Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Allergy Clin Immunol ; 142(1): 235-245.e6, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29705247

RESUMEN

BACKGROUND: X-linked lymphoproliferative disease 1 arises from mutations in the SH2D1A gene encoding SLAM-associated protein (SAP), an adaptor protein expressed in T, natural killer (NK), and NKT cells. Defects lead to abnormalities of T-cell and NK cell cytotoxicity and T cell-dependent humoral function. Clinical manifestations include hemophagocytic lymphohistiocytosis, lymphoma, and dysgammaglobulinemia. Curative treatment is limited to hematopoietic stem cell transplantation, with outcomes reliant on a good donor match. OBJECTIVES: Because most symptoms arise from defective T-cell function, we investigated whether transfer of SAP gene-corrected T cells could reconstitute known effector cell defects. METHODS: CD3+ lymphocytes from Sap-deficient mice were transduced with a gammaretroviral vector encoding human SAP cDNA before transfer into sublethally irradiated Sap-deficient recipients. After immunization with the T-dependent antigen 4-hydroxy-3-nitrophenylacetly chicken gammaglobulin (NP-CGG), recovery of humoral function was evaluated through germinal center formation and antigen-specific responses. To efficiently transduce CD3+ cells from patients, we generated an equivalent lentiviral SAP vector. Functional recovery was demonstrated by using in vitro cytotoxicity and T follicular helper cell function assays alongside tumor clearance in an in vivo lymphoblastoid cell line lymphoma xenograft model. RESULTS: In Sap-deficient mice 20% to 40% engraftment of gene-modified T cells led to significant recovery of germinal center formation and NP-specific antibody responses. Gene-corrected T cells from patients demonstrated improved cytotoxicity and T follicular helper cell function in vitro. Adoptive transfer of gene-corrected cytotoxic T lymphocytes from patients reduced tumor burden to a level comparable with that seen in healthy donor cytotoxic T lymphocytes in an in vivo lymphoma model. CONCLUSIONS: These data demonstrate that autologous T-cell gene therapy corrects SAP-dependent defects and might offer an alternative therapeutic option for patients with X-linked lymphoproliferative disease 1.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Trastornos Linfoproliferativos , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria/genética , Linfocitos T Citotóxicos/trasplante , Animales , Xenoinjertos , Humanos , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/inmunología , Ratones
2.
J Cell Sci ; 129(8): 1734-49, 2016 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-26940916

RESUMEN

Rab8 is a small Ras-related GTPase that regulates polarized membrane transport to the plasma membrane. Here, we developed a high-content analysis (HCA) tool to dissect Rab8-mediated actin and focal adhesion reorganization that revealed that Rab8 activation significantly induced Rac1 and Tiam1 to mediate cortical actin polymerization and RhoA-dependent stress fibre disassembly. Rab8 activation increased Rac1 activity, whereas its depletion activated RhoA, which led to reorganization of the actin cytoskeleton. Rab8 was also associated with focal adhesions, promoting their disassembly in a microtubule-dependent manner. This Rab8 effect involved calpain, MT1-MMP (also known as MMP14) and Rho GTPases. Moreover, we demonstrate the role of Rab8 in the cell migration process. Indeed, Rab8 is required for EGF-induced cell polarization and chemotaxis, as well as for the directional persistency of intrinsic cell motility. These data reveal that Rab8 drives cell motility by mechanisms both dependent and independent of Rho GTPases, thereby regulating the establishment of cell polarity, turnover of focal adhesions and actin cytoskeleton rearrangements, thus determining the directionality of cell migration.


Asunto(s)
Calpaína/metabolismo , Adhesiones Focales/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Metaloproteinasa 14 de la Matriz/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Citoesqueleto de Actina/metabolismo , Movimiento Celular , Polaridad Celular , Células HeLa , Humanos , ARN Interferente Pequeño/genética , Fibras de Estrés/metabolismo , Proteína 1 de Invasión e Inducción de Metástasis del Linfoma-T , Proteínas de Unión al GTP rab/genética , Proteína de Unión al GTP rhoA/metabolismo
3.
Mult Scler ; 23(13): 1762-1771, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27903935

RESUMEN

OBJECTIVE: To assess the decision-making impairment in patients with multiple sclerosis (MS) and how they relate to other cognitive domains. METHODS: We performed a cross-sectional analysis in 84 patients with MS, and 21 matched healthy controls using four tasks taken from behavioral economics: (1) risk preferences, (2) choice consistency, (3) delay of gratification, and (4) rate of learning. All tasks were conducted using real-world reward outcomes (food or money) in different real-life conditions. Participants underwent cognitive examination using the Brief Repeatable Battery-Neuropsychology. RESULTS: Patients showed higher risk aversion (general propensity to choose the lottery was 0.51 vs 0.64, p = 0.009), a trend to choose more immediate rewards over larger but delayed rewards ( p = 0.108), and had longer reactions times ( p = 0.033). Choice consistency and learning rates were not different between groups. Progressive patients chose slower than relapsing patients. In relation to general cognitive impairments, we found correlations between impaired decision-making and impaired verbal memory ( r = 0.29, p = 0.009), visual memory ( r = -0.37, p = 0.001), and reduced processing speed ( r = -0.32, p = 0.001). Normalized gray matter volume correlated with deliberation time ( r = -0.32, p = 0.005). CONCLUSION: Patients with MS suffer significant decision-making impairments, even at the early stages of the disease, and may affect patients' quality and social life.


Asunto(s)
Disfunción Cognitiva/fisiopatología , Toma de Decisiones/fisiología , Aprendizaje/fisiología , Esclerosis Múltiple/fisiopatología , Asunción de Riesgos , Adulto , Conducta de Elección/fisiología , Disfunción Cognitiva/etiología , Estudios Transversales , Descuento por Demora/fisiología , Economía del Comportamiento , Femenino , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/complicaciones
4.
J Pathol ; 236(2): 219-28, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25664618

RESUMEN

BCR-JAK2 is an infrequent gene fusion found in chronic/acute, myeloid/lymphoid Philadelphia chromosome-negative leukaemia. In this study, we demonstrated that in vivo expression of BCR-JAK2 in mice induces neoplasia, with fatal consequences. Transplantation of BCR-JAK2 bone marrow progenitors promoted splenomegaly, with megakaryocyte infiltration and elevated leukocytosis of myeloid origin. Analysis of peripheral blood revealed the presence of immature myeloid cells, platelet aggregates and ineffective erythropoiesis. A possible molecular mechanism for these observations involved inhibition of apoptosis by deregulated expression of the anti-apoptotic mediator Bcl-xL and the serine/threonine kinase Pim1. Together, these data provide a suitable in vivo molecular mechanism for leukaemia induction by BCR-JAK2 that validates the use of this model as a relevant preclinical tool for the design of new targeted therapies in Philadelphia chromosome-negative leukaemia involving BCR-JAK2-driven activation of the JAK2 pathway.


Asunto(s)
Janus Quinasa 2/fisiología , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Proteínas Proto-Oncogénicas c-bcr/fisiología , Animales , Femenino , Reordenamiento Génico , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/fisiología , Janus Quinasa 2/genética , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/mortalidad , Leucocitosis/etiología , Masculino , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-bcr/genética , Retroviridae , Factor de Transcripción STAT5/metabolismo , Esplenomegalia/etiología , Transducción Genética/métodos , Transgenes
5.
Mol Ther Methods Clin Dev ; 26: 459-470, 2022 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-36092365

RESUMEN

Leukocyte adhesion deficiency type I (LAD-I) is a primary immunodeficiency caused by mutations in the ITGB2 gene, which encodes for the CD18 subunit of ß2-integrins. Deficient expression of ß2-integrins results in impaired neutrophil migration in response to bacterial and fungal infections. Using a lentiviral vector (LV) that mediates a preferential myeloid expression of human CD18 (Chim.hCD18-LV), we first demonstrated that gene therapy efficiently corrected the phenotype of mice with severe LAD-I. Next, we investigated if the ectopic hCD18 expression modified the phenotypic characteristics of human healthy donor hematopoietic stem cells and their progeny. Significantly, transduction of healthy CD34+ cells with the Chim.hCD18-LV did not modify the membrane expression of CD18 nor the adhesion of physiological ligands to transduced cells. Additionally, we observed that the repopulating properties of healthy CD34+ cells were preserved following transduction with the Chim.hCD18-LV, and that a safe polyclonal repopulation pattern was observed in transplanted immunodeficient NOD scid gamma (NSG) mice. In a final set of experiments, we demonstrated that transduction of CD34+ cells from a severe LAD-I patient with the Chim.hCD18-LV restores the expression of ß2-integrins in these cells. These results offer additional preclinical safety and efficacy evidence supporting the gene therapy of patients with severe LAD-I.

6.
Cell Stem Cell ; 25(5): 607-621.e7, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31543367

RESUMEN

Non-homologous end-joining (NHEJ) is the preferred mechanism used by hematopoietic stem cells (HSCs) to repair double-stranded DNA breaks and is particularly increased in cells deficient in the Fanconi anemia (FA) pathway. Here, we show feasible correction of compromised functional phenotypes in hematopoietic cells from multiple FA complementation groups, including FA-A, FA-C, FA-D1, and FA-D2. NHEJ-mediated repair of targeted CRISPR-Cas9-induced DNA breaks generated compensatory insertions and deletions that restore the coding frame of the mutated gene. NHEJ-mediated editing efficacy was initially verified in FA lymphoblastic cell lines and then in primary FA patient-derived CD34+ cells, which showed marked proliferative advantage and phenotypic correction both in vitro and after transplantation. Importantly, and in contrast to homologous directed repair, NHEJ efficiently targeted primitive human HSCs, indicating that NHEJ editing approaches may constitute a sound alternative for editing self-renewing human HSCs and consequently for treatment of FA and other monogenic diseases affecting the hematopoietic system.


Asunto(s)
Sistemas CRISPR-Cas/genética , Reparación del ADN por Unión de Extremidades/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Edición Génica/métodos , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas , Alelos , Animales , Antígenos CD34/metabolismo , Línea Celular , Proliferación Celular/genética , Roturas del ADN de Doble Cadena , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación INDEL , Ratones , Ratones Endogámicos NOD , Ratones Desnudos
7.
Neurotherapeutics ; 16(3): 808-827, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30815844

RESUMEN

The development of neuroprotective therapies is a sought-after goal. By screening combinatorial chemical libraries using in vitro assays, we identified the small molecule BN201 that promotes the survival of cultured neural cells when subjected to oxidative stress or when deprived of trophic factors. Moreover, BN201 promotes neuronal differentiation, the differentiation of precursor cells to mature oligodendrocytes in vitro, and the myelination of new axons. BN201 modulates several kinases participating in the insulin growth factor 1 pathway including serum-glucocorticoid kinase and midkine, inducing the phosphorylation of NDRG1 and the translocation of the transcription factor Foxo3 to the cytoplasm. In vivo, BN201 prevents axonal and neuronal loss, and it promotes remyelination in models of multiple sclerosis, chemically induced demyelination, and glaucoma. In summary, we provide a new promising strategy to promote neuroaxonal survival and remyelination, potentially preventing disability in brain diseases.


Asunto(s)
Amidas/uso terapéutico , Axones/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Vaina de Mielina/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Peptoides/uso terapéutico , Pirrolidinonas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Técnica del Anticuerpo Fluorescente , Glaucoma/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Nervio Óptico/efectos de los fármacos , Proguanil , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Triazinas
8.
EMBO Mol Med ; 9(11): 1574-1588, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28899930

RESUMEN

Gene targeting constitutes a new step in the development of gene therapy for inherited diseases. Although previous studies have shown the feasibility of editing fibroblasts from Fanconi anemia (FA) patients, here we aimed at conducting therapeutic gene editing in clinically relevant cells, such as hematopoietic stem cells (HSCs). In our first experiments, we showed that zinc finger nuclease (ZFN)-mediated insertion of a non-therapeutic EGFP-reporter donor in the AAVS1 "safe harbor" locus of FA-A lymphoblastic cell lines (LCLs), indicating that FANCA is not essential for the editing of human cells. When the same approach was conducted with therapeutic FANCA donors, an efficient phenotypic correction of FA-A LCLs was obtained. Using primary cord blood CD34+ cells from healthy donors, gene targeting was confirmed not only in in vitro cultured cells, but also in hematopoietic precursors responsible for the repopulation of primary and secondary immunodeficient mice. Moreover, when similar experiments were conducted with mobilized peripheral blood CD34+ cells from FA-A patients, we could demonstrate for the first time that gene targeting in primary hematopoietic precursors from FA patients is feasible and compatible with the phenotypic correction of these clinically relevant cells.


Asunto(s)
Antígenos CD34/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/patología , Edición Génica/métodos , Células Madre Hematopoyéticas/metabolismo , Animales , Secuencia de Bases , Células Cultivadas , Dependovirus/genética , Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Sangre Fetal/citología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Especies Reactivas de Oxígeno/metabolismo , Nucleasas con Dedos de Zinc/genética , Nucleasas con Dedos de Zinc/metabolismo
9.
EMBO Mol Med ; 6(6): 835-48, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24859981

RESUMEN

Gene targeting is progressively becoming a realistic therapeutic alternative in clinics. It is unknown, however, whether this technology will be suitable for the treatment of DNA repair deficiency syndromes such as Fanconi anemia (FA), with defects in homology-directed DNA repair. In this study, we used zinc finger nucleases and integrase-defective lentiviral vectors to demonstrate for the first time that FANCA can be efficiently and specifically targeted into the AAVS1 safe harbor locus in fibroblasts from FA-A patients. Strikingly, up to 40% of FA fibroblasts showed gene targeting 42 days after gene editing. Given the low number of hematopoietic precursors in the bone marrow of FA patients, gene-edited FA fibroblasts were then reprogrammed and re-differentiated toward the hematopoietic lineage. Analyses of gene-edited FA-iPSCs confirmed the specific integration of FANCA in the AAVS1 locus in all tested clones. Moreover, the hematopoietic differentiation of these iPSCs efficiently generated disease-free hematopoietic progenitors. Taken together, our results demonstrate for the first time the feasibility of correcting the phenotype of a DNA repair deficiency syndrome using gene-targeting and cell reprogramming strategies.


Asunto(s)
Reprogramación Celular , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Marcación de Gen , Células Cultivadas , Fibroblastos/citología , Fibroblastos/metabolismo , Terapia Genética/métodos , Hematopoyesis , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo
10.
PLoS One ; 9(3): e90671, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599318

RESUMEN

BACKGROUND: 5'-deoxy-5'-methylthioadenosine (MTA) is an endogenous compound produced through the metabolism of polyamines. The therapeutic potential of MTA has been assayed mainly in liver diseases and, more recently, in animal models of multiple sclerosis. The aim of this study was to determine the neuroprotective effect of this molecule in vitro and to assess whether MTA can cross the blood brain barrier (BBB) in order to also analyze its potential neuroprotective efficacy in vivo. METHODS: Neuroprotection was assessed in vitro using models of excitotoxicity in primary neurons, mixed astrocyte-neuron and primary oligodendrocyte cultures. The capacity of MTA to cross the BBB was measured in an artificial membrane assay and using an in vitro cell model. Finally, in vivo tests were performed in models of hypoxic brain damage, Parkinson's disease and epilepsy. RESULTS: MTA displays a wide array of neuroprotective activities against different insults in vitro. While the data from the two complementary approaches adopted indicate that MTA is likely to cross the BBB, the in vivo data showed that MTA may provide therapeutic benefits in specific circumstances. Whereas MTA reduced the neuronal cell death in pilocarpine-induced status epilepticus and the size of the lesion in global but not focal ischemic brain damage, it was ineffective in preserving dopaminergic neurons of the substantia nigra in the 1-methyl-4-phenyl-1,2,3,6-tetrahydro-pyridine (MPTP)-mice model. However, in this model of Parkinson's disease the combined administration of MTA and an A2A adenosine receptor antagonist did produce significant neuroprotection in this brain region. CONCLUSION: MTA may potentially offer therapeutic neuroprotection.


Asunto(s)
Desoxiadenosinas/farmacología , Fármacos Neuroprotectores/farmacología , Tionucleósidos/farmacología , Enfermedad Aguda , Antagonistas Adrenérgicos/farmacología , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Permeabilidad de la Membrana Celular , Células Cultivadas , Enfermedad Crónica , Desoxiadenosinas/uso terapéutico , Modelos Animales de Enfermedad , Glucosa/deficiencia , Masculino , Ratones , N-Metilaspartato/toxicidad , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/patología , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Oxígeno , Pilocarpina , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/patología , Tionucleósidos/uso terapéutico , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/toxicidad
11.
PLoS One ; 8(2): e54722, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23431360

RESUMEN

BACKGROUND: Demyelination and axonal damage are critical processes in the pathogenesis of multiple sclerosis (MS). Oxidative stress and pro-inflammatory cytokines elicited by inflammation mediates tissue damage. METHODS/PRINCIPAL FINDINGS: To monitor the demyelination and axonal injury associated with microglia activation we employed a model using cerebellar organotypic cultures stimulated with lipopolysaccharide (LPS). Microglia activated by LPS released pro-inflammatory cytokines (IL-1ß, IL-6 and TNFα), and increased the expression of inducible nitric oxide synthase (iNOS) and production of reactive oxygen species (ROS). This activation was associated with demyelination and axonal damage in cerebellar cultures. Axonal damage, as revealed by the presence of non-phosphorylated neurofilaments, mitochondrial accumulation in axonal spheroids, and axonal transection, was associated with stronger iNOS expression and concomitant increases in ROS. Moreover, we analyzed the contribution of pro-inflammatory cytokines and oxidative stress in demyelination and axonal degeneration using the iNOS inhibitor ethyl pyruvate, a free-scavenger and xanthine oxidase inhibitor allopurinol, as well as via blockage of pro-inflammatory cytokines using a Fc-TNFR1 construct. We found that blocking microglia activation with ethyl pyruvate or allopurinol significantly decreased axonal damage, and to a lesser extent, demyelination. Blocking TNFα significantly decreased demyelination but did not prevented axonal damage. Moreover, the most common therapy for MS, interferon-beta, was used as an example of an immunomodulator compound that can be tested in this model. In vitro, interferon-beta treatment decreased oxidative stress (iNOS and ROS levels) and the release of pro-inflammatory cytokines after LPS stimulation, reducing axonal damage. CONCLUSION: The model of neuroinflammation using cerebellar culture stimulated with endotoxin mimicked myelin and axonal damage mediated by the combination of oxidative stress and pro-inflammatory cytokines. This model may both facilitate understanding of the events involved in neuroinflammation and aid in the development of neuroprotective therapies for the treatment of MS and other neurodegenerative diseases.


Asunto(s)
Citocinas/metabolismo , Enfermedades Desmielinizantes/metabolismo , Mediadores de Inflamación/metabolismo , Neuritis/metabolismo , Estrés Oxidativo , Alopurinol/farmacología , Animales , Axones/inmunología , Axones/patología , Cerebelo/inmunología , Cerebelo/metabolismo , Cerebelo/patología , Enfermedades Desmielinizantes/inmunología , Depuradores de Radicales Libres/farmacología , Interferón beta/farmacología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/inmunología , Vaina de Mielina/patología , Neuritis/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oligodendroglía/fisiología , Piruvatos/farmacología , Técnicas de Cultivo de Tejidos , Factor de Necrosis Tumoral alfa/metabolismo
13.
Eur J Immunol ; 38(2): 576-86, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18200504

RESUMEN

T regulatory cells type 1 (Tr1 cells) are excellent candidates for cell therapy in multiple sclerosis (MS). The aim of our study was to assess the functional state of Tr1 cells and IL-10R signaling in patients with MS. Tr1 cells were induced in vitro by activation with anti-CD46 antibodies in controls and patients with MS. Cells were phenotyped by cytometry and suppression assays, and the expression of cytokines and transcription factors was evaluated by real-time PCR, ELISA, cytometry and Western blotting. We found that the activity of Tr1 cells and IL-10R signaling is impaired in MS patients since Tr1 cells isolated from MS patients produced less IL-10 than those obtained from controls. Indeed, the supernatants from Tr1 cells from controls did not suppress the proliferation of stimulated CD4(+) cells from patients with MS. Furthermore, the IL-10R signaling pathway was not fully active in CD4(+) cells from MS patients and these cells had higher baseline levels of SOCS3 transcripts than controls. Indeed, after in vitro IL-10 stimulation, the expression levels of the STAT1, STAT3 and IL-10RA genes were higher in MS patients than in controls. Moreover, Stat-3 phosphorylation was lower in controls than in patients after IL-10 stimulation. These results indicate that IL-10 regulatory function is impaired in patients with MS.


Asunto(s)
Interleucina-10/fisiología , Esclerosis Múltiple/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Adulto , Células Cultivadas , Regulación hacia Abajo/inmunología , Femenino , Humanos , Tolerancia Inmunológica , Interleucina-10/antagonistas & inhibidores , Masculino , Esclerosis Múltiple/patología , Receptores de Interleucina-10/antagonistas & inhibidores , Receptores de Interleucina-10/metabolismo , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA