Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
PLoS Pathog ; 15(2): e1007574, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742696

RESUMEN

TDP-43, an RNA-binding protein that is primarily nuclear and important in splicing and RNA metabolism, is mislocalized from the nucleus to the cytoplasm of neural cells in amyotrophic lateral sclerosis (ALS), and contributes to disease. We sought to investigate whether TDP-43 is mislocalized in infections with the acute neuronal GDVII strain and the persistent demyelinating DA strain of Theiler's virus murine encephalomyelitis virus (TMEV), a member of the Cardiovirus genus of Picornaviridae because: i) L protein of both strains is known to disrupt nucleocytoplasmic transport, including transport of polypyrimidine tract binding protein, an RNA-binding protein, ii) motor neurons and oligodendrocytes are targeted in both TMEV infection and ALS. TDP-43 phosphorylation, cleavage, and cytoplasmic mislocalization to an aggresome were observed in wild type TMEV-infected cultured cells, with predicted splicing abnormalities. In contrast, cells infected with DA and GDVII strains that have L deletion had rare TDP-43 mislocalization and no aggresome formation. TDP-43 mislocalization was also present in neural cells of TMEV acutely-infected mice. Of note, TDP-43 was mislocalized six weeks after DA infection to the cytoplasm of oligodendrocytes and other glial cells in demyelinating lesions of spinal white matter. A recent study showed that TDP-43 knock down in oligodendrocytes in mice led to demyelination and death of this neural cell [1], suggesting that TMEV infection mislocalization of TDP-43 and other RNA-binding proteins is predicted to disrupt key cellular processes and contribute to the pathogenesis of TMEV-induced diseases. Drugs that inhibit nuclear export may have a role in antiviral therapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteinopatías TDP-43/metabolismo , Theilovirus/metabolismo , Animales , Autopsia , Línea Celular , Núcleo Celular , Células Cultivadas , Citoplasma , Proteínas de Unión al ADN/fisiología , Humanos , Ratones , Transporte de Proteínas/fisiología , Proteinopatías TDP-43/fisiopatología , Theilovirus/patogenicidad
2.
PLoS Biol ; 16(6): e2006459, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29953453

RESUMEN

Mutation rates can evolve through genetic drift, indirect selection due to genetic hitchhiking, or direct selection on the physicochemical cost of high fidelity. However, for many systems, it has been difficult to disentangle the relative impact of these forces empirically. In RNA viruses, an observed correlation between mutation rate and virulence has led many to argue that their extremely high mutation rates are advantageous because they may allow for increased adaptability. This argument has profound implications because it suggests that pathogenesis in many viral infections depends on rare or de novo mutations. Here, we present data for an alternative model whereby RNA viruses evolve high mutation rates as a byproduct of selection for increased replicative speed. We find that a poliovirus antimutator, 3DG64S, has a significant replication defect and that wild-type (WT) and 3DG64S populations have similar adaptability in 2 distinct cellular environments. Experimental evolution of 3DG64S under selection for replicative speed led to reversion and compensation of the fidelity phenotype. Mice infected with 3DG64S exhibited delayed morbidity at doses well above the lethal level, consistent with attenuation by slower growth as opposed to reduced mutational supply. Furthermore, compensation of the 3DG64S growth defect restored virulence, while compensation of the fidelity phenotype did not. Our data are consistent with the kinetic proofreading model for biosynthetic reactions and suggest that speed is more important than accuracy. In contrast with what has been suggested for many RNA viruses, we find that within-host spread is associated with viral replicative speed and not standing genetic diversity.


Asunto(s)
Tasa de Mutación , Virus ARN/genética , Virus ARN/patogenicidad , Virulencia/genética , Células 3T3 , Sustitución de Aminoácidos , Animales , Evolución Molecular Dirigida , Femenino , Interacciones Microbiota-Huesped/genética , Cinética , Masculino , Ratones , Ratones Transgénicos , Modelos Genéticos , Mutagénesis Sitio-Dirigida , Polimorfismo de Nucleótido Simple , Virus ARN/fisiología , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Theilovirus/genética , Theilovirus/patogenicidad , Theilovirus/fisiología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
3.
Arch Virol ; 166(4): 1015-1033, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33582855

RESUMEN

Multiple sclerosis (MS) is a common inflammatory demyelinating disease of the central nervous system. Although the etiology of MS is unknown, genetics and environmental factors, such as infections, play a role. Viral infections of mice have been used as model systems to study this demyelinating disease of humans. Three viruses that have long been studied in this capacity are Theiler's murine encephalomyelitis virus, mouse hepatitis virus, and Semliki Forest virus. This review describes the viruses themselves, the infection process, the disease caused by infection and its accompanying pathology, and the model systems and their usefulness in studying MS.


Asunto(s)
Modelos Animales de Enfermedad , Esclerosis Múltiple/patología , Esclerosis Múltiple/virología , Infecciones por Virus ARN/patología , Infecciones por Virus ARN/virología , Animales , Sistema Nervioso Central/patología , Sistema Nervioso Central/fisiología , Sistema Nervioso Central/virología , Humanos , Ratones , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/fisiopatología , Virus de la Hepatitis Murina/patogenicidad , Virus de la Hepatitis Murina/fisiología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/fisiopatología , Virus de los Bosques Semliki/patogenicidad , Virus de los Bosques Semliki/fisiología , Theilovirus/patogenicidad , Theilovirus/fisiología
4.
Int J Mol Sci ; 22(10)2021 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-34067536

RESUMEN

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


Asunto(s)
Enfermedades Desmielinizantes/inmunología , Esclerosis Múltiple/inmunología , Theilovirus/fisiología , Inmunidad Adaptativa/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Astrocitos/metabolismo , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/metabolismo , Infecciones por Cardiovirus/virología , Citocinas , Enfermedades Desmielinizantes/patología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/inmunología , Ratones , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Oligodendroglía/metabolismo , Transducción de Señal/inmunología , Theilovirus/patogenicidad
5.
Brain Behav Immun ; 77: 110-126, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30582962

RESUMEN

Remyelination is an endogenous process by which functional recovery of damaged neurons is achieved by reinstating the myelin sheath around axons. Remyelination has been documented in multiple sclerosis (MS) lesions and experimental models, although it is often incomplete or fails to affect the integrity of the axon, thereby leading to progressive disability. Microglia play a crucial role in the clearance of the myelin debris produced by demyelination and in inflammation-dependent OPC activation, two processes necessary for remyelination to occur. We show here that following corpus callosum demyelination in the TMEV-IDD viral murine model of MS, there is spontaneous and partial remyelination that involves a temporal discordance between OPC mobilization and microglia activation. Pharmacological treatment with the endocannabinoid 2-AG enhances the clearance of myelin debris by microglia and OPC differentiation, resulting in complete remyelination and a thickening of the myelin sheath. These results highlight the importance of targeting microglia during the repair processes in order to enhance remyelination.


Asunto(s)
Ácidos Araquidónicos/farmacología , Endocannabinoides/farmacología , Glicéridos/farmacología , Microglía/efectos de los fármacos , Remielinización/efectos de los fármacos , Animales , Ácidos Araquidónicos/metabolismo , Axones/metabolismo , Diferenciación Celular/fisiología , Cuerpo Calloso/patología , Cuerpo Calloso/fisiología , Enfermedades Desmielinizantes/fisiopatología , Modelos Animales de Enfermedad , Endocannabinoides/metabolismo , Femenino , Glicéridos/metabolismo , Masculino , Ratones , Ratones Endogámicos , Microglía/metabolismo , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/fisiopatología , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/fisiología , Oligodendroglía/metabolismo , Theilovirus/patogenicidad
6.
Int J Mol Sci ; 20(13)2019 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-31262031

RESUMEN

(1) Background: Canine distemper virus (CDV)-induced demyelinating leukoencephalitis (CDV-DL) in dogs and Theiler's murine encephalomyelitis (TME) virus (TMEV)-induced demyelinating leukomyelitis (TMEV-DL) are virus-induced demyelinating conditions mimicking Multiple Sclerosis (MS). Reactive oxygen species (ROS) can induce the degradation of lipids and nucleic acids to characteristic metabolites such as oxidized lipids, malondialdehyde, and 8-hydroxyguanosine. The hypothesis of this study is that ROS are key effector molecules in the pathogenesis of myelin membrane breakdown in CDV-DL and TMEV-DL. (2) Methods: ROS metabolites and antioxidative enzymes were assessed using immunofluorescence in cerebellar lesions of naturally CDV-infected dogs and spinal cord tissue of TMEV-infected mice. The transcription of selected genes involved in ROS generation and detoxification was analyzed using gene-expression microarrays in CDV-DL and TMEV-DL. (3) Results: Immunofluorescence revealed increased amounts of oxidized lipids, malondialdehyde, and 8-hydroxyguanosine in CDV-DL while TMEV-infected mice did not reveal marked changes. In contrast, microarray-analysis showed an upregulated gene expression associated with ROS generation in both diseases. (4) Conclusion: In summary, the present study demonstrates a similar upregulation of gene-expression of ROS generation in CDV-DL and TMEV-DL. However, immunofluorescence revealed increased accumulation of ROS metabolites exclusively in CDV-DL. These results suggest differences in the pathogenesis of demyelination in these two animal models.


Asunto(s)
Moquillo/metabolismo , Encefalitis Viral/metabolismo , Vaina de Mielina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Catalasa/metabolismo , Moquillo/patología , Perros , Encefalitis Viral/patología , Encefalitis Viral/virología , Femenino , Masculino , Ratones , Vaina de Mielina/patología , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa/metabolismo , Theilovirus/patogenicidad
7.
Cytokine ; 102: 83-93, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28800924

RESUMEN

Macrophages are common targets for infection and innate immune activation by many pathogenic viruses including the neurotropic Theiler's Murine Encephalomyelitis Virus (TMEV). As both infection and innate activation of macrophages are key determinants of viral pathogenesis especially in the central nervous system (CNS), an analysis of macrophage growth factors on these events was performed. C3H mouse bone-marrow cells were differentiated in culture using either recombinant macrophage colony stimulating factor (M-CSF) or granulocyte-macrophage colony-stimulating factor (GM-CSF), inoculated with TMEV (BeAn) and analyzed at various times thereafter. Cytokine RNA and protein analysis, virus titers, and flow cytometry were performed to characterize virological parameters under these culture conditions. GM-CSF-differentiated macrophages showed higher levels of TMEV viral RNA and proinflammatory molecules compared to infected M-CSF-differentiated cells. Thus, GM-CSF increases both TMEV infection and TMEV-induced activation of macrophages compared to that seen with M-CSF. Moreover, while infectious viral particles decreased from a peak at 12h to undetectable levels at 48h post infection, TMEV viral RNA remained higher in GM-CSF- compared to M-CSF-differentiated macrophages in concert with increased proinflammatory gene expression. Analysis of a possible basis for these differences determined that glycolytic rates contributed to heightened virus replication and proinflammatory cytokine secretion in GM-CSF compared to M-CSF-differentiated macrophages. In conclusion, we provide evidence implicating a role for GM-CSF in promoting virus replication and proinflammatory cytokine expression in macrophages, indicating that GM-CSF may be a key factor for TMEV infection and the induction of chronic TMEV-induced immunopathogenesis in the CNS.


Asunto(s)
Infecciones por Cardiovirus/etiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Activación de Macrófagos , Factor Estimulante de Colonias de Macrófagos/inmunología , Theilovirus/patogenicidad , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Diferenciación Celular/inmunología , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Glucólisis , Macrófagos/inmunología , Macrófagos/patología , Macrófagos/virología , Ratones , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , Theilovirus/genética , Theilovirus/aislamiento & purificación , Replicación Viral/inmunología
8.
Neurobiol Dis ; 99: 121-132, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28017800

RESUMEN

Following intracerebral inoculation, the BeAn 8386 strain of Theiler's virus causes persistent infection and inflammatory demyelinating encephalomyelitis in the spinal cord of T-cell defective SJL/J mice, which is widely used as a model of multiple sclerosis. In contrast, C57BL/6 (B6) mice clear the virus and develop inflammation and lesions in the hippocampus, associated with acute and chronic seizures, representing a novel model of viral encephalitis-induced epilepsy. Here we characterize the geno- and phenotype of two naturally occurring variants of BeAn (BeAn-1 and BeAn-2) that can be used to further understand the viral and host factors involved in the neuropathogenesis in B6 and SJL/J mice. Next generation sequencing disclosed 15 single nucleotide differences between BeAn-1 and BeAn-2, of which 4 are coding changes and 3 are in the 5'-UTR (5'-untranslated region). The relatively minor variations in the nucleotide sequence of the two BeAn substrains led to marked differences in neurovirulence. In SJL/J mice, inflammatory demyelination in the spinal cord and its clinical consequences were significantly more marked following infection with BeAn-1 than with BeAn-2. Both BeAn substrains caused lymphocyte infiltration and increase of MAC3-positive cells in the hippocampus, but hippocampal damage and seizures were only observed in B6 mice. Seizures occurred in one third of BeAn-2 infected B6 mice, but not in BeAn-1 infected B6 mice. By comparing individual mice by receiver operating characteristic (ROC) curve analysis, the severity of hippocampal neurodegeneration and amount of MAC3-positive microglia/macrophages discriminated seizing from non-seizing B6 mice, whereas T-lymphocyte brain infiltration was not found to be a crucial factor. These data add novel evidence to the view that differential outcome of infection may be not invariably linked to a distinct viral burden but to a finely tuned balance between antiviral immune responses that although essential for host resistance can also contribute to immunopathology.


Asunto(s)
Encefalitis Viral/patología , Encefalomielitis Aguda Diseminada/patología , Epilepsia/patología , Esclerosis Múltiple/patología , Theilovirus , Animales , Encéfalo/inmunología , Encéfalo/patología , Encéfalo/virología , Modelos Animales de Enfermedad , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Encefalomielitis Aguda Diseminada/inmunología , Encefalomielitis Aguda Diseminada/virología , Epilepsia/inmunología , Epilepsia/virología , Femenino , Interacciones Huésped-Patógeno , Ratones Endogámicos C57BL , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/virología , Fenotipo , Polimorfismo de Nucleótido Simple , ARN Viral/metabolismo , Especificidad de la Especie , Theilovirus/genética , Theilovirus/patogenicidad , Virulencia
9.
Int Immunol ; 28(12): 575-584, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27803063

RESUMEN

Natalizumab, which is an antibody against α4 integrin, has been used for the treatment of multiple sclerosis. In the present study, we investigated both the role of α4 integrin and the therapeutic effect of HCA3551, a newly synthesized orally active small molecule α4 integrin antagonist, in the development of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). The mRNA levels of α4 integrins were significantly up-regulated in the central nervous system (CNS) of mice with TMEV-IDD as compared with naive mice (*P < 0.05). HCA3551 treatment in the effector phase significantly suppressed both the clinical and histological development of TMEV-IDD. The number of infiltrating mononuclear inflammatory cells in the CNS was significantly decreased in the mice treated with HCA3551 (**P < 0.01). The labeling indices for CD68 antigen and the absolute cell numbers of TNF-α-producing CD4+ T cells and IFN-γ-producing CD8+ T cells were significantly decreased in the CNS of mice treated with HCA3551 (*P < 0.05). HCA3551 treatment in the effector phase might inhibit the binding of α4 integrin to vascular cell adhesion molecule-1, thereby decreasing the number of mononuclear cells in the CNS.


Asunto(s)
Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/virología , Integrina alfa4/metabolismo , Esclerosis Múltiple/metabolismo , Theilovirus/patogenicidad , Animales , Modelos Animales de Enfermedad , Femenino , Integrina alfa4/genética , Integrina alfa4/inmunología , Ratones , Ratones Endogámicos , Esclerosis Múltiple/virología
10.
J Neurovirol ; 22(1): 66-79, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26260496

RESUMEN

Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) is an important model of the progressive disability caused by irreversible CNS tissue injury, and provides an example of how a CNS pathogen can cause inflammation, demyelination, and neuronal damage. We were interested in which molecules, especially inflammatory mediators, might be upregulated in the CNS throughout TMEV-IDD. We quantitated by a real-time RT-PCR multi-gene system the expression of a pathway-focused panel of genes at 30 and 165 days post infection, characterizing both the early inflammatory and the late neurodegenerative stages of TMEV-IDD. Also, we measured 32 cytokines/chemokines by multiplex Luminex analysis in CSF specimens from early and late TMEV-IDD as well as sham-treated mice. Results indicate that, in the later stage of TMEV-IDD, activation of the innate immune response is most prominent: TLRs, type I IFN response genes, and innate immunity-associated cytokines were highly expressed in late TMEV-IDD compared to sham (p ≤ 0.0001) and early TMEV-IDD (p < 0.05). Conversely, several molecular mediators of adaptive immune response were highly expressed in early TMEV-IDD (all p ≤ 0.001). Protein detection in the CSF was broadly concordant with mRNA abundance of the corresponding gene measured by real-time RT-PCR in the spinal cord, since several cytokines/chemokines were increased in the CSF of TMEV-IDD mice. Results show a clear shift from adaptive to innate immunity from early to late TMEV-IDD, indicating that adaptive and innate immune pathways are likely involved in the development and progression of the disease to different extents. CSF provides an optimal source of biomarkers of CNS neuroinflammation.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Cardiovirus/inmunología , Enfermedades Desmielinizantes/inmunología , Interacciones Huésped-Patógeno , Inmunidad Innata , Animales , Infecciones por Cardiovirus/líquido cefalorraquídeo , Infecciones por Cardiovirus/genética , Infecciones por Cardiovirus/virología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Citocinas/líquido cefalorraquídeo , Citocinas/genética , Citocinas/inmunología , Enfermedades Desmielinizantes/líquido cefalorraquídeo , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/virología , Progresión de la Enfermedad , Regulación de la Expresión Génica , Inflamación , Ratones , Anotación de Secuencia Molecular , ARN Mensajero/líquido cefalorraquídeo , ARN Mensajero/genética , ARN Mensajero/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Theilovirus/crecimiento & desarrollo , Theilovirus/inmunología , Theilovirus/patogenicidad , Factores de Tiempo , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología
11.
Epilepsia ; 57(12): 1958-1967, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27739576

RESUMEN

OBJECTIVE: Infection with Theiler's murine encephalomyelitis virus (TMEV) in C57Bl/6J mice induces acute seizures and development of spontaneous recurrent seizures and behavioral comorbidities weeks later. The present studies sought to determine whether acute therapeutic intervention with an anti-inflammatory-based approach could prevent or modify development of TMEV-induced long-term behavioral comorbidities. Valproic acid (VPA), in addition to its prototypical anticonvulsant properties, inhibits histone deacetylase (HDAC) activity, which may alter expression of the inflammasome. Minocycline (MIN) has previously demonstrated an antiseizure effect in the TMEV model via direct anti-inflammatory mechanisms, but the long-term effect of MIN treatment on the development of chronic behavioral comorbidities is unknown. METHODS: Mice infected with TMEV were acutely administered MIN (50 mg/kg, b.i.d. and q.d.) or VPA (100 mg/kg, q.d.) during the 7-day viral infection period. Animals were evaluated for acute seizure severity and subsequent development of chronic behavioral comorbidities and seizure threshold. RESULTS: Administration of VPA reduced the proportion of mice with seizures, delayed onset of symptomatic seizures, and reduced seizure burden during the acute infection. This was in contrast to the effects of administration of once-daily MIN, which did not affect the proportion of mice with seizures or delay onset of acute symptomatic seizures. However, VPA-treated mice were no different from vehicle (VEH)-treated mice in long-term behavioral outcomes, including open field activity and seizure threshold. Once-daily MIN treatment, despite no effect on the maximum observed Racine stage seizure severity, was associated with improved long-term behavioral outcomes and normalized seizure threshold. SIGNIFICANCE: Acute seizure control alone is insufficient to modify chronic disease comorbidities in the TMEV model. This work further supports the role of an inflammatory response in the development of chronic behavioral comorbidities and further highlights the utility of this platform for the development of mechanistically novel pharmacotherapies for epilepsy.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Conducta Animal/efectos de los fármacos , Epilepsia del Lóbulo Temporal , Minociclina/uso terapéutico , Theilovirus/patogenicidad , Ácido Valproico/uso terapéutico , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/etiología , Peso Corporal/efectos de los fármacos , Distribución de Chi-Cuadrado , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Epilepsia del Lóbulo Temporal/etiología , Epilepsia del Lóbulo Temporal/virología , Conducta Exploratoria/efectos de los fármacos , Ratones , Actividad Motora/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante
12.
Int Immunol ; 26(7): 369-81, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24486565

RESUMEN

Infection by Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) induces an immune-mediated demyelinating disease in susceptible mouse strains and serves as a relevant infection model for human multiple sclerosis. T-cell immunoglobulin and mucin domain-3 (TIM-3) has been demonstrated to play a crucial role in the maintenance of peripheral tolerance. In this study, we examined the regulatory role of the TIM-3 pathway in the development of TMEV-induced demyelinating disease (TMEV-IDD). The expression of TIM-3 was increased at both protein and mRNA levels in the spinal cords of mice with TMEV-IDD compared with naive controls. In addition, by utilizing a blocking mAb, we demonstrate that TIM-3 negatively regulates TMEV-specific ex vivo production of IFN-γ and IL-10 by CD4(+) T cells and IFN-γ by CD8(+) T cells from the CNS of mice with TMEV-IDD at 36 days post-infection (dpi). In vivo blockade of TIM-3 by using the anti-TIM-3 mAb resulted in significant exacerbation of the development of TMEV-IDD both clinically and histologically. The number of infiltrating mononuclear cells in the CNS was also increased in mice administered with anti-TIM-3 mAb both at the induction phase (10 dpi) and at the effector phase (36 dpi). Flow cytometric analysis of intracellular cytokines revealed that the number of CD4(+) T cells producing TNF, IL-4, IL-10 and IL-17 was significantly increased at the effector phase in the CNS of anti-TIM-3 mAb-treated mice. These results suggest that the TIM-3 pathway plays a critical role in the regulation of TMEV-IDD.


Asunto(s)
Poliomielitis/genética , ARN Mensajero/inmunología , Receptores Virales/inmunología , Médula Espinal/inmunología , Theilovirus/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucina-10/biosíntesis , Interleucina-10/metabolismo , Interleucina-17/biosíntesis , Interleucina-17/metabolismo , Interleucina-4/biosíntesis , Interleucina-4/metabolismo , Ratones , Esclerosis Múltiple , Tolerancia Periférica , Poliomielitis/inmunología , Poliomielitis/patología , Poliomielitis/virología , ARN Mensajero/genética , Receptores Virales/genética , Transducción de Señal , Médula Espinal/patología , Theilovirus/patogenicidad
13.
J Virol ; 87(3): 1849-60, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23236075

RESUMEN

Viral infections of the central nervous system (CNS) can trigger an antiviral immune response, which initiates an inflammatory cascade to control viral replication and dissemination. The extent of the proinflammatory response in the CNS and the timing of the release of proinflammatory cytokines can lead to neuronal excitability. Tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), two proinflammatory cytokines, have been linked to the development of acute seizures in Theiler's murine encephalomyelitis virus-induced encephalitis. It is unclear the extent to which the infiltrating macrophages versus resident CNS cells, such as microglia, contribute to acute seizures, as both cell types produce TNF-α and IL-6. In this study, we show that following infection a significantly higher number of microglia produced TNF-α than did infiltrating macrophages. In contrast, infiltrating macrophages produced significantly more IL-6. Mice treated with minocycline or wogonin, both of which limit infiltration of immune cells into the CNS and their activation, had significantly fewer macrophages infiltrating the brain, and significantly fewer mice had seizures. Therefore, our studies implicate infiltrating macrophages as an important source of IL-6 that contributes to the development of acute seizures.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/patología , Interleucina-6/metabolismo , Macrófagos/inmunología , Macrófagos/virología , Convulsiones , Theilovirus/patogenicidad , Animales , Infecciones por Cardiovirus/inmunología , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Theilovirus/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
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.
Exp Neurol ; 379: 114851, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38876197

RESUMEN

Multiple sclerosis (MS) is a chronic disabling disease of the central nervous system affecting over 2.5 million people worldwide. Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) is a murine model that reproduces the progressive form of MS and serves as a reference model for studying virus-induced demyelination. Certain mouse strains such as SJL are highly susceptible to this virus and serve as a prototype strain for studying TMEV infection. Other strains such as SWR are also susceptible, but their disease course following TMEV infection differs from SJL's. The quantification of motor and behavioral deficits following the induction of TMEV-IDD could help identify the differences between the two strains. Motor deficits have commonly been measured with the rotarod apparatus, but a multicomponent assessment tool has so far been lacking. For that purpose, we present a novel way of quantifying locomotor deficits, gait alterations and behavioral changes in this well-established mouse model of multiple sclerosis by employing automated video analysis technology (The PhenoTyper, Noldus Information Technology). We followed 12 SJL and 12 SWR female mice and their mock-infected counterparts over a period of 9 months following TMEV-IDD induction. We demonstrated that SJL and SWR mice both suffer significant gait alterations and reduced exploration following TMEV infection. However, SJL mice also display an earlier and more severe decline in spontaneous locomotion, especially in velocity, as well as in overall activity. Maintenance behaviors such as eating and grooming are not affected in either of the two strains. The system also showed differences in mock-infected mice from both strains, highlighting an age-related decline in spontaneous locomotion in the SJL strain, as opposed to hyperactivity in the SWR strain. Our study confirms that this automated video tracking system can reliably track the progression of TMEV-IDD for 9 months. We have also shown how this system can be utilized for longitudinal phenotyping in mice by describing useful parameters that quantify locomotion, gait and behavior.


Asunto(s)
Modelos Animales de Enfermedad , Esclerosis Múltiple , Fenotipo , Theilovirus , Animales , Ratones , Theilovirus/patogenicidad , Femenino , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Ratones Endogámicos , Infecciones por Cardiovirus , Grabación en Video/métodos , Estudios Longitudinales , Especificidad de la Especie , Actividad Motora/fisiología
16.
J Neuroinflammation ; 9: 60, 2012 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-22452799

RESUMEN

BACKGROUND: The extent to which susceptibility to brain hemorrhage is derived from blood-derived factors or stromal tissue remains largely unknown. We have developed an inducible model of CD8 T cell-initiated blood-brain barrier (BBB) disruption using a variation of the Theiler's murine encephalomyelitis virus (TMEV) model of multiple sclerosis. This peptide-induced fatal syndrome (PIFS) model results in severe central nervous system (CNS) vascular permeability and death in the C57BL/6 mouse strain, but not in the 129 SvIm mouse strain, despite the two strains' having indistinguishable CD8 T-cell responses. Therefore, we hypothesize that hematopoietic factors contribute to susceptibility to brain hemorrhage, CNS vascular permeability and death following induction of PIFS. METHODS: PIFS was induced by intravenous injection of VP2121-130 peptide at 7 days post-TMEV infection. We then investigated brain inflammation, astrocyte activation, vascular permeability, functional deficit and microhemorrhage formation using T2*-weighted magnetic resonance imaging (MRI) in C57BL/6 and 129 SvIm mice. To investigate the contribution of hematopoietic cells in this model, hemorrhage-resistant 129 SvIm mice were reconstituted with C57BL/6 or autologous 129 SvIm bone marrow. Gadolinium-enhanced, T1-weighted MRI was used to visualize the extent of CNS vascular permeability after bone marrow transfer. RESULTS: C57BL/6 and 129 SvIm mice had similar inflammation in the CNS during acute infection. After administration of VP2121-130 peptide, however, C57BL/6 mice had increased astrocyte activation, CNS vascular permeability, microhemorrhage formation and functional deficits compared to 129 SvIm mice. The 129 SvIm mice reconstituted with C57BL/6 but not autologous bone marrow had increased microhemorrhage formation as measured by T2*-weighted MRI, exhibited a profound increase in CNS vascular permeability as measured by three-dimensional volumetric analysis of gadolinium-enhanced, T1-weighted MRI, and became moribund in this model system. CONCLUSION: C57BL/6 mice are highly susceptible to microhemorrhage formation, severe CNS vascular permeability and morbidity compared to the 129 SvIm mouse. This susceptibility is transferable with the bone marrow compartment, demonstrating that hematopoietic factors are responsible for the onset of brain microhemorrhage and vascular permeability in immune-mediated fatal BBB disruption.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Infecciones por Cardiovirus/complicaciones , Hemorragias Intracraneales/etiología , Animales , Astrocitos/efectos de los fármacos , Barrera Hematoencefálica , Trasplante de Médula Ósea/métodos , Linfocitos T CD8-positivos/efectos de los fármacos , Permeabilidad Capilar/efectos de los fármacos , Proteínas de la Cápside/efectos adversos , Modelos Animales de Enfermedad , Citometría de Flujo , Fluoresceína-5-Isotiocianato/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Hematínicos , Hemorragias Intracraneales/cirugía , Hemorragias Intracraneales/virología , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos , Actividad Motora/fisiología , Prueba de Desempeño de Rotación con Aceleración Constante , Theilovirus/patogenicidad , Proteínas Virales/efectos adversos
17.
J Virol ; 85(18): 9614-22, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752908

RESUMEN

Stress granules (SG) are cytoplasmic aggregates of stalled translation preinitiation complexes that form in cells exposed to various environmental stresses. Here, we show that stress granules assemble in cells infected with Theiler's murine encephalomyelitis virus (TMEV) mutants carrying alterations in the leader (L) protein, but not in cells infected with wild-type TMEV. Stress granules also formed in STAT1-deficient cells, suggesting that SG formation was not a consequence of increased type I interferon (IFN) production when cells were infected with the mutant virus. Ectopic expression of the wild-type L protein was sufficient to inhibit stress granule formation induced by sodium arsenite or thapsigargin treatment. In conclusion, TMEV infection induces stress granule assembly, but this process is inhibited by the L protein. Unlike poliovirus-induced stress granules, TMEV-induced stress granules did not contain the nuclear protein Sam68 but contained polypyrimidine tract binding protein (PTB), an internal ribosome entry site (IRES)-interacting protein. Moreover, G3BP was not degraded and was found in SG after TMEV infection, suggesting that SG content could be virus specific. Despite the colocalization of PTB with SG and the known interaction of PTB with viral RNA, in situ hybridization and immunofluorescence assays failed to detect viral RNA trapped in infection-induced SG. Recombinant Theiler's viruses expressing the L protein of Saffold virus 2 (SAFV-2), a closely related human theilovirus, or the L protein of mengovirus, an encephalomyocarditis virus (EMCV) strain, also inhibited infection-induced stress granule assembly, suggesting that stress granule antagonism is a common feature of cardiovirus L proteins.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Theilovirus/patogenicidad , Proteínas de la Matriz Viral/metabolismo , Animales , Línea Celular , Humanos , Ratones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas de la Matriz Viral/genética
18.
J Virol ; 85(14): 6893-905, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21543488

RESUMEN

Theiler's murine encephalitis viruses (TMEV) are divided into two subgroups based on their neurovirulence. Persistent strains resemble Theiler's original viruses (referred to as the TO subgroup), which largely induce a subclinical polioencephalomyelitis during the acute phase of the disease and can persist in the spinal cord of susceptible animals, inducing a chronic demyelinating disease. In contrast, members of the neurovirulent subgroup cause an acute encephalitis characterized by the rapid onset of paralysis and death within days following intracranial inoculation. We report herein the characterization of a novel neurovirulent strain of TMEV, identified using pyrosequencing technology and referred to as NIHE. Complete coverage of the NIHE viral genome was obtained, and it shares <90% nucleotide sequence identity to known TMEV strains irrespective of subgroup, with the greatest sequence variability being observed in genes encoding the leader and capsid proteins. The histopathological analysis of infected brain and spinal cord demonstrate inflammatory lesions and neuronal necrosis during acute infection with no evidence of viral persistence or chronic disease. Intriguingly, genetic analysis indicates the putative expression of the L protein, considered a hallmark of strains within the persistent subgroup. Thus, the identification and characterization of a novel neurovirulent TMEV strain sharing features previously associated with both subgroups will lead to a deeper understanding of the evolution of TMEV strains and new insights into the determinants of neurovirulence.


Asunto(s)
Theilovirus/aislamiento & purificación , Secuencia de Aminoácidos , Animales , Encéfalo/patología , Encéfalo/virología , Cápside/química , Genoma Viral , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Filogenia , Homología de Secuencia de Aminoácido , Médula Espinal/patología , Médula Espinal/virología , Theilovirus/clasificación , Theilovirus/patogenicidad , Tropismo Viral
19.
J Virol ; 85(16): 8149-57, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21680509

RESUMEN

Infection of C57BL/6 mice by the intracerebral route with the Daniels (DA) strain of Theiler's murine encephalomyelitis virus (TMEV) resulted in acute behavioral seizures in approximately 50% of the mice. By titration, the viral dose correlated with the percentage of mice developing seizures; however, neuropathological changes were similar over the dose range, and viral clearance from the brains occurred uniformly by day 14 postinfection (p.i.). Other TMEV strains and mutants (GDVII, WW, BeAn 8386 [BeAn], DApBL2M, H101) induced seizures in C57BL/6 mice to various degrees. The BeAn strain and DApBL2M mutant were similar to the DA strain in the percentages of mice developing seizures and neuropathological changes and in the extent of infected cells. The GDVII and WW strains caused 100% mortality by days 5 and 6 p.i., respectively, at which time neuropathological changes and neuronal infection were extensive. The H101 mutant induced seizures and caused 100% mortality by day 7 p.i.; however, only minor neuropathological changes and few infected cells were observed. Thus, in H101 mutant infections, it appears that elevated levels of cytokines, rather than neuronal cell death, play the dominant role in seizure induction.


Asunto(s)
Infecciones por Cardiovirus/virología , Encefalitis Viral/virología , Convulsiones/virología , Theilovirus/genética , Theilovirus/patogenicidad , Animales , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/patología , Citocinas/biosíntesis , Citocinas/sangre , Encefalitis , Encefalitis Viral/inmunología , Encefalitis Viral/patología , Ratones , Ratones Endogámicos C57BL , Theilovirus/clasificación
20.
J Virol ; 85(18): 9377-84, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752920

RESUMEN

The DA strain and other members of the TO subgroup of Theiler's murine encephalomyelitis virus (TMEV) induce an early transient subclinical neuronal disease followed by a chronic progressive inflammatory demyelination, with persistence of the virus in the central nervous system (CNS) for the life of the mouse. Although TMEV-induced demyelinating disease (TMEV-IDD) is thought to be immune mediated, there is also evidence that supports a role for the virus in directly inducing demyelination. In order to clarify the function of DA virus genes, we generated a transgenic mouse that had tamoxifen-inducible expression of the DA L-coding region in oligodendrocytes (and Schwann cells), a cell type in which the virus is known to persist. Tamoxifen-treated young transgenic mice usually developed an acute progressive fatal paralysis, with abnormalities of the oligodendrocytes and Schwann cells and demyelination, but without significant lymphocytic infiltration; later treatment led to transient weakness with demyelination and persistent expression of the recombined transgene. These findings demonstrate that a high level of expression of DA L can cause the death of myelin-synthesizing cells and death of the mouse, while a lower level of L expression (which can persist) can lead to cellular dysfunction with survival. The results suggest that expression of DA L plays an important role in the pathogenesis of TMEV-IDD. Virus-induced infection and death of oligodendrocytes may play a part in the demyelination of other diseases in which an immune-mediated mechanism has been stressed, including multiple sclerosis.


Asunto(s)
Muerte Celular , Neuronas/patología , Neuronas/virología , ARN Viral/genética , Theilovirus/patogenicidad , Proteínas Virales/metabolismo , Factores de Virulencia/metabolismo , Animales , Enfermedades Desmielinizantes , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Oligodendroglía/patología , Oligodendroglía/virología , Poliomielitis/patología , Poliomielitis/virología , Enfermedades de los Roedores/patología , Enfermedades de los Roedores/virología , Células de Schwann/patología , Células de Schwann/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA