Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Int J Mol Sci ; 22(10)2021 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-34067536

RESUMO

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.


Assuntos
Doenças Desmielinizantes/imunologia , Esclerose Múltipla/imunologia , Theilovirus/fisiologia , Imunidade Adaptativa/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Astrócitos/metabolismo , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/virologia , Citocinas , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Humanos , Imunidade Inata/imunologia , Camundongos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais/imunologia , Theilovirus/patogenicidade
2.
Sci Rep ; 11(1): 9188, 2021 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-33911136

RESUMO

Eukaryotic translation initiation factor 2 alpha kinase 2 (EIF2AK2), better known as PKR, plays a key role in the response to viral infections and cellular homeostasis by regulating mRNA translation. Upon binding dsRNA, PKR is activated through homodimerization and subsequent autophosphorylation on residues Thr446 and Thr451. In this study, we identified a novel PKR phosphorylation site, Ser6, located 3 amino acids upstream of the first double-stranded RNA binding motif (DRBM1). Another Ser residue occurs in PKR at position 97, the very same position relative to the DRBM2. Ser or Thr residues also occur 3 amino acids upstream DRBMs of other proteins such as ADAR1 or DICER. Phosphoinhibiting mutations (Ser-to-Ala) introduced at Ser6 and Ser97 spontaneously activated PKR. In contrast, phosphomimetic mutations (Ser-to-Asp) inhibited PKR activation following either poly (I:C) transfection or virus infection. These mutations moderately affected dsRNA binding or dimerization, suggesting a model where negative charges occurring at position 6 and 97 tighten the interaction of DRBMs with the kinase domain, thus keeping PKR in an inactive closed conformation even in the presence of dsRNA. This study provides new insights on PKR regulation mechanisms and identifies Ser6 and Ser97 as potential targets to modulate PKR activity for therapeutic purposes.


Assuntos
Motivo de Ligação ao RNA de Cadeia Dupla , Serina/genética , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo , Alanina/genética , Substituição de Aminoácidos , Infecções por Cardiovirus/metabolismo , Células HeLa , Interações Hospedeiro-Patógeno , Humanos , Mutação , Fosforilação , Multimerização Proteica , Proteínas de Ligação a RNA/metabolismo , Serina/química , Theilovirus/patogenicidade
3.
Arch Virol ; 166(4): 1015-1033, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33582855

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Infecções por Vírus de RNA/patologia , Infecções por Vírus de RNA/virologia , Animais , Sistema Nervoso Central/patologia , Sistema Nervoso Central/fisiologia , Sistema Nervoso Central/virologia , Humanos , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/fisiopatologia , Vírus da Hepatite Murina/patogenicidade , Vírus da Hepatite Murina/fisiologia , Infecções por Vírus de RNA/imunologia , Infecções por Vírus de RNA/fisiopatologia , Vírus da Floresta de Semliki/patogenicidade , Vírus da Floresta de Semliki/fisiologia , Theilovirus/patogenicidade , Theilovirus/fisiologia
4.
Cells ; 9(8)2020 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32727036

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated inflammatory demyelinating disease in susceptible mice that is similar to human multiple sclerosis (MS). In light of anti-CD20 therapies for MS, the susceptibility of B cells to TMEV infection is particularly important. In our study, direct viral exposure to macrophages and lymphocytes resulted in viral replication and cellular stimulation in the order of DCs, macrophages, B cells, and T cells. Notably, B cells produced viral proteins and expressed elevated levels of CD69, an activation marker. Similarly, the expression of major histocompatibility complex class II and costimulatory molecules in B cells was upregulated. Moreover, TMEV-infected B cells showed elevated levels of antigen-presenting function and antibody production. TMEV infection appeared to polyclonally activate B cells to produce autoantibodies and further T cell stimulation. Thus, the viral infection might potentially affect the outcome of autoimmune diseases, and/or the development of other chronic infections, including the protection and/or pathogenesis of TMEV-induced demyelinating disease.


Assuntos
Autoanticorpos/metabolismo , Linfócitos B/imunologia , Esclerose Múltipla/imunologia , Theilovirus/patogenicidade , Animais , Feminino , Camundongos
5.
Sci Rep ; 10(1): 6740, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317718

RESUMO

To protect the audiosensory organ from tissue damage from the immune system, the inner ear is separated from the circulating immune system by the blood-labyrinth barrier, which was previously considered an immune-privileged site. Recent studies have shown that macrophages are distributed in the cochlea, especially in the spiral ligament, spiral ganglion, and stria vascularis; however, the direct pathogen defence mechanism used by audiosensory receptor hair cells (HCs) has remained obscure. Here, we show that HCs are protected from pathogens by surrounding accessory supporting cells (SCs) and greater epithelial ridge (GER or Kölliker's organ) cells (GERCs). In isolated murine cochlear sensory epithelium, we established Theiler's murine encephalomyelitis virus, which infected the SCs and GERCs, but very few HCs. The virus-infected SCs produced interferon (IFN)-α/ß, and the viruses efficiently infected the HCs in the IFN-α/ß receptor-null sensory epithelium. Interestingly, the virus-infected SCs and GERCs expressed macrophage marker proteins and were eliminated from the cell layer by cell detachment. Moreover, lipopolysaccharide induced phagocytosis of the SCs without cell detachment, and the SCs phagocytosed the bacteria. These results reveal that SCs function as macrophage-like cells, protect adjacent HCs from pathogens, and provide a novel anti-infection inner ear immune system.


Assuntos
Células Ciliadas Auditivas Internas/fisiologia , Células Ciliadas Auditivas Externas/fisiologia , Células Labirínticas de Suporte/imunologia , Macrófagos/imunologia , Gânglio Espiral da Cóclea/fisiologia , Estria Vascular/fisiologia , Animais , Animais Recém-Nascidos , Escherichia coli/imunologia , Células Ciliadas Auditivas Internas/citologia , Células Ciliadas Auditivas Externas/citologia , Imunidade Inata , Interferon-alfa/biossíntese , Interferon-alfa/imunologia , Interferon beta/biossíntese , Interferon beta/imunologia , Células Labirínticas de Suporte/citologia , Células Labirínticas de Suporte/efeitos dos fármacos , Células Labirínticas de Suporte/virologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos ICR , Técnicas de Cultura de Órgãos , Fagocitose/efeitos dos fármacos , Saccharomyces cerevisiae/imunologia , Gânglio Espiral da Cóclea/citologia , Estria Vascular/citologia , Theilovirus/crescimento & desenvolvimento , Theilovirus/patogenicidade
6.
Int J Mol Sci ; 20(13)2019 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-31262031

RESUMO

(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.


Assuntos
Cinomose/metabolismo , Encefalite Viral/metabolismo , Bainha de Mielina/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Catalase/metabolismo , Cinomose/patologia , Cães , Encefalite Viral/patologia , Encefalite Viral/virologia , Feminino , Masculino , Camundongos , Bainha de Mielina/patologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase/metabolismo , Theilovirus/patogenicidade
7.
PLoS Pathog ; 15(2): e1007574, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30742696

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteinopatias TDP-43/metabolismo , Theilovirus/metabolismo , Animais , Autopsia , Linhagem Celular , Núcleo Celular , Células Cultivadas , Citoplasma , Proteínas de Ligação a DNA/fisiologia , Humanos , Camundongos , Transporte Proteico/fisiologia , Proteinopatias TDP-43/fisiopatologia , Theilovirus/patogenicidade
8.
Brain Behav Immun ; 77: 110-126, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30582962

RESUMO

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.


Assuntos
Ácidos Araquidônicos/farmacologia , Endocanabinoides/farmacologia , Glicerídeos/farmacologia , Microglia/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Animais , Ácidos Araquidônicos/metabolismo , Axônios/metabolismo , Diferenciação Celular/fisiologia , Corpo Caloso/patologia , Corpo Caloso/fisiologia , Doenças Desmielinizantes/fisiopatologia , Modelos Animais de Doenças , Endocanabinoides/metabolismo , Feminino , Glicerídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Células Precursoras de Oligodendrócitos/fisiologia , Oligodendroglia/metabolismo , Theilovirus/patogenicidade
9.
PLoS One ; 13(9): e0202590, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30231069

RESUMO

Extracellular vesicles (EVs) have emerged as important mediators of intercellular communication and as possible therapeutic agents in inflammation-mediated demyelinating diseases, including multiple sclerosis (MS). In the present study, we investigated whether intravenously administered EVs derived from mesenchymal stem cells (MSCs) from human adipose tissue might mediate recovery in Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease, a progressive model of MS. SJL/J mice were subjected to EV treatment once the disease was established. We found that intravenous EV administration improved motor deficits, reduced brain atrophy, increased cell proliferation in the subventricular zone and decreased inflammatory infiltrates in the spinal cord in mice infected with TMEV. EV treatment was also capable of modulating neuroinflammation, given glial fibrillary acidic protein and Iba-1 staining were reduced in the brain, whereas myelin protein expression was increased. Changes in the morphology of microglial cells in the spinal cord suggest that EVs also modulate the activation state of microglia. The clear reduction in plasma cytokine levels, mainly in the Th1 and Th17 phenotypes, in TMEV mice treated with EVs confirms the immunomodulatory ability of intravenous EVs. According to our results, EV administration attenuates motor deficits through immunomodulatory actions, diminishing brain atrophy and promoting remyelination. Further studies are necessary to establish EV delivery as a possible therapy for the neurodegenerative phase of MS.


Assuntos
Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/citologia , Esclerose Múltipla/terapia , Theilovirus/patogenicidade , Tecido Adiposo/citologia , Administração Intravenosa , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Proteínas da Mielina/metabolismo , Resultado do Tratamento
10.
PLoS Biol ; 16(6): e2006459, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29953453

RESUMO

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.


Assuntos
Taxa de Mutação , Vírus de RNA/genética , Vírus de RNA/patogenicidade , Virulência/genética , Células 3T3 , Substituição de Aminoácidos , Animais , Evolução Molecular Direcionada , Feminino , Interações entre Hospedeiro e Microrganismos/genética , Cinética , Masculino , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Mutagênese Sítio-Dirigida , Polimorfismo de Nucleotídeo Único , Vírus de RNA/fisiologia , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/metabolismo , Theilovirus/genética , Theilovirus/patogenicidade , Theilovirus/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral/genética
11.
Neurosci Lett ; 677: 88-93, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29705539

RESUMO

Multiple sclerosis (MS) is a high prevalence degenerative disease characterized at the cellular level by glial and neuronal cell death. The causes of cell death during the disease course are not fully understood. In this work we demonstrate that in a MS model induced by Theiler's murine encephalomyelitis virus (TMEV) infection, the inward rectifier (Kir) 4.1 potassium channel subunit is overexpressed in astrocytes. In voltage clamp experiments the inward current density from TMEV-infected astrocytes was significantly larger than in mock-infected ones. The cRNA hybridization analysis from mock- and TMEV-infected cells showed an upregulation of a potassium transport channel coding sequence. We validated this mRNA increase by RT-PCR and quantitative PCR using Kir 4.1 specific primers. Western blotting experiments confirmed the upregulation of Kir 4.1, and alignment between sequences provided the demonstration that the over-expressed gene encodes for a Kir family member. Flow cytometry showed that the Kir 4.1 protein is located mainly in the cell membrane in mock and TMEV-infected astrocytes. Our results demonstrate an increase in K+ inward current in TMEV-infected glial cells, this increment may reduce the neuronal depolarization, contributing to cell resilience mechanisms.


Assuntos
Astrócitos/metabolismo , Esclerose Múltipla/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Animais , Infecções por Cardiovirus/complicações , Infecções por Cardiovirus/metabolismo , Linhagem Celular , Modelos Animais de Doenças , Potenciais da Membrana , Mesocricetus , Esclerose Múltipla/virologia , RNA Mensageiro , Theilovirus/patogenicidade , Regulação para Cima
12.
Cytokine ; 102: 83-93, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28800924

RESUMO

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.


Assuntos
Infecções por Cardiovirus/etiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Ativação de Macrófagos , Fator Estimulador de Colônias de Macrófagos/imunologia , Theilovirus/patogenicidade , Animais , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Diferenciação Celular/imunologia , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Glicólise , Macrófagos/imunologia , Macrófagos/patologia , Macrófagos/virologia , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Viral/genética , Theilovirus/genética , Theilovirus/isolamento & purificação , Replicação Viral/imunologia
13.
J Neuroimmunol ; 313: 34-40, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29153606

RESUMO

We evaluated the effects of pegylated-interferonß-1a (pegIFNß) therapy on intrathecal antibody responses, disability progression, and viral load in the CNS in mice infected with the Theiler's virus (TMEV), an animal model of progressive disability in Multiple Sclerosis (MS). The lack of a direct antiviral activity in the CNS, the absence of any effect upon the intrathecal immune response, and the failure to treat disease progression, indicate that the immunomodulatory effects of pegIFNß-1a likely occur in the systemic circulation rather than within the CNS. These results may be relevant to the relative lack of effect of IFNß in progressive MS relative to relapsing MS.


Assuntos
Fatores Imunológicos/uso terapêutico , Interferon beta/uso terapêutico , Esclerose Múltipla/tratamento farmacológico , Esclerose Múltipla/virologia , Theilovirus/patogenicidade , Animais , Anticorpos Antivirais/sangue , Avaliação da Deficiência , Modelos Animais de Doenças , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Camundongos , RNA Mensageiro/metabolismo , Teste de Desempenho do Rota-Rod , Estatísticas não Paramétricas , Theilovirus/imunologia , Carga Viral
14.
Sci Rep ; 7(1): 12194, 2017 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-28939838

RESUMO

Infection by Theiler's murine encephalomyelitis virus (TMEV) is a model for neurological outcomes caused by virus infection because it leads to diverse neurological conditions in mice, depending on the strain infected. To extend knowledge on the heterogeneous neurological outcomes caused by TMEV and identify new models of human neurological diseases associated with antecedent infections, we analyzed the phenotypic consequences of TMEV infection in the Collaborative Cross (CC) mouse population. We evaluated 5 different CC strains for outcomes of long-term infection (3 months) and acute vs. early chronic infection (7 vs. 28 days post-infection), using neurological and behavioral phenotyping tests and histology. We correlated phenotypic observations with haplotypes of genomic regions previously linked to TMEV susceptibility to test the hypothesis that genomic diversity within CC mice results in variable disease phenotypes in response to TMEV. None of the 5 strains analyzed had a response identical to that of any other CC strain or inbred strain for which prior data are available, indicating that strains of the CC can produce novel models of neurological disease. Thus, CC strains can be a powerful resource for studying how viral infection can cause different neurological outcomes depending on host genetic background.


Assuntos
Doenças Desmielinizantes/genética , Modelos Animais de Doenças , Patrimônio Genético , Camundongos Endogâmicos/genética , Theilovirus/patogenicidade , Animais , Comportamento Animal , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Feminino , Humanos , Masculino , Camundongos , Fenótipo , Viroses
15.
Sci Rep ; 7: 44377, 2017 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-28290524

RESUMO

Recent studies have begun to point out the contribution of microbiota to multiple sclerosis (MS) pathogenesis. Theiler's murine encephalomyelitis virus induced demyelinating disease (TMEV-IDD) is a model of progressive MS. Here, we first analyze the effect of intracerebral infection with TMEV on commensal microbiota and secondly, whether the early microbiota depletion influences the immune responses to TMEV on the acute phase (14 dpi) and its impact on the chronic phase (85 dpi). The intracranial inoculation of TMEV was associated with a moderate dysbiosis. The oral administration of antibiotics (ABX) of broad spectrum modified neuroimmune responses to TMEV dampening brain CD4+ and CD8+ T infiltration during the acute phase. The expression of cytokines, chemokines and VP2 capsid protein was enhanced and accompanied by clusters of activated microglia disseminated throughout the brain. Furthermore, ABX treated mice displayed lower levels of CD4+ and CD8+T cells in cervical and mesenteric lymph nodes. Increased mortality to TMEV was observed after ABX cessation at day 28pi. On the chronic phase, mice that survived after ABX withdrawal and recovered microbiota diversity showed subtle changes in brain cell infiltrates, microglia and gene expression of cytokines. Accordingly, the surviving mice of the group ABX-TMEV displayed similar disease severity than TMEV mice.


Assuntos
Encéfalo/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Esclerose Múltipla/imunologia , Animais , Encéfalo/microbiologia , Encéfalo/fisiopatologia , Encéfalo/virologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Disbiose/microbiologia , Disbiose/patologia , Disbiose/virologia , Humanos , Linfonodos/imunologia , Linfonodos/microbiologia , Linfonodos/virologia , Ativação Linfocitária/imunologia , Camundongos , Esclerose Múltipla/microbiologia , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Neuroimunomodulação , Medula Espinal/imunologia , Medula Espinal/microbiologia , Medula Espinal/patologia , Medula Espinal/virologia , Theilovirus/imunologia , Theilovirus/patogenicidade
16.
J Neuroimmunol ; 308: 30-42, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28237622

RESUMO

Mouse models are great tools to study the mechanisms of disease development. Theiler's murine encephalomyelitis virus is used in two distinct viral infection mouse models to study the human diseases multiple sclerosis (MS) and epilepsy. Intracerebral (i.c.) infection of the SJL/J mouse strain results in persistent viral infection of the central nervous system and a MS-like disease, while i.c. infection of the C57BL/6J mouse strain results in acute seizures and epilepsy. Our understanding of how the immune system contributes to the development of two disparate diseases caused by the same virus is presented.


Assuntos
Epilepsia , Esclerose Múltipla , Poliomielite/complicações , Theilovirus/patogenicidade , Imunidade Adaptativa , Animais , Antígenos CD/metabolismo , Linfócitos B/patologia , Linfócitos B/virologia , Modelos Animais de Doenças , Progressão da Doença , Epilepsia/genética , Epilepsia/imunologia , Epilepsia/patologia , Epilepsia/virologia , Macrófagos/patologia , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microglia/patologia , Microglia/virologia , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Linfócitos T/patologia , Linfócitos T/virologia
17.
Neurobiol Dis ; 99: 121-132, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28017800

RESUMO

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.


Assuntos
Encefalite Viral/patologia , Encefalomielite Aguda Disseminada/patologia , Epilepsia/patologia , Esclerose Múltipla/patologia , Theilovirus , Animais , Encéfalo/imunologia , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Encefalite Viral/imunologia , Encefalite Viral/virologia , Encefalomielite Aguda Disseminada/imunologia , Encefalomielite Aguda Disseminada/virologia , Epilepsia/imunologia , Epilepsia/virologia , Feminino , Interações Hospedeiro-Patógeno , Camundongos Endogâmicos C57BL , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Fenótipo , Polimorfismo de Nucleotídeo Único , RNA Viral/metabolismo , Especificidade da Espécie , Theilovirus/genética , Theilovirus/patogenicidade , Virulência
18.
Int Immunol ; 28(12): 575-584, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27803063

RESUMO

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.


Assuntos
Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/virologia , Integrina alfa4/metabolismo , Esclerose Múltipla/metabolismo , Theilovirus/patogenicidade , Animais , Modelos Animais de Doenças , Feminino , Integrina alfa4/genética , Integrina alfa4/imunologia , Camundongos , Camundongos Endogâmicos , Esclerose Múltipla/virologia
19.
Epilepsia ; 57(12): 1958-1967, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27739576

RESUMO

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.


Assuntos
Anticonvulsivantes/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Epilepsia do Lobo Temporal , Minociclina/uso terapêutico , Theilovirus/patogenicidade , Ácido Valproico/uso terapêutico , Animais , Transtornos de Ansiedade/tratamento farmacológico , Transtornos de Ansiedade/etiologia , Peso Corporal/efeitos dos fármacos , Distribuição de Qui-Quadrado , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/etiologia , Epilepsia do Lobo Temporal/virologia , Comportamento Exploratório/efeitos dos fármacos , Camundongos , Atividade Motora/efeitos dos fármacos , Desempenho Psicomotor/efeitos dos fármacos , Teste de Desempenho do Rota-Rod
20.
J Immunol Methods ; 439: 23-28, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27623324

RESUMO

Effective recovery of activated brain infiltrating lymphocytes is critical for investigations involving murine neurological disease models. To optimize lymphocyte recovery, we compared two isolation methods using brains harvested from seven-day Theiler's murine encephalomyelitis virus (TMEV) and TMEV-OVA infected mice. Brains were processed using either a manual dounce based approach or enzymatic digestion using type IV collagenase. The resulting cell suspensions from these two techniques were transferred to a percoll gradient, centrifuged, and lymphocytes were recovered. Flow cytometric analysis of CD45hi cells showed greater percentage of CD44hiCD62lo activated lymphocytes and CD19+ B cells using the dounce method. In addition, we achieved a 3-fold greater recovery of activated virus-specific CD8 T cells specific for the immunodominant Db:VP2121-130 and engineered Kb:OVA257-264 epitopes through manual dounce homogenization approach as compared to collagenase digest. A greater percentage of viable cells was also achieved through dounce homogenization. Therefore, we conclude that manual homogenization is a superior approach to isolate activated T cells from the mouse brain.


Assuntos
Antígenos/imunologia , Encéfalo/imunologia , Linfócitos T CD8-Positivos/imunologia , Infecções por Cardiovirus/imunologia , Separação Celular/métodos , Ativação Linfocitária , Ovalbumina/imunologia , Theilovirus/imunologia , Animais , Antígenos CD19/imunologia , Linfócitos B/imunologia , Encéfalo/metabolismo , Proteínas do Capsídeo/imunologia , Infecções por Cardiovirus/virologia , Sobrevivência Celular , Centrifugação com Gradiente de Concentração , Colágeno Tipo IV/metabolismo , Colagenases/metabolismo , Modelos Animais de Doenças , Epitopos , Feminino , Citometria de Fluxo , Receptores de Hialuronatos/imunologia , Masculino , Camundongos Endogâmicos C57BL , Povidona/química , Selectinas/imunologia , Dióxido de Silício/química , Theilovirus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA