Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 8(8)2019 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-31362466

RESUMEN

NK cells (natural killer cells) being a part of the innate immune system have been shown to be involved in immunoregulation of autoimmune diseases. Previously we have shown that HINT1/Hsp70 treatment induced regulatory NK cells ameliorating experimental autoimmune encephalomyelitis (EAE) course and CD4+ T cells proliferation. NK cells were isolated from mice treated with HINT1/Hsp70 and co-cultured with proteolipid protein (PLP)-stimulated CD4+ T cells isolated from EAE mice. Cell proliferation was assessed by thymidine uptake, cytotoxicity by lactate dehydrogenase (LDH) release assay and fluorescence activated cell sorting (FACS) analysis, protein expression by Western blot, mRNA by quantitative RT-PCR. Gene related to anergy in lymphocytes (GRAIL) expression was downregulated by specific siRNA and GRAIL overexpression was induced by pcDNA-GRAIL transfection. HINT1/Hsp70 pretreatment of EAE SJL/J mice ameliorated EAE course, suppressed PLP-induced T cell proliferation by enhancing T cell expression of GRAIL as GRAIL downregulation restored T cell proliferation. HINT1/Hsp70 treatment induced immunoregulatory NK cells which inhibited PLP-stimulated T cell proliferation not depending on T cell necrosis and apoptosis. This immunoregulatory NK cell function depended on NK cell expression of GRAIL as GRAIL downregulation diminished inhibition of NK cell suppression of T cell proliferation. Similarly GRAIL overexpression in NK cells induced their regulatory function. HINT1/Hsp70 treatment generated regulatory NK cells characterized by expression of GRAIL.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Encefalomielitis Autoinmune Experimental/inmunología , Proteínas HSP70 de Choque Térmico/administración & dosificación , Células Asesinas Naturales/citología , Proteínas del Tejido Nervioso/administración & dosificación , Ubiquitina-Proteína Ligasas/genética , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Anergia Clonal , Técnicas de Cocultivo , Encefalomielitis Autoinmune Experimental/etiología , Femenino , Proteínas HSP70 de Choque Térmico/farmacología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/metabolismo , Proteínas del Tejido Nervioso/farmacología , Proteolípidos/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
2.
Bioorg Med Chem ; 26(9): 2221-2228, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29681483

RESUMEN

We report the novel synthesis of cyclic PLP139-151 (cPLP) and its application in SJL/J mice to study its encephalitogenic effects. Our results indicate that the cPLP analog is minimally encephalitogenic when administered to induce experimental autoimmune encephalomyelitis (low disease burden, minimal inflammatory, demyelinating and axonopathic pathology compared to its linear counterpart). Proliferation assays confirmed the low stimulatory potential of the cPLP compared to linPLP (2.5-fold lower proliferation) as well as inducing lower antibody responses. Molecular modeling showed a completely different TCR recognition profile of cPLP in regard to linPLP, where H147 replaces W144 and F151-K150 replace H147 as TCR contacts, which may explain the difference on each peptide's response.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Proteína Proteolipídica de la Mielina/inmunología , Fragmentos de Péptidos/inmunología , Péptidos Cíclicos/inmunología , Secuencia de Aminoácidos , Animales , Ciclización , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/patología , Epítopos/efectos adversos , Epítopos/química , Epítopos/inmunología , Epítopos/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Ratones Endogámicos , Simulación del Acoplamiento Molecular , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/química , Proteína Proteolipídica de la Mielina/metabolismo , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Péptidos Cíclicos/efectos adversos , Péptidos Cíclicos/síntesis química , Péptidos Cíclicos/metabolismo , Unión Proteica , Conformación Proteica , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T/inmunología , Médula Espinal/patología , Linfocitos T/metabolismo
3.
J Neuroimmunol ; 313: 77-81, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29153612

RESUMEN

In patients with multiple sclerosis, the selective serotonin reuptake inhibitor, fluoxetine, resulted in less acute disease activity. We tested the immune modulating effects of fluoxetine in a mouse model of multiple sclerosis, i.e. experimental autoimmune encephalomyelitis (EAE). We show that fluoxetine delayed the onset of disease and reduced clinical paralysis in mice with established disease. Fluoxetine had abrogating effects on proliferation of immune cells and inflammatory cytokine production by both antigen-presenting cells and T cells. Specifically, in CD4 T cells, fluoxetine increased Fas-induced apoptosis. We conclude that fluoxetine possesses immune-modulating effects resulting in the amelioration of symptoms in EAE.


Asunto(s)
Antiinflamatorios/uso terapéutico , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/prevención & control , Fluoxetina/uso terapéutico , Animales , Apoptosis , Antígeno CD11b/metabolismo , Muerte Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Adyuvante de Freund/efectos adversos , Adyuvante de Freund/inmunología , Macrófagos/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Transgénicos , Proteína Básica de Mielina/genética , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/inmunología , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Receptor fas/metabolismo
4.
Autoimmunity ; 48(4): 208-21, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25427822

RESUMEN

The regulation of cytokine expression by immune deviation from a pro-inflammatory to anti-inflammatory or "regulatory" milieu is crucial to the prevention of permanent central nervous system (CNS) damage in neuroinflammation. Earlier studies in the murine experimental autoimmune encephalomyelitis (EAE) model pointed to an anti-inflammatory role for the Th2 cytokine, IL-4, which was not confirmed in IL-4Rα-deficient mice (IL-4Rα(-/-)). To examine the pathological consequences of loss of responsiveness to Th2 cytokines, we compared lesion evolution in IL-4Rα(-/-) and wild type (WT) BALB/c mice immunized with PLP180-199 and investigated how altering the magnitude of the antigen-specific autoimmune response in this model affected the pathology. We found that while changing the magnitude of the peripheral antigen-specific response differentially affected the incidence of clinical disease in WT BALB/c relative to IL-4Rα(-/-) mice, the differences in incidence did not correlate to differences in pro-inflammatory cytokine production. Additionally, although only approximately 75% of WT mice developed clinical disease, lesions were observed in 100% of the mice, principally in the cerebellum, mid-brain and cerebral hemispheres, and lesion load increased with increasing pro-inflammatory cytokine production. Despite being resistant to disease induction with increasing pro-inflammatory cytokine production, lesion incidence in IL-4Rα-deficient animals was equal to their WT counterparts. However, lesion severity in IL-4Rα-deficient animals was preferentially reduced in the mid-brain and cerebral hemispheres. From these studies, we conclude that signaling through IL-4Rα has little effect on regulating the peripheral pro-inflammatory cytokine profile in this EAE variant but has distinct effects on the determination of lesion topography.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/inmunología , Eliminación de Gen , Predisposición Genética a la Enfermedad , Receptores de Superficie Celular/genética , Animales , Antígenos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , Proteína Proteolipídica de la Mielina/efectos adversos , Nervio Óptico/inmunología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Médula Espinal/inmunología , Médula Espinal/metabolismo , Médula Espinal/patología
5.
J Autoimmun ; 38(4): 344-53, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22459490

RESUMEN

Previously we showed that transgenic mice expressing human HLA-DR3 gene are susceptible to PLP(91-110) induced experimental autoimmune encephalomyelitis (EAE) and can serve as an animal model of multiple sclerosis (MS). HLA-DR3 mice with EAE showed increased number of CD8 T cells indicating their important role in disease pathogenesis. The role of CD8 T cells in MS, an inflammatory demyelinating disease of CNS, has been enigmatic as it has been assigned both regulatory and pathogenic roles. Therefore, to evaluate the role of CD8 T cells, we generated CD8 deficient HLA-DR3 transgenic mice (DR3.CD8(-/-)). Immunization with PLP(91-110) led to more severe EAE in DR3.CD8(-/-) mice compared to HLA-DR3 mice indicating a regulatory role for CD8 T cells. Interestingly, DR3.CD8(-/-) mice with EAE showed decreased CNS pathology compared to DR3 mice thus suggesting a pathogenic role for CD8 T cells. We show that these two subsets of CD8 T cells can be differentiated based on the surface expression of CD122 (IL-2 Rß chain). CD8 T cells expressing CD122 (CD8+CD122+) play a regulatory role while CD8+CD122- T cells act as a pathogenic subset. CD122 expressing CD8 T cells are the regulatory subset of CD8 T cells and regulate the encephalitogenic CD4 T cells through direct modulation of antigen presenting cells and/or through the release of immunoregulatory cytokines such as IL-10, IFNγ and TGFß. We also showed that adoptive transfer of CD8CD122- T cells caused increased spinal cord demyelination indicating that these are pathogenic subset of CD8 T cells. Our study suggests that CD8+ T cells play both regulatory as well as pathogenic role in disease pathogenesis of EAE. A better understanding of these subsets could aid in designing novel therapy for MS patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Antígeno HLA-DR3/genética , Proteína Proteolipídica de la Mielina/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Comunicación Celular/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/inmunología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Antígeno HLA-DR3/inmunología , Subunidad beta del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/química , Neuroglía/inmunología
6.
PLoS One ; 6(10): e25721, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21998684

RESUMEN

We report here the design, development and performance of a novel formulation of liposome- encapsulated glucocorticoids (GCs). A highly efficient (>90%) and stable GC encapsulation was obtained based on a transmembrane calcium acetate gradient driving the active accumulation of an amphipathic weak acid GC pro-drug into the intraliposome aqueous compartment, where it forms a GC-calcium precipitate. We demonstrate fabrication principles that derive from the physicochemical properties of the GC and the liposomal lipids, which play a crucial role in GC release rate and kinetics. These principles allow fabrication of formulations that exhibit either a fast, second-order (t(1/2) ~1 h), or a slow, zero-order release rate (t(1/2) ~ 50 h) kinetics. A high therapeutic efficacy was found in murine models of experimental autoimmune encephalomyelitis (EAE) and hematological malignancies.


Asunto(s)
Química Farmacéutica/métodos , Glucocorticoides/administración & dosificación , Glucocorticoides/química , Nanoestructuras/química , Animales , Cápsulas , Fenómenos Químicos , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Glucocorticoides/farmacocinética , Glucocorticoides/uso terapéutico , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Leucemia/tratamiento farmacológico , Liposomas , Linfoma de Células T/tratamiento farmacológico , Hemisuccinato de Metilprednisolona/administración & dosificación , Hemisuccinato de Metilprednisolona/química , Hemisuccinato de Metilprednisolona/farmacocinética , Hemisuccinato de Metilprednisolona/uso terapéutico , Ratones , Esclerosis Múltiple/tratamiento farmacológico , Proteína Proteolipídica de la Mielina/efectos adversos , Solubilidad , Agua/química
7.
J Autoimmun ; 37(3): 228-41, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21752599

RESUMEN

The respective roles of inflammatory and neurodegenerative processes in the pathology of multiple sclerosis (MS) and in its animal model experimental autoimmune encephalomyelitis (EAE) are controversial. Novel treatment strategies aim to operate within the CNS to induce neuroprotection and repair processes in addition to their anti-inflammatory properties. In this study we analyzed and compared the in situ pathological manifestations of EAE utilizing two different models, namely the relapsing-remitting PLP-induced and the chronic MOG-induced diseases. To characterize pathological changes, both transmission electron microscopy (TEM) and immunohistochemistry were employed. The effect of the approved MS drug glatiramer acetate (GA, Copaxone) on myelin damage/repair and on motor neuron loss/preservation was studied in both EAE models. Ultrastructural spinal cord analysis revealed multiple white matter damage foci, with different patterns in the two EAE models. Thus, the relapsing-remitting model was characterized mainly by widespread myelin damage and by remyelinating fibers, whereas in the chronic model axonal degeneration was more prevalent. Loss of lower motor neurons was manifested only in mice with chronic MOG-induced disease. In the GA-treated mice, smaller lesions, increased axonal density and higher prevalence of normal appearing axons were observed, as well as decreased demyelination and degeneration. Furthermore, quantitative analysis of the relative remyelination versus demyelination, provides for the first time evidence of significant augmentation of remyelination after GA treatment. The loss of motor neurons in GA-treated mice was also reduced in comparison to that of EAE untreated mice. These effects were obtained even when GA treatment was applied in a therapeutic schedule, namely after the appearance of clinical symptoms. Hence, the remyelination and neuronal preservation induced by GA are in support of the neuroprotective consequences of this treatment.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Inflamación/patología , Esclerosis Múltiple Recurrente-Remitente/patología , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Péptidos/administración & dosificación , Médula Espinal/patología , Animales , Axones/efectos de los fármacos , Axones/ultraestructura , Enfermedad Crónica , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Acetato de Glatiramer , Humanos , Inmunohistoquímica , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/ultraestructura , Esclerosis Múltiple Recurrente-Remitente/inducido químicamente , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Esclerosis Múltiple Recurrente-Remitente/inmunología , Proteínas de la Mielina/administración & dosificación , Proteínas de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/administración & dosificación , Proteína Proteolipídica de la Mielina/efectos adversos , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/ultraestructura , Glicoproteína Asociada a Mielina/ultraestructura , Glicoproteína Mielina-Oligodendrócito , Fármacos Neuroprotectores/uso terapéutico , Péptidos/uso terapéutico , Médula Espinal/efectos de los fármacos , Médula Espinal/ultraestructura
8.
APMIS ; 119(6): 336-46, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21569091

RESUMEN

In this study we demonstrate that experimental autoimmune encephalomyelitis (EAE) induced by the MBP-PLP fusion protein MP4, MOG peptide 35-55, or PLP peptide 178-191 in C57BL/6 mice, respectively, displays distinct features of CNS pathology. Major differences between the three models resided in (i) the region-/tract-specificity and disseminated nature of spinal cord degeneration, (ii) the extent and kinetics of demyelination, and (iii) the involvement of motoneurons in the disease. In contrast, axonal damage was present in all models and to a similar extent, proposing this feature as a possible morphological correlate for the comparable chronic clinical course of the disease induced by the three antigens. The data suggest that the antigen targeted in autoimmune encephalomyelitis is crucial to the induction of differential histopathological disease manifestations. The use of MP4-, MOG:35-55-, and PLP:178-191-induced EAE on the C57BL/6 background can be a valuable tool when it comes to reproducing and studying the structural-morphological diversity of multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Proteína Proteolipídica de la Mielina/inmunología , Glicoproteína Asociada a Mielina/inmunología , Fragmentos de Péptidos/inmunología , Médula Espinal/patología , Animales , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Ratones , Ratones Endogámicos C57BL , Enfermedad de la Neurona Motora/patología , Proteína Básica de Mielina/inmunología , Proteínas de la Mielina , Proteína Proteolipídica de la Mielina/efectos adversos , Glicoproteína Asociada a Mielina/efectos adversos , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos/efectos adversos , Fenotipo , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/inmunología
9.
Ann Neurol ; 69(5): 878-91, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21391234

RESUMEN

OBJECTIVE: The subventricular zone (SVZ) of the brain constitutes a niche for neural stem and progenitor cells that can initiate repair after central nervous system (CNS) injury. In a relapsing-remitting model of experimental autoimmune encephalomyelitis (EAE), the neural stem cells (NSCs) become activated and initiate regeneration during acute disease, but lose this ability during the chronic phases of disease. We hypothesized that chronic microglia activation contributes to the failure of the NSC repair potential in the SVZ. METHODS: Using bromodeoxyuridine injections at different time points during EAE, we quantified the number of proliferating and differentiating progenitors, and evaluated the structure of the SVZ by electron microscopy. In vivo minocycline treatment during EAE was used to address the effect of microglia inactivation on SVZ dysfunction. RESULTS: In vivo treatment with minocycline, an inhibitor of microglia activation, increases stem cell proliferation in both naive and EAE animals. Minocycline treatment decreases cortical and periventricular pathology in the chronic phase of EAE, improving the proliferation of Sox2 stem cells and NG2 oligodendrocyte precursors cells originating in the SVZ and their differentiation into mature oligodendrocytes. INTERPRETATION: These data suggest that failure of repair observed during chronic EAE correlates with microglia activation and that treatments targeting chronic microglial activation have the potential for enhancing repair in the CNS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/patología , Células-Madre Neurales/fisiología , Nicho de Células Madre/patología , Animales , Antibacterianos/farmacología , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/fisiopatología , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Minociclina/farmacología , Esclerosis Múltiple , Proteína Proteolipídica de la Mielina/efectos adversos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/ultraestructura , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Fragmentos de Péptidos/efectos adversos , Prevención Secundaria , Nicho de Células Madre/efectos de los fármacos , Factores de Tiempo
10.
J Neurosci ; 30(27): 9074-83, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20610741

RESUMEN

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS and remyelination in MS ultimately fails. Although strategies to promote myelin repair are eagerly sought, mechanisms underlying remyelination in vivo have been elusive. CXCR2 is expressed on neutrophils and oligodendrocyte lineage cells in the CNS. CXCR2-positive neutrophils facilitate inflammatory demyelination in demyelination models such as experimental autoimmune encephalomyelitis (EAE) and cuprizone intoxication. Systemic injection of a small molecule CXCR2 antagonist at the onset of EAE decreased demyelinated lesions. These results left the cellular target of the CXCR2 antagonist uncertain and did not clarify whether CXCR2 blockade prevented demyelination or promoted remyelination. Here, we show that the actions of CXCR2 on nonhematopoietic cells unexpectedly delay myelin repair. Bone marrow chimeric mice (Cxcr2(+/-)-->Cxcr2(-/-) and Cxcr2(+/-)-->Cxcr2(+/+)) were subjected to two distinct models of myelin injury. In all cases, myelin repair was more efficient in Cxcr2(+/-)-->Cxcr2(-/-) animals. Oligodendrocyte progenitor cells (OPCs) in demyelinated lesions of Cxcr2(+/-)-->Cxcr2(-/-) mice proliferated earlier and more vigorously than in tissues from Cxcr2(+/-)--> Cxcr2(+/+) animals. In vitro demyelinated CNS slice cultures also showed better myelin repair when CXCR2 was blocked with neutralizing antibodies or was genetically deleted. Our results suggest that CXCR2 inactivation permits optimal spatiotemporal positioning of OPCs in demyelinating lesions to receive local proliferative and differentiating signals. Given that CXCR2 exerts dual functions that promote demyelination and decrease remyelination by actions toward hematopoietic cells and nonhematopoietic cells, respectively, our findings identify CXCR2 as a promising drug target for clinical demyelinating disorders.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/fisiopatología , Vaina de Mielina/fisiología , Regeneración Nerviosa/fisiología , Receptores de Interleucina-8B/metabolismo , Recuperación de la Función/fisiología , Animales , Animales Recién Nacidos , Anticuerpos/farmacología , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Estimulantes del Sistema Nervioso Central/farmacología , Cerebelo/efectos de los fármacos , Cuprizona , Enfermedades Desmielinizantes/inducido químicamente , Enfermedades Desmielinizantes/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Citometría de Flujo , Adyuvante de Freund/efectos adversos , Glicoproteínas/efectos adversos , Técnicas In Vitro , Antígenos Comunes de Leucocito/metabolismo , Lipopolisacáridos/efectos adversos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión/métodos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/metabolismo , Proteína Proteolipídica de la Mielina/efectos adversos , Vaina de Mielina/ultraestructura , Glicoproteína Mielina-Oligodendrócito , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/genética , Examen Neurológico , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Fragmentos de Péptidos/efectos adversos , Picrotoxina/farmacología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Receptores de Interleucina-8B/deficiencia , Receptores de Interleucina-8B/genética , Receptores de Interleucina-8B/inmunología , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/genética , Índice de Severidad de la Enfermedad , Células Madre/efectos de los fármacos , Factores de Tiempo
11.
Neurobiol Dis ; 32(2): 273-80, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18778774

RESUMEN

We investigated the treatment of experimental autoimmune encephalomyelitis (EAE) in mice with Niaspan, an agent used to elevate high-density lipoprotein (HDL). EAE mice were treated with Niaspan starting on the immunization or clinical onset day. Neurological functional recovery was significantly increased in the Niaspan treated mice (100 mg/kgbw) compared to the controls. Inflammatory infiltrates were significantly reduced in the Niaspan treatment group compared to the EAE controls. HDL level, intact myelin area, newly formed oligodendrocytes, regenerating axons, gene and protein levels of sonic hedgehog (Shh)/Gli1 were significantly increased in the Niaspan treated mice compared to EAE controls. These data indicate that Niaspan treatment improved functional recovery after EAE, possibly, via reducing inflammatory infiltrates and demyelination areas, and stimulating oligodendrogenesis and axonal regeneration. Niaspan-mediated activation of Shh/Gli1 pathway may promote functional recovery post-EAE.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/fisiopatología , Niacina/uso terapéutico , Recuperación de la Función/efectos de los fármacos , Complejo Vitamínico B/uso terapéutico , Animales , Línea Celular Transformada , Proliferación Celular/efectos de los fármacos , HDL-Colesterol/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/efectos adversos , Regeneración Nerviosa/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Transducción de Señal/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Médula Espinal/fisiopatología , Factores de Tiempo
12.
Clin Immunol ; 129(1): 69-79, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18676182

RESUMEN

The objective was to optimize and evaluate the in vivo activities of our novel bifunctional peptide inhibitor (BPI), which alters immune response in autoimmune diseases by modulating the immunological synapse formation. Previously, we have designed PLP-BPI and GAD-BPI by conjugating myelin proteolipid protein (PLP)(139-151) and glutamic acid decarboxylase (GAD)(208-217), respectively, with CD11a(237-246) via a spacer peptide. PLP-BPI and GAD-BPI suppressed the disease progression in experimental autoimmune encephalomyelitis (EAE) and in type-1 diabetes, respectively. In this study, various PLP-BPI derivatives were synthesized and evaluated in the EAE model. Intravenous injections of PLP-BPI derivatives prevented the disease progression more efficiently than did unmodified PLP-BPI. Production of IL-17, a potent proinflammatory cytokine found commonly among MS patients, was significantly low in Ac-PLP-BPI-NH(2)-2-treated mice. Treatment given after the disease onset could dramatically ameliorate the disease. BPI induced anaphylactic responses at a lower incidence than PLP(139-151). In conclusion, PLP-BPI derivatives can effectively suppress the disease severity and morbidity of EAE by post-onset therapeutic treatment as well as prophylactic use.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/prevención & control , Interleucina-17/sangre , Proteína Proteolipídica de la Mielina/uso terapéutico , Fragmentos de Péptidos/uso terapéutico , Anafilaxia/inducido químicamente , Animales , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Ratones , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/inmunología , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/inmunología
13.
J Immunol ; 178(8): 4749-56, 2007 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-17404254

RESUMEN

Administration of autoantigens under conditions that induce type 2 immunity frequently leads to protection from T cell-mediated autoimmune diseases. Such treatments, however, are inherently linked to the induction of IgG1 Abs and to the risk of triggering anaphylactic reactions. We studied the therapeutic benefit vs risk of immune deviation in experimental allergic encephalomyelitis of SJL mice induced by MP4, a myelin basic protein-proteolipid protein (PLP) fusion protein. MP4 administration in IFA induced type 2 T cell immunity, IgG1 Abs, and experimental allergic encephalomyelitis protection, and all three were enhanced by repeat injections. Despite high Ab titers, anaphylactic side reactions were not observed when MP4 was repeatedly injected in IFA or as soluble Ag s.c. In contrast, lethal anaphylaxis was seen after s.c. injection of soluble PLP:139-151 peptide, but not when the peptide was reinjected in IFA. Therefore, the Ab response accompanying the immune therapy constituted an anaphylactic risk factor only when the autoantigen was not retained in an adjuvant and when it was small enough to be readily disseminated within the body. Taken together, our data show that treatment regimens can be designed to boost the protective type 2 T cell response while avoiding the risk of Ab-mediated allergic side effects.


Asunto(s)
Anafilaxia/etiología , Autoantígenos/uso terapéutico , Encefalomielitis Autoinmune Experimental/terapia , Proteína Básica de Mielina/uso terapéutico , Proteína Proteolipídica de la Mielina/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Autoantígenos/administración & dosificación , Autoantígenos/efectos adversos , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Inmunización , Inmunoglobulina G/biosíntesis , Interleucina-2/biosíntesis , Interleucina-4/biosíntesis , Ratones , Proteína Básica de Mielina/administración & dosificación , Proteína Básica de Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/administración & dosificación , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/inmunología , Proteínas del Tejido Nervioso/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Linfocitos T/inmunología
14.
J Immunol ; 178(3): 1372-8, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17237384

RESUMEN

In experimental autoimmune encephalomyelitis (EAE), the production of proinflammatory cytokines by neuroantigen-specific T cells is thought to initiate and maintain the inflammatory autoimmune pathology. Because gene knockout strategies have shown that IFN-gamma and TNF are not essential for EAE development, there is increasing interest in establishing the role of other proinflammatory cytokines, primarily IL-17 in EAE. We used an IL-17 ELISPOT assay to track the neuroantigen-specific IL-17-producing T cells at single-cell resolution in various organs of SJL mice undergoing PLP 139-151-induced EAE. Overall, the migration patterns and population kinetics of the PLP 139-151-specific IL-17-producing CD4 cells were reminiscent of the IFN-gamma-producing cells, with the exception of IL-17 producers far outnumbering the IFN-gamma and IL-2 producers in the inflamed CNS. The selective enrichment of IL-17-producing CD4 cells in the CNS is suggestive of the pathogenic role of an independent (non-Th1) IL-17-producing proinflammatory effector T cell class in EAE.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Movimiento Celular , Encefalomielitis Autoinmune Experimental/inmunología , Interleucina-17/análisis , Animales , Sistema Nervioso Central/citología , Encefalomielitis Autoinmune Experimental/inducido químicamente , Inflamación/etiología , Interferón gamma , Interleucina-2 , Cinética , Ratones , Ratones Endogámicos , Proteína Proteolipídica de la Mielina/efectos adversos , Especificidad de Órganos , Fragmentos de Péptidos/efectos adversos , Linfocitos T Colaboradores-Inductores
15.
J Immunol ; 169(1): 117-25, 2002 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12077236

RESUMEN

Pertussis toxin (PT) has been widely used to facilitate the induction of experimental autoimmune encephalomyelitis (EAE) in rodents. It has been suggested that this microbial product promotes EAE by opening up the blood-brain barrier and thereby facilitates the migration of pathogenic T cells to the CNS. However, PT has other biological effects that could contribute to its activity in EAE, such as enhancing the cytokine production by T cells and induction of lymphocytosis. In this work, we investigated the effects of PT on the pathogenicity, cytokine differentiation, and clonal sizes of neuroantigen-reactive T cells in EAE in mice. Our results show that PT prevented the protection from EAE conferred by injection of PLPp139-151 in IFA and induced high frequencies of peptide-specific Th1 cells and disease. Interestingly, the mice developed EAE despite the simultaneous vigorous clonal expansion of PLPp139-151-specific Th2 cells. The data indicate that the Th2 cells in this model neither were protective against EAE nor promoted the disease. Furthermore, the results suggested that the effects of the toxin on neuroantigen-reactive T cells were promoted by the PT-induced activation of APCs in lymphoid tissues and the CNS. Together, the results suggest that microbial products, such as PT, could contribute to the initiation of autoimmune disease by modulating the interaction between the innate and adaptive immune system in the response to self Ags.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Adyuvante de Freund/administración & dosificación , Lípidos , Activación de Linfocitos/inmunología , Proteína Básica de Mielina/inmunología , Proteína Proteolipídica de la Mielina/inmunología , Fragmentos de Péptidos/inmunología , Toxina del Pertussis , Células TH1/inmunología , Células Th2/citología , Factores de Virulencia de Bordetella/farmacología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/inmunología , Movimiento Celular/inmunología , Células Clonales/citología , Células Clonales/inmunología , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/prevención & control , Epítopos de Linfocito T/administración & dosificación , Epítopos de Linfocito T/inmunología , Femenino , Adyuvante de Freund/efectos adversos , Adyuvante de Freund/inmunología , Adyuvante de Freund/farmacología , Tolerancia Inmunológica , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Recuento de Linfocitos , Ratones , Ratones Endogámicos , Proteína Básica de Mielina/administración & dosificación , Proteína Básica de Mielina/uso terapéutico , Proteína Proteolipídica de la Mielina/administración & dosificación , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/uso terapéutico , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/efectos adversos , Fragmentos de Péptidos/uso terapéutico , Médula Espinal/citología , Médula Espinal/inmunología , Médula Espinal/patología , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/trasplante , Células TH1/citología , Células Th2/inmunología , Células Th2/trasplante
16.
Nat Immunol ; 2(7): 605-11, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11429544

RESUMEN

The inducible costimulatory molecule (ICOS) is expressed on activated T cells and participates in a variety of important immunoregulatory functions. After the induction of experimental allergic encephalomyelitis in SJL mice with proteolipid protein (PLP), brain ICOS mRNA and protein were up-regulated on infiltrating CD3+ T cells before disease onset. ICOS blockade during the efferent immune response (9-20 days after immunization) abrogated disease, but blockade during antigen priming (1-10 days after immunization) exacerbated disease. Upon culture with PLP and compared with immunized controls, splenocytes produced either decreased interferon-gamma (IFN-gamma, in efferent blockade) or excessive IFN-gamma (in priming blockade). PLP-specific immunoglobulin G1 was decreased in animals treated with anti-ICOS during antigen priming, but not in other groups.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Encéfalo/inmunología , Encéfalo/patología , Citocinas/biosíntesis , Citocinas/genética , Encefalomielitis Autoinmune Experimental/patología , Femenino , Inmunoglobulina G/biosíntesis , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Interferón gamma/biosíntesis , Ratones , Proteína Proteolipídica de la Mielina/efectos adversos , Proteína Proteolipídica de la Mielina/inmunología , Linfocitos T/inmunología , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...