Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Eur J Immunol ; 50(1): 73-85, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31621069

RESUMEN

Antibody production by the B cell compartment is a crucial part of the adaptive immune response. Dysregulated antibody production in the form of autoantibodies can cause autoimmune disease. To date, B-cell depletion with anti-CD20 antibodies is commonly applied in autoimmunity, but pre-existing plasma cells are not eliminated in this way. Alternative ways of more selective inhibition of antibody production would add to the treatment of these autoimmune diseases. To explore novel therapeutic targets in signaling pathways essential for plasmablast formation and/or immunoglobulin production, we performed a compound screen of almost 200 protein kinase inhibitors in a robust B-cell differentiation culture system. This study yielded 35 small cell-permeable compounds with a reproducible inhibitory effect on B-cell activation and plasmablast formation, among which was the clinically applied mammalian target of rapamycin (mTOR) inhibitor rapamycin. Two additional compounds targeting the phosphoinositide 3-kinase-AKT-mTOR pathway (BKM120 and WYE-354) did not affect proliferation and plasmablast formation, but specifically reduced the immunoglobulin production. With this compound screen we successfully applied a method to investigate therapeutic targets for B-cell differentiation and identified compounds in the phosphoinositide 3-kinase-AKT-mTOR pathway that could specifically inhibit immunoglobulin production only. These drugs may well be explored to be of value in current B-cell-depleting treatment regimens in autoimmune disorders.


Asunto(s)
Autoanticuerpos/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Células Plasmáticas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Aminopiridinas/farmacología , Formación de Anticuerpos/efectos de los fármacos , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/inmunología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Diferenciación Celular/efectos de los fármacos , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos , Morfolinas/farmacología , Células Plasmáticas/inmunología , Purinas/farmacología , Sirolimus/farmacología
2.
J Clin Immunol ; 36(7): 656-66, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27484504

RESUMEN

Primary antibody deficiencies (PADs) are the most common immunodeficiency in humans, characterized by low levels of immunoglobulins and inadequate antibody responses upon immunization. These PADs may result from an early block in B cell development with a complete absence of peripheral B cells and lack of immunoglobulins. In the presence of circulating B cells, some PADs are genetically caused by a class switch recombination (CSR) defect, but in the most common PAD, common variable immunodeficiency (CVID), very few gene defects have as yet been characterized despite various phenotypic classifications. Using a functional read-out, we previously identified a functional subgroup of CVID patients with plasmablasts (PBs) producing IgM only. We have now further characterized such CVID patients by a direct functional comparison with patients having genetically well-characterized CSR defects in CD40L, activation-induced cytidine deaminase (AID) and uracil N-glycosylase activity (UNG). The CSR-like CVID patients showed a failure in B cell activation patterns similar to the classical AID/UNG defects in three out of five CVID patients and distinct more individual defects in the two other CVID cases when tested for cellular activation and PB differentiation. Thus, functional categorization of B cell activation and differentiation pathways extends the expected variation in CVID to CSR-like defects of as yet unknown genetic etiology.


Asunto(s)
Inmunodeficiencia Variable Común/genética , Inmunodeficiencia Variable Común/inmunología , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Fenotipo , Adolescente , Adulto , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Inmunodeficiencia Variable Común/diagnóstico , Femenino , Predisposición Genética a la Enfermedad , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Inmunofenotipificación , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Adulto Joven
3.
J Immunol ; 190(10): 5012-9, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23585684

RESUMEN

Primary immunodeficiencies consist to a large extent of B cell defects, as indicated by inadequate Ab levels or response upon immunization. Many B cell defects have not yet been well characterized. Our objective was to create reliable in vivo and in vitro assays to routinely analyze human B cell differentiation, proliferation, and Ig production and to define reference ranges for different age categories. The in vitro assays were applied to classify the developmental and/or functional B cell defects in patients previously diagnosed with common variable immunodeficiency. Apart from standard immunophenotyping of circulating human B cell subsets, an in vitro CFSE dilution assay was used for the assessment of proliferative capacity comparing T cell-dependent and T cell-independent B cell activation. Plasmablast/plasma cell differentiation was assessed by staining for CD20, CD38, and CD138, and measurement of in vitro Ig secretion. At young age, B cells proliferate upon in vitro activation, but neither differentiate nor produce IgG. These latter functions reached adult levels at 5 and 10 y of age for T cell-dependent versus T cell-independent stimulations, respectively. The capacity of B cells to differentiate into plasmablasts and to produce IgG appeared to be contained within the switched memory B cell pool. Using these assays, we could categorize common variable immunodeficiency patients into subgroups and identified a class-switch recombination defect caused by an UNG mutation in one of the patients. We defined age-related reference ranges for human B cell differentiation. Our findings indicate that in vivo B cell functionality can be tested in vitro and helps to diagnose suspected B cell defects.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Linfocitos B/inmunología , Inmunodeficiencia Variable Común/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , ADP-Ribosil Ciclasa 1/análisis , Adulto , Anticuerpos/sangre , Antígenos CD20/análisis , Subgrupos de Linfocitos B/clasificación , Subgrupos de Linfocitos B/citología , Linfocitos B/clasificación , Linfocitos B/citología , Diferenciación Celular/inmunología , Proliferación Celular , Células Cultivadas , Inmunodeficiencia Variable Común/clasificación , Femenino , Humanos , Inmunoglobulinas/biosíntesis , Inmunofenotipificación , Masculino , Valores de Referencia , Sindecano-1/análisis
4.
Clin Immunol ; 149(1): 25-31, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23891736

RESUMEN

Mutations in the DOCK8 gene define the most common form of autosomal-recessive Hyper-IgE-syndrome (AR-HIES/OMIM#243700). In a patient with extensive molluscum contagiosum lesions, a homozygous DOCK8 gene deletion was demonstrated. In-vivo 18-FDG uptake showed multiple non-enlarged lymph nodes without uptake in the spleen. Lymph node biopsies for subsequent immunohistochemistry showed clear differences with the mouse model of DOCK8 deficiency in which these mice show no GCs. Unexpectedly, the patient's lymph nodes demonstrated lymphocyte polyclonality, follicular hyperplasia and an unusual IgE(+) plasma cell expansion. In contrast, the proliferative capacity of circulating B-cells was almost absent with little in-vitro Ig production or plasmablast formation. Also the T-cell proliferation indicated a partial defect. Hematopoietic stem cell transplantation (HSCT) was performed resulting in the disappearance of the molluscum contagiosum lesions. In sum, DOCK8 deficiency results in defective antibody responses and undirected plasma cell expansion in the lymph nodes, as part of a combined immunodeficiency cured by HSCT.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/deficiencia , Síndromes de Inmunodeficiencia/inmunología , Molusco Contagioso/inmunología , Adolescente , Anticuerpos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Proliferación Celular , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/inmunología , Trasplante de Células Madre Hematopoyéticas , Humanos , Hiperplasia/genética , Hiperplasia/inmunología , Hiperplasia/patología , Hiperplasia/terapia , Inmunidad Humoral , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/terapia , Ganglios Linfáticos/patología , Recuento de Linfocitos , Molusco Contagioso/genética , Molusco Contagioso/patología , Molusco Contagioso/terapia , Linfocitos T/inmunología , Linfocitos T/patología
5.
Haematologica ; 98(7): 1030-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23403317

RESUMEN

Mutations in the common gamma chain (γc, CD132, encoded by the IL2RG gene) can lead to B(+)T(-)NK(-) X-linked severe combined immunodeficiency, as a consequence of unresponsiveness to γc-cytokines such as interleukins-2, -7 and -15. Hypomorphic mutations in CD132 may cause combined immunodeficiencies with a variety of clinical presentations. We analyzed peripheral blood mononuclear cells of a 6-year-old boy with normal lymphocyte counts, who suffered from recurrent pneumonia and disseminated mollusca contagiosa. Since proliferative responses of T cells and NK cells to γc -cytokines were severely impaired, we performed IL2RG gene analysis, showing a heterozygous mutation in the presence of a single X-chromosome. Interestingly, an IL2RG reversion to normal predominated in both naïve and antigen-primed CD8(+) T cells and increased over time. Only the revertant CD8(+) T cells showed normal expression of CD132 and the various CD8(+) T cell populations had a different T-cell receptor repertoire. Finally, a fraction of γδ(+) T cells and differentiated CD4(+)CD27(-) effector-memory T cells carried the reversion, whereas NK or B cells were repeatedly negative. In conclusion, in a patient with a novel IL2RG mutation, gene-reverted CD8(+) T cells accumulated over time. Our data indicate that selective outgrowth of particular T-cell subsets may occur following reversion at the level of committed T progenitor cells.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Subunidad gamma Común de Receptores de Interleucina/genética , Mutación/genética , Inmunodeficiencia Combinada Grave/genética , Linfocitos T CD8-positivos/patología , Proliferación Celular , Niño , Humanos , Subunidad gamma Común de Receptores de Interleucina/fisiología , Recuento de Linfocitos , Masculino , Linaje , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA