Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 113(36): 10145-50, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27555590

RESUMEN

T helper 17 (TH17) cells represent a discrete TH cell subset instrumental in the immune response to extracellular bacteria and fungi. However, TH17 cells are considered to be detrimentally involved in autoimmune diseases like multiple sclerosis (MS). In contrast to TH17 cells, regulatory T (Treg) cells were shown to be pivotal in the maintenance of peripheral tolerance. Thus, the balance between Treg cells and TH17 cells determines the severity of a TH17 cell-driven disease and therefore is a promising target for treating autoimmune diseases. However, the molecular mechanisms controlling this balance are still unclear. Here, we report that pharmacological inhibition as well as genetic ablation of the protein kinase CK2 (CK2) ameliorates experimental autoimmune encephalomyelitis (EAE) severity and relapse incidence. Furthermore, CK2 inhibition or genetic ablation prevents TH17 cell development and promotes the generation of Treg cells. Molecularly, inhibition of CK2 leads to reduced STAT3 phosphorylation and strongly attenuated expression of the IL-23 receptor, IL-17, and GM-CSF. Thus, these results identify CK2 as a nodal point in TH17 cell development and suggest this kinase as a potential therapeutic target to treat TH17 cell-driven autoimmune responses.


Asunto(s)
Quinasa de la Caseína II/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Animales , Quinasa de la Caseína II/deficiencia , Quinasa de la Caseína II/genética , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Factores de Transcripción Forkhead , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Interleucina-17 , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/patología , Glicoproteína Mielina-Oligodendrócito , Fragmentos de Péptidos , Fosforilación , Receptores de Interleucina , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Índice de Severidad de la Enfermedad , Transducción de Señal , Linfocitos T Reguladores/citología , Células Th17/patología
2.
Mult Scler ; 24(5): 632-641, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28436295

RESUMEN

BACKGROUND: The effect of dimethyl fumarate (DMF) on circulating lymphocyte subsets and their contribution as predictors of clinical efficacy have not yet been investigated in multiple sclerosis (MS). OBJECTIVE: To evaluate lymphocytes and lymphocyte subsets (analyzed 6 months after DMF start) in MS patients with and without disease activity after 1 year of treatment in a retrospective study. METHODS: Peripheral blood lymphocyte subsets were analyzed by flow cytometry. Untreated MS patients ( n = 40) were compared to those 6 months after onset of DMF treatment ( n = 51). Clinical and magnetic resonance imaging (MRI) disease activity of DMF-treated patients were assessed in the first year under treatment. RESULTS: Stable patients showed significantly lower lymphocytes, CD4+ and CD8+ T cells as well as CD19+ B cells compared to active patients under DMF treatment. Furthermore, an increased CD4/CD8 ratio ( p < 0.025) in stable patients indicated a disproportionate reduction of CD8+ T cells relative to CD4+ T cells. Reduced lymphocytes, CD8+ T cells, and CD19+ B cells 6 months after DMF start allowed prediction of the treatment response in the first year. CONCLUSION: DMF treatment response is reflected by lower circulating lymphocytes and specific lymphocyte subsets. Changes in the cellular immune profiles under DMF treatment are clinically relevant and might serve as a surrogate marker of treatment response.


Asunto(s)
Linfocitos T CD8-positivos , Dimetilfumarato/uso terapéutico , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Adolescente , Adulto , Antígenos CD19 , Linfocitos B/inmunología , Relación CD4-CD8 , Estudios Transversales , Dimetilfumarato/efectos adversos , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Inmunosupresores/efectos adversos , Estudios Longitudinales , Recuento de Linfocitos , Linfopenia/etiología , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/sangre , Esclerosis Múltiple/patología , Curva ROC , Estudios Retrospectivos , Estadísticas no Paramétricas , Resultado del Tratamiento , Adulto Joven
3.
Cell Mol Life Sci ; 73(13): 2425-50, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26970979

RESUMEN

Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disorder of the central nervous system characterized by infiltration of immune cells and progressive damage to myelin sheaths and neurons. There is still no cure for the disease, but drug regimens can reduce the frequency of relapses and slightly delay progression. Myeloid cells or antigen-presenting cells (APCs) such as dendritic cells (DC), macrophages, and resident microglia, are key players in both mediating immune responses and inducing immune tolerance. Mounting evidence indicates a contribution of these myeloid cells to the pathogenesis of multiple sclerosis and to the effects of treatment, the understanding of which might provide strategies for more potent novel therapeutic interventions. Here, we review recent insights into the role of APCs, with specific focus on DCs in the modulation of neuroinflammation in MS.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Descubrimiento de Drogas , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/inmunología , Animales , Células Presentadoras de Antígenos/efectos de los fármacos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/patología , Células Dendríticas/patología , Descubrimiento de Drogas/métodos , Encefalomielitis Autoinmune Experimental/patología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/patología , Terapia Molecular Dirigida/métodos , Esclerosis Múltiple/patología
4.
Int J Mol Sci ; 18(9)2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28914804

RESUMEN

Extracellular signal-regulated kinase (ERK) signaling plays a crucial role in regulating immune cell function and has been implicated in autoimmune disorders. To date, all commercially available inhibitors of ERK target upstream components, such as mitogen-activated protein (MAP) kinase/ERK kinase (MEKs), but not ERK itself. Here, we directly inhibit nuclear ERK translocation by a novel pharmacological approach (Glu-Pro-Glu (EPE) peptide), leading to an increase in cytosolic ERK phosphorylation during T helper (Th)17 cell differentiation. This was accompanied by diminished secretion of granulocyte-macrophage colony-stimulating factor (GM-CSF), a cytokine influencing the encephalitogenicity of Th17 cells. Neither the production of the cytokine interleukin (IL)-17 nor the proliferation rate of T cells was affected by the EPE peptide. The in vivo effects of ERK inhibition were challenged in two independent variants of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Overall, ERK inhibition had only a very minor impact on the clinical disease course of EAE. This indicates that while ERK translocation might promote encephalitogenicity in T cells in vitro by facilitating GM-CSF production, this effect is overcome in more complex in vivo animal models of central nervous system (CNS) autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Sistema de Señalización de MAP Quinasas , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Activación de Linfocitos/inmunología , Ratones , Modelos Biológicos , Esclerosis Múltiple/etiología , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Fosforilación , Transporte de Proteínas , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
5.
Acta Neuropathol ; 132(5): 703-720, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27544757

RESUMEN

In multiple sclerosis (MS), the immune cell attack leads to axonal injury as a major cause for neurological disability. Here, we report a novel role of the cell adhesion molecule L1 in the crosstalk between the immune and nervous systems. L1 was found to be expressed by CNS axons of MS patients and human T cells. In MOG35-55-induced murine experimental neuroinflammation, CD4+ T cells were associated with degenerating axons in the spinal cord, both expressing L1. However, neuronal L1 expression in the spinal cord was reduced, while levels of the transcriptional repressor REST (RE1-Silencing Transcription Factor) were up-regulated. In PLP139-151-induced relapsing-remitting neuroinflammation, L1 expression was low at the peak stage of disease, reached almost normal levels in the remission stage, but decreased again during disease relapse indicating adaptive expression regulation of L1. In vitro, activated CD4+ T cells caused contact-dependent down-regulation of L1, up-regulation of its repressor REST and axonal injury in co-cultured neurons. T cell adhesion to neurons and axonal injury were prevented by an antibody blocking L1 suggesting that down-regulation of L1 ameliorates neuroinflammation. In support of this hypothesis, antibody-mediated blocking of L1 in C57BL/6 mice as well as neuron-specific depletion of L1 in synapsinCre × L1fl/fl mice reduces disease severity and axonal pathology despite unchanged immune cell infiltration of the CNS. Our data suggest that down-regulation of neuronal L1 expression is an adaptive process of neuronal self-defense in response to pro-inflammatory T cells, thereby alleviating immune-mediated axonal injury.


Asunto(s)
Regulación hacia Abajo/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/metabolismo , Linfocitos T/fisiología , Anciano , Animales , Axones/efectos de los fármacos , Axones/patología , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/inducido químicamente , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Proteína Proteolipídica de la Mielina/farmacología , Glicoproteína Mielina-Oligodendrócito/farmacología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/genética , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Sinapsinas/genética , Sinapsinas/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/patología
6.
Mult Scler ; 21(14): 1811-22, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25732840

RESUMEN

OBJECTIVE: We aimed to clarify whether fingolimod has direct effects on antigen-presenting cells in multiple sclerosis patients. METHODS: Frequency and phenotype of directly ex vivo dendritic cells and monocytes were analyzed in 43 individuals, including fingolimod-treated and untreated multiple sclerosis patients as well as healthy subjects. These cells were further stimulated with lipopolysaccharide to determine functional effects of fingolimod treatment. RESULTS: Absolute numbers of CD1c+ dendritic cells and monocytes were not significantly reduced in fingolimod-treated patients indicating that fingolimod did not block the migration of antigen-presenting cells to peripheral blood. CD86 was upregulated on CD1c+ dendritic cells and thus their activation was not impaired under fingolimod treatment. Quantitative analyses of gene transcription in cells and protein content in supernatants from ex vivo CD1c+ dendritic cells and monocytes, however, showed lower secretion of TNFα, IL1-ß and IL-6 upon lipopolysaccharide-stimulation. These results could be matched with CD4+MOG-specific transgenic T cells exhibiting reduced levels of TNFα and IFN-γ but not IL-4 upon stimulation with murine dendritic cells loaded with MOG, when treated with fingolimod. CONCLUSIONS: Our data indicate that fingolimod - apart from trapping lymphocytes in lymph nodes - exerts its disease-modulating activity by rebalancing the immune tolerance networks by modulation of antigen-presenting cells.


Asunto(s)
Citocinas/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Clorhidrato de Fingolimod/farmacología , Inmunosupresores/farmacología , Monocitos/efectos de los fármacos , Esclerosis Múltiple/sangre , Esclerosis Múltiple/tratamiento farmacológico , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
7.
Vet Immunol Immunopathol ; 259: 110578, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36965292

RESUMEN

In drug development, the dog is often used as a model for non-rodent preclinical safety studies. In particular, immunophenotyping in dogs can be important to characterize the toxicological profile of a test item. A wide range of antibodies specific to surface antigens is needed, however, commercially available antibodies to dog are scarce. To date, numerous studies have reported the cross-reactivity of human monoclonal antibodies with canine peripheral blood mononuclear cells (PBMC). In this study, we aimed to increase the number of canine-specific antibodies and took a rather novel approach to further determine cross-reactivity of 378 human recombinant antibodies lacking Fc regions to surface antigens on canine PBMC. The screening resulted in 30 human monoclonal antibodies well reactive to canine PBMC. Sequence homology of the targeted human and canine antigens was analyzed with Basic Local Alignment Search Tool. Thirteen human cross-reactive antibodies of interest were analyzed with cells from canine whole blood in combination with lineage markers. Finally, ten antibodies were identified as useful markers for the application in dog. Except for CD27, the remaining nine antibodies are already commercially available human cross-reactive antibodies. This study provides a new source for all ten antibodies described here.


Asunto(s)
Anticuerpos Monoclonales , Leucocitos Mononucleares , Humanos , Perros , Animales , Reacciones Cruzadas , Antígenos de Superficie , Inmunofenotipificación/veterinaria , Citometría de Flujo/veterinaria
8.
J Exp Med ; 203(7): 1761-72, 2006 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-16801398

RESUMEN

The chronic graft-versus-host (cGvH) reaction is a model of induced lupus caused by alloreactive CD4(+) T cells from a Bm-12 mouse in a C57BL/6 recipient. We used this cGvH reaction in C57BL/6 anti-DNA H chain transgenic mice, 56R/B6, to understand the structure, specificity, and origin of the induced autoantibodies (auto-Abs). We found anti-DNA Abs that reacted to several different antigens, such as phosphatidylserine, myelin basic protein, thyroglobulin, histone, insulin, cytochrome C, and beta-galactosidase. This polyreactivity was found for Abs from B cells that expressed the 56R H chain transgene with "editor" L chains that did not completely veto autoreactivity. We suggest that such incomplete editing results in polyreactivity and that incompletely edited polyreactive B cells influence the subsequent expression of pathogenic auto-Abs in disease. We also found B cells that coexpress kappa and lambda L chain. These B cells contributed to the autoimmune response and are possibly in the marginal zone of the spleen.


Asunto(s)
Autoanticuerpos/biosíntesis , Reacción Injerto-Huésped/inmunología , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/genética , Edición de ARN/fisiología , Animales , Enfermedad Crónica , Reacción Injerto-Huésped/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Int Immunol ; 23(6): 391-403, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21602175

RESUMEN

The erbb-2 gene receptor is often over-expressed in human cancer and its overexpression is accompanied by worse prognosis. Targeting erbb-2 gene with antibodies is an effective approach to curtail the progression of erbb-2 gene-expressing cancer types. Two monoclonal antibodies, L-26 and N-12, previously generated in our laboratory, have shown effective tumor inhibition in mice, especially when used in combination. Here, we describe novel peptide mimics of erbb-2 gene protein epitopes, also called mimotopes, that were selected from a constraint random 12-mer peptide phage library, specific for the antibodies L-26 and N-12. Initial sequencing analyses revealed little sequence conservation among the peptide mimotopes, and no sequence homology with the erbb-2 gene protein. However, computational analyses of the two groups of peptides, specific for L-26 and N-12, suggested different epitopes on the erbb-2 gene extracellular domain. In vitro assays showed that the phage displayed peptide mimotopes were specific to their respective antibodies. Selected cyclic peptide mimotopes, but not their corresponding linear equivalents, were able to inhibit binding of the antibodies L-26 and N-12 to the surface of erbb-2 gene-expressing cancer cells in a concentration-dependent manner. In line with this observation, phage-displayed cyclic peptides successfully competed in vitro with recombinant erbb-2 gene protein for binding to their respective antibodies L-26 or N-12. Consistent with the antibody inhibition experiments, we detected specific anti-erbb-2 gene antibodies following vaccination with KLH-coupled cyclic peptides but not with multiple antigenic linear peptides. Potentially, the selected peptides could serve as a starting point for the development of a vaccine against erbb-2 gene over-expressing cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Especificidad de Anticuerpos/inmunología , Epítopos/química , Imitación Molecular/inmunología , Oligopéptidos/química , Oligopéptidos/inmunología , Receptor ErbB-2/inmunología , Animales , Línea Celular Tumoral , Epítopos/análisis , Epítopos/genética , Epítopos/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Oligopéptidos/análisis , Oligopéptidos/genética , Biblioteca de Péptidos , Receptor ErbB-2/química
11.
Physiology (Bethesda) ; 25(2): 85-101, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20430953

RESUMEN

Under physiological conditions, cells receive fate-determining signals from their tissue surroundings, primarily in the form of polypeptide growth factors. Integration of these extracellular signals underlies tissue homeostasis. Although departure from homeostasis and tumor initiation are instigated by oncogenic mutations rather than by growth factors, the latter are the major regulators of all subsequent steps of tumor progression, namely clonal expansion, invasion across tissue barriers, angiogenesis, and colonization of distant niches. Here, we discuss the relevant growth factor families, their roles in tumor biology, as well as the respective downstream signaling pathways. Importantly, cancer-associated activating mutations that impinge on these pathways often relieve, in part, the reliance of tumors on growth factors. On the other hand, growth factors are frequently involved in evolvement of resistance to therapeutic regimens, which extends the roles for polypeptide factors to very late phases of tumor progression and offers opportunities for cancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal/fisiología , Progresión de la Enfermedad , Humanos , Neoplasias/terapia
12.
Proc Natl Acad Sci U S A ; 105(10): 3861-6, 2008 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-18310318

RESUMEN

Tolerance to dsDNA is achieved through editing of Ig receptors that react with dsDNA. Nevertheless, some B cells with anti-dsDNA receptors escape editing and migrate to the spleen. Certain anti-dsDNA B cells that are recovered as hybridomas from the spleens of anti-dsDNA H chain transgenic mice also bind an additional, Golgi-associated antigen. B cells that bind this antigen accumulate intracellular IgM. The intracellular accumulation of IgM is incomplete, because IgM clusters are observed at the cell surface. In the spleen, B cells that express the heavy and light chains encoding this IgM are surface IgM-bright and acquire the CD21-high/CD23-low phenotype of marginal zone B cells. Our data imply that expression of an Ig that binds dsDNA and an additional antigen expressed in the secretory compartment renders B cells resistant to central tolerance. In the periphery, these B cells may be sequestered in the splenic marginal zone.


Asunto(s)
Linfocitos B/inmunología , ADN/inmunología , Reordenamiento Génico de Linfocito B , Animales , Linfocitos B/citología , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Humanos , Hibridomas , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/inmunología , Región Variable de Inmunoglobulina/inmunología , Células Jurkat , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fosfatidilserinas/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Autotolerancia
13.
J Neuroimmune Pharmacol ; 12(2): 305-313, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27832402

RESUMEN

Multiple sclerosis is a chronic, disseminated inflammation of the central nervous system which is thought to be driven by autoimmune T cells. Genetic association studies in multiple sclerosis and a large number of studies in the animal model of the disease support a role for effector/memory T helper cells. However, the mechanisms underlying relapses, remission and chronic progression in multiple sclerosis or the animal model experimental autoimmune encephalomyelitis, are not clear. In particular, there is only scarce information on the role of central nervous system-invading naive T helper cells in these processes. By applying two-photon laser scanning microscopy we could show in vivo that antigen unexperienced T helper cells migrated into the deep parenchyma of the inflamed central nervous system in experimental autoimmune encephalomyelitis, independent of their antigen specificity. Using flow cytometric analyses of central nervous system-derived lymphocytes we found that only antigen-specific, formerly naive T helper cells became activated during inflammation of the central nervous system encountering their corresponding antigen.


Asunto(s)
Antígenos/metabolismo , Sistema Nervioso Central/metabolismo , Encefalomielitis Autoinmune Experimental/metabolismo , Fenotipo , Linfocitos T Colaboradores-Inductores/metabolismo , Animales , Antígenos/inmunología , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Técnicas de Cocultivo , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Linfocitos T Colaboradores-Inductores/inmunología
14.
J Neuroimmune Pharmacol ; 11(4): 721-732, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27423192

RESUMEN

The importance of CD11c+ antigen-presenting cells (APCs) in the pathogenesis of experimental autoimmune encephalomyelitis (EAE) is well accepted and the gate keeper function of perivascular CD11c+ APCs has been demonstrated. CD11c can be expressed by APCs from external sources or by central nervous system (CNS) resident APCs such as microglia. Yet, changes in the gene expression pattern of CNS CD11c+ APCs during disease are still unclear and differentially expressed genes might play a decisive role in EAE progression. Due to their low numbers in the diseased brain and due to the absence of considerable numbers in the healthy CNS, analysis of CNS CD11c+ cells is technically difficult. To ask whether the CD11c+ APC population contributes to remission of EAE disease, we used Illumina deep mRNA sequencing (RNA-Seq) and quantitative real time polymerase chain reaction (qRT-PCR) analyses to identify the transcriptome of CD11c+ APCs during disease course. We identified a battery of genes that were significantly regulated during the exacerbation of the disease compared to remission and relapse. Three of these genes, Arginase-1, Chi3l3 and Ms4a8a, showed a higher expression at the exacerbation than at later time points during the disease, both in SJL/J and in C57BL/6 mice, and could be attributed to alternatively activated APCs. Expression of Arginase-1, Chi3l3 and Ms4a8a genes was linked to the disease phase of EAE rather than to disease score. Expression of these genes suggested that APCs resembling alternatively activated macrophages are involved during the first wave of neuroinflammation and can be directly associated with the disease progress.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Antígeno CD11c/biosíntesis , Encefalomielitis Autoinmune Experimental/metabolismo , Redes Reguladoras de Genes/fisiología , Animales , Antígeno CD11c/genética , Células Dendríticas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/patología , Ratones , Ratones Endogámicos C57BL
15.
J Immunol ; 180(6): 3708-18, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18322176

RESUMEN

Most mature B lymphocytes express one BCR L chain, kappa or lambda, but recent work has shown that there are exceptions in that some B lymphocytes express both kappa and lambda and some even bear two different kappa L chains. Using the anti-DNA H chain-transgenic mouse, 56R, we find that B cells with pre-existing autoreactivity are especially subject to L chain inclusion. Specifically, we show that isotypic and allelic inclusion enables autoreactive B cells to bypass central tolerance giving rise to B cells that retain dangerous features. One receptor in dual receptor B cells is an editor L chain, i.e., neutralizes or alters self-reactivity of the 56R H chain transgene. We compare the 56R mouse when on the C57/BL/6 background, a strain prone to autoimmunity, with that of 56R when on the BALB/c background, a strain that resists autoimmunity. In the B6.56R mouse, polyreactive B cells with dual L chain move to the follicular B cell compartment. Their localization in the follicular compartment may explain the ease with which B cells in the B6.56R differentiate into autoantibody-producing plasma cells. Likewise, in the BALB/c.56R mouse, dual L chain B cells are found in the follicular B cell compartment. Yet, the lack of autoantibody-producing plasma cells in the BALB/c.56R suggests that postfollicular tolerance checkpoints are intact. The Jkappa usage in dual kappa L chain B cells suggests increased receptor editing activity and is consistent with the expected distribution of Jkappa genes in our computational model for random selection of Jkappa.


Asunto(s)
Alelos , Anticuerpos Antinucleares/genética , Subgrupos de Linfocitos B/inmunología , Reordenamiento Génico de Cadena Ligera de Linfocito B , Isotipos de Inmunoglobulinas/genética , Cadenas kappa de Inmunoglobulina/genética , Cadenas lambda de Inmunoglobulina/genética , Edición de ARN/genética , Animales , Subgrupos de Linfocitos B/metabolismo , Regiones Constantes de Inmunoglobulina/biosíntesis , Regiones Constantes de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/biosíntesis , Región Variable de Inmunoglobulina/biosíntesis , Región Variable de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/biosíntesis , Cadenas lambda de Inmunoglobulina/biosíntesis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Edición de ARN/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Bazo/citología , Bazo/inmunología , Bazo/metabolismo
16.
Immunology ; 118(3): 353-60, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16827896

RESUMEN

CD4+ CD45RO+ T cells could mature freshly isolated human plasmacytoid dendritic cells (PDC) in a superantigen-driven culture in a similar way to recombinant interleukin-3 (IL-3). Mature PDC expressed significantly higher levels of inducible costimulator ligand (ICOS-L), but similar levels of CD80 and CD86, when compared to mature monocyte-derived DC (moDC). We therefore directly compared the capacities of mature PDC and moDC to activate T cells. A similar T helper type 1 (Th1)/Th2 pattern of cytokines was generated in both systems, but significantly higher levels of IL-3, IL-4 and IL-10 were induced by PDC. In T cells interacting with PDC, the ICOS/ICOS-L costimulatory pathway played a pre-eminent role in the generation of IL-3 and IL-10, CD28 was central to the induction of IL-2, and both pathways were equally important for the generation of other cytokines. In cocultures with moDC, the CD28 pathway was dominant over ICOS under all circumstances, except for the ICOS-mediated release of IL-10. In general, our data demonstrate an eminent role of ICOS in the interaction of T cells with PDC, and thus modify the current paradigm of CD28 dominance for the costimulation of T cells interacting with professional antigen-presenting cells. In particular, our data highlight the role of ICOS in the generation of IL-3, a factor central to the biology of human PDC.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Antígenos CD , Antígenos CD28/inmunología , Ligando de CD40/inmunología , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Técnicas de Cocultivo , Citocinas/biosíntesis , Humanos , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Interleucina-10/biosíntesis , Interleucina-3/biosíntesis , Interleucina-3/inmunología , Antígenos Comunes de Leucocito/análisis , Activación de Linfocitos/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas/metabolismo , Regulación hacia Arriba/inmunología
17.
Eur J Immunol ; 32(9): 2680-6, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12207353

RESUMEN

With newly generated ICOS-ligand (ICOS-L)-specific monoclonal antibodies we determined that human Langerhans cells in situ express similar levels of ICOS-L, CD80, and CD86, compared to immature dendritic cells (DC) derived from monocytes in vitro. Maturation of DC strongly up-regulated CD80 and CD86 but did not significantly change ICOS-L levels. On coculture of "naive"CD4(+) T cells with mature DC in the presence of superantigen, ICOS was highly up-regulated on T cells, but played only a secondary role in the CD28-dominated release of TNF-alpha and IFN-gamma, and did not participate in the induction of IL-2. Cocultures of "effector" CD4(+) T cells with mature DC revealed CD28 as the driving force for the secretion of IL-2, IFN-gamma, IL-6, and IL-13, with no apparent contribution of ICOS. In contrast, the release of IL-10 was differentially regulated. Interaction of ICOS with ICOS-L strongly promoted IL-10 secretion, whereas the CD28/B7 pathway acted as a potent attenuator of IL-10 release. Our data thus indicate a selective regulation of IL-10 secretion by ICOS on re-activation of effector T cells with professional antigen-presenting cells (bearing CD80 and CD86) in lymphoid tissue.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/fisiología , Antígenos CD28/fisiología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Inmunoconjugados , Interleucina-10/biosíntesis , Activación de Linfocitos , Proteínas/fisiología , Subgrupos de Linfocitos T/inmunología , Abatacept , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antígenos CD/biosíntesis , Antígenos CD/genética , Antígenos de Diferenciación/inmunología , Antígeno B7-1/biosíntesis , Antígeno B7-1/genética , Antígeno B7-2 , Linfocitos T CD4-Positivos/metabolismo , Antígeno CTLA-4 , Diferenciación Celular/efectos de los fármacos , Células Cultivadas/efectos de los fármacos , Células Dendríticas/citología , Células Epidérmicas , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hibridomas/inmunología , Inmunoglobulina G/inmunología , Ligando Coestimulador de Linfocitos T Inducibles , Proteína Coestimuladora de Linfocitos T Inducibles , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucinas/metabolismo , Células de Langerhans/química , Células de Langerhans/citología , Células de Langerhans/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Ratones , Monocitos/citología , Monocitos/efectos de los fármacos , Biosíntesis de Proteínas , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA