Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Front Immunol ; 10: 829, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31040853

RESUMEN

Determining antigen specificity is vital for understanding B cell biology and for producing human monoclonal antibodies. We describe here a powerful method for identifying B cells that recognize membrane antigens expressed on cells. The technique depends on two characteristics of the interaction between a B cell and an antigen-expressing cell: antigen-receptor-mediated extraction of antigen from the membrane of the target cell, and B cell activation. We developed the method using influenza hemagglutinin as a model viral membrane antigen, and tested it using acetylcholine receptor (AChR) as a model membrane autoantigen. The technique involves co-culturing B cells with adherent, bioorthogonally labeled cells expressing GFP-tagged antigen, and sorting GFP-capturing, newly activated B cells. Hemagglutinin-specific B cells isolated this way from vaccinated human donors expressed elevated CD20, CD27, CD71, and CD11c, and reduced CD21, and their secreted antibodies blocked hemagglutination and neutralized viral infection. Antibodies cloned from AChR-capturing B cells derived from patients with myasthenia gravis bound specifically to the receptor on cell membrane. The approach is sensitive enough to detect antigen-specific B cells at steady state, and can be adapted for any membrane antigen.


Asunto(s)
Antígenos de Superficie/inmunología , Linfocitos B/inmunología , Separación Celular/métodos , Adulto , Anciano , Animales , Antígenos de Superficie/aislamiento & purificación , Autoantígenos/inmunología , Autoantígenos/aislamiento & purificación , Subgrupos de Linfocitos B/inmunología , Línea Celular Tumoral , Células Clonales , Epítopos de Linfocito B/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Inmunofenotipificación , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miastenia Gravis/inmunología , Receptores Colinérgicos/inmunología
2.
J Neuroimmunol ; 326: 19-27, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30447419

RESUMEN

Two validated assays, a bridging ELISA and a luciferase-based bioassay, were compared for detection of anti-drug antibodies (ADA) against interferon-beta (IFN-ß) in patients with multiple sclerosis. Serum samples were tested from patients enrolled in a prospective study of 18 months. In contrast to the ELISA, when IFN-ß-specific rabbit polyclonal and human monoclonal antibodies were tested, the bioassay was the more sensitive to detect IFN-ß ADA in patients' sera. For clinical samples, selection of method of ELISA should be evaluated prior to the use of a multi-tiered approach. A titer threshold value is reported that may be used as a predictor for persistently positive neutralizing ADA.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Esclerosis Múltiple/sangre , Pruebas de Neutralización/métodos , Bioensayo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Factores Inmunológicos/inmunología , Factores Inmunológicos/uso terapéutico , Interferón beta/inmunología , Interferón beta/uso terapéutico , Masculino , Esclerosis Múltiple/tratamiento farmacológico
3.
J Autoimmun ; 86: 39-50, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28958667

RESUMEN

INTRODUCTION: The mode of action of dimethyl fumarate (DMF), an immunomodulatory treatment for relapsing-remitting multiple sclerosis (RRMS), has not yet been fully elucidated. While in-vitro experiments and animal studies suggest effects on immune cell survival, proliferation, migration and oxidative stress response, corresponding observations from human studies are lacking. This study aims to characterize ex-vivo and in-vivo effects in a cohort of DMF treated RRMS patients. METHODS: Blood samples were collected from twenty well-characterized RRMS patients at baseline and after 3, 6 and 12 months of DMF treatment and an age- and gender-matched cohort of 20 healthy individuals at 0 and 3 months. Leukocyte subpopulations, immunoglobulin levels and cytokine secretion were measured. T cells were assessed for their levels of reactive oxygen species (ROS), metabolic status and their proliferative capacity. Levels of antioxidants were determined in serum by mass spectrometry. Responses of monocyte activation markers as well as NFkB and MAPK pathways to DMF were analysed. RESULTS: Upon DMF treatment, all lymphocyte subpopulations dropped significantly over the course of 12 months with cytotoxic and effector T cells being affected most significantly. DMF induced cell death and inhibited proliferation of T cells in-vitro. Interestingly, this anti-proliferative effect decreased under treatment. In-vivo DMF treatment led to decreased T cell glycolysis and higher turn-over of antioxidants. In line with these results a significant increase of cytosolic ROS levels after 3 months treatment was detected in T cells. In-vitro DMF treatment reduced NFkB (p65) translocation to the nucleus and MAPK (p38) levels decreased upon stimulation with monomethyl fumarate (MMF) in-vitro and ex-vivo. Consequently, the expression of co-stimulatory molecules like CD40 and CD150 was decreased in antigen presenting cells both in-vitro and ex-vivo. CONCLUSION: This study translates knowledge from in-vitro and animal studies on DMF into the clinical setting. Our data suggest that DMF not only alters lymphocyte composition, but also has profound effects on proliferation and induces oxidative stress in T cells. It also acts on innate immunity by reducing the activation status of antigen presenting cells (APCs) via NFkB and MAPK inactivation.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Dimetilfumarato/uso terapéutico , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Inmunidad Adaptativa , Adulto , Presentación de Antígeno , Células Presentadoras de Antígenos/efectos de los fármacos , Muerte Celular , Proliferación Celular , Células Cultivadas , Estudios de Cohortes , Femenino , Glucólisis , Humanos , Inmunidad Innata , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/efectos de los fármacos
4.
Proc Natl Acad Sci U S A ; 114(4): 734-739, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28057865

RESUMEN

Autoantibodies against myelin oligodendrocyte glycoprotein (MOG) are associated with autoimmune central nervous system diseases like acute disseminated encephalomyelitis (ADEM). For ADEM, it is speculated that a preceding infection is the trigger of the autoimmune response, but the mechanism connecting the infection to the production of MOG antibodies remains a mystery. We reasoned that the ability of B cells to capture cognate antigen from cell membranes, along with small quantities of coexpressed "bystander" antigens, might enable B-cell escape from tolerance. We tested this hypothesis using influenza hemagglutinin as a model viral antigen and transgenic, MOG-specific B cells. Using flow cytometry and live and fixed cell microscopy, we show that MOG-specific B cells take up large amounts of MOG from cell membranes. Uptake of the antigen from the membrane leads to a strong activation of the capturing B cell. When influenza hemagglutinin is also present in the membrane of the target cell, it can be cocaptured with MOG by MOG-specific B cells via the B-cell receptor. Hemagglutinin and MOG are both presented to T cells, which in turn are activated and proliferate. As a consequence, MOG-specific B cells get help from hemagglutinin-specific T cells to produce anti-MOG antibodies. In vivo, the transfer of MOG-specific B cells into recipient mice after the cocapture of MOG and hemagglutinin leads to the production of class-switched anti-MOG antibodies, dependent on the presence of hemagglutinin-specific T cells. This mechanism offers a link between infection and autoimmunity.


Asunto(s)
Antígenos Virales/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Animales , Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Línea Celular , Membrana Celular/inmunología , Células HEK293 , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Linfocitos T/inmunología
5.
Neurol Neuroimmunol Neuroinflamm ; 3(3): e223, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27088119

RESUMEN

OBJECTIVES: To assess messenger RNA (mRNA) expression of POU2AF1 and Spi-B and their potential regulatory microRNAs (miRNAs) in natalizumab-treated patients with multiple sclerosis and in therapy-associated progressive multifocal leukoencephalopathy (PML). METHODS: Expression of POU2AF1/Spi-B was analyzed by using real-time reverse transcription PCR assays on isolated B/CD8(+) T lymphocytes and peripheral blood mononuclear cells (PBMCs) from cohorts of untreated and natalizumab-treated patients with and without PML. Longitudinal expression analysis was performed on CD4(+), CD8(+) T and B cells from 14 patients who interrupted natalizumab therapy for 8 weeks. The miRNA profiling was conducted in PBMCs from 5 untreated and 5 natalizumab-treated patients using low-density arrays followed by validation with single miRNAs assays in untreated and natalizumab-treated patients. RESULTS: POU2AF1 and Spi-B mRNAs were upregulated in B and CD8(+) T cells from natalizumab-treated patients, which was validated in PBMCs from different cohorts of natalizumab-treated patients with and without PML, with a noteworthy higher expression of Spi-B in patients with PML. In contrast, downregulation of POU2AF1/Spi-B expression was measured in B and CD8(+) T cells after natalizumab discontinuation. Seventeen differentially expressed miRNAs including miR-10b, a regulator of POU2AF1 mRNA, were identified in long-term natalizumab-treated patients compared with untreated ones. CONCLUSIONS: Upregulation of POU2AF1 and Spi-B, known transactivators of the JC virus, the causative agent for PML, and its association with occurrence of PML in natalizumab-treated patients, corroborates POU2AF1/Spi-B as potential biomarkers for PML risk, which merits further evaluation.

6.
Neurol Neuroimmunol Neuroinflamm ; 3(2): e202, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26894205

RESUMEN

OBJECTIVE: To evaluate serum cytokine profiles for their utility to determine the heterogeneous responses to interferon (IFN)-ß treatment in patients with multiple sclerosis (MS). METHODS: Patients with relapsing-remitting MS (RRMS) or clinically isolated syndrome receiving de novo IFN-ß treatment were included in this prospective, observational study. Number of relapses and changes in disability were assessed 2 years prior to and 2 years after initiation of treatment. Sera were collected at baseline and after 3 months on therapy. Cytokine levels in sera were assessed by Luminex multiplex assays. Baseline cytokine profiles were grouped by hierarchical clustering analysis. Demographic features, changes in cytokines, and clinical outcome were then assessed in the clustered patient groups. RESULTS: A total of 157 patients were included in the study and clustered into 6 distinct subsets by baseline cytokine profiles. These subsets differed significantly in their clinical and biological response to IFN-ß therapy. Two subsets were associated with patients who responded poorly to therapy. Two other subsets, associated with a good response to therapy, showed a significant reduction in relapse rates and no worsening of disability. Each subset also had differential changes in cytokine levels after 3 months of IFN-ß treatment. CONCLUSIONS: There is heterogeneity in the immunologic pathways of the RRMS population, which correlates with IFN-ß response.

7.
PLoS One ; 11(1): e0146190, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26734938

RESUMEN

BACKGROUND: Fingolimod is a first in class oral compound approved for the treatment of relapsing-remitting multiple sclerosis (RR-MS). The aim of this study was to evaluate clinical and neuroradiological responses to fingolimod as well as the safety and tolerability in RR-MS patients in clinical practice. In addition, a panel of pro-inflammatory serum cytokines was explored as potential biomarker for treatment response. METHODS: We conducted a retrospective, non-randomized, open-label, observational study in 105 patients with RR-MS and measured cytokines in longitudinal serum samples. RESULTS: Compared to the year before fingolimod start the annualized relapse rate was reduced by 44%. Also, the percentage of patients with a worsening of the EDSS decreased. Accordingly, the fraction of patients with no evidence of disease activity (no relapse, stable EDSS, no new active lesions in MRI) increased from 11% to 38%. The efficacy and safety were comparable between highly active patients or patients with relevant comorbidities and our general patient population. CONCLUSIONS: The efficacy in reducing relapses was comparable to that observed in the phase III trials. In our cohort fingolimod was safe and efficacious irrespective of comorbidities and previous treatment.


Asunto(s)
Clorhidrato de Fingolimod/efectos adversos , Inmunosupresores/efectos adversos , Esclerosis Múltiple Recurrente-Remitente/patología , Adulto , Anciano , Estudios de Casos y Controles , Citocinas/sangre , Disnea/etiología , Edema/etiología , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Recurrencia , Estudios Retrospectivos
8.
Neurol Neuroimmunol Neuroinflamm ; 2(5): e144, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26380353

RESUMEN

OBJECTIVE: To monitor the systemic immune responses of patients with multiple sclerosis (MS) under treatment with GNbAC1, a monoclonal antibody against the envelope protein of the MS- associated retrovirus, during a phase 2a trial. METHODS: We analyzed the composition of immune cell subsets and the activation level of monocytes by flow cytometry and the response against viral and vaccine antigens by ELISpot. RESULTS: None of the endpoints measured revealed any immunosuppressive effect of the therapeutic antibody. Activation of monocytes slightly decreased during treatment as predicted by the hypothesized mechanism of action of GNbAC1. CONCLUSION: These results support the conclusion that the antibody is safe for use in patients with MS. CLASSIFICATION OF EVIDENCE: This study provides Class III evidence that in patients with MS GNbAC1 does not significantly affect several biomarkers of systemic immune response.

9.
J Neuroinflammation ; 12: 46, 2015 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-25889963

RESUMEN

BACKGROUND: Antibodies against myelin oligodendrocyte glycoprotein (MOG) have been identified in a subgroup of pediatric patients with inflammatory demyelinating disease of the central nervous system (CNS) and in some patients with neuromyelitis optica spectrum disorder (NMOSD). The aim of this study was to examine the frequency, clinical features, and long-term disease course of patients with anti-MOG antibodies in a European cohort of NMO/NMOSD. FINDINGS: Sera from 48 patients with NMO/NMOSD and 48 patients with relapsing-remitting multiple sclerosis (RR-MS) were tested for anti-aquaporin-4 (AQP4) and anti-MOG antibodies with a cell-based assay. Anti-MOG antibodies were found in 4/17 patients with AQP4-seronegative NMO/NMOSD, but in none of the AQP4-seropositive NMO/NMOSD (n = 31) or RR-MS patients (n = 48). MOG-seropositive patients tended towards younger disease onset with a higher percentage of patients with pediatric (<18 years) disease onset (MOG+, AQP4+, MOG-/AQP4-: 2/4, 3/31, 0/13). MOG-seropositive patients presented more often with positive oligoclonal bands (OCBs) (3/3, 5/29, 1/13) and brain magnetic resonance imaging (MRI) lesions during disease course (2/4, 5/31, 1/13). Notably, the mean time to the second attack affecting a different CNS region was longer in the anti-MOG antibody-positive group (11.3, 3.2, 3.4 years). CONCLUSIONS: MOG-seropositive patients show a diverse clinical phenotype with clinical features resembling both NMO (attacks mainly confined to the spinal cord and optic nerves) and MS with an opticospinal presentation (positive OCBs, brain lesions). Anti-MOG antibodies can serve as a diagnostic and maybe prognostic tool in patients with an AQP4-seronegative NMO phenotype and should be tested in those patients.


Asunto(s)
Autoanticuerpos/sangre , Glicoproteína Mielina-Oligodendrócito/inmunología , Neuromielitis Óptica/sangre , Adulto , Anciano , Acuaporina 4/inmunología , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/sangre , Estudios Retrospectivos , Adulto Joven
10.
J Immunol Res ; 2014: 897249, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24901013

RESUMEN

MicroRNAs (miRNAs) are a family of noncoding RNAs that play critical roles in the posttranscriptional regulation of gene expression. Accumulating evidence supports their involvement in the pathogenesis of multiple sclerosis (MS). Here, we compare miR-17 expressions in CD4+ T cells from relapsing-remitting (RR) MS patients treated with natalizumab versus untreated patients. miR-17 was downregulated under natalizumab treatment and upregulated during relapse, therefore supporting a possible role of miR-17 in MS immunopathogenesis. Downregulation of miR-17 was associated with upregulation of PTEN, BIM, E2F1, and p21 target genes. In vitro miR-17 inhibition was associated with upregulation of the same targets and resulted in impaired CD4+ T cell activation and proliferation. We further describe deregulated TGFBR2 expression in untreated patients versus healthy volunteers (HVs) and confirm in vitro the link between miR-17 and TGFBR2 expressions. These findings support an effect of natalizumab on expression of specific miRNA and subsequent expression of genes involved in proliferation and control of the cell cycle.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Adulto , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Proteína 11 Similar a Bcl2 , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Estudios de Casos y Controles , Ciclo Celular/efectos de los fármacos , Inhibición de Migración Celular , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/inmunología , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/inmunología , Femenino , Humanos , Activación de Linfocitos/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , MicroARNs/inmunología , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Esclerosis Múltiple Recurrente-Remitente/patología , Natalizumab , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/inmunología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/inmunología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA