Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Mult Scler Relat Disord ; 30: 257-261, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30851639

RESUMEN

BACKGROUND: Nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) is a master transcriptional regulator of the protective cellular response to oxidative stress. Sulforaphane is a Nrf2 activator but is unstable at ambient temperature. SFX-01 is a novel composition comprised of synthetic sulforaphane stabilised within the pocket of an α-cyclodextrin complex. Here we tested the efficacy of SFX-01 in murine relapsing experimental autoimmune encephalomyelitis (EAE), a model of relapsing-remitting MS (RRMS). METHODS: Relapsing EAE was induced in female SJL mice using immunization against PLP139-151. In the therapeutic experiment, the aim was to model initiation of treatment after diagnosis in RRMS, so treatment was started at day 19, one day prior to the expected relapse onset. In the prophylactic experiment, mice were treated from the time of immunization and followed for three weeks. RESULTS: SFX-01 reduced residual disability in both experiments. Most of this effect was mediated by a decrease in maximum severity of relapses and improved recovery during follow-up. Histological examination of the spinal cord was consistent with the clinical findings, with improvement in demyelination and the number of apoptotic cells, but not inflammatory cell infiltration, compared to the vehicle group. CONCLUSIONS: SFX-01 is efficacious in EAE. In first-in-man and phase II clinical trials for other indications, SFX-01 was found to be well-tolerated. A trial comparing BG-12 and SFX-01 would address whether SFX-01 can offer RRMS patients a better option with respect to efficacy and tolerability.


Asunto(s)
Anticarcinógenos/uso terapéutico , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Isotiocianatos/uso terapéutico , Resultado del Tratamiento , Animales , Evaluación de la Discapacidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Adyuvante de Freund/toxicidad , Estimación de Kaplan-Meier , Ratones , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Sulfóxidos , Factores de Tiempo
2.
Acta Neuropathol Commun ; 6(1): 86, 2018 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-30176931

RESUMEN

While hypoxic pre-conditioning protects against neurological disease the underlying mechanisms have yet to be fully defined. As chronic mild hypoxia (CMH, 10% O2) triggers profound vascular remodeling in the central nervous system (CNS), the goal of this study was to examine the protective potential of hypoxic pre-conditioning in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS) and then determine how CMH influences vascular integrity and the underlying cellular and molecular mechanisms during EAE. We found that mice exposed to CMH at the same time as EAE induction were strongly protected against the development of EAE progression, as assessed both at the clinical level and at the histopathological level by reduced levels of inflammatory leukocyte infiltration, vascular breakdown and demyelination. Mechanistically, our studies indicate that CMH protects, at least in part, by enhancing several properties of blood vessels that contribute to vascular integrity, including reduced expression of the endothelial activation molecules VCAM-1 and ICAM-1, maintained expression of endothelial tight junction proteins ZO-1 and occludin, and upregulated expression of the leukocyte inhibitory protein laminin-111 in the vascular basement membrane. Taken together, these data suggest that optimization of BBB integrity is an important mechanism underlying the protective effect of hypoxic pre-conditioning.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/terapia , Precondicionamiento Isquémico/métodos , Animales , Antígenos CD/metabolismo , Antígenos de Superficie/metabolismo , Colágeno Tipo IV/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Fibrinógeno/metabolismo , Adyuvante de Freund/toxicidad , Regulación de la Expresión Génica/fisiología , Laminina/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/toxicidad , Ocludina/metabolismo , Fragmentos de Péptidos/toxicidad , Molécula 1 de Adhesión Celular Vascular/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
3.
Nat Neurosci ; 21(10): 1392-1403, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30258239

RESUMEN

Multiple sclerosis (MS) patients exhibit neuropsychological symptoms in early disease despite the immune attack occurring predominantly in white matter and spinal cord. It is unclear why neurodegeneration may start early in the disease and is prominent in later stages. We assessed cortical microcircuit activity by employing spiking-specific two-photon Ca2+ imaging in proteolipid protein-immunized relapsing-remitting SJL/J mice in vivo. We identified the emergence of hyperactive cortical neurons in remission only, independent of direct immune-mediated damage and paralleled by elevated anxiety. High levels of neuronal activity were accompanied by increased caspase-3 expression. Cortical TNFα expression was mainly increased by excitatory neurons in remission; blockade with intraventricular infliximab restored AMPA spontaneous excitatory postsynaptic current frequencies, completely recovered normal neuronal network activity patterns and alleviated elevated anxiety. This suggests a dysregulation of cortical networks attempting to achieve functional compensation by synaptic plasticity mechanisms, indicating a link between immune attack and early start of neurodegeneration.


Asunto(s)
Corteza Cerebral/fisiopatología , Encefalomielitis Autoinmune Experimental/complicaciones , Encefalomielitis Autoinmune Experimental/patología , Hipercinesia/etiología , Recuperación de la Función/fisiología , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Carbazoles/uso terapéutico , Células Cultivadas , Corteza Cerebral/ultraestructura , Cuprizona/toxicidad , Modelos Animales de Enfermedad , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacocinética , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Adyuvante de Freund/toxicidad , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Transgénicos , Microglía/patología , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Quinoxalinas/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Tetrodotoxina/farmacología
4.
J Neuroinflammation ; 15(1): 225, 2018 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-30098594

RESUMEN

BACKGROUND: Multiple sclerosis (MS) is a chronic autoimmune disease of the central nervous system (CNS) for which several new treatment options were recently introduced. Among them is the monoclonal anti-CD52 antibody alemtuzumab that depletes mainly B cells and T cells in the immune periphery. Considering the ongoing controversy about the involvement of B cells and in particular the formation of B cell aggregates in the brains of progressive MS patients, an in-depth understanding of the effects of anti-CD52 antibody treatment on the B cell compartment in the CNS itself is desirable. METHODS: We used myelin basic protein (MBP)-proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) in C57BL/6 (B6) mice as B cell-dependent model of MS. Mice were treated intraperitoneally either at the peak of EAE or at 60 days after onset with 200 µg murine anti-CD52 vs. IgG2a isotype control antibody for five consecutive days. Disease was subsequently monitored for 10 days. The antigen-specific B cell/antibody response was measured by ELISPOT and ELISA. Effects on CNS infiltration and B cell aggregation were determined by immunohistochemistry. Neurodegeneration was evaluated by Luxol Fast Blue, SMI-32, and Olig2/APC staining as well as by electron microscopy and phosphorylated heavy neurofilament serum ELISA. RESULTS: Treatment with anti-CD52 antibody attenuated EAE only when administered at the peak of disease. While there was no effect on the production of MP4-specific IgG, the treatment almost completely depleted CNS infiltrates and B cell aggregates even when given as late as 60 days after onset. On the ultrastructural level, we observed significantly less axonal damage in the spinal cord and cerebellum in chronic EAE after anti-CD52 treatment. CONCLUSION: Anti-CD52 treatment abrogated B cell infiltration and disrupted existing B cell aggregates in the CNS.


Asunto(s)
Anticuerpos/uso terapéutico , Linfocitos B/patología , Antígeno CD52/inmunología , Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Antígenos CD/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Enfermedades Desmielinizantes/tratamiento farmacológico , Enfermedades Desmielinizantes/etiología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/complicaciones , Femenino , Citometría de Flujo , Adyuvante de Freund/toxicidad , Antígenos Comunes de Leucocito/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/toxicidad , Proteína Proteolipídica de la Mielina/toxicidad , Proteínas de Neurofilamentos/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Factores de Tiempo
5.
Curr Protoc Neurosci ; 81: 9.61.1-9.61.20, 2017 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-29058769

RESUMEN

In this unit, we describe in detail the most common methods used to break immunological tolerance for central myelin antigens and induce experimental autoimmune encephalomyelitis (EAE) in Lewis rats as an animal model of multiple sclerosis. The resulting disease course ranges from an acute monophasic disease to a chronic relapsing or chronic progressive course, which strongly resembles the human disease. These models enable the study of cellular and humoral autoimmunity against major antigenic epitopes of the myelin basic protein, myelin oligodendrocyte glycoprotein, or proteolipid protein. We provide an overview of common immunization protocols for induction of active and passive EAE, assessment and analysis of clinical score, preparation and purification of myelin basic protein, and derivation of neuroantigen-specific rat T cell lines. Finally, we describe the major clinical characteristics of these models. © 2017 by John Wiley & Sons, Inc.


Asunto(s)
Autoinmunidad/fisiología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple , Linfocitos T/patología , Animales , Linfocitos T CD4-Positivos/fisiología , Encefalomielitis Autoinmune Experimental/fisiopatología , Adyuvante de Freund/toxicidad , Cobayas , Metabolismo de los Lípidos , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/toxicidad , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/inmunología , Glicoproteína Mielina-Oligodendrócito/toxicidad , Examen Neurológico , Ratas , Ratas Endogámicas Lew , Médula Espinal/patología , Linfocitos T/inmunología
6.
J Neuroinflammation ; 14(1): 148, 2017 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-28738885

RESUMEN

BACKGROUND: MP4-induced experimental autoimmune encephalomyelitis (EAE) is a mouse model of multiple sclerosis (MS), which enables targeted research on B cells, currently much discussed protagonists in MS pathogenesis. Here, we used this model to study the impact of the S1P1 receptor modulator FTY720 (fingolimod) on the autoreactive B cell and antibody response both in the periphery and the central nervous system (CNS). METHODS: MP4-immunized mice were treated orally with FTY720 for 30 days at the peak of disease or 50 days after EAE onset. The subsequent disease course was monitored and the MP4-specific B cell/antibody response was measured by ELISPOT and ELISA. RNA sequencing was performed to determine any effects on B cell-relevant gene expression. S1P1 receptor expression by peripheral T and B cells, B cell subset distribution in the spleen and B cell infiltration into the CNS were studied by flow cytometry. The formation of B cell aggregates and of tertiary lymphoid organs (TLOs) was evaluated by histology and immunohistochemistry. Potential direct effects of FTY720 on B cell aggregation were studied in vitro. RESULTS: FTY720 significantly attenuated clinical EAE when treatment was initiated at the peak of EAE. While there was a significant reduction in the number of T cells in the blood after FTY720 treatment, B cells were only slightly diminished. Yet, there was evidence for the modulation of B cell receptor-mediated signaling upon FTY720 treatment. In addition, we detected a significant increase in the percentage of B220+ B cells in the spleen both in acute and chronic EAE. Whereas acute treatment completely abrogated B cell aggregate formation in the CNS, the numbers of infiltrating B cells and plasma cells were comparable between vehicle- and FTY720-treated mice. In addition, there was no effect on already developed aggregates in chronic EAE. In vitro B cell aggregation assays suggested the absence of a direct effect of FTY720 on B cell aggregation. However, FTY720 impacted the evolution of B cell aggregates into TLOs. CONCLUSIONS: The data suggest differential effects of FTY720 on the B cell compartment in MP4-induced EAE.


Asunto(s)
Linfocitos B/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Clorhidrato de Fingolimod/uso terapéutico , Inmunosupresores/uso terapéutico , Animales , Antígenos CD19/metabolismo , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/patología , Proteínas de Unión al Calcio/metabolismo , Agregación Celular/efectos de los fármacos , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ensayo de Immunospot Ligado a Enzimas , Femenino , Citometría de Flujo , Adyuvante de Freund/toxicidad , Ganglios Linfáticos/patología , Ratones , Proteína Básica de Mielina/inmunología , Proteína Básica de Mielina/toxicidad , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Bazo/patología , Factores de Tiempo
7.
J Neuroinflammation ; 14(1): 152, 2017 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-28754118

RESUMEN

BACKGROUND: In multiple sclerosis, coagulation factors have been shown to modulate inflammation. In this translational study, we investigated whether long-term anticoagulation with warfarin or rivaroxaban has beneficial effects on the course of autoimmune experimental encephalomyelitis (EAE). METHODS: Female SJL/J mice treated with anticoagulants namely warfarin or rivaroxaban were immunized with PLP139-151. Stable anticoagulation was maintained throughout the entire experiment. Mice without anticoagulation treated with the vehicle only were used as controls. The neurological deficit was recorded during the course of EAE, and histopathological analyses of inflammatory lesions were performed. RESULTS: In preventive settings, both treatment with warfarin and rivaroxaban reduced the maximum EAE score as compared to the control group and led to a reduction of inflammatory lesions in the spinal cord. In contrast, therapeutic treatment with warfarin had no beneficial effects on the clinical course of EAE. Signs of intraparenchymal hemorrhage at the site of the inflammatory lesions were not observed. CONCLUSION: We developed long-term anticoagulation models that allowed exploring the course of EAE under warfarin and rivaroxaban treatment. We found a mild preventive effect of both warfarin and rivaroxaban on neurological deficits and local inflammation, indicating a modulation of the disease induction by anticoagulation.


Asunto(s)
Anticoagulantes/uso terapéutico , Encefalomielitis Autoinmune Experimental/prevención & control , Rivaroxabán/uso terapéutico , Warfarina/uso terapéutico , Animales , Encéfalo/anatomía & histología , Modelos Animales de Enfermedad , Impedancia Eléctrica , Encefalomielitis Autoinmune Experimental/sangre , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Células Endoteliales/efectos de los fármacos , Adyuvante de Freund/toxicidad , Ratones , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Distribución Aleatoria , Rivaroxabán/sangre , Porcinos , Trombina/metabolismo , Factores de Tiempo
8.
J Neuroimmunol ; 307: 18-26, 2017 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-28495132

RESUMEN

Altered peptide ligands (APLs) have routinely been studied in clonal populations of Th cells that express a single T cell receptor (TCR), but results generated in this manner poorly predict the effects of APLs on polyclonal Th cells in vivo, contributing to the failure of phase II clinical trials of APLs in autoimmune diseases such as multiple sclerosis (MS). We have used a panel of APLs derived from an encephalitogenic epitope of myelin proteolipid protein to investigate the relationship between antigen cross-reactivity in a polyclonal environment, encephalitogenicity, and the capacity of an APL to provide protection against experimental autoimmune encephalomyelitis (EAE) in SJL mice. In general, polyclonal Th cell lines specific for encephalitogenic APLs cross-reacted with other encephalitogenic APLs, but not with non-encephalitogenic APLs, and vice versa. This, alongside analysis of TCR Vß usage, suggested that encephalitogenic and non-encephalitogenic subgroups of APLs expand largely non-cross-reactive Th cell populations. As an exception to the rule, one non-encephalitogenic APL, L188, induced proliferation in polyclonal CD4+ T cells specific for the native encephalitogen, with minimal induction of cytokine production. Co-immunization of L188 alongside the native encephalitogen slightly enhanced disease development. In contrast, another APL, A188, which induced IL-10 production without proliferation in CD4+ T cells specific for the native encephalitogen, was able to protect against development of EAE in a dose-dependent fashion when co-immunized alongside the native encephalitogen. These results suggest that testing against polyclonal Th cell lines in vitro may be an effective strategy for distinguishing between potentially therapeutic and non-therapeutic APLs.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/efectos de los fármacos , Células TH1/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Proliferación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Epítopos/química , Epítopos/inmunología , Femenino , Citometría de Flujo , Adyuvante de Freund/toxicidad , Ratones , Proteína Proteolipídica de la Mielina/química , Proteína Proteolipídica de la Mielina/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , ARN Mensajero , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/inmunología
9.
CNS Neurol Disord Drug Targets ; 16(6): 714-723, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28240190

RESUMEN

BACKGROUND: In April 2015, the US Food and Drug Administration approved the first generic glatiramer acetate, Glatopa® (M356), as fully substitutable for Copaxone® 20 mg/mL for relapsing forms of multiple sclerosis (MS). This approval was accomplished through an Abbreviated New Drug Application that demonstrated equivalence to Copaxone. METHOD: This article will provide an overview of the methods used to establish the biological and immunological equivalence of the two glatiramer acetate products, including methods evaluating antigenpresenting cell (APC) biology, T-cell biology, and other immunomodulatory effects. RESULTS: In vitro and in vivo experiments from multiple redundant orthogonal assays within four biological processes (aggregate biology, APC biology, T-cell biology, and B-cell biology) modulated by glatiramer acetate in MS established the biological and immunological equivalence of Glatopa and Copaxone and are described. The following were observed when comparing Glatopa and Copaxone in these experiments: equivalent delays in symptom onset and reductions in "disease" intensity in experimental autoimmune encephalomyelitis; equivalent dose-dependent increases in Glatopa- and Copaxone- induced monokine-induced interferon-gamma release from THP-1 cells; a shift to a T helper 2 phenotype resulting in the secretion of interleukin (IL)-4 and downregulation of IL-17 release; no differences in immunogenicity and the presence of equivalent "immunofingerprints" between both versions of glatiramer acetate; and no stimulation of histamine release with either glatiramer acetate in basophilic leukemia 2H3 cell lines. CONCLUSION: In summary, this comprehensive approach across different biological and immunological pathways modulated by glatiramer acetate consistently supported the biological and immunological equivalence of Glatopa and Copaxone.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Acetato de Glatiramer/uso terapéutico , Inmunosupresores/uso terapéutico , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Histamina/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Linfocitos T/efectos de los fármacos , Equivalencia Terapéutica
10.
J Neuroimmunol ; 301: 7-11, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27836179

RESUMEN

The lack of biomarkers is a major obstacle for investigating myelin repair. We used metabolic incorporation of the choline analog - propargyl-choline (P-Cho) to label and visualize newly synthesized myelin in the CNS of mice induced with experimental autoimmune encephalomyelitis (EAE). We further developed unbiased colocalization analysis to quantify P-Cho incorporation specifically into the myelin. Our findings indicate that P-Cho injection to mice recovering from EAE, either spontaneously or following glatiramer acetate treatment, results in significant elevation of its incorporation into the myelin, offering a novel strategy for assessing remyelination in animal models and the remyelination potential of candidate drugs.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Vaina de Mielina/metabolismo , Recuperación de la Función/fisiología , Médula Espinal/patología , Alquinos/metabolismo , Animales , Nucléolo Celular/metabolismo , Nucléolo Celular/patología , Colina/análogos & derivados , Colina/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/diagnóstico por imagen , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Acetato de Glatiramer/uso terapéutico , Inmunosupresores/uso terapéutico , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Microscopía Inmunoelectrónica , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/toxicidad , Vaina de Mielina/ultraestructura , Fragmentos de Péptidos/toxicidad , Recuperación de la Función/efectos de los fármacos , Médula Espinal/ultraestructura
11.
J Neurosci ; 36(35): 9253-66, 2016 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-27581464

RESUMEN

UNLABELLED: T-helper 17 (Th17) cells play an important role in the pathogenesis of multiple sclerosis (MS), an autoimmune demyelinating disease that affects the CNS. In the present study, MicroRNA sequencing (miRNA-seq) was performed in mouse Th0 and Th17 cells to determine the critical miRNAs that are related to Th17 differentiation. We found that miR-30a was significantly downregulated during mouse Th17 differentiation. In addition, the level of miR-30a in CD4(+) T cells from peripheral blood of MS patients and experimental autoimmune encephalomyelitis (EAE) animal models was also decreased and inversely correlated with the expression of interleukin 17a, the canonical cytokine of Th17 cells. Moreover, overexpression of miR-30a inhibited Th17 differentiation and prevented the full development of EAE, whereas interference of miR-30a promoted Th17 differentiation. Mechanism studies showed that miR-30a reduced IRF4 expression by specifically binding with the 3'-untranslated region. Through screening of 640 different Food and Drug Administration (FDA)-approved drugs, we found that disulfiram and diphenhydramine hydrochloride were effective candidates for inhibiting Th17 differentiation and ameliorating EAE development through upregulating miR-30a. To our knowledge, the present work is not only the first miRNA-seq study focusing on Th17 differentiation, but also the first chemical screening for FDA-approved drugs that inhibit Th17 differentiation through regulating miRNA expression. SIGNIFICANCE STATEMENT: The present work is the first miRNA sequencing (miRNA-seq) study focusing on T-helper 17 (Th17) differentiation. By miRNA deep sequencing, we found that miR-30a was downregulated during Th17 differentiation. miR-30a was also decreased in CD4(+) T cells from multiple sclerosis patients and experimental autoimmune encephalomyelitis (EAE) mice. miR-30a reduced IRF4 expression by specific binding with the 3'-untranslated region and thus suppressed Th17 differentiation and prevented the full development of EAE. Interestingly, by performing a chemical screen with Food and Drug Administration-approved small-molecule drugs, we found that disulfiram and diphenhydramine upregulated miR-30a and suppressed Th17-associated autoimmune demyelination.


Asunto(s)
Difenhidramina/farmacología , Disulfiram/farmacología , Encefalomielitis Autoinmune Experimental/metabolismo , Interleucina-17/metabolismo , MicroARNs/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Estudios de Casos y Controles , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/prevención & control , Femenino , Células HEK293 , Humanos , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Proteolipídica de la Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Estadísticas no Paramétricas , Transfección
12.
J Neurosci Res ; 94(10): 907-14, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27312674

RESUMEN

Previous research has examined the effects of exercise in experimental autoimmune encephalomyelitis (EAE), the animal model of multiple sclerosis. However, all previous studies have utilized a chronic model of EAE, with exercise delivered prior to or immediately after induction of EAE. To our knowledge, no study has examined the effects of exercise delivered during a remission period after initial disease onset in a relapsing-remitting model of EAE (RR-EAE). The current study examines the effects of both voluntary wheel running and forced treadmill exercise on clinical disability and hippocampal brain-derived neurotrophic factor (BDNF) in SJL mice with RR-EAE. The results demonstrate no significant effects of exercise delivered during remission after initial disease onset on clinical disability scores or levels of hippocampal BDNF in mice with RR-EAE. Furthermore, our results demonstrate no significant increase in citrate synthase activity in the gastrocnemius and soleus muscles of mice in the running wheel or treadmill conditions compared with the sedentary condition. These results suggest that the exercise stimuli might have been insufficient to elicit differences in clinical disability or hippocampal BDNF among treatment conditions. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/fisiopatología , Condicionamiento Físico Animal/fisiología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citrato (si)-Sintasa/metabolismo , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Prueba de Esfuerzo , Femenino , Adyuvante de Freund/toxicidad , Hipocampo/metabolismo , Ratones , Actividad Motora/fisiología , Esclerosis Múltiple/inducido químicamente , Esclerosis Múltiple/patología , Esclerosis Múltiple/fisiopatología , Músculos/patología , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Tamaño de los Órganos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Distribución Aleatoria , Recurrencia , Carrera/fisiología , Médula Espinal/metabolismo , Médula Espinal/patología
13.
J Neuroinflammation ; 13: 48, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26920550

RESUMEN

BACKGROUND: Conditions of inflammatory tissue distress are associated with high extracellular levels of adenosine, due to increased adenosine triphosphate (ATP) degradation upon cellular stress or the release of extracellular ATP upon cell death, which can be degraded to adenosine by membrane-bound ecto-enzymes like CD39 and CD73. Adenosine is recognised to mediate anti-inflammatory effects via the adenosine A2a receptor (A2aR), as shown in experimental models of arthritis. Here, using pharmacological interventions and genetic inactivation, we investigated the roles of A2aR in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). METHODS: We used two independent mouse EAE variants, i.e. active immunization in C57BL/6 with myelin oligodendrocyte glycoprotein (MOG)35-55 or transfer-EAE by proteolipid protein (PLP)139-155-stimulated T lymphocytes and EAE in mice treated with A2aR-agonist CGS21680 at different stages of disease course and in mice lacking A2aR (A2aR(-/-)) compared to direct wild-type littermates. In EAE, we analysed myelin-specific proliferation and cytokine synthesis ex vivo, as well as inflammation and demyelination by immunohistochemistry. In vitro, we investigated the effect of A2aR on migration of CD4(+) T cells, macrophages and microglia, as well as the impact of A2aR on phagocytosis of macrophages and microglia. Statistical tests were Mann-Whitney U and Student's t test. RESULTS: We found an upregulation of A2aR in the central nervous system (CNS) in EAE, predominantly detected on T cells and macrophages/microglia within the inflamed tissue. Preventive EAE treatment with A2aR-specific agonist inhibited myelin-specific T cell proliferation ex vivo and ameliorated disease, while application of the same agonist after disease onset exacerbated non-remitting EAE progression and resulted in more severe tissue destruction. Accordingly, A2aR-deficient mice showed accelerated and exacerbated disease manifestation with increased frequencies of IFN-γ-, IL-17- and GM-CSF-producing CD4(+) T helper cells and higher numbers of inflammatory lesions in the early stage. However, EAE quickly ameliorated and myelin debris accumulation was lower in A2aR(-/-) mice. In vitro, activation of A2aR inhibited phagocytosis of myelin by macrophages and primary microglia as well as migration of CD4(+) T cells, macrophages and primary microglia. CONCLUSIONS: A2aR activation exerts a complex pattern in chronic autoimmune neurodegeneration: while providing anti-inflammatory effects on T cells and thus protection at early stages, A2aR seems to play a detrimental role during later stages of disease and may thus contribute to sustained tissue damage within the inflamed CNS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Receptor de Adenosina A2A/metabolismo , Adenosina/análogos & derivados , Adenosina/uso terapéutico , Animales , Antiinflamatorios/uso terapéutico , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Microglía/metabolismo , Proteína Proteolipídica de la Mielina/toxicidad , Glicoproteína Mielina-Oligodendrócito/toxicidad , Fragmentos de Péptidos/toxicidad , Fagocitosis/efectos de los fármacos , Fagocitosis/genética , Fenetilaminas/uso terapéutico , Receptor de Adenosina A2A/genética , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
14.
J Neuroimmunol ; 291: 59-69, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26857496

RESUMEN

IL-12Rß2 is a common receptor subunit of heterodimeric receptors for IL-12 and IL-35, two cytokines that are implicated in immunopathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. We evaluated the role of IL-12Rß2 in relapsing-remitting EAE (RR-EAE). IL-12Rß2-deficient SJL/J mice developed markedly more severe clinical EAE, and had greater mortality and more severe relapses compared with wild-type controls. IL-12Rß2-deficient EAE mice also had more infiltrating mononuclear cells in the CNS, as well as higher T cell proliferative capacity and decreased IFN-γ production at the periphery. These findings demonstrate a protective role of IL-12Rß2 in RR-EAE.


Asunto(s)
Sistema Nervioso Central/patología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/prevención & control , Receptores de Interleucina-12/deficiencia , Animales , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Citometría de Flujo , Adyuvante de Freund , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Proteolipídica de la Mielina/toxicidad , Infiltración Neutrófila/genética , Fragmentos de Péptidos/toxicidad , Receptores de Interleucina-12/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo
15.
J Neuroinflammation ; 13: 7, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26747276

RESUMEN

BACKGROUND: Demyelination and axonal degeneration, hallmarks of multiple sclerosis (MS), are associated with the central nervous system (CNS) inflammation facilitated by C-X-C motif chemokine 12 (CXCL12) chemokine. Both in MS and in experimental autoimmune encephalomyelitis (EAE), the deleterious CNS inflammation has been associated with upregulation of CXCL12 expression in the CNS. We investigated the expression dynamics of CXCL12 in the CNS with progression of clinical EAE and following spontaneous recovery, with a focus on CXCL12 expression in the hippocampal neurogenic dentate gyrus (DG) and in the corpus callosum (CC) of spontaneously recovered mice, and its potential role in promoting the endogenous myelin/neuronal repair capacity. METHODS: CNS tissue sections from mice with different clinical EAE phases or following spontaneous recovery and in vitro differentiated adult neural stem cell cultures were analyzed by immunofluorescent staining and confocal imaging for detecting and enumerating neuronal progenitor cells (NPCs) and oligodendrocyte precursor cells (OPCs) and for expression of CXCL12. RESULTS: Our expression dynamics analysis of CXCL12 in the CNS with EAE progression revealed elevated CXCL12 expression in the DG and CC, which persistently increases following spontaneous recovery even though CNS inflammation has subsided. Correspondingly, the numbers of NPCs and OPCs in the DG and CC, respectively, of EAE-recovered mice increased compared to that of naïve mice (NPCs, p < 0.0001; OPCs, p < 0.00001) or mice with active disease (OPCs, p < 0.0005). Notably, about 30 % of the NPCs and unexpectedly also OPCs (~50 %) express CXCL12, and their numbers in DG and CC, respectively, are higher in EAE-recovered mice compared with naïve mice and also compared with mice with ongoing clinical EAE (CXCL12(+) NPCs, p < 0.005; CXCL12(+) OPCs, p < 0.0005). Moreover, a significant proportion (>20 %) of the CXCL12(+) NPCs and OPCs co-express the CXCL12 receptor, CXCR4, and their numbers significantly increase with recovery from EAE not only relative to naïve mice (p < 0.0002) but also to mice with ongoing EAE (p < 0.004). CONCLUSIONS: These data link CXCL12 expression in the DG and CC of EAE-recovering mice to the promotion of neuro/oligodendrogenesis generating CXCR4(+) CXCL12(+) neuronal and oligodendrocyte progenitor cells endowed with intrinsic neuro/oligondendroglial differentiation potential. These findings highlight the post-CNS-inflammation role of CXCL12 in augmenting the endogenous myelin/neuronal repair capacity in MS-like disease, likely via CXCL12/CXCR4 autocrine signaling.


Asunto(s)
Sistema Nervioso Central/metabolismo , Quimiocina CXCL12/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Vaina de Mielina/metabolismo , Recuperación de la Función/fisiología , Animales , Recuento de Células , Diferenciación Celular , Células Cultivadas , Ventrículos Cerebrales/citología , Quimiocina CXCL12/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteínas de Dominio Doblecortina , Encefalomielitis Autoinmune Experimental/inducido químicamente , Regulación de la Expresión Génica/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Proteína Proteolipídica de la Mielina/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Células-Madre Neurales/metabolismo , Neuronas/patología , Neuropéptidos/metabolismo , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/toxicidad , Cicatrización de Heridas/fisiología
16.
Neurobiol Dis ; 88: 85-95, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26805386

RESUMEN

Multiple sclerosis (MS) is a demyelinating disease of the central nervous system (CNS). No effective remyelination therapies are in use. We hypothesized that thymosin beta4 (Tß4) is an effective remyelination treatment by promoting differentiation of oligodendrocyte progenitor cells (OPCs), and that the epidermal growth factor receptor (EGFR) signaling pathway contributes to this process. Two demyelination animal models were employed in this study: 1) experimental autoimmune encephalomyelitis (EAE), an animal model of MS. EAE mice were treated daily for 30days, with Tß4 or saline treatment initiated on the day of EAE onset; and 2) cuprizone diet model, a non-inflammatory demyelination model. The mice were treated daily for 4weeks with Tß4 or saline after fed a cuprizone diet for 5weeks. Immunofluorescent staining and Western blot were performed to measure the differentiation of OPCs, myelin and axons, respectively. To obtain insight into mechanisms of action, the expression and activation of the EGFR pathway was measured. AG1478, an EGFR inhibitor, was employed in a loss-of-function study. Data revealed that animals in both demyelination models exhibited significant reduction of myelin basic protein (MBP(+)) levels and CNPase(+) oligodendrocytes. Treatment of EAE mice with Tß4 significantly improved neurological outcome. Double immunofluorescent staining showed that Tß4 significantly increased the number of newly generated oligodendrocytes identified by BrdU(+)/CNPase(+) cells and MBP(+) mature oligodendrocytes, and reduced axonal damage in the EAE mice compared with the saline treatment. The newly generated mature oligodendrocytes remyelinated axons, and the increased mature oligodendrocytes significantly correlated with functional improvement (r=0.73, p<0.05). Western blot analysis revealed that Tß4 treatment increased expression and activation of the EGFR pathway. In the cuprizone demyelination model, Tß4 treatment was confirmed that significantly increased OPC differentiation and remyelination, and increased the expression of EGFR and activated the EGFR pathway in the demyelinating corpus callosum. In cultured OPCs, blockage of the activation of the EGFR pathway with AG1478 abolished the Tß4-increased OPC differentiation. Collectively, these findings indicate that: 1) Tß4 increases proliferation of OPCs and the maturation of OPCs to myelinating oligodendrocytes which in concert, likely contribute to the beneficial effect of Tß4 on EAE, 2) EGFR upregulated and activated by Tß4 may mediate the process of OPC differentiation, and 3) Tß4 could potentially be developed as a therapy for MS patients, and for other demyelinating neurological disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Oligodendroglía/efectos de los fármacos , Timosina/uso terapéutico , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quelantes/toxicidad , Cuprizona/toxicidad , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Inhibidores Enzimáticos/farmacología , Receptores ErbB/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Proteoglicanos/metabolismo , Quinazolinas/farmacología , Células Madre/efectos de los fármacos , Factores de Tiempo , Tirfostinos/farmacología
17.
J Neuroimmunol ; 287: 36-42, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26439960

RESUMEN

Gelsolin is the fourth most abundant protein in the body and its depletion in the blood has been found in multiple sclerosis (MS) patients. How gelsolin affects the MS brain has not been studied. We found that while the secreted form of gelsolin (pGSN) decreased in the blood of experimental autoimmune encephalomyelitis (EAE) mice, pGSN concentration increased in the EAE brain. Recombinant human pGSN (rhp-GSN) decreased extracellular actin and myeloperoxidase activity in the brain, resulting in reduced disease activity and less severe clinical disease, suggesting that gelsolin could be a potential therapeutic target for MS.


Asunto(s)
Actinas/toxicidad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/metabolismo , Gelsolina/metabolismo , Gelsolina/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Actinas/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Adyuvante de Freund/toxicidad , Glioma/patología , Humanos , Ratones , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Mycobacterium tuberculosis/inmunología , Proteína Proteolipídica de la Mielina/toxicidad , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fragmentos de Péptidos/toxicidad , Peroxidasa/metabolismo , Factores de Tiempo
18.
Exp Neurol ; 273: 234-42, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342755

RESUMEN

Toll-like receptor 2 (TLR2) is expressed on immune cells in the periphery and the CNS and mediates both innate and adaptive immune responses. Recent studies have implicated TLR2 in systemic pathogenesis of adaptive immunity in experimental autoimmune encephalomyelitis (EAE). In addition, TLR2 is expressed on oligodendrocyte progenitor cells and its activation inhibits their differentiation and myelination. We investigated the roles of CNS TLR2 activation in mediating neuro-inflammatory responses in intact versus EAE animals. We examined the effects of intra-cerebro-ventricular (ICV) injection of Zymosan, a TLR2 agonist, on naive versus EAE animals. The neuro-inflammatory response was characterized by immune-fluorescent staining for IBA-1+ microglia/macrophages and CD3+ T cells, and by semi-quantitative real time PCR for TLR2 and immune cytokines. The nature of the immune cells isolated from EAE brain tissue was assessed by their proliferative response to the PLP peptide autoantigen. Survival and clinical scores were monitored; demyelination and axonal loss were quantified by Gold-Black and Bielschowsky stains. Our findings showed that Zymosan injection in naïve mice induced a massive neuro-inflammatory response without any clinical manifestations. In EAE mice, ICV Zymosan induced a severe acute toxic response with 80% mortality. Surviving animals returned to pre-injection clinical score, and their course of disease was not altered as compared to control EAE group. Demyelination and axonal loss were not affected by ICV Zymosan injection. Quantification of immune response in the brain by real time PCR, immunofluorescent stains and proliferative response to PLP peptide suggested that TLR2 activation induces innate but not adaptive immune response. We conclude that EAE mice are hypersensitive to CNS TLR2 activation with a severe toxic response. This might represent the susceptibility of multiple sclerosis patients to even trivial infections. As CNS TLR2 activation does not alter the clinical and pathological course of EAE, it implies that CNS TLR2 activation affects the innate but not adaptive brain immune responses.


Asunto(s)
Inmunidad Adaptativa/fisiología , Sistema Nervioso Central , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Inmunidad Innata/fisiología , Receptor Toll-Like 2/metabolismo , Inmunidad Adaptativa/efectos de los fármacos , Animales , Complejo CD3/metabolismo , Proliferación Celular/efectos de los fármacos , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Citocinas/genética , Modelos Animales de Enfermedad , Encefalitis/inducido químicamente , Encefalitis/inmunología , Encefalitis/metabolismo , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Inmunidad Innata/efectos de los fármacos , Inyecciones Intraventriculares , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Microglía/metabolismo , Microglía/patología , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , ARN Mensajero/metabolismo , Linfocitos T/metabolismo , Linfocitos T/patología , Factores de Tiempo , Receptor Toll-Like 2/agonistas , Zimosan/toxicidad
19.
J Neuroimmunol ; 279: 71-4, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25670003

RESUMEN

Multiple sclerosis (MS) is widely viewed as a prototypic human autoimmune disease involving proinflammatory T cells that induce lesions in the central nervous system (CNS) in response to myelin self proteins. Although the impact of sex hormones on MS is well recognized, the converse effects of autoimmunity on sex hormones are still unclear. The current study was designed to assess the impact of CNS autoimmunity on female reproductive physiology. In order to identify subtle hormonal disturbances as a result of autoimmunity, we analyzed the estrous cycle in SJL/J mice after active induction of experimental autoimmune encephalomyelitis (EAE), an animal model with substantial similarities to MS. Here we show that CNS autoimmunity significantly shortens the murine estrous cycle. This shortening of the estrous cycle is characterized by a dramatic decrease in the length of the metestrus-diestrus luteal phase partially offset by a highly significant but less dramatic elongation of the proestrus-estrus follicular phase of the uterine cycle. Thus, our study provides experimental evidence for a direct causal link between CNS autoimmunity and disruption of the homeostatic balance of the uterine cycle often observed in women with MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/fisiopatología , Ciclo Estral/fisiología , Homeostasis/fisiología , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Ciclo Estral/efectos de los fármacos , Ciclo Estral/inmunología , Femenino , Homeostasis/efectos de los fármacos , Homeostasis/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad
20.
J Neuroimmunol ; 278: 112-22, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25595260

RESUMEN

Inflammation in the meninges, tissues surrounding the brain and spinal cord that enclose the cerebrospinal fluid, closely parallels clinical exacerbations in relapsing-remitting experimental autoimmune encephalomyelitis (EAE). In preclinical disease, an influx of innate immune cells precedes loss of blood brain barrier (BBB) integrity and large-scale inflammation in the central nervous system (CNS). T cell infiltration into the meninges is observed in acute disease as well as during relapse, when neither BBB permeability nor significant increases in peripherally-derived immune cell numbers in the CNS are observed. These findings support the idea that the meninges are a gateway for immune cell access into the CNS, a finding that has important therapeutic implications.


Asunto(s)
Sistema Nervioso Central/patología , Meninges/patología , Meningitis/complicaciones , Esclerosis Múltiple Recurrente-Remitente/complicaciones , Animales , Barrera Hematoencefálica/fisiopatología , Sistema Nervioso Central/inmunología , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Adyuvante de Freund/toxicidad , Expresión Génica/efectos de los fármacos , Meningitis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Esclerosis Múltiple Recurrente-Remitente/inducido químicamente , Proteína Proteolipídica de la Mielina/toxicidad , Fragmentos de Péptidos/toxicidad , Permeabilidad , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Linfocitos T/metabolismo , Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA