Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 43(5): 884-95, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26546282

RESUMEN

Activation-induced cytidine deaminase (AID), the enzyme-mediating class-switch recombination (CSR) and somatic hypermutation (SHM) of immunoglobulin genes, is essential for the removal of developing autoreactive B cells. How AID mediates central B cell tolerance remains unknown. We report that AID enzymes were produced in a discrete population of immature B cells that expressed recombination-activating gene 2 (RAG2), suggesting that they undergo secondary recombination to edit autoreactive antibodies. However, most AID+ immature B cells lacked anti-apoptotic MCL-1 and were deleted by apoptosis. AID inhibition using lentiviral-encoded short hairpin (sh)RNA in B cells developing in humanized mice resulted in a failure to remove autoreactive clones. Hence, B cell intrinsic AID expression mediates central B cell tolerance potentially through its RAG-coupled genotoxic activity in self-reactive immature B cells.


Asunto(s)
Tolerancia Central/genética , Tolerancia Central/inmunología , Citidina Desaminasa/genética , Activación de Linfocitos/inmunología , Células Precursoras de Linfocitos B/inmunología , Adolescente , Adulto , Anciano , Animales , Apoptosis/genética , Apoptosis/inmunología , Estudios de Casos y Controles , Niño , Preescolar , Proteínas de Unión al ADN/genética , Femenino , Genes de Inmunoglobulinas/genética , Genes de Inmunoglobulinas/inmunología , Humanos , Activación de Linfocitos/genética , Masculino , Ratones , Persona de Mediana Edad , Proteínas Nucleares/genética , Recombinación Genética/genética , Recombinación Genética/inmunología , Hipermutación Somática de Inmunoglobulina/genética , Hipermutación Somática de Inmunoglobulina/inmunología , Adulto Joven
2.
Cell ; 136(3): 420-34, 2009 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-19203578

RESUMEN

The biological response to DNA double-strand breaks acts to preserve genome integrity. Individuals bearing inactivating mutations in components of this response exhibit clinical symptoms that include cellular radiosensitivity, immunodeficiency, and cancer predisposition. The archetype for such disorders is Ataxia-Telangiectasia caused by biallelic mutation in ATM, a central component of the DNA damage response. Here, we report that the ubiquitin ligase RNF168 is mutated in the RIDDLE syndrome, a recently discovered immunodeficiency and radiosensitivity disorder. We show that RNF168 is recruited to sites of DNA damage by binding to ubiquitylated histone H2A. RNF168 acts with UBC13 to amplify the RNF8-dependent histone ubiquitylation by targeting H2A-type histones and by promoting the formation of lysine 63-linked ubiquitin conjugates. These RNF168-dependent chromatin modifications orchestrate the accumulation of 53BP1 and BRCA1 to DNA lesions, and their loss is the likely cause of the cellular and developmental phenotypes associated with RIDDLE syndrome.


Asunto(s)
Daño del ADN , Síndromes de Inmunodeficiencia/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Línea Celular , Histonas/metabolismo , Humanos , Síndromes de Inmunodeficiencia/genética , Tolerancia a Radiación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Nucleic Acids Res ; 49(9): 5057-5073, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33950194

RESUMEN

Activation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naïve and memory B cells both display widespread DNA methylation alterations, of which ∼25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naïve B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/fisiología , Metilación de ADN , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Síndrome de Inmunodeficiencia con Hiper-IgM/inmunología , Autoinmunidad , Linfocitos B/metabolismo , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Centro Germinal/inmunología , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/metabolismo , Tolerancia Inmunológica , Memoria Inmunológica , Receptores de Antígenos de Linfocitos B/genética , Transcriptoma , Secuenciación Completa del Genoma
4.
Blood ; 135(9): 638-643, 2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-31942637

RESUMEN

Activated phosphatidylinositol 3-kinase-δ (PI3K-δ) syndrome (APDS) is a rare primary combined immunodeficiency caused by either dominant gain-of-function mutations in the PIK3CD gene encoding the catalytic subunit p110δ of PI3K-δ (referred to as type 1 APDS) or dominant loss-of-function mutations in the PIK3R1 gene encoding the p85α, p55α, and p50α regulatory subunits (type 2 APDS). In types 1 and 2 APDS, the PI3K-δ hyperactivity resulting from the gene mutations leads to similar clinical presentations, characterized by increased susceptibility to bacterial and viral infections and (to a lesser extent) autoimmune manifestations. A hallmark of this disease is lymphoproliferation, which may even be life threatening and require repeated surgical treatment. A major complication of APDS is malignancy (especially B-cell lymphomas), which greatly worsens the prognosis. Here, we review the different neoplastic conditions observed in patients with APDS and discuss the uncontrolled PI3K-δ activity in B and T cells that leads to malignant transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Linfoma de Células B/etiología , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades de Inmunodeficiencia Primaria/complicaciones , Enfermedades de Inmunodeficiencia Primaria/metabolismo , Linfocitos B/inmunología , Linfocitos B/metabolismo , Transformación Celular Neoplásica/inmunología , Humanos , Enfermedades de Inmunodeficiencia Primaria/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
PLoS Genet ; 15(6): e1007721, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31199803

RESUMEN

B-cell activation yields abundant cell death in parallel to clonal amplification and remodeling of immunoglobulin (Ig) genes by activation-induced deaminase (AID). AID promotes affinity maturation of Ig variable regions and class switch recombination (CSR) in mature B lymphocytes. In the IgH locus, these processes are under control of the 3' regulatory region (3'RR) super-enhancer, a region demonstrated in the mouse to be both transcribed and itself targeted by AID-mediated recombination. Alternatively to CSR, IgH deletions joining Sµ to "like-switch" DNA repeats that flank the 3' super-enhancer can thus accomplish so-called "locus suicide recombination" (LSR) in mouse B-cells. Using an optimized LSR-seq high throughput method, we now show that AID-mediated LSR is evolutionarily conserved and also actively occurs in humans, providing an activation-induced cell death pathway in multiple conditions of B-cell activation. LSR either focuses on the functional IgH allele or is bi-allelic, and its signature is mainly detected when LSR is ongoing while it vanishes from fully differentiated plasma cells or from "resting" blood memory B-cells. Highly diversified breakpoints are distributed either within the upstream (3'RR1) or downstream (3'RR2) copies of the IgH 3' super-enhancer and all conditions activating CSR in vitro also seem to trigger LSR although TLR ligation appeared the most efficient. Molecular analysis of breakpoints and junctions confirms that LSR is AID-dependent and reveals junctional sequences somehow similar to CSR junctions but with increased usage of microhomologies.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/genética , Región de Cambio de la Inmunoglobulina/genética , Inmunoglobulinas/inmunología , Alelos , Animales , Diferenciación Celular/genética , Citidina Desaminasa/inmunología , Marcación de Gen , Humanos , Región de Cambio de la Inmunoglobulina/inmunología , Tejido Linfoide/inmunología , Ratones , Tonsila Palatina/inmunología , Tonsila Palatina/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Secuencias Reguladoras de Ácidos Nucleicos
6.
Clin Immunol ; 222: 108638, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33276124

RESUMEN

To assess how B cell phenotype analysis correlates with antigen responses in patients with class switch recombination defects (CSRD) we quantified memory B cells by flow-cytometry and immunized CSRD patients with the neoantigen bacteriophage phiX174 (phage). CSRD patients showed uniformly absent or markedly reduced switched memory B cells (IgM-IgD-CD27+). CD40L patients had reduced CD27+ memory B cells (both non-switched and switched). In NEMO patients, results varied depending on the IKKγ gene variant. Three of four AID patients had normal percentages of CD27+ memory B cells while CD27+IgM-IgD- switched memory B cells were markedly reduced in all AID patients. Antibody response to phage was remarkably decreased with lack of memory amplification and class-switching in immunized CD40L, UNG deficient, and NEMO patients. Distinct B-cell phenotype pattern correlated with abnormal antibody responses to a T-cell dependent neoantigen, representing a powerful tool to identify CSRD patients.


Asunto(s)
Linfocitos B/citología , Bacteriófago phi X 174/inmunología , Cambio de Clase de Inmunoglobulina/genética , Cambio de Clase de Inmunoglobulina/inmunología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Adolescente , Adulto , Formación de Anticuerpos/genética , Formación de Anticuerpos/inmunología , Ligando de CD40/deficiencia , Niño , Preescolar , Femenino , Citometría de Flujo , Humanos , Proteínas I-kappa B/genética , Inmunización , Inmunoglobulina D/inmunología , Inmunoglobulina M/inmunología , Síndromes de Inmunodeficiencia/patología , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Lactante , Masculino , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
7.
J Clin Immunol ; 41(6): 1272-1290, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33929673

RESUMEN

Biallelic inactivating mutations in IL21R causes a combined immunodeficiency that is often complicated by cryptosporidium infections. While eight IL-21R-deficient patients have been reported previously, the natural course, immune characteristics of disease, and response to hematopoietic stem cell transplantation (HSCT) remain to be comprehensively examined. In our study, we have collected clinical histories of 13 patients with IL-21R deficiency from eight families across seven centers worldwide, including five novel patients identified by exome or NGS panel sequencing. Eight unique mutations in IL21R were identified in these patients, including two novel mutations. Median age at disease onset was 2.5 years (0.5-7 years). The main clinical manifestations were recurrent bacterial (84.6%), fungal (46.2%), and viral (38.5%) infections; cryptosporidiosis-associated cholangitis (46.2%); and asthma (23.1%). Inflammatory skin diseases (15.3%) and recurrent anaphylaxis (7.9%) constitute novel phenotypes of this combined immunodeficiency. Most patients exhibited hypogammaglobulinemia and reduced proportions of memory B cells, circulating T follicular helper cells, MAIT cells and terminally differentiated NK cells. However, IgE levels were elevated in 50% of IL-21R-deficient patients. Overall survival following HSCT (6 patients, mean follow-up 1.8 year) was 33.3%, with pre-existing organ damage constituting a negative prognostic factor. Mortality of non-transplanted patients (n = 7) was 57.1%. Our detailed analysis of the largest cohort of IL-21R-deficient patients to date provides in-depth clinical, immunological and immunophenotypic features of these patients, thereby establishing critical non-redundant functions of IL-21/IL-21R signaling in lymphocyte differentiation, humoral immunity and host defense against infection, and mechanisms of disease pathogenesis due to IL-21R deficiency. Outcome following HSCT depends on prior chronic infections and organ damage, which should thus be considered as early as possible following molecular diagnosis.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-21/deficiencia , Subunidad alfa del Receptor de Interleucina-21/genética , Adolescente , Linfocitos B/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Niño , Preescolar , Criptosporidiosis/genética , Criptosporidiosis/inmunología , Cryptosporidium/inmunología , Femenino , Genómica/métodos , Humanos , Inmunidad Humoral/genética , Inmunidad Humoral/inmunología , Lactante , Subunidad alfa del Receptor de Interleucina-21/inmunología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Células B de Memoria/inmunología , Infección Persistente/genética , Infección Persistente/inmunología , Fenotipo , Transducción de Señal/genética , Transducción de Señal/inmunología , Adulto Joven
8.
Pediatr Allergy Immunol ; 32(8): 1601-1615, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34181780

RESUMEN

Predominantly antibody deficiencies (PADs) encompass a heterogeneous group of disorders characterized by low immunoglobulin serum levels in the presence or absence of peripheral B cells. Clinical presentation of affected patients may include recurrent respiratory and gastrointestinal infections, invasive infections, autoimmune manifestations, allergic reactions, lymphoproliferation, and increased susceptibility to malignant transformation. In the last decades, several genetic alterations affecting B-cell development/maturation have been identified as causative of several forms of PADs, adding important information on the genetic background of PADs, which in turn should lead to a better understanding of these disorders and precise clinical management of affected patients. This review aimed to present a comprehensive overview of the known and potentially involved molecules in the etiology of PADs to elucidate the pathogenesis of these disorders and eventually offer a better prognosis for affected patients.


Asunto(s)
Síndromes de Inmunodeficiencia , Neoplasias , Linfocitos B , Humanos , Síndromes de Inmunodeficiencia/genética , Activación de Linfocitos , Pronóstico
9.
J Pediatr ; 223: 207-211.e1, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32423680

RESUMEN

The genetic investigation of a family presenting with a dominant form of hyper IgM syndrome published in 1963 and 1975 revealed a R190X nonsense mutation in activation-induced cytidine deaminase. This report illustrates the progress made over 6 decades in the characterization of primary immunodeficiencies, from immunochemistry to whole-exome sequencing.


Asunto(s)
Citidina Desaminasa/genética , Disgammaglobulinemia/genética , Predicción , Síndromes de Inmunodeficiencia/complicaciones , Mutación , Citidina Desaminasa/metabolismo , Análisis Mutacional de ADN , Disgammaglobulinemia/complicaciones , Disgammaglobulinemia/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Persona de Mediana Edad
10.
J Allergy Clin Immunol ; 143(1): 266-275, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29778502

RESUMEN

BACKGROUND: Activated phosphatidylinositol-3-OH kinase δ syndrome type 1 (APDS1) is a recently described primary immunodeficiency syndrome characterized by recurrent respiratory tract infections, lymphoid hyperplasia, and Herpesviridae infections caused by germline gain-of-function mutations of PIK3CD. Hematopoietic stem cell transplantation (HSCT) can be considered to ameliorate progressive immunodeficiency and associated malignancy, but appropriate indications, methods, and outcomes of HSCT for APDS1 remain undefined. OBJECTIVE: Our objective was to analyze the clinical manifestations, laboratory findings, prognosis, and treatment of APDS1 and explore appropriate indications and methods of HSCT. METHODS: We reviewed retrospectively the medical records of cohorts undergoing HSCT at collaborating facilities. RESULTS: Thirty-year overall survival was 86.1%, but event-free survival was 39.6%. Life-threatening events, such as severe infections or lymphoproliferation, were frequent in childhood and adolescence and were common indications for HSCT. Nine patients underwent HSCT with fludarabine-based reduced-intensity conditioning. Seven patients survived after frequent adverse complications and engraftment failure. Most symptoms improved after HSCT. CONCLUSION: Patients with APDS1 showed variable clinical manifestations. Life-threatening progressive combined immunodeficiency and massive lymphoproliferation were common indications for HSCT. Fludarabine-based reduced-intensity conditioning-HSCT ameliorated clinical symptoms, but transplantation-related complications were frequent, including graft failure.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Síndromes de Inmunodeficiencia , Trastornos Linfoproliferativos , Adolescente , Adulto , Aloinjertos , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Supervivencia sin Enfermedad , Femenino , Humanos , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/mortalidad , Síndromes de Inmunodeficiencia/patología , Síndromes de Inmunodeficiencia/terapia , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/mortalidad , Trastornos Linfoproliferativos/patología , Trastornos Linfoproliferativos/terapia , Masculino , Enfermedades de Inmunodeficiencia Primaria , Tasa de Supervivencia
11.
Immunity ; 33(3): 400-11, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20832341

RESUMEN

Tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3) functions downstream of multiple TNF receptors and receptors that induce interferon-α (IFN-α), IFN-ß, and IFN-λ production, including Toll-like receptor 3 (TLR3), which is deficient in some patients with herpes simplex virus-1 encephalitis (HSE). Mice lacking TRAF3 die in the neonatal period, preventing direct investigation of the role of TRAF3 in immune responses and host defenses in vivo. Here, we report autosomal dominant, human TRAF3 deficiency in a young adult with a history of HSE in childhood. The TRAF3 mutant allele is loss-of-expression, loss-of-function, dominant-negative and associated with impaired, but not abolished, TRAF3-dependent responses upon stimulation of both TNF receptors and receptors that induce IFN production. TRAF3 deficiency is associated with a clinical phenotype limited to HSE resulting from the impairment of TLR3-dependent induction of IFN. Thus, TLR3-mediated immunity against primary infection by HSV-1 in the central nervous system is critically dependent on TRAF3.


Asunto(s)
Encefalitis por Herpes Simple/inmunología , Factor 3 Asociado a Receptor de TNF/fisiología , Receptor Toll-Like 3/fisiología , Células Cultivadas , Susceptibilidad a Enfermedades , Humanos , Interferones/fisiología , Mutación , Receptores del Factor de Necrosis Tumoral/fisiología , Factor 3 Asociado a Receptor de TNF/genética
12.
J Allergy Clin Immunol ; 139(2): 597-606.e4, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27555459

RESUMEN

BACKGROUND: Activated phosphoinositide 3-kinase δ syndrome (APDS) is a recently described combined immunodeficiency resulting from gain-of-function mutations in PIK3CD, the gene encoding the catalytic subunit of phosphoinositide 3-kinase δ (PI3Kδ). OBJECTIVE: We sought to review the clinical, immunologic, histopathologic, and radiologic features of APDS in a large genetically defined international cohort. METHODS: We applied a clinical questionnaire and performed review of medical notes, radiology, histopathology, and laboratory investigations of 53 patients with APDS. RESULTS: Recurrent sinopulmonary infections (98%) and nonneoplastic lymphoproliferation (75%) were common, often from childhood. Other significant complications included herpesvirus infections (49%), autoinflammatory disease (34%), and lymphoma (13%). Unexpectedly, neurodevelopmental delay occurred in 19% of the cohort, suggesting a role for PI3Kδ in the central nervous system; consistent with this, PI3Kδ is broadly expressed in the developing murine central nervous system. Thoracic imaging revealed high rates of mosaic attenuation (90%) and bronchiectasis (60%). Increased IgM levels (78%), IgG deficiency (43%), and CD4 lymphopenia (84%) were significant immunologic features. No immunologic marker reliably predicted clinical severity, which ranged from asymptomatic to death in early childhood. The majority of patients received immunoglobulin replacement and antibiotic prophylaxis, and 5 patients underwent hematopoietic stem cell transplantation. Five patients died from complications of APDS. CONCLUSION: APDS is a combined immunodeficiency with multiple clinical manifestations, many with incomplete penetrance and others with variable expressivity. The severity of complications in some patients supports consideration of hematopoietic stem cell transplantation for severe childhood disease. Clinical trials of selective PI3Kδ inhibitors offer new prospects for APDS treatment.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Síndromes de Inmunodeficiencia/genética , Trastornos Linfoproliferativos/genética , Mutación/genética , Infecciones del Sistema Respiratorio/genética , Adolescente , Adulto , Animales , Profilaxis Antibiótica , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Estudios de Cohortes , Inhibidores Enzimáticos/uso terapéutico , Femenino , Trasplante de Células Madre Hematopoyéticas , Infecciones por Herpesviridae/genética , Infecciones por Herpesviridae/mortalidad , Infecciones por Herpesviridae/terapia , Humanos , Inmunoglobulinas Intravenosas/uso terapéutico , Síndromes de Inmunodeficiencia/mortalidad , Síndromes de Inmunodeficiencia/terapia , Lactante , Cooperación Internacional , Trastornos Linfoproliferativos/mortalidad , Trastornos Linfoproliferativos/terapia , Masculino , Ratones , Persona de Mediana Edad , Recurrencia , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/terapia , Encuestas y Cuestionarios , Análisis de Supervivencia , Adulto Joven
14.
Proc Natl Acad Sci U S A ; 111(11): E988-97, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24591601

RESUMEN

Activation-induced deaminase (AID) triggers antibody class switch recombination (CSR) in B cells by initiating DNA double strand breaks that are repaired by nonhomologous end-joining pathways. A role for AID at the repair step is unclear. We show that specific inactivation of the C-terminal AID domain encoded by exon 5 (E5) allows very efficient deamination of the AID target regions but greatly impacts the efficiency and quality of subsequent DNA repair. Specifically eliminating E5 not only precludes CSR but also, causes an atypical, enzymatic activity-dependent dominant-negative effect on CSR. Moreover, the E5 domain is required for the formation of AID-dependent Igh-cMyc chromosomal translocations. DNA breaks at the Igh switch regions induced by AID lacking E5 display defective end joining, failing to recruit DNA damage response factors and undergoing extensive end resection. These defects lead to nonproductive resolutions, such as rearrangements and homologous recombination that can antagonize CSR. Our results can explain the autosomal dominant inheritance of AID variants with truncated E5 in patients with hyper-IgM syndrome 2 and establish that AID, through the E5 domain, provides a link between DNA damage and repair during CSR.


Asunto(s)
Citidina Desaminasa/metabolismo , Roturas del ADN , Reparación del ADN por Unión de Extremidades/fisiología , Cambio de Clase de Inmunoglobulina/genética , Análisis de Varianza , Animales , Linfocitos B/inmunología , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Reparación del ADN por Unión de Extremidades/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Translocación Genética/genética , Uracil-ADN Glicosidasa/genética
15.
J Allergy Clin Immunol ; 138(6): 1681-1689.e8, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27405666

RESUMEN

BACKGROUND: We investigated 7 male patients (from 5 different families) presenting with profound lymphopenia, hypogammaglobulinemia, fluctuating monocytopenia and neutropenia, a poor immune response to vaccine antigens, and increased susceptibility to bacterial and varicella zoster virus infections. OBJECTIVE: We sought to characterize the genetic defect involved in a new form of X-linked immunodeficiency. METHODS: We performed genetic analyses and an exhaustive phenotypic and functional characterization of the lymphocyte compartment. RESULTS: We observed hemizygous mutations in the moesin (MSN) gene (located on the X chromosome and coding for MSN) in all 7 patients. Six of the latter had the same missense mutation, which led to an amino acid substitution (R171W) in the MSN four-point-one, ezrin, radixin, moesin domain. The seventh patient had a nonsense mutation leading to a premature stop codon mutation (R533X). The naive T-cell counts were particularly low for age, and most CD8+ T cells expressed the senescence marker CD57. This phenotype was associated with impaired T-cell proliferation, which was rescued by expression of wild-type MSN. MSN-deficient T cells also displayed poor chemokine receptor expression, increased adhesion molecule expression, and altered migration and adhesion capacities. CONCLUSION: Our observations establish a causal link between an ezrin-radixin-moesin protein mutation and a primary immunodeficiency that could be referred to as X-linked moesin-associated immunodeficiency.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cromosomas Humanos X/genética , Síndromes de Inmunodeficiencia/genética , Infecciones/genética , Proteínas de Microfilamentos/genética , Mutación/genética , Adolescente , Adulto , Anciano , Adhesión Celular , Movimiento Celular , Niño , Preescolar , Estudios de Asociación Genética , Humanos , Recuento de Linfocitos , Masculino , Linaje
16.
J Allergy Clin Immunol ; 138(1): 210-218.e9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27221134

RESUMEN

BACKGROUND: Activated phosphoinositide 3-kinase δ syndrome (APDS) 2 (p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency [PASLI]-R1), a recently described primary immunodeficiency, results from autosomal dominant mutations in PIK3R1, the gene encoding the regulatory subunit (p85α, p55α, and p50α) of class IA phosphoinositide 3-kinases. OBJECTIVES: We sought to review the clinical, immunologic, and histopathologic phenotypes of APDS2 in a genetically defined international patient cohort. METHODS: The medical and biological records of 36 patients with genetically diagnosed APDS2 were collected and reviewed. RESULTS: Mutations within splice acceptor and donor sites of exon 11 of the PIK3R1 gene lead to APDS2. Recurrent upper respiratory tract infections (100%), pneumonitis (71%), and chronic lymphoproliferation (89%, including adenopathy [75%], splenomegaly [43%], and upper respiratory tract lymphoid hyperplasia [48%]) were the most common features. Growth retardation was frequently noticed (45%). Other complications were mild neurodevelopmental delay (31%); malignant diseases (28%), most of them being B-cell lymphomas; autoimmunity (17%); bronchiectasis (18%); and chronic diarrhea (24%). Decreased serum IgA and IgG levels (87%), increased IgM levels (58%), B-cell lymphopenia (88%) associated with an increased frequency of transitional B cells (93%), and decreased numbers of naive CD4 and naive CD8 cells but increased numbers of CD8 effector/memory T cells were predominant immunologic features. The majority of patients (89%) received immunoglobulin replacement; 3 patients were treated with rituximab, and 6 were treated with rapamycin initiated after diagnosis of APDS2. Five patients died from APDS2-related complications. CONCLUSION: APDS2 is a combined immunodeficiency with a variable clinical phenotype. Complications are frequent, such as severe bacterial and viral infections, lymphoproliferation, and lymphoma similar to APDS1/PASLI-CD. Immunoglobulin replacement therapy, rapamycin, and, likely in the near future, selective phosphoinositide 3-kinase δ inhibitors are possible treatment options.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/etiología , Fenotipo , Adolescente , Adulto , Alelos , Biopsia , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Niño , Preescolar , Estudios de Cohortes , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Síndromes de Inmunodeficiencia/mortalidad , Masculino , Persona de Mediana Edad , Mutación , Sitios de Empalme de ARN , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Adulto Joven
17.
Blood ; 124(10): 1597-609, 2014 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-24970930

RESUMEN

Autoimmune lymphoproliferative syndrome (ALPS) caused by impaired FAS-mediated apoptosis of lymphocytes is characterized by lymphoproliferation, autoimmunity, but also an increased risk of invasive bacterial infection, notably following splenectomy. We surveyed a cohort of 100 ALPS patients (including 33 splenectomized) and found that 12 (10 splenectomized) had experienced 23 invasive bacterial infections mainly caused by Streptococcus pneumoniae. This vulnerability was associated with evidence of defective B-cell function characterized by low serum immunoglobulin (Ig) M, low IgM antibody production in response to S pneumoniae following nonconjugated immunization, and low blood memory B-cells counts (including marginal zone [MZ] B-cell counts). This immunodeficiency strongly correlated with intensity of lymphoproliferation. Spleen sections from 9 ALPS patients revealed double-negative T-cell (DN-T) infiltration of the MZ, which was depleted of B cells. MZ in ALPS patients contained an abnormally thick layer of MAdCAM-1((+)) stromal cells and an excess of DN-Ts. DN-Ts were shown to express MAdCAM-1 ligand, the α4ß7 integrin. These observations suggest that accumulating DN-Ts are trapped within stromal cell meshwork and interfere with correct localization of MZ B cells. Similar observations were made in spleens of fas-deficient mice. Our data revealed an unexpected mechanism by which ALPS results in anti-polysaccharide IgM antibody production-specific defect. Splenectomy should be avoided.


Asunto(s)
Formación de Anticuerpos , Síndrome Linfoproliferativo Autoinmune/inmunología , Síndrome Linfoproliferativo Autoinmune/patología , Lipopolisacáridos/inmunología , Bazo/inmunología , Bazo/patología , Adolescente , Adulto , Animales , Síndrome Linfoproliferativo Autoinmune/epidemiología , Síndrome Linfoproliferativo Autoinmune/cirugía , Niño , Preescolar , Humanos , Lactante , Recién Nacido , Ratones , Ratones Transgénicos , Bazo/cirugía , Esplenectomía/efectos adversos , Esplenectomía/estadística & datos numéricos , Infecciones Estreptocócicas/epidemiología , Infecciones Estreptocócicas/inmunología , Streptococcus pneumoniae/crecimiento & desarrollo , Streptococcus pneumoniae/inmunología , Adulto Joven
18.
J Med Genet ; 52(5): 348-52, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25691505

RESUMEN

BACKGROUND: Inherited mutations in DNA mismatch repair genes predispose to different cancer syndromes depending on whether they are mono-allelic or bi-allelic. This supports a causal relationship between expression level in the germline and phenotype variation. As a model to study this relationship, our study aimed to define the pathogenic characteristics of a recurrent homozygous coding variant in PMS2 displaying an attenuated phenotype identified by clinical genetic testing in seven Inuit families from Northern Quebec. METHODS: Pathogenic characteristics of the PMS2 mutation NM_000535.5:c.2002A>G were studied using genotype-phenotype correlation, single-molecule expression detection and single genome microsatellite instability analysis. RESULTS: This PMS2 mutation generates a de novo splice site that competes with the authentic site. In homozygotes, expression of the full-length protein is reduced to a level barely detectable by conventional diagnostics. Median age at primary cancer diagnosis is 22 years among 13 NM_000535.5:c.2002A>G homozygotes, versus 8 years in individuals carrying bi-allelic truncating mutations. Residual expression of full-length PMS2 transcript was detected in normal tissues from homozygotes with cancers in their 20s. CONCLUSIONS: Our genotype-phenotype study of c.2002A>G illustrates that an extremely low level of PMS2 expression likely delays cancer onset, a feature that could be exploited in cancer preventive intervention.


Asunto(s)
Adenosina Trifosfatasas/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/genética , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Efecto Fundador , Homocigoto , Mutación , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Fenotipo , Adolescente , Adulto , Anciano , Alelos , Niño , Preescolar , Mapeo Cromosómico , Exones , Femenino , Expresión Génica , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Adulto Joven
19.
J Allergy Clin Immunol ; 135(4): 998-1007.e6, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25312759

RESUMEN

BACKGROUND: Immunoglobulin class-switch recombination defects (CSR-D) are rare primary immunodeficiencies characterized by impaired production of switched immunoglobulin isotypes and normal or elevated IgM levels. They are caused by impaired T:B cooperation or intrinsic B cell defects. However, many immunoglobulin CSR-Ds are still undefined at the molecular level. OBJECTIVE: This study's objective was to delineate new causes of immunoglobulin CSR-Ds and thus gain further insights into the process of immunoglobulin class-switch recombination (CSR). METHODS: Exome sequencing in 2 immunoglobulin CSR-D patients identified variations in the INO80 gene. Functional experiments were performed to assess the function of INO80 on immunoglobulin CSR. RESULTS: We identified recessive, nonsynonymous coding variations in the INO80 gene in 2 patients affected by defective immunoglobulin CSR. Expression of wild-type INO80 in patients' fibroblastic cells corrected their hypersensitivity to high doses of γ-irradiation. In murine CH12-F3 cells, the INO80 complex accumulates at Sα and Eµ regions of the IgH locus, and downregulation of INO80 as well as its partners Reptin and Pontin impaired CSR. In addition, Reptin and Pontin were shown to interact with activation-induced cytidine deaminase. Finally, an abnormal separation of sister chromatids was observed upon INO80 downregulation in CH12-F3 cells, pinpointing its role in cohesin activity. CONCLUSION: INO80 deficiency appears to be associated with defective immunoglobulin CSR. We propose that the INO80 complex modulates cohesin function that may be required during immunoglobulin switch region synapsis.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/genética , Reordenamiento Génico , Cambio de Clase de Inmunoglobulina , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Supervivencia Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Variación Genética , Humanos , Isotipos de Inmunoglobulinas/genética , Región de Cambio de la Inmunoglobulina , Síndromes de Inmunodeficiencia/metabolismo , Modelos Biológicos , Unión Proteica , Transporte de Proteínas , Cohesinas
20.
Blood ; 122(24): 3940-50, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24159173

RESUMEN

B-cell responses are guided by the integration of signals through the B-cell receptor (BCR), CD40, and cytokine receptors. The common γ chain (γc)-binding cytokine interleukin (IL)-21 drives humoral immune responses via STAT3-dependent induction of transcription factors required for plasma cell generation. We investigated additional mechanisms by which IL-21/STAT3 signaling modulates human B-cell responses by studying patients with STAT3 mutations. IL-21 strongly induced CD25 (IL-2Rα) in normal, but not STAT3-deficient, CD40L-stimulated naïve B cells. Chromatin immunoprecipitation confirmed IL2RA as a direct target of STAT3. IL-21-induced CD25 expression was also impaired on B cells from patients with IL2RG or IL21R mutations, confirming a requirement for intact IL-21R signaling in this process. IL-2 increased plasmablast generation and immunoglobulin secretion from normal, but not CD25-deficient, naïve B cells stimulated with CD40L/IL-21. IL-2 and IL-21 were produced by T follicular helper cells, and neutralizing both cytokines abolished the B-cell helper capacity of these cells. Our results demonstrate that IL-21, via STAT3, sensitizes B cells to the stimulatory effects of IL-2. Thus, IL-2 may play an adjunctive role in IL-21-induced B-cell differentiation. Lack of this secondary effect of IL-21 may amplify the humoral immunodeficiency in patients with mutations in STAT3, IL2RG, or IL21R due to impaired responsiveness to IL-21.


Asunto(s)
Linfocitos B/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Interleucina-2/farmacología , Interleucinas/farmacología , Células Plasmáticas/efectos de los fármacos , Factor de Transcripción STAT3/genética , Linfocitos B/citología , Linfocitos B/metabolismo , Ligando de CD40/farmacología , Diferenciación Celular/genética , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Expresión Génica/efectos de los fármacos , Humanos , Interleucina-10/farmacología , Subunidad alfa del Receptor de Interleucina-2/genética , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcriptoma/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA