice were infected with TMEV and fed a control diet or a diet containing resveratrol during the chronic phase of TMEV-IDD. In EAE, all groups of mice treated with resveratrol had more severe clinical signs than the control group. In particular, resveratrol treatment during the induction phase resulted in the most severe EAE, both clinically and histologically. Similarly, in the viral model, the mice treated with resveratrol developed significantly more severe TMEV-IDD than the control group. Thus, surprisingly, the resveratrol treatment significantly exacerbated demyelination and inflammation without neuroprotection in the central nervous system in both models. Our findings indicate that caution should be exercised in potential therapeutic applications of resveratrol in human inflammatory demyelinating diseases, including multiple sclerosis.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Progresión de la Enfermedad , Esclerosis Múltiple/patología , Esclerosis Múltiple/virología , Estilbenos/efectos adversos , Theilovirus/fisiología , Animales , Axones/efectos de los fármacos , Axones/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/virología , Humanos , Ratones , Ratones Endogámicos C57BL , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/inmunología , Glicoproteína Mielina-Oligodendrócito/inmunología , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/inmunología , Degeneración Nerviosa/patología , Degeneración Nerviosa/virología , Fármacos Neuroprotectores/efectos adversos , Resveratrol , Theilovirus/patogenicidad , Virulencia
15.
Methods Mol Biol ; 900: 381-401, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22933080

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) and Theiler's Murine Encephalitis Virus-Induced Demyelinating Disease (TMEV-IDD) are two clinically relevant murine models of multiple sclerosis (MS). Like MS, both are characterized by mononuclear cell infiltration into the CNS and demyelination. EAE is induced by either the administration of myelin protein or peptide in adjuvant or by the adoptive transfer of encephalitogenic T cell blasts into naïve recipients. The relative merits of each of these protocols are compared. Depending on the type of question being asked, different mouse strains and peptides are used. Different disease courses are observed with different strains and different peptides in active EAE. These variations are also addressed. Additionally, issues relevant to clinical grading of EAE in mice are discussed. In addition to EAE induction, useful references for other disease indicators such as DTH, in vitro proliferation, and immunohistochemistry are provided. TMEV-IDD is a useful model for understanding the possible viral etiology of MS. This section provides detailed information on the preparation of viral stocks and subsequent intracerebral infection of mice. Additionally, virus plaque assay and clinical disease assessment are discussed. Recently, recombinant TMEV strains have been created for the study of molecular mimicry which incorporate various 30 amino acid myelin epitopes within the leader region of TMEV.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/virología , Técnicas Inmunológicas/métodos , Esclerosis Múltiple/patología , Theilovirus/fisiología , Traslado Adoptivo , Secuencia de Aminoácidos , Animales , Progresión de la Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Esclerosis Múltiple/inmunología , Péptidos/química , Vacunación
16.
Sci Rep ; 2: 545, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22848791

RESUMEN

Neuronal injury during acute viral infection of the brain is associated with the development of persistent cognitive deficits and seizures in humans. In C57BL/6 mice acutely infected with the Theiler's murine encephalomyelitis virus, hippocampal CA1 neurons are injured by a rapid innate immune response, resulting in profound memory deficits. In contrast, infected SJL and B6xSJL F1 hybrid mice exhibit essentially complete hippocampal and memory preservation. Analysis of brain-infiltrating leukocytes revealed that SJL mice mount a sharply attenuated inflammatory monocyte response as compared to B6 mice. Bone marrow transplantation experiments isolated the attenuation to the SJL immune system. Adoptive transfer of B6 inflammatory monocytes into acutely infected B6xSJL hosts converted these mice to a hippocampal damage phenotype and induced a cognitive deficit marked by failure to recognize a novel object. These findings show that inflammatory monocytes are the critical cellular mediator of hippocampal injury during acute picornavirus infection of the brain.


Asunto(s)
Hipocampo/inmunología , Hipocampo/virología , Monocitos/inmunología , Poliomielitis/inmunología , Poliomielitis/virología , Theilovirus/fisiología , Traslado Adoptivo , Animales , Apoptosis , Trasplante de Médula Ósea , Trastornos del Conocimiento/etiología , Modelos Animales de Enfermedad , Femenino , Hipocampo/patología , Inmunofenotipificación , Masculino , Ratones , Monocitos/citología , Monocitos/metabolismo , Neuronas/patología , Poliomielitis/patología , Tropismo Viral , Replicación Viral
17.
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
18.
Cell Commun Adhes ; 17(3): 57-68, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20825262

RESUMEN

The present article reports the up-regulation of the expression of the vascular cell adhesion molecule-1 (VCAM-1) by SJL/J mouse brain astrocytes infected with Theiler's murine encephalomyelitis virus (TMEV). Complementary RNA (cRNA) from mock- and TMEV-infected cells was hybridized to the Affymetrix whole murine genome U74v2 DNA microarray. Hybridization data analysis revealed background expression in untreated cells and the up-regulation of three sequences coding for VCAM-1, as described by the SCOP (Structural Classification Of Proteins) database. The authors further studied its regulation, confirming and validating their mRNA increase by reverse transcriptase-polymerase chain reaction (RT-PCR) and quantitative real-time RT-PCR. The presence of the 100-kDa VCAM-1 protein in mock- and TMEV-infected cells was demonstrated in the cell membrane by a specific cell-based enzyme-linked immunosorbent assay (ELISA), in addition to flow cytometry and confocal immunohistochemistry. Further, Western blots were used to quantify the amount of VCAM-1 molecules in cell extracts. All these data demonstrated a mean 75% increase in the expression of VCAM-1 on the surface of TMEV-infected cells. Three inflammatory cytokines, interleukin-1alpha (IL-1alpha), interferon gamma (IFNgumma), and specially tumor necrosis factor alpha (TNF-α), some of which are also induced by TMEV in astrocytes (IL-1alpha and TNF-alpha), were potent inducers of VCAM-1 expression. To demonstrate whether the VCAM-1 molecules were biologically active, mediating adhesion to other cells as the integrin alpha4-expressing CD4+ T lymphocytes, the authors used a cell adhesion test. It was also demonstrated by immunohistochemistry that in vivo VCAM-1 expression is enhanced after TMEV intracraneal infection. The present data show a small but statistically significant overexpression of VCAM-1 after astrocyte infection with TMEV that could play a significant role in vivo.


Asunto(s)
Astrocitos/metabolismo , Astrocitos/virología , Encéfalo/citología , Theilovirus/fisiología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/genética , Animales , Animales Recién Nacidos , Encéfalo/virología , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Inmunoquímica , Ratones , Ratones Endogámicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Virol ; 84(3): 1348-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19923182

RESUMEN

The DA strain and other members of the TO subgroup of Theiler's murine encephalomyelitis virus (TMEV) induce a persistent central nervous system infection associated with an inflammatory white matter demyelinating disease. TO subgroup strains synthesize an 18-kDa protein, L*, out of frame with the polyprotein from an initiation codon 13 nucleotides downstream from the polyprotein's AUG codon. We previously generated a mutant virus from our infectious DA full-length clone that has a change of the L* AUG codon to ACG (with no change in the polyprotein's amino acid sequence). Studies of this mutant virus showed that L* was key to the TO subgroup phenotype because the mutant had a decreased ability to persist and demyelinate. This work was initially called into question because a similar mutant derived from a different full-length DA infectious clone persisted and demyelinated similarly to wild-type DA virus (O. van Eyll and T. Michiels, J. Virol. 74:9071-9077, 2000). We now report that (i) the sequence of the L* coding region differs in the two infectious clones, resulting in a Ser or Leu as the predicted amino acid at position 93 of L* (with no change in the polyprotein's amino acid sequence), (ii) the difference in this amino acid is key to the phenotypic differences between the two mutants, and (iii) the change in amino acid 93 may affect L* phosphorylation. It is of interest that this amino acid only appears critical in determining the virus phenotype when L* is present in a significantly reduced amount (i.e., following translation from an ACG initiating codon).


Asunto(s)
Enfermedades Desmielinizantes/virología , Theilovirus/fisiología , Proteínas Virales/fisiología , Animales , Secuencia de Bases , Western Blotting , Línea Celular , Codón , Cricetinae , Cartilla de ADN , Ratones , Theilovirus/química , Proteínas Virales/química , Proteínas Virales/genética
20.
Viral Immunol ; 22(6): 371-87, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19951174

RESUMEN

SHP-1 is a protein tyrosine phosphatase that negatively regulates cytokine signaling and inflammatory gene expression. Mice genetically lacking SHP-1 (me/me) display severe inflammatory demyelinating disease following intracranial inoculation with the BeAn strain of Theiler's murine encephalomyelitis virus (TMEV) compared to infected wild-type mice. Furthermore, SHP-1-deficient mice show a profound and predominant infiltration of blood-derived macrophages into the CNS following intracerebral injection of TMEV, and these macrophages are concentrated in areas of demyelination in brain and spinal cord. In the present study we investigated the role of SHP-1 in controlling CNS inflammatory demyelination following a peripheral instead of an intracerebral inoculation of TMEV. Surprisingly, we found that while wild-type mice were entirely refractory to intraperitoneal (IP) infection by TMEV, in agreement with previous studies, all SHP-1-deficient mice displayed profound macrophage neuroinvasion and macrophage-mediated inflammatory demyelination. Moreover, SHP-1 deficiency led to increased expression of inflammatory molecules in macrophages, serum, and CNS following IP infection with TMEV. Importantly, pharmacological depletion of peripheral macrophages significantly decreased both paralysis and CNS viral loads in SHP-1-deficient mice. In addition, peripheral MCP-1 neutralization attenuated disease severity, decreased macrophage infiltration into the CNS, and decreased monocyte numbers in the blood of SHP-1-deficient mice, implicating MCP-1 as an important mediator of monocyte migration between multiple tissues. These results demonstrate that peripheral TMEV infection results in a unique evolution of macrophage-mediated demyelination in SHP-1-deficient mice, implicating SHP-1 in the control of neuroinvasion of inflammatory macrophages and neurotropic viruses into the CNS.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/etiología , Macrófagos/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/fisiología , Theilovirus/patogenicidad , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Quimiocina CCL2/fisiología , Quimiocinas/biosíntesis , Quimiocinas/genética , Quimiotaxis de Leucocito , Ácido Clodrónico/farmacología , Citocinas/biosíntesis , Citocinas/genética , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/virología , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Inyecciones Intraperitoneales , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Mutantes , Parálisis/etiología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/deficiencia , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Theilovirus/fisiología , Carga Viral , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA