Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
J Immunol ; 210(7): 926-934, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36883849

RESUMO

The proinflammatory microRNA-155 (miR-155) is highly expressed in the serum and CNS lesions of patients with multiple sclerosis (MS). Global knockout (KO) of miR-155 in mice confers resistance to a mouse model of MS, experimental autoimmune encephalomyelitis (EAE), by reducing the encephalogenic potential of CNS-infiltrating Th17 T cells. However, cell-intrinsic roles for miR-155 during EAE have not been formally determined. In this study, we use single-cell RNA sequencing and cell-specific conditional miR-155 KOs to determine the importance of miR-155 expression in distinct immune cell populations. Time-course single-cell sequencing revealed reductions in T cells, macrophages, and dendritic cells (DCs) in global miR-155 KO mice compared with wild-type controls at day 21 after EAE induction. Deletion of miR-155 in T cells, driven by CD4 Cre, significantly reduced disease severity similar to global miR-155 KOs. CD11c Cre-mediated deletion of miR-155 in DCs also resulted in a modest yet significant reduction in the development of EAE, with both T cell- and DC-specific KOs showing a reduction in Th17 T cell infiltration into the CNS. Although miR-155 is highly expressed in infiltrating macrophages during EAE, deletion of miR-155 using LysM Cre did not impact disease severity. Taken together, these data show that although miR-155 is highly expressed in most infiltrating immune cells, miR-155 has distinct roles and requirements depending on the cell type, and we have demonstrated this using the gold standard conditional KO approach. This provides insights into which functionally relevant cell types should be targeted by the next generation of miRNA therapeutics.


Assuntos
Encefalomielite Autoimune Experimental , MicroRNAs , Esclerose Múltipla , Animais , Camundongos , Doenças Neuroinflamatórias , Células Th17/metabolismo , Encéfalo/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
J Neuroinflammation ; 17(1): 291, 2020 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-33023618

RESUMO

BACKGROUND: Multiple sclerosis (MS) is an inflammatory demyelinating disease that affects 2.5 million people worldwide. Growing evidence suggests that perturbation of the gut microbiota, the dense collection of microorganisms that colonize the gastrointestinal tract, plays a functional role in MS. Indeed, specific gut-resident bacteria are altered in patients with MS compared to healthy individuals, and colonization of gnotobiotic mice with MS-associated microbiota exacerbates preclinical models of MS. However, defining the molecular mechanisms by which gut commensals can remotely affect the neuroinflammatory process remains a critical gap in the field. METHODS: We utilized monophasic experimental autoimmune encephalomyelitis (EAE) in C57BL/6J mice and relapse-remitting EAE in SJL/J mice to test the effects of the products from a human gut-derived commensal strain of Lactobacillus paracasei (Lb). RESULTS: We report that Lb can ameliorate preclinical murine models of MS with both prophylactic and therapeutic administrations. Lb ameliorates disease through a Toll-like receptor 2-dependent mechanism via its microbe-associated molecular patterns that can be detected in the systemic circulation, are sufficient to downregulate chemokine production, and can reduce immune cell infiltration into the central nervous system (CNS). In addition, alterations in the gut microbiota mediated by Lb-associated molecular patterns are sufficient to provide partial protection against neuroinflammatory diseases. CONCLUSIONS: Local Lb modulation of the gut microbiota and the shedding of Lb-associated molecular patterns into the circulation may be important physiological signals to prevent aberrant peripheral immune cell infiltration into the CNS and have relevance to the development of new therapeutic strategies for MS.


Assuntos
Sistema Nervoso Central/imunologia , Microbioma Gastrointestinal/imunologia , Lacticaseibacillus paracasei/imunologia , Leucócitos/imunologia , Animais , Sistema Nervoso Central/patologia , Feminino , Humanos , Leucócitos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
Proc Natl Acad Sci U S A ; 114(21): 5497-5502, 2017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28487480

RESUMO

Symbiotic microbes impact the severity of a variety of diseases through regulation of T-cell development. However, little is known regarding the molecular mechanisms by which this is accomplished. Here we report that a secreted factor, Erdr1, is regulated by the microbiota to control T-cell apoptosis. Erdr1 expression was identified by transcriptome analysis to be elevated in splenic T cells from germfree and antibiotic-treated mice. Suppression of Erdr1 depends on detection of circulating microbial products by Toll-like receptors on T cells, and this regulation is conserved in human T cells. Erdr1 was found to function as an autocrine factor to induce apoptosis through caspase 3. Consistent with elevated levels of Erdr1, germfree mice have increased splenic T-cell apoptosis. RNA sequencing of Erdr1-overexpressing cells identified the up-regulation of genes involved in Fas-mediated cell death, and Erdr1 fails to induce apoptosis in Fas-deficient cells. Importantly, forced changes in Erdr1 expression levels dictate the survival of auto-reactive T cells and the clinical outcome of neuro-inflammatory autoimmune disease. Cellular survival is a fundamental feature regulating appropriate immune responses. We have identified a mechanism whereby the host integrates signals from the microbiota to control T-cell apoptosis, making regulation of Erdr1 a potential therapeutic target for autoimmune disease.


Assuntos
Apoptose , Proteínas de Membrana/fisiologia , Microbiota , Linfócitos T/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Animais , Encefalomielite Autoimune Experimental/metabolismo , Homeostase , Humanos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Receptor fas/metabolismo
4.
Eur J Immunol ; 48(7): 1199-1210, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29697856

RESUMO

Increasing evidence points to an important role for neutrophils in participating in the pathogenesis of the human demyelinating disease MS and the animal model EAE. Therefore, a better understanding of the signals controlling migration of neutrophils as well as evaluating the role of these cells in demyelination is important to define cellular components that contribute to disease in MS patients. In this study, we examined the functional role of the chemokine CXCL1 in contributing to neuroinflammation and demyelination in EAE. Using transgenic mice in which expression of CXCL1 is under the control of a tetracycline-inducible promoter active within glial fibrillary acidic protein-positive cells, we have shown that sustained CXCL1 expression within the CNS increased the severity of clinical and histologic disease that was independent of an increase in the frequency of encephalitogenic Th1 and Th17 cells. Rather, disease was associated with enhanced recruitment of CD11b+ Ly6G+ neutrophils into the spinal cord. Targeting neutrophils resulted in a reduction in demyelination arguing for a role for these cells in myelin damage. Collectively, these findings emphasize that CXCL1-mediated attraction of neutrophils into the CNS augments demyelination suggesting that this signaling pathway may offer new targets for therapeutic intervention.


Assuntos
Sistema Nervoso Central/imunologia , Quimiocina CXCL1/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Esclerose Múltipla/imunologia , Neutrófilos/imunologia , Medula Espinal/imunologia , Animais , Autoantígenos/imunologia , Antígeno CD11b/metabolismo , Células Cultivadas , Quimiocina CXCL1/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Terapia de Alvo Molecular , Glicoproteína Mielina-Oligodendrócito/imunologia , Inflamação Neurogênica , Infiltração de Neutrófilos , Fragmentos de Peptídeos/imunologia , Transdução de Sinais , Medula Espinal/patologia
5.
J Neuroinflammation ; 16(1): 152, 2019 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-31325960

RESUMO

BACKGROUND: In the healthy central nervous system (CNS), microglia are found in a homeostatic state and peripheral macrophages are absent from the brain. Microglia play key roles in maintaining CNS homeostasis and acting as first responders to infection and inflammation, and peripheral macrophages infiltrate the CNS during neuroinflammation. Due to their distinct origins and functions, discrimination between these cell populations is essential to the comprehension of neuroinflammatory disorders. Studies comparing the gene profiles of microglia and peripheral macrophages, or macrophages in vitro-derived from bone marrow, under non-infectious conditions of the CNS, have revealed valuable microglial-specific genes. However, studies comparing gene profiles between CNS-infiltrating macrophages and microglia, when both are isolated from the CNS during viral-induced neuroinflammation, are lacking. METHODS: We isolated, via flow cytometry, microglia and infiltrating macrophages from the brains of Theiler's murine encephalomyelitis virus-infected C57BL/6 J mice and used RNA-Seq, followed by validation with qPCR, to examine the differential transcriptional profiles of these cells. We utilized primary literature defining subcellular localization to determine whether or not particular proteins extracted from the transcriptional profiles were expressed at the cell surface. The surface expression and cellular specificity of triggering receptor expressed on myeloid cells 1 (TREM-1) protein were examined via flow cytometry. We also examined the immune response gene profile within the transcriptional profiles of these isolated microglia and infiltrating macrophages. RESULTS: We have identified and validated new microglial- and macrophage-specific genes, encoding cell surface proteins, expressed at the peak of neuroinflammation. TREM-1 protein was confirmed to be expressed by infiltrating macrophages, not microglia, at the peak of neuroinflammation. We also identified both unique and redundant immune functions, through examination of the immune response gene profiles, of microglia and infiltrating macrophages during neurotropic viral infection. CONCLUSIONS: The differential expression of cell surface-specific genes during neuroinflammation can potentially be used to discriminate between microglia and macrophages as well as provide a resource that can be further utilized to target and manipulate specific cell responses during neuroinflammation.


Assuntos
Encéfalo/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Microglia/imunologia , Animais , Infecções por Cardiovirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Theilovirus/imunologia , Transcrição Gênica , Transcriptoma
6.
J Neurovirol ; 25(3): 415-421, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30859497

RESUMO

Microglia are the only resident myeloid cell in the central nervous system (CNS) parenchyma, but the role of microglia in the context of neurotropic viral infection is poorly understood. Using different amounts of Theiler's murine encephalomyelitis virus (TMEV) in a preclinical model of epilepsy and PLX5622, a colony stimulating factor-1 receptor inhibitor that selectively depletes microglia in the CNS, we report that microglia-depleted, TMEV-infected mice develop seizures, manifest paralysis, and uniformly succumb to fatal encephalitis regardless of viral amount. CNS demyelination correlates with viral amount; however, viral amount does not correlate with axon damage and TMEV antigen in the CNS.


Assuntos
Infecções por Cardiovirus/imunologia , Encefalite Viral/imunologia , Microglia/imunologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Theilovirus/imunologia
7.
J Neurovirol ; 24(4): 464-477, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29687406

RESUMO

Viral infection of the central nervous system can result in encephalitis. About 20% of individuals who develop viral encephalitis go on to develop epilepsy. We have established an experimental model where virus infection of mice with Theiler's murine encephalomyelitis virus (TMEV) leads to acute seizures, followed by a latent period (no seizures/epileptogenesis phase) and then spontaneous recurrent seizures-epilepsy. Infiltrating macrophages (CD11b+CD45hi) present in the brain at day 3 post-infection are an important source of interleukin-6, which contributes to the development of acute seizures in the TMEV-induced seizure model. Time course analysis of viral infection and inflammatory [CD11b+CD45hiLy-6Chi] and patrolling [CD11b+CD45hiLy-6Clow] monocyte and T cell infiltration into the brains of TMEV-infected C57BL/6J mice over the entire course of the acute viral infection was performed to elucidate the role of virus and the immune response to virus in seizures and viral clearance. The infiltrating inflammatory macrophages were present early following infection but declined over the course of acute viral infection, supporting a role in seizure development, while the lymphocyte infiltration increased rapidly and plateaued, advocating that they play a role in viral clearance. In addition, we showed for the first time that, while TMEV infection of RAG1-/- mice did not alter the number of mice experiencing acute seizures, TMEV infection of C57BL/6J mice depleted of macrophages resulted in a significant decrease in the number of mice experiencing seizures, again supporting a role for infiltrating macrophages in the development of acute seizures in the TMEV-induced seizure model.


Assuntos
Infecções por Cardiovirus/imunologia , Encefalite Viral/imunologia , Convulsões/virologia , Animais , Encefalite Viral/virologia , Camundongos , Camundongos Endogâmicos C57BL , Theilovirus/imunologia
9.
J Neurovirol ; 23(5): 696-703, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28741149

RESUMO

Seizure disorders are often associated with infectious etiologies. Infection, via the intracerebral (i.c.) route, of C57BL/6J mice with the Daniels (DA) strain of Theiler's murine encephalomyelitis virus (TMEV) results in approximately 50% of the mice developing acute behavioral seizures. TMEV-DA is the wild-type strain of the virus that replicates within the parenchyma of the brain. A variant of TMEV-DA, TMEV-H101, does not replicate within the parenchyma of the brain. However, infection with TMEV-H101 via the i.c. route still results in approximately 40% of the mice developing acute behavioral seizures. Infiltrating macrophages producing interleukin-6 (IL-6) have been implicated in the induction of acute seizures following TMEV-DA infection. We examined macrophage infiltration and microglial activation within the brain and cytokine levels in the periphery in mice infected with TMEV-DA or TMEV-H101 and assessed the effects of the addition of recombinant IL-6 to the periphery in wild-type and IL-6 knockout mice infected with TMEV-DA. We found that pathologic levels of IL-6 in the periphery may play a role in the development of seizures when viral replication within the brain is limited. Examination of the role played by the peripheral immune system in the development of seizures/epilepsy in the TMEV-induced seizure model, the first viral infection driven model for epilepsy, could lead to the elucidation of novel therapeutics.


Assuntos
Infecções por Cardiovirus/complicações , Infecções por Cardiovirus/imunologia , Interleucina-6/imunologia , Convulsões/metabolismo , Convulsões/virologia , Animais , Infecções por Cardiovirus/metabolismo , Interleucina-6/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Microglia/metabolismo , Theilovirus/imunologia
10.
Acta Neuropathol ; 131(2): 211-234, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26423537

RESUMO

Epilepsy is the tendency to have unprovoked epileptic seizures. Anything causing structural or functional derangement of brain physiology may lead to seizures, and different conditions may express themselves solely by recurrent seizures and thus be labelled "epilepsy." Worldwide, epilepsy is the most common serious neurological condition. The range of risk factors for the development of epilepsy varies with age and geographic location. Congenital, developmental and genetic conditions are mostly associated with the development of epilepsy in childhood, adolescence and early adulthood. Head trauma, infections of the central nervous system (CNS) and tumours may occur at any age and may lead to the development of epilepsy. Infections of the CNS are a major risk factor for epilepsy. The reported risk of unprovoked seizures in population-based cohorts of survivors of CNS infections from developed countries is between 6.8 and 8.3 %, and is much higher in resource-poor countries. In this review, the various viral, bacterial, fungal and parasitic infectious diseases of the CNS which result in seizures and epilepsy are discussed. The pathogenesis of epilepsy due to brain infections, as well as the role of experimental models to study mechanisms of epileptogenesis induced by infectious agents, is reviewed. The sterile (non-infectious) inflammatory response that occurs following brain insults is also discussed, as well as its overlap with inflammation due to infections, and the potential role in epileptogenesis. Furthermore, autoimmune encephalitis as a cause of seizures is reviewed. Potential strategies to prevent epilepsy resulting from brain infections and non-infectious inflammation are also considered.


Assuntos
Infecções do Sistema Nervoso Central/complicações , Epilepsia/etiologia , Epilepsia/imunologia , Animais , Infecções do Sistema Nervoso Central/patologia , Epilepsia/patologia , Humanos
11.
J Virol ; 87(3): 1849-60, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23236075

RESUMO

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.


Assuntos
Infecções por Cardiovirus/complicações , Infecções por Cardiovirus/patologia , Interleucina-6/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Convulsões , Theilovirus/patogenicidade , Animais , Infecções por Cardiovirus/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia , Theilovirus/imunologia , Fator de Necrose Tumoral alfa/metabolismo
12.
NMR Biomed ; 27(7): 843-52, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24816651

RESUMO

Clinicopathological paradox has hampered significantly the effective assessment of the efficacy of therapeutic intervention for multiple sclerosis. Neuroimaging biomarkers of tissue injury could guide more effective treatment by accurately reflecting the underlying subclinical pathologies. Diffusion tensor imaging-derived directional diffusivity and anisotropy indices have been applied to characterize white matter disorders. However, these biomarkers are sometimes confounded by the complex pathologies seen in multiple sclerosis and its animal models. Recently, a novel technique of diffusion basis spectrum imaging has been developed to quantitatively assess axonal injury, demyelination and inflammation in a mouse model of inflammatory demyelination. Lenaldekar, which inhibits T-cell expansion in a non-cytolytic manner, has been shown to suppress relapses and preserve white matter integrity in mice with experimental autoimmune encephalomyelitis. In this study, relapsing-remitting experimental autoimmune encephalomyelitis was induced through active immunization of SJL/J mice with a myelin proteolipid protein peptide. The therapeutic efficacy of Lenaldekar treatment was evaluated via daily clinical score, cross-sectional ex vivo diffusion basis spectrum imaging examination and histological analysis. Lenaldekar greatly reduced relapse severity and protected white matter integrity in these experimental autoimmune encephalomyelitis mice. Diffusion basis spectrum imaging-derived axial diffusivity, radial diffusivity and restricted diffusion tensor fraction accurately reflected axonal injury, myelin integrity and inflammation-associated cellularity change, respectively. These results support the potential use of diffusion basis spectrum imaging as an effective outcome measure for preclinical drug evaluation.


Assuntos
Axônios/patologia , Doenças Desmielinizantes/diagnóstico , Imagem de Tensor de Difusão , Encefalomielite Autoimune Experimental/diagnóstico , Inflamação/diagnóstico , Animais , Biomarcadores/metabolismo , Doenças Desmielinizantes/complicações , Doenças Desmielinizantes/patologia , Encefalomielite Autoimune Experimental/complicações , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/patologia , Feminino , Hidrazonas/uso terapêutico , Indóis/metabolismo , Inflamação/complicações , Inflamação/patologia , Camundongos , Proteína Básica da Mielina/metabolismo , Quinolinas/uso terapêutico , Recidiva
13.
Curr Opin Rheumatol ; 25(4): 496-501, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23656710

RESUMO

PURPOSE OF REVIEW: This review will explore two new aspects of the involvement of viruses in multiple sclerosis pathogenesis. The first aspect is the complex interactions between viruses. The second aspect is the proposal of a mechanism by which autoreactive T cells are able to escape thymic selection and potentially recognize self and a pathogen. RECENT FINDINGS: With regard to viruses, recent work has demonstrated that one virus may enhance the replication of another virus, potentially leading to an increase in inflammation and disease progression. Also, interactions between human endogenous retroviruses, which likely do not replicate, and certain herpes viruses, may also play a role in disease pathogenesis. Mechanistically, T cells expressing dual T-cell receptors would be able to recognize self and a foreign antigen specifically. Therefore, human endogenous retroviruses potentially play a role in multiple sclerosis pathogenesis, and both interactions between multiple viruses and autoreactive CD8(+) T cells with dual T-cell receptors may play a role in the pathogenesis of the disease. SUMMARY: The complex interactions between multiple viral infections, either within the central nervous system or in the periphery, and the host immune response to viral infection may be such that a variety of viral specificities result in the activation of T cells that recognize self and induce multiple sclerosis. Therefore, it is unlikely that any one microbe will be determined to be the causative agent of multiple sclerosis as reflected by the number of potential triggering mechanisms of the disease.


Assuntos
Autoimunidade/imunologia , Esclerose Múltipla/imunologia , Esclerose Múltipla/virologia , Viroses/complicações , Linfócitos T CD8-Positivos/imunologia , Humanos , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Viroses/imunologia , Replicação Viral
14.
Heliyon ; 9(7): e18095, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37483821

RESUMO

Over the course of human history, numerous diseases have been caused by the transmission of viruses from an animal reservoir into the human population. The viruses of the genus Morbillivirus are human and animal pathogens that emerged from a primordial ancestor a millennia ago and have been transmitting to new hosts, adapting, and evolving ever since. Through interaction with susceptible individuals, as yet undiscovered morbilliviruses or existing morbilliviruses in animal hosts could cause future zoonotic diseases in humans.

15.
J Virol ; 85(16): 8149-57, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21680509

RESUMO

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.


Assuntos
Infecções por Cardiovirus/virologia , Encefalite Viral/virologia , Convulsões/virologia , Theilovirus/genética , Theilovirus/patogenicidade , Animais , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/patologia , Citocinas/biossíntese , Citocinas/sangue , Encefalite , Encefalite Viral/imunologia , Encefalite Viral/patologia , Camundongos , Camundongos Endogâmicos C57BL , Theilovirus/classificação
16.
J Virol ; 85(14): 6913-22, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21543484

RESUMO

Cells that can participate in an innate immune response within the central nervous system (CNS) include infiltrating cells (polymorphonuclear leukocytes [PMNs], macrophages, and natural killer [NK] cells) and resident cells (microglia and sometimes astrocytes). The proinflammatory cytokine interleukin-6 (IL-6) is produced by all of these cells and has been implicated in the development of behavioral seizures in the Theiler's murine encephalomyelitis virus (TMEV)-induced seizure model. The assessment, via PCR arrays, of the mRNA expression levels of a large number of chemokines (ligands and receptors) in TMEV-infected and mock-infected C57BL/6 mice both with and without seizures did not clearly demonstrate the involvement of PMNs, monocytes/macrophages, or NK cells in the development of seizures, possibly due to overlapping function of the chemokines. Additionally, C57BL/6 mice unable to recruit or depleted of infiltrating PMNs and NK cells had seizure rates comparable to those of controls following TMEV infection, and therefore PMNs and NK cells do not significantly contribute to seizure development. In contrast, C57BL/6 mice treated with minocycline, which affects monocytes/macrophages, microglial cells, and PMNs, had significantly fewer seizures than controls following TMEV infection, indicating monocytes/macrophages and resident microglial cells are important in seizure development. Irradiated bone marrow chimeric mice that were either IL-6-deficient mice reconstituted with wild-type bone marrow cells or wild-type mice reconstituted with IL-6-deficient bone marrow cells developed significantly fewer behavioral seizures following TMEV infection. Therefore, both resident CNS cells and infiltrating cells are necessary for seizure development.


Assuntos
Sistema Nervoso Central/metabolismo , Encefalite Viral/complicações , Interleucina-6/fisiologia , Convulsões/etiologia , Theilovirus/isolamento & purificação , Animais , Anticorpos Monoclonais/administração & dosagem , Sistema Nervoso Central/patologia , Quimiocinas/fisiologia , Encefalite Viral/virologia , Imuno-Histoquímica , Interleucina-6/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Minociclina/administração & dosagem , Neutrófilos/fisiologia , Reação em Cadeia da Polimerase
17.
J Neurovirol ; 18(2): 127-37, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22403027

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) causes a demyelinating disease similar to multiple sclerosis in the central nervous system (CNS) of susceptible SJL/J mice. Immune responses to TMEV contribute to viral clearance as well as to demyelination. We constructed recombinant vaccinia viruses (VV) that encode each or all of the capsid proteins (VV(VP1), VV(VP2), VV(VP3), VV(VP4), and VV(all)) or non-structural proteins (VV(P2), VV(P2P3), and VV(3'P3)) of the Daniels strain of TMEV. To determine the role of each of the coding regions of TMEV in vivo, we immunized SJL/J mice with each recombinant VV, with or without subsequent TMEV infection. The groups of mice were compared clinically, immunologically, and histologically. No mice immunized with any recombinant VV without subsequent TMEV infection developed demyelination. However, antibody responses to TMEV were detected in mice immunized with VV(all). In addition, in some mice, VV(P2) immunization induced mild meningitis. VV(VP3) or VV(VP4) immunization of mice prior to TMEV infection ameliorated TMEV-induced pathology or clinical signs of disease. The beneficial effect of VP4 immunization was also seen through DNA immunization with a plasmid encoding VP4 and leader prior to TMEV infection. Therefore, vaccination against not only surface capsid proteins (VV(VP3) and VV(all)) but also non-surface capsid protein (VV(VP4)), and non-structural proteins (VV(P2)) can elicit immune responses to virus or modulate subsequent viral-induced CNS disease.


Assuntos
Proteínas do Capsídeo/imunologia , Sistema Nervoso Central/imunologia , Doenças Desmielinizantes/prevenção & controle , Theilovirus/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/administração & dosagem , Animais , Proteínas do Capsídeo/genética , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Feminino , Engenharia Genética , Imunização , Meningite Viral/etiologia , Meningite Viral/virologia , Camundongos , Fases de Leitura Aberta , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Theilovirus/imunologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Proteínas não Estruturais Virais/genética , Vacinas Virais/efeitos adversos , Vacinas Virais/uso terapêutico
18.
J Neurovirol ; 18(6): 471-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22991336

RESUMO

No one single pathogen has been identified as the causative agent of multiple sclerosis (MS). Alternately, the likelihood of an autoimmune event may be nonspecifically enhanced by different infectious agents. In a novel animal model of MS, SJL/J mice primed through infection with a recombinant vaccinia virus (VV) encoding myelin proteolipid protein (PLP) (VV(PLP)) were susceptible to a central nervous system (CNS) inflammatory disease following administration of a nonspecific immunostimulant [complete Freund's adjuvant (CFA) plus Bordetella pertussis (BP)]. Mononuclear cells isolated from the brains, but not the spleens, of VV(PLP)-primed CFA/BP challenged mice produced interleukin (IL)-17 and interferon-γ and transferred a CNS inflammatory disease to naïve SJL/J mice. Administration of curdlan, a T helper 17 cell inducer, unexpectedly resulted in less severe clinical and histological signs of disease, compared to CFA/BP challenged mice, despite the induction of IL-17 in the periphery. Further examination of the VV(PLP)-prime CFA/BP challenge model may suggest new mechanisms for how different pathogens associated with MS can protect or enhance disease.


Assuntos
Encéfalo/imunologia , Interleucina-17/imunologia , Leucócitos Mononucleares/imunologia , Esclerose Múltipla/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Vetores Genéticos , Imunomodulação , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-17/biossíntese , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Leucócitos Mononucleares/transplante , Camundongos , Esclerose Múltipla/metabolismo , Esclerose Múltipla/patologia , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/imunologia , Especificidade de Órgãos , Polissacarídeos Bacterianos/administração & dosagem , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Índice de Gravidade de Doença , Baço/imunologia , Baço/metabolismo , Baço/patologia , Vaccinia virus/genética , beta-Glucanas/administração & dosagem
19.
J Neurovirol ; 18(1): 45-54, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22281874

RESUMO

Major histocompatibility complex class I-restricted CD8(+) cytotoxic T lymphocytes are involved in the pathogenesis of multiple sclerosis (MS) and both autoimmune, experimental autoimmune encephalomyelitis, and viral, Theiler's murine encephalomyelitis virus (TMEV) infection, animal models of MS. Following TMEV infection, certain T cell hybridomas, generated from cloned TMEV-induced CD8(+) T cells, were able to produce clinical signs of disease (flaccid hind limb paralysis) upon adoptive transfer into naive mice. Dual T cell receptors (TCR) are present on the surface of these cells as both Vß3 and Vß6 were detected by polymerase chain reaction (PCR) screening and flow cytometry and multiple Vα mRNAs were detected by PCR screening. This is the first demonstration of antiviral CD8(+) T cells having more than one TCR initiating an autoimmune disease in the natural host of the virus. We hypothesize that this is a potential mechanism for virus-induced autoimmune disease initiated by CD8(+) T cells.


Assuntos
Infecções por Cardiovirus/imunologia , Encefalomielite Autoimune Experimental/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Linfócitos T Citotóxicos/imunologia , Theilovirus , Transferência Adotiva , Animais , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Encefalomielite Autoimune Experimental/virologia , Feminino , Citometria de Fluxo , Hibridomas/imunologia , Hibridomas/transplante , Hibridomas/virologia , Injeções Intravenosas , Camundongos , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Linfócitos T Citotóxicos/transplante , Linfócitos T Citotóxicos/virologia
20.
J Neurovirol ; 18(1): 30-44, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22328242

RESUMO

Viral infections of the central nervous system (CNS) are associated with an increased risk for seizures during the acute infection period and the subsequent development of chronic epilepsy that is often difficult to treat. In previous work, we have shown that mice of the C57BL/6 strain infected with Theiler's murine encephalomyelitis virus (TMEV) exhibit a similar sequence, thereby providing a potential useful model of virus-induced epilepsy. The present study examines spontaneous and miniature excitatory postsynaptic currents in CA3 pyramidal cells recorded from brain slices prepared during both the acute phase during encephalitis and 2 months following TMEV infection. Animals that develop chronic epilepsy following TMEV infection exhibit considerable hippocampal sclerosis, directly implicating this brain region in the process of epileptogenesis. There are significant increases in amplitude and frequency of spontaneous and miniature excitatory currents in CA3 cells recorded in brain slices prepared during the acute infection period and 2 months after infection. However, the patterns of changes observed are markedly different during these two periods, suggesting that there are underlying changes in the network over time. These differences have implications for the treatment used during the acute infection and after chronic seizures develop.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Encefalite/fisiopatologia , Epilepsia/fisiopatologia , Convulsões/fisiopatologia , Theilovirus , Doença Aguda , Animais , Região CA3 Hipocampal/virologia , Doença Crônica , Modelos Animais de Doenças , Encefalite/complicações , Encefalite/virologia , Epilepsia/complicações , Epilepsia/virologia , Potenciais Pós-Sinápticos Excitadores , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Convulsões/complicações , Convulsões/virologia , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA