Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 9: 2399, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30410484

RESUMEN

The guanine nucleotide exchange factor Vav1 is essential for transducing T cell receptor (TCR) signals and plays an important role in T cell development and activation. Previous genetic studies identified a natural variant of Vav1 characterized by the substitution of an arginine (R) residue by a tryptophane (W) at position 63 (Vav1R63W). This variant impacts Vav1 adaptor functions and controls susceptibility to T cell-mediated neuroinflammation. To assess the implication of this Vav1 variant on the susceptibility to antibody-mediated diseases, we used the animal model of myasthenia gravis, experimental autoimmune myasthenia gravis (EAMG). To this end, we generated a knock-in (KI) mouse model bearing a R to W substitution in the Vav1 gene (Vav1R63W) and immunized it with either torpedo acetylcholine receptor (tAChR) or the α146-162 immunodominant peptide. We observed that the Vav1R63W conferred increased susceptibility to EAMG, revealed by a higher AChR loss together with an increased production of effector cytokines (IFN-γ, IL-17A, GM-CSF) by antigen-specific CD4+ T cells, as well as an increased frequency of antigen-specific CD4+ T cells. This correlated with the emergence of a dominant antigen-specific T cell clone in KI mice that was not present in wild-type mice, suggesting an impact on thymic selection and/or a different clonal selection threshold following antigen encounter. Our results highlight the key role of Vav1 in the pathophysiology of EAMG and this was associated with an impact on the TCR repertoire of AChR reactive T lymphocytes.


Asunto(s)
Variación Genética , Miastenia Gravis Autoinmune Experimental/etiología , Miastenia Gravis Autoinmune Experimental/metabolismo , Proteínas Proto-Oncogénicas c-vav/genética , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Ratones , Miastenia Gravis Autoinmune Experimental/patología , Fenotipo , Receptores Nicotínicos/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T
2.
Eur J Immunol ; 47(8): 1295-1304, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28605013

RESUMEN

Antibody production is key for effective immune response and relies on follicular helper T (Tfh) cells. B cell-Tfh cell interactions result either in an extra-follicular low affinity B-cell response or in germinal center reactions producing high-affinity memory B cells and long-lived plasma cells. As Tfh cells influence B-cell commitment, it also became clear that B cells influence these interactions in ways that still remain unresolved. We observed that strong BCR signals decreased Tfh-cell differentiation in vitro, which correlated with decreased expression of ICOS-L at the surface of stimulated B cells. Further, we comprehensively demonstrated that ICOS-L expression correlated with the level of Tfh differentiation irrespective of antigen presentation at the surface of activated B cells. Our in vivo experiments could show that immunization with a high-affinity antigen for B cells resulted in much less Tfh development than immunization with low-affinity antigen. Furthermore, blocking ICOS-L in vivo inhibited Tfh development when using low-affinity antigen. Altogether, these results indicate that BCR affinity shapes Tfh-cell development in part through ICOS/ICOS-L interactions. Ultimately, we reveal new depths in the B cell-Tfh cell crosstalk that could eventually result in better vaccine protocols.


Asunto(s)
Linfocitos B/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/metabolismo , Activación de Linfocitos , Receptores de Antígenos de Linfocitos B/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Presentación de Antígeno , Linfocitos B/metabolismo , Diferenciación Celular , Citometría de Flujo , Centro Germinal/inmunología , Ligando Coestimulador de Linfocitos T Inducibles/genética , Ratones , Receptores de Antígenos de Linfocitos B/inmunología , Receptores CXCR5/genética , Receptores CXCR5/inmunología , Transducción de Señal
3.
Cell Rep ; 17(1): 193-205, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681431

RESUMEN

CXCR4 plays a central role in B cell immune response, notably by promoting plasma cell (PC) migration and maintenance in the bone marrow (BM). Gain-of-function mutations in CXCR4 affecting receptor desensitization have been reported in the rare immunodeficiency called WHIM syndrome (WS). Despite lymphopenia, patients mount an immune response but fail to maintain it over time. Using a knockin mouse model phenocopying WS, we showed that, counter-intuitively, a gain of Cxcr4 function inhibited the maintenance of antibody titers after immunization. Although the Cxcr4 mutation intrinsically and locally promoted germinal center response and PC differentiation, antigen-specific PCs were barely detected in the BM, a defect mirrored by early accumulation of immature plasmablasts potentially occupying the survival niches for long-lived PCs. Therefore, fine-tuning of Cxcr4 desensitization is critically required for efficient PC differentiation and maintenance, and absence of such a regulatory process may account for the defective humoral immunity observed in WS patients.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Médula Ósea/inmunología , Desensibilización Inmunológica , Síndromes de Inmunodeficiencia/inmunología , Células Plasmáticas/inmunología , Receptores CXCR4/inmunología , Verrugas/inmunología , Animales , Anticuerpos/sangre , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/patología , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Diferenciación Celular , Movimiento Celular , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Sustitución del Gen , Centro Germinal , Haptenos , Hemocianinas/administración & dosificación , Humanos , Inmunidad Humoral , Inmunización , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Ratones , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/patología , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Transducción de Señal , Verrugas/genética , Verrugas/patología
4.
Brain Behav Immun ; 26(6): 919-30, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22522067

RESUMEN

Prion diseases are caused by the transconformation of the host cellular prion protein PrP(c) into an infectious neurotoxic isoform called PrP(Sc). While vaccine-induced PrP-specific CD4(+) T cells and antibodies partially protect scrapie-infected mice from disease, the potential autoreactivity of CD8(+) cytotoxic T lymphocytes (CTLs) received little attention. Beneficial or pathogenic influence of PrP(c)-specific CTL was evaluated by stimulating a CD8(+) T-cell-only response against PrP in scrapie-infected C57BL/6 mice. To circumvent immune tolerance to PrP, five PrP-derived nonamer peptides identified using prediction algorithms were anchored-optimized to improve binding affinity for H-2D(b) and immunogenicity (NP-peptides). All of the NP-peptides elicited a significant number of IFNγ secreting CD8(+) T cells that better recognized the NP-peptides than the natives; three of them induced T cells that were lytic in vivo for NP-peptide-loaded target cells. Peptides 168 and 192 were naturally processed and presented by the 1C11 neuronal cell line. Minigenes encoding immunogenic NP-peptides inserted into adenovirus (rAds) vectors enhanced the specific CD8(+) T-cell responses. Immunization with rAd encoding 168NP before scrapie inoculation significantly prolonged the survival of infected mice. This effect was attributable to a significant lengthening of the symptomatic phase and was associated with enhanced CD3(+) T cell recruitment to the CNS. However, immunization with Ad168NP in scrapie-incubating mice induced IFNγ-secreting CD8(+) T cells that were not cytolytic in vivo and did not influence disease progression nor infiltrated the brain. In conclusion, the data suggest that vaccine-induced PrP-specific CD8(+) T cells interact with prions into the CNS during the clinical phase of the disease.


Asunto(s)
Sistema Nervioso Central/patología , Enfermedades por Prión/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Scrapie/patología , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Sistema Nervioso Central/inmunología , Radioisótopos de Cromo , Técnica del Anticuerpo Fluorescente , Inmunización , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Péptidos/inmunología , Plásmidos/genética , Proteínas PrPC/genética , Proteínas PrPC/inmunología , Scrapie/inmunología
5.
PLoS One ; 4(3): e4917, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19295917

RESUMEN

In prion diseases, PrP(c), a widely expressed protein, is transformed into a pathogenic form called PrP(Sc), which is in itself infectious. Antibodies directed against PrP(c) have been shown to inhibit PrP(c) to PrP(Sc) conversion in vitro and protect in vivo from disease. Other effectors with potential to eliminate PrPSc-producing cells are cytotoxic T cells directed against PrP-derived peptides but their ability to protect or to induce deleterious autoimmune reactions is not known. The natural tolerance to PrP(c) makes difficult to raise efficient adaptive responses. To break tolerance, adenovirus (Ad) encoding human PrP (hPrP) or control Ad were administered to wild-type mice by direct injection or by transfer of Ad-transduced dendritic cells (DCs). Control Ad-transduced DCs from Tg650 mice overexpressing hPrP were also used for immunization. DC-mediated but not direct administration of AdhPrP elicited antibodies that bound to murine native PrP(c). Frequencies of PrP-specific IFNgamma-secreting T cells were low and in vivo lytic activity only targeted cells strongly expressing hPrP. Immunohistochemical analysis revealed that CD3(+) T cell infiltration was similar in the brain of vaccinated and unvaccinated 139A-infected mice suggesting the absence of autoimmune reactions. Early splenic PrP(Sc) replication was strongly inhibited ten weeks post infection and mean survival time prolonged from 209 days in untreated 139A-infected mice to 246 days in mice vaccinated with DCs expressing the hPrP. The efficacy appeared to be associated with antibody but not with cytotoxic cell-mediated PrP-specific responses.


Asunto(s)
Adenoviridae , Células Dendríticas/inmunología , Vectores Genéticos , Priones/inmunología , Scrapie/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Secuencia de Aminoácidos , Animales , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Humanos , Inmunización , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Priones/genética , Scrapie/patología , Scrapie/prevención & control , Alineación de Secuencia , Tasa de Supervivencia
6.
J Immunol ; 181(1): 768-75, 2008 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-18566443

RESUMEN

Prion diseases are associated with the conversion of the normal host cellular prion protein to an abnormal protease-resistant (PrPres) associated with infectivity. No specific immune response against prions develops during infection due to the strong tolerance to cellular prion protein. We examined the protective potential on prion diseases of immune responses elicited in C57BL/6 mice with PrP peptides 98-127 (P5) or 158-187 (P9) with CpG. After immunization, P5-treated mice developed high titer and long-lasting Abs, and P9-treated mice developed transient IFN-gamma secreting T cells and poor and variable Ab responses. Both treatments impaired early accumulation of PrPres in the spleen and prolonged survival of mice infected with 139A scrapie. Additional P9 boosts after 139A infection sustained the T cell response and partially inhibited PrPres early accumulation but did not improve the survival. Surprisingly, when P9 injections were started 1 mo after infection and repeated subsequently, specific T cell and Ab responses were impaired and no beneficial effect on prion disease was observed. After a single injection of P9, the number of IFN-gamma secreting CD4+ T cells was also reduced in mice 8- to 10-wk postinfection compared with healthy mice. In vivo and in vitro removal of CD4+CD25+ T cells restored the T cell response to P9 in infected mice. In conclusion, CD4+ T cells as well as Abs might participate to the protection against scrapie. Of importance, the peripheral accumulation of PrPres during infection negatively interferes with the development of T and B cell responses to PrP and regulatory T cells might contribute to this phenomenon.


Asunto(s)
Anticuerpos/inmunología , Péptidos/inmunología , Priones/inmunología , Scrapie/inmunología , Scrapie/patología , Linfocitos T/inmunología , Vacunas contra el SIDA , Animales , Progresión de la Enfermedad , Femenino , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos C57BL , Priones/patogenicidad , Scrapie/clasificación , Tasa de Supervivencia , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA