Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 171
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 529: 162-171, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37598833

RESUMEN

In this study, we examined infection with the highly neurovirulent GDVII, the less neurovirulent DA strains, and with a mutant DA, which lacks the L* protein (L*-1) involved in viral persistence and demyelinating disease, to analyze the direct effects of Theiler's murine encephalomyelitis virus (TMEV) replication using primary cultures of mouse brain hippocampal neurons. All viruses replicate in cultured neurons, with GDVII having the highest titers and L*-1 the lowest. Accordingly, all were positive for viral antigen staining 3 days postinfection (dpi), and DA and L*-1 were also positive after 12 dpi. NeuN + immunostaining showed an early and almost complete absence of positive cells in cultures infected with GDVII, an approximately 50% reduction in cultures infected with DA, and fewer changes in L*-1 strains at 3 dpi. Accordingly, staining with chloromethyltetramethylrosamine orange (Mitotracker OrangeTM) as a parameter for cell viability showed similar results. Moreover, at 1 dpi, the strain DA induced higher transcript levels of neuroprotective genes such as IFN-Iß, IRF7, and IRF8. At 3 dpi, strains GDVII and DA, but not the L*-1 mutant, showed lower PKR expression. In addition, confocal analysis showed that L*-1-infected neurons exhibited a decrease in spine density. Treatment with poly (I:C), which is structurally related to dsRNA and is known to trigger IFN type I synthesis, reduced spine density even more. These results confirmed the use of mouse hippocampal neuron cultures as a model to study neuronal responses after TMEV infection, particularly in the formation of spine density.


Asunto(s)
Theilovirus , Ratones , Animales , Theilovirus/fisiología , Neuronas , Columna Vertebral
2.
Sci Rep ; 13(1): 10960, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37414913

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) induces an acute polioencephalomyelitis and a chronic demyelinating leukomyelitis in SJL mice. C57BL/6 (B6) mice generally do not develop TMEV-induced demyelinating disease (TMEV-IDD) due to virus elimination. However, TMEV can persist in specific immunodeficient B6 mice such as IFNß-/- mice and induce a demyelinating process. The proinflammatory cytokines IL-1ß and IL-18 are activated by the inflammasome pathway, which consists of a pattern recognition receptor molecule sensing microbial pathogens, the adaptor molecule Apoptosis-associated speck-like protein containing a CARD (ASC), and the executioner caspase-1. To analyze the contribution of the inflammasome pathway to the resistance of B6 mice to TMEV-IDD, ASC- and caspase-1-deficient mice and wild type littermates were infected with TMEV and investigated using histology, immunohistochemistry, RT-qPCR, and Western Blot. Despite the antiviral activity of the inflammasome pathway, ASC- and caspase-1-deficient mice eliminated the virus and did not develop TMEV-IDD. Moreover, a similar IFNß and cytokine gene expression was found in the brain of immunodeficient mice and their wild type littermates. Most importantly, Western Blot showed cleavage of IL-1ß and IL-18 in all investigated mice. Consequently, inflammasome-dependent activation of IL-1ß and IL-18 does not play a major role in the resistance of B6 mice to TMEV-IDD.


Asunto(s)
Enfermedades Desmielinizantes , Theilovirus , Animales , Ratones , Caspasa 1/genética , Citocinas , Enfermedades Desmielinizantes/patología , Inflamasomas , Interleucina-18/genética , Ratones Endogámicos C57BL , Ratones Endogámicos , Theilovirus/fisiología
3.
J Vis Exp ; (184)2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35816000

RESUMEN

One of the main causes of epilepsy is an infection of the central nervous system (CNS); approximately 8% of patients who survive such an infection develop epilepsy as a consequence, with rates being significantly higher in less economically developed countries. This work provides an overview of modeling epilepsy of infectious etiology and using it as a platform for novel antiseizure compound testing. A protocol of epilepsy induction by non-stereotactic intracerebral injection of Theiler's murine encephalomyelitis virus (TMEV) in C57BL/6 mice is presented, which replicates many of the early and chronic clinical symptoms of viral encephalitis and subsequent epilepsy in human patients. The clinical evaluation of mice during encephalitis to monitor seizure activity and detect the potential antiseizure effects of novel compounds is described. Furthermore, histopathological consequences of viral encephalitis and seizures such as hippocampal damage and neuroinflammation are shown, as well as long-term consequences such as spontaneous epileptic seizures. The TMEV model is one of the first translational, infection-driven, experimental platforms to allow for the investigation of the mechanisms of epilepsy development as a consequence of CNS infection. Thus, it also serves to identify potential therapeutic targets and compounds for patients at risk of developing epilepsy following a CNS infection.


Asunto(s)
Encefalitis Viral , Epilepsia , Theilovirus , Animales , Modelos Animales de Enfermedad , Epilepsia/etiología , Humanos , Ratones , Ratones Endogámicos C57BL , Convulsiones/diagnóstico , Theilovirus/fisiología
4.
PLoS One ; 17(6): e0270239, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35714111

RESUMEN

Astrocytes produce extracellular matrix (ECM) glycoproteins contributing to the blood-brain barrier and regulating the immune response in the central nervous system (CNS). The aim of this study was to investigate the impact of astrocyte depletion upon the clinical outcome and the composition of ECM glycoproteins in a virus-induced animal model of demyelination. Glial fibrillary acidic protein (GFAP)-thymidine-kinase transgenic SJL (GFAP-knockout) and wildtype mice were infected with Theiler's murine encephalomyelitis virus (TMEV). Astrocyte depletion was induced during the progressive, demyelinating disease phase by ganciclovir administration once daily between 56 and 77 days post infection (dpi). At 77 dpi GFAP-knockout mice showed a significant deterioration of clinical signs associated with a reduction of azan and picrosirius red stained ECM-molecules in the thoracic spinal cord. Basement-membrane-associated ECM-molecules including laminin, entactin/nidogen-1 and Kir4.1 as well as non-basement membrane-associated ECM-molecules like collagen I, decorin, tenascin-R and CD44 were significantly reduced in the spinal cord of GFAP-knockout mice. The reduction of the investigated ECM-molecules demonstrates that astrocytes play a key role in the production of ECM-molecules. The present findings indicate that the detected loss of Kir4.1 and CD44 as well as the disruption of the integrity of perineuronal nets led to the deterioration of clinical signs in GFAP-knockout mice.


Asunto(s)
Encefalomielitis , Theilovirus , Animales , Astrocitos , Matriz Extracelular , Glicoproteínas , Ratones , Ratones Noqueados , Theilovirus/fisiología
5.
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
6.
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
7.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34445189

RESUMEN

Tamoxifen is frequently used in murine knockout systems with CreER/LoxP. Besides possible neuroprotective effects, tamoxifen is described as having a negative impact on adult neurogenesis. The present study investigated the effect of a high-dose tamoxifen application on Theiler's murine encephalomyelitis virus (TMEV)-induced hippocampal damage. Two weeks after TMEV infection, 42% of the untreated TMEV-infected mice were affected by marked inflammation with neuronal loss, whereas 58% exhibited minor inflammation without neuronal loss. Irrespective of the presence of neuronal loss, untreated mice lacked TMEV antigen expression within the hippocampus at 14 days post-infection (dpi). Interestingly, tamoxifen application 0, 2 and 4, or 5, 7 and 9 dpi decelerated virus elimination and markedly increased neuronal loss to 94%, associated with increased reactive astrogliosis at 14 dpi. T cell infiltration, microgliosis and expression of water channels were similar within the inflammatory lesions, regardless of tamoxifen application. Applied at 0, 2 and 4 dpi, tamoxifen had a negative impact on the number of doublecortin (DCX)-positive cells within the dentate gyrus (DG) at 14 dpi, without a long-lasting effect on neuronal loss at 147 dpi. Thus, tamoxifen application during a TMEV infection is associated with transiently increased neuronal loss in the hippocampus, increased reactive astrogliosis and decreased neurogenesis in the DG.


Asunto(s)
Antagonistas de Estrógenos/efectos adversos , Hipocampo/efectos de los fármacos , Neuronas/efectos de los fármacos , Tamoxifeno/efectos adversos , Animales , Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/patología , Infecciones por Cardiovirus/veterinaria , Muerte Celular/efectos de los fármacos , Proteína Doblecortina , Hipocampo/patología , Ratones Endogámicos C57BL , Neuronas/patología , Theilovirus/fisiología
8.
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
9.
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
10.
Int J Mol Sci ; 21(20)2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33086489

RESUMEN

The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) induces a T cell-mediated demyelinating disease. This system has been studied as a relevant infection model for multiple sclerosis (MS). Therefore, defining the type of T cell responses and their functions is critically important for understanding the relevant pathogenic mechanisms. In this study, we adoptively transferred naive VP2-specific TCR-Tg CD4+ T cells into syngeneic susceptible SJL mice and monitored the development of the disease and the activation and proliferation of CD4+ T cells during the early stages of viral infection. The preexisting VP2-specific naive CD4+ T cells promoted the pathogenesis of the disease in a dose-dependent manner. The transferred VP2-specific CD4+ T cells proliferated rapidly in the CNS starting at 2-3 dpi. High levels of FoxP3+CD4+ T cells were found in the CNS early in viral infection (3 dpi) and persisted throughout the infection. Activated VP2-specific FoxP3+CD4+ T cells inhibited the production of IFN-γ, but not IL-17, via the same VP2-specific CD4+ T cells without interfering in proliferation. Thus, the early presence of regulatory T cells in the CNS with viral infection may favor the induction of pathogenic Th17 cells over protective Th1 cells in susceptible mice, thereby establishing the pathogenesis of virus-induced demyelinating disease.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Infecciones por Cardiovirus/inmunología , Infecciones por Cardiovirus/virología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Theilovirus/fisiología , Traslado Adoptivo , Animales , Proliferación Celular , Sistema Nervioso Central/patología , Citocinas/biosíntesis , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/virología , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Interleucina-17/biosíntesis , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/metabolismo , Especificidad de la Especie
11.
J Vis Exp ; (159)2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32510499

RESUMEN

The central nervous system (CNS) is comprised of the brain and spinal cord and is enveloped by the meninges, membranous layers serving as a barrier between the periphery and the CNS. The CNS is an immunologically specialized site, and in steady state conditions, immune privilege is most evident in the CNS parenchyma. In contrast, the meninges harbor a diverse array of resident cells, including innate and adaptive immune cells. During inflammatory conditions triggered by CNS injury, autoimmunity, infection, or even neurodegeneration, peripherally derived immune cells may enter the parenchyma and take up residence within the meninges. These cells are thought to perform both beneficial and detrimental actions during CNS disease pathogenesis. Despite this knowledge, the meninges are often overlooked when analyzing the CNS compartment, because conventional CNS tissue extraction methods omit the meningeal layers. This protocol presents two distinct methods for the rapid isolation of murine CNS tissues (i.e., brain, spinal cord, and meninges) that are suitable for downstream analysis via single-cell techniques, immunohistochemistry, and in situ hybridization methods. The described methods provide a comprehensive analysis of CNS tissues, ideal for assessing the phenotype, function, and localization of cells occupying the CNS compartment under homeostatic conditions and during disease pathogenesis.


Asunto(s)
Sistema Nervioso Central/citología , Sistema Nervioso Central/inmunología , Meninges/citología , Meninges/inmunología , Animales , Encéfalo/citología , Encéfalo/inmunología , Agregación Celular , Criopreservación , Enfermedades Desmielinizantes/patología , Enfermedades Desmielinizantes/virología , Femenino , Antígenos Comunes de Leucocito/metabolismo , Ratones , Adhesión en Parafina , Médula Espinal/citología , Médula Espinal/inmunología , Theilovirus/fisiología , Fijación del Tejido
12.
Sci Rep ; 10(1): 7918, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404926

RESUMEN

Antecedent viral infection may contribute to increased susceptibility to several neurological diseases, such as multiple sclerosis and Parkinson's disease. Variation in clinical presentations of these diseases is often associated with gender, genetic background, or a combination of these and other factors. The complicated etiologies of these virally influenced diseases are difficult to study in conventional laboratory mouse models, which display a very limited number of phenotypes. We have used the genetically and phenotypically diverse Collaborative Cross mouse panel to examine complex neurological phenotypes after viral infection. Female and male mice from 18 CC strains were evaluated using a multifaceted phenotyping pipeline to define their unique disease profiles following infection with Theiler's Murine Encephalomyelitis Virus, a neurotropic virus. We identified 4 distinct disease progression profiles based on limb-specific paresis and paralysis, tremors and seizures, and other clinical signs, along with separate gait profiles. We found that mice of the same strain had more similar profiles compared to those of different strains, and also identified strains and phenotypic parameters in which sex played a significant role in profile differences. These results demonstrate the value of using CC mice for studying complex disease subtypes influenced by sex and genetic background. Our findings will be useful for developing novel mouse models of virally induced neurological diseases with heterogenous presentation, an important step for designing personalized, precise treatments.


Asunto(s)
Cruzamientos Genéticos , Estudios de Asociación Genética , Antígenos H-2/genética , Fenotipo , Animales , Análisis por Conglomerados , Encefalitis/etiología , Femenino , Marcha , Masculino , Ratones , Poliomielitis/etiología , Convulsiones/etiología , Factores Sexuales , Especificidad de la Especie , Theilovirus/fisiología , Carga Viral
13.
Front Immunol ; 10: 1374, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31258540

RESUMEN

Gut microbiota dysbiosis has been implicated in MS and other immune diseases, although it remains unclear how manipulating the gut microbiota may affect the disease course. Using a well-established model of progressive MS triggered by intracranial infection with Theiler's murine encephalomyelitis virus (TMEV), we sought to determine whether dysbiosis induced by oral antibiotics (ABX) administered on pre-symptomatic and symptomatic phases of the disease influences its course. We also addressed the effects of microbiota recolonization after ABX withdrawn in the presence or absence of probiotics. Central and peripheral immunity, plasma acetate and butyrate levels, axon damage and motor disability were evaluated. The cocktail of ABX prevented motor dysfunction and limited axon damage in mice, which had fewer CD4+ and CD8+ T cells in the CNS, while gut microbiota recolonization worsened motor function and axonal integrity. The underlying mechanisms of ABX protective effects seem to involve CD4+CD39+ T cells and CD5+CD1d+ B cells into the CNS. In addition, microglia adopted a round amoeboid morphology associated to an anti-inflammatory gene profile in the spinal cord of TMEV mice administered ABX. The immune changes in the spleen and mesenteric lymph nodes were modest, yet ABX treatment of mice limited IL-17 production ex vivo. Collectively, our results provide evidence of the functional relevance of gut microbiota manipulation on the neurodegenerative state and disease severity in a model of progressive MS and reinforce the role of gut microbiota as target for MS treatment.


Asunto(s)
Antibacterianos/uso terapéutico , Axones/patología , Linfocitos B/inmunología , Infecciones por Cardiovirus/inmunología , Microbioma Gastrointestinal/inmunología , Trastornos Motores/inmunología , Esclerosis Múltiple/inmunología , Probióticos/uso terapéutico , Linfocitos T/inmunología , Theilovirus/fisiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunidad , Activación de Linfocitos , Ratones , Trastornos Motores/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico
14.
J Virol ; 93(19)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31292248

RESUMEN

Leader (L) proteins encoded by cardioviruses are multifunctional proteins that contribute to innate immunity evasion. L proteins of Theiler's murine encephalomyelitis virus (TMEV), Saffold virus (SAFV), and encephalomyocarditis virus (EMCV) were reported to inhibit stress granule assembly in infected cells. Here, we show that TMEV L can act at two levels in the stress granule formation pathway: on the one hand, it can inhibit sodium arsenite-induced stress granule assembly without preventing eIF2α phosphorylation and, thus, acts downstream of eIF2α; on the other hand, it can inhibit eucaryotic translation initiation factor 2 alpha kinase 2 (PKR) activation and the consequent PKR-mediated eIF2α phosphorylation. Interestingly, coimmunostaining experiments revealed that PKR colocalizes with viral double-stranded RNA (dsRNA) in cells infected with L-mutant viruses but not in cells infected with the wild-type virus. Furthermore, PKR coprecipitated with dsRNA from cells infected with L-mutant viruses significantly more than from cells infected with the wild-type virus. These data strongly suggest that L blocks PKR activation by preventing the interaction between PKR and viral dsRNA. In infected cells, L also rendered PKR refractory to subsequent activation by poly(I·C). However, no interaction was observed between L and either dsRNA or PKR. Taken together, our results suggest that, unlike other viral proteins, L indirectly acts on PKR to negatively regulate its responsiveness to dsRNA.IMPORTANCE The leader (L) protein encoded by cardioviruses is a very short multifunctional protein that contributes to evasion of the host innate immune response. This protein notably prevents the formation of stress granules in infected cells. Using Theiler's virus as a model, we show that L proteins can act at two levels in the stress response pathway leading to stress granule formation, the most striking one being the inhibition of eucaryotic translation initiation factor 2 alpha kinase 2 (PKR) activation. Interestingly, the leader protein appears to inhibit PKR via a novel mechanism by rendering this kinase unable to detect double-stranded RNA, its typical activator. Unlike other viral proteins, such as influenza virus NS1, the leader protein appears to interact with neither PKR nor double-stranded RNA, suggesting that it acts indirectly to trigger the inhibition of the kinase.


Asunto(s)
Activación Enzimática , Interacciones Huésped-Patógeno , Evasión Inmune , Theilovirus/fisiología , Proteínas Virales/metabolismo , eIF-2 Quinasa/antagonistas & inhibidores , Animales , Línea Celular , Humanos , Unión Proteica , ARN Bicatenario/metabolismo , ARN Viral/metabolismo
15.
Sci Rep ; 9(1): 3444, 2019 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-30837498

RESUMEN

Epilepsy is a complex neurological disease characterized by recurrent seizures. Patients with viral encephalitis have a 16-fold increased risk of developing epilepsy, and this risk can persist for about 15 years after the occurrence of initial viral infection. Theiler's murine encephalomyelitis virus (TMEV) infection induces a well-characterized experimental model of epilepsy in C57BL/6 mice. In response to intracerebral (I.C.) injection of Daniel's (DA) strain of TMEV, there is vigorous immune response, which is detrimental to neurons and contributes to acute seizures, rendering mice susceptible to epilepsy. A comparative in vivo challenge study with either one of the two variants of the DA strain, small (DA-DS) or large (DA-CL) plaque forming variants, revealed differences in the diseases they induced in C57BL/6 mice. Compared to DA-CL-, DA-DS-infected mice exhibited significantly more seizures, higher clinical scores, neuroinflammation, and neuronal damage (mainly in the CA1-CA2 regions of hippocampus). Moreover, the brains of DA-DS infected mice contained approximately five-fold higher virus than those of DA-CL infected mice. A sequence comparison of the DA-CL and DA-DS genome sequences showed mutations in the leader (L) and L* proteins of DA-CL variant, which may be the cause of attenuating phenotype of DA-CL variant in the C57BL/6 mouse model of epilepsy.


Asunto(s)
Infecciones por Cardiovirus/complicaciones , Infecciones por Cardiovirus/virología , Epilepsia/etiología , Epilepsia/patología , Theilovirus/fisiología , Animales , Antígenos Virales/inmunología , Modelos Animales de Enfermedad , Epilepsia/diagnóstico , Hipocampo/inmunología , Hipocampo/metabolismo , Hipocampo/virología , Ratones , Convulsiones/diagnóstico , Convulsiones/etiología , Índice de Severidad de la Enfermedad , Carga Viral
16.
Sci Rep ; 9(1): 4588, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30872675

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV)-induces a demyelinating disease in the spinal cord (SC) of susceptible but not in resistant (B6) mouse strains. The aim of the present study was to induce SC demyelination and a peripheral neuropathy in resistant mice by switching the infection site from cerebrum to SC. B6 mice were intraspinally inoculated with TMEV. Infected mice showed clinical signs starting at 7 days post infection (dpi). Histopathology revealed a mononuclear myelitis, centred on the injection site at 3 dpi with subsequent antero- and retrograde spread, accompanied by demyelination and axonal damage within the SC. Virus protein was detected in the SC at all time points. SC inflammation decreased until the end of the investigation period (28 dpi). Concurrent with the amelioration of SC inflammation, the emergence of a peripheral neuropathy, characterized by axonal damage, demyelination and macrophage infiltration, contributing to persistent clinical sings, was observed. Intraspinal TMEV infection of resistant mice induced inflammation, demyelination and delayed viral clearance in the spinal cord and more interestingly, subsequent, virus-triggered inflammation and degeneration within the PN associated with dramatic and progressive clinical signs. The lesions observed in the PN resemble important features of Guillain-Barré syndrome, especially of acute motor/motor-sensory axonal forms.


Asunto(s)
Enfermedades Desmielinizantes/etiología , Enfermedades Desmielinizantes/patología , Síndrome de Guillain-Barré/diagnóstico , Síndrome de Guillain-Barré/etiología , Enfermedades del Sistema Nervioso Periférico/etiología , Enfermedades del Sistema Nervioso Periférico/patología , Médula Espinal/patología , Médula Espinal/virología , Animales , Biomarcadores , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Inmunohistoquímica , Ratones , Poliomielitis/complicaciones , Poliomielitis/virología , Médula Espinal/metabolismo , Theilovirus/fisiología , Virosis/complicaciones
17.
Int J Mol Sci ; 20(2)2019 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-30669615

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV), a naturally occurring, enteric pathogen of mice is a Cardiovirus of the Picornaviridae family. Low neurovirulent TMEV strains such as BeAn cause a severe demyelinating disease in susceptible SJL mice following intracerebral infection. Furthermore, TMEV infections of C57BL/6 mice cause acute polioencephalitis initiating a process of epileptogenesis that results in spontaneous recurrent epileptic seizures in approximately 50% of affected mice. Moreover, C3H mice develop cardiac lesions after an intraperitoneal high-dose application of TMEV. Consequently, TMEV-induced diseases are widely used as animal models for multiple sclerosis, epilepsy, and myocarditis. The present review summarizes morphological lesions and pathogenic mechanisms triggered by TMEV with a special focus on the development of hippocampal degeneration and seizures in C57BL/6 mice as well as demyelination in the spinal cord in SJL mice. Furthermore, a detailed description of innate and adaptive immune responses is given. TMEV studies provide novel insights into the complexity of organ- and mouse strain-specific immunopathology and help to identify factors critical for virus persistence.


Asunto(s)
Enfermedades de los Animales/virología , Infecciones por Cardiovirus/veterinaria , Theilovirus/fisiología , Inmunidad Adaptativa , Enfermedades de los Animales/inmunología , Enfermedades de los Animales/patología , Enfermedades de los Animales/transmisión , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epilepsia/etiología , Epilepsia/patología , Epilepsia/fisiopatología , Humanos , Inmunidad Innata , Ratones , Esclerosis Múltiple/etiología , Esclerosis Múltiple/patología , Miocarditis/etiología , Miocarditis/patología , Miocarditis/fisiopatología , Convulsiones/etiología , Convulsiones/patología , Convulsiones/fisiopatología , Tropismo Viral
18.
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
19.
Apoptosis ; 23(2): 170-186, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29435686

RESUMEN

Theiler's murine encephalomyelitis (TME) is caused by the TME virus (TMEV) and represents an important animal model for multiple sclerosis (MS). Oligodendroglial apoptosis and reduced apoptotic elimination of encephalitogenic leukocytes seem to participate in autoimmune demyelination in MS. The present study quantified apoptotic cells in BeAn-TMEV-induced spinal cord white matter lesions at 14, 42, 98, and 196 days post infection (dpi) using immunostaining. Apoptotic cells were identified by transmission electron microscopy and double-immunofluorescence. The mRNA expression of apoptosis-related genes was investigated using microarray analysis. Oligodendroglial apoptosis was already detected in the predemyelinating phase at 14 dpi. Apoptotic cell numbers peaked at 42 dpi and decreased until 196 dpi partly due to reduced T cell apoptosis. In addition to genes involved in the classical pathways of apoptosis induction, microarray analysis detected the expression of genes related to alternative mechanisms of cell death such as pyroptosis, necroptosis, and endoplasmic reticulum stress. Consequently, oligodendroglial apoptosis is involved in the initiation of the TME demyelination process, whereas the development of apoptosis resistance of T cells potentially favors the maintenance of inflammation and myelin loss.


Asunto(s)
Apoptosis , Esclerosis Múltiple/virología , Médula Espinal/virología , Theilovirus/fisiología , Animales , Muerte Celular , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Médula Espinal/metabolismo , Médula Espinal/patología , Médula Espinal/fisiopatología , Linfocitos T/citología , Linfocitos T/metabolismo
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA