Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 324
Filtrar
1.
Front Immunol ; 12: 737427, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777346

RESUMEN

Activation-induced deaminase (AID) is the major actor of immunoglobulin (Ig) gene diversification in germinal center B-cells. From its first description, it was considered as mandatory for class switch recombination (CSR), and this discovery initiated a long quest for all of the AID-interacting factors controlling its activity. The mechanisms focusing AID-mediated DNA lesions to given target sequences remain incompletely understood with regards the detailed characterization of optimal substrates in which cytidine deamination will lead to double strand breaks (DSBs) and chromosomal cleavage. In an effort to reconsider whether such CSR breaks absolutely require AID, we herein provide evidence, based on deep-sequencing approaches, showing that this dogma is not absolute in both human and mouse B lymphocytes. In activated B-cells from either AID-deficient mice or human AID-deficient patients, we report an intrinsic ability of the IgH locus to undergo "on-target" cleavage and subsequent synapsis of broken regions in conditions able to yield low-level CSR. DNA breaks occur in such conditions within the same repetitive S regions usually targeted by AID, but their repair follows a specific pathway with increased usage of microhomology-mediated repair. These data further demonstrate the role of AID machinery as not initiating de novo chromosomal cleavage but rather catalyzing a process which spontaneously initiates at low levels in an appropriately conformed IgH locus.


Asunto(s)
Linfocitos B/enzimología , Citidina Desaminasa/deficiencia , Cambio de Clase de Inmunoglobulina , Cadenas Pesadas de Inmunoglobulina/genética , Síndromes de Inmunodeficiencia/genética , Activación de Linfocitos , Animales , Linfocitos B/inmunología , Citidina Desaminasa/genética , Roturas del ADN , Reparación del ADN por Unión de Extremidades , Modelos Animales de Enfermedad , Sitios Genéticos , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/inmunología , Ratones Noqueados
2.
Front Immunol ; 12: 667430, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34093563

RESUMEN

Background: Autoinflammatory phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) is a rare autoinflammatory disease caused by gain-of-function mutations in the PLCG2 gene. Here we report a rare case of APLAID patient carrying a novel heterozygous missense PLCG2 I169V mutation with gangrenous pyoderma and concomitant high serum immunoglobulin (Ig) E level. Methods: The patient was diagnosed as APLAID and has been treated in our department. His phenotype and genotype were carefully documented and studied. We also conducted a comprehensive literature review on APLAID. Results: A 23-year-old Chinese Han man presented with recurrent fever for 18 years and vesiculopustular rashes for 9 years, along with chronic bronchitis, leukocytosis, increased C-reactive protein, immunodeficiency and high serum IgE. Skin biopsy showed chronic inflammatory cells infiltration. A paternal heterozygous missense variant in exon 6 of the PLCG2 gene p. I169V was identified. His vesiculopustular and IgE level responded to medium dose corticosteroids. After withdrawal of steroids, he developed severe arthritis and a large deteriorating ulceration resembling pyoderma gangrenosum on the left knee. Large dose corticosteroids were suboptimal. Then he received adalimumab with satisfactory response for arthritis and skin lesion. But he got an immunodeficiency-associated lymphoproliferative disorder 2 months later. Through literature review, there were a total of 10 APLAID patients reported by six English-language publications. Vesiculopustular rashes, sinopulmonary infection and immunodeficiency were the most frequent symptoms of APLAID patients. Glucocorticoids, intravenous immunoglobulin and biologics were clinically used to treat APLAID but none of these patients had a complete recovery. Conclusions: The rarity and diversity of APLAID make it difficult to be diagnosed. Our study reported the first case of APLAID with gangrenous pyoderma and concomitant high IgE carrying a novel PLCG2 mutation, which may expand the clinical phenotype and genotype of APLAID.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Fosfolipasa C gamma/genética , Piodermia Gangrenosa/genética , Exones , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/enzimología , Masculino , Mutación Missense , Fenotipo , Fosfolipasa C gamma/metabolismo , Piodermia Gangrenosa/complicaciones , Adulto Joven
3.
J Clin Invest ; 129(10): 4194-4206, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31449058

RESUMEN

Polymerase δ is essential for eukaryotic genome duplication and synthesizes DNA at both the leading and lagging strands. The polymerase δ complex is a heterotetramer comprising the catalytic subunit POLD1 and the accessory subunits POLD2, POLD3, and POLD4. Beyond DNA replication, the polymerase δ complex has emerged as a central element in genome maintenance. The essentiality of polymerase δ has constrained the generation of polymerase δ-knockout cell lines or model organisms and, therefore, the understanding of the complexity of its activity and the function of its accessory subunits. To our knowledge, no germline biallelic mutations affecting this complex have been reported in humans. In patients from 2 independent pedigrees, we have identified what we believe to be a novel syndrome with reduced functionality of the polymerase δ complex caused by germline biallelic mutations in POLD1 or POLD2 as the underlying etiology of a previously unknown autosomal-recessive syndrome that combines replicative stress, neurodevelopmental abnormalities, and immunodeficiency. Patients' cells showed impaired cell-cycle progression and replication-associated DNA lesions that were reversible upon overexpression of polymerase δ. The mutations affected the stability and interactions within the polymerase δ complex or its intrinsic polymerase activity. We believe our discovery of human polymerase δ deficiency identifies the central role of this complex in the prevention of replication-related DNA lesions, with particular relevance to adaptive immunity.


Asunto(s)
ADN Polimerasa III/deficiencia , ADN Polimerasa III/genética , Mutación de Línea Germinal , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Adolescente , Alelos , Sustitución de Aminoácidos , ADN Polimerasa III/química , Replicación del ADN/genética , Estabilidad de Enzimas/genética , Genes Recesivos , Humanos , Masculino , Modelos Moleculares , Complejos Multienzimáticos/química , Complejos Multienzimáticos/deficiencia , Complejos Multienzimáticos/genética , Mutación Missense , Trastornos del Neurodesarrollo/enzimología , Trastornos del Neurodesarrollo/genética , Linaje , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Adulto Joven
5.
Front Immunol ; 9: 979, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29867957

RESUMEN

Patients with primary immunodeficiency can be prone to severe Epstein-Barr virus (EBV) associated immune dysregulation. Individuals with mutations in the interleukin-2-inducible T-cell kinase (ITK) gene experience Hodgkin and non-Hodgkin lymphoma, EBV lymphoproliferative disease, hemophagocytic lymphohistiocytosis, and dysgammaglobulinemia. In this review, we give an update on further reported patients. We believe that current clinical data advocate early definitive treatment by hematopoietic stem cell transplantation, as transplant outcome in primary immunodeficiency disorders in general has gradually improved in recent years. Furthermore, we summarize experimental data in the murine model to provide further insight of pathophysiology in ITK deficiency.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Síndromes de Inmunodeficiencia/enzimología , Trastornos Linfoproliferativos/inmunología , Proteínas Tirosina Quinasas/deficiencia , Adolescente , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Infecciones por Virus de Epstein-Barr/inmunología , Femenino , Trasplante de Células Madre Hematopoyéticas , Herpesvirus Humano 4/inmunología , Enfermedad de Hodgkin , Humanos , Síndromes de Inmunodeficiencia/diagnóstico , Recién Nacido , Linfoma no Hodgkin/complicaciones , Masculino , Ratones , Ratones Noqueados , Proteínas Tirosina Quinasas/inmunología , Linfocitos T/inmunología
6.
Front Immunol ; 9: 2863, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619256

RESUMEN

Background: The auto-inflammation and phospholipase Cγ2 (PLCγ2)-associated antibody deficiency and immune dysregulation (APLAID) syndrome is a rare primary immunodeficiency caused by a gain-of-function mutation S707Y in the PLCG2 gene previously described in two patients from one family. The APLAID patients presented with early-onset blistering skin lesions, posterior uveitis, inflammatory bowel disease (IBD) and recurrent sinopulmonary infections caused by a humoral defect, but lacked circulating autoantibodies and had no cold-induced urticaria, contrary to the patients with the related PLAID syndrome. Case: We describe a new APLAID patient who presented with vesiculopustular rash in the 1st weeks of life, followed by IBD, posterior uveitis, recurrent chest infections, interstitial pneumonitis, and also had sensorineural deafness and cutis laxa. Her disease has been refractory to most treatments, including IL1 blockers and a trial with ruxolitinib has been attempted. Results: In this patient, we found a unique de novo heterozygous missense L848P mutation in the PLCG2 gene, predicted to affect the PLCγ2 structure. Similarly to S707Y, the L848P mutation led to the increased basal and EGF-stimulated PLCγ2 activity in vitro. Whole blood assays showed reduced production of IFN-γ and IL-17 in response to polyclonal T-cell stimulation and reduced production of IL-10 and IL-1ß after LPS stimulation. Reduced IL-1ß levels and the lack of clinical response to treatment with IL-1 blockers argue against NLRP3 inflammasome hyperactivation being the main mechanism mediating the APLAID pathogenesis. Conclusion: Our findings indicate that L848P is novel a gain-of-function mutation that leads to PLCγ2 activation and suggest cutis laxa as a possible clinical manifestations of the APLAID syndrome.


Asunto(s)
Cutis Laxo/genética , Enfermedades Autoinflamatorias Hereditarias/genética , Síndromes de Inmunodeficiencia/genética , Mutación Missense , Fosfolipasa C gamma/genética , Secuencia de Aminoácidos , Secuencia de Bases , Cutis Laxo/complicaciones , Cutis Laxo/enzimología , Análisis Mutacional de ADN , Femenino , Enfermedades Autoinflamatorias Hereditarias/complicaciones , Enfermedades Autoinflamatorias Hereditarias/enzimología , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/enzimología , Recién Nacido , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/genética , Masculino , Linaje , Fosfolipasa C gamma/química , Fosfolipasa C gamma/metabolismo , Homología de Secuencia de Aminoácido
7.
Blood ; 130(21): 2307-2316, 2017 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-28972011

RESUMEN

Pathogenic gain-of-function variants in the genes encoding phosphoinositide 3-kinase δ (PI3Kδ) lead to accumulation of transitional B cells and senescent T cells, lymphadenopathy, and immune deficiency (activated PI3Kδ syndrome [APDS]). Knowing the genetic etiology of APDS afforded us the opportunity to explore PI3Kδ inhibition as a precision-medicine therapy. Here, we report in vitro and in vivo effects of inhibiting PI3Kδ in APDS. Treatment with leniolisib (CDZ173), a selective PI3Kδ inhibitor, caused dose-dependent suppression of PI3Kδ pathway hyperactivation (measured as phosphorylation of AKT/S6) in cell lines ectopically expressing APDS-causative p110δ variants and in T-cell blasts derived from patients. A clinical trial with 6 APDS patients was conducted as a 12-week, open-label, multisite, within-subject, dose-escalation study of oral leniolisib to assess safety, pharmacokinetics, and effects on lymphoproliferation and immune dysregulation. Oral leniolisib led to a dose-dependent reduction in PI3K/AKT pathway activity assessed ex vivo and improved immune dysregulation. We observed normalization of circulating transitional and naive B cells, reduction in PD-1+CD4+ and senescent CD57+CD4- T cells, and decreases in elevated serum immunoglobulin M and inflammatory markers including interferon γ, tumor necrosis factor, CXCL13, and CXCL10 with leniolisib therapy. After 12 weeks of treatment, all patients showed amelioration of lymphoproliferation with lymph node sizes and spleen volumes reduced by 39% (mean; range, 26%-57%) and 40% (mean; range, 13%-65%), respectively. Thus, leniolisib was well tolerated and improved laboratory and clinical parameters in APDS, supporting the specific inhibition of PI3Kδ as a promising new targeted therapy in APDS and other diseases characterized by overactivation of the PI3Kδ pathway. This trial was registered at www.clinicaltrials.gov as #NCT02435173.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Síndromes de Inmunodeficiencia/tratamiento farmacológico , Síndromes de Inmunodeficiencia/enzimología , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Animales , Quimiocinas/sangre , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Demografía , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunoglobulina M/sangre , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Lactante , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Activación de Linfocitos/efectos de los fármacos , Masculino , Mutación/genética , Tamaño de los Órganos , Fenotipo , Enfermedades de Inmunodeficiencia Primaria , Piridinas/farmacocinética , Pirimidinas/farmacocinética , Ratas , Bazo/efectos de los fármacos , Bazo/patología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Transfección
8.
Blood ; 130(24): 2682-2688, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-28974505

RESUMEN

Deficiency of adenosine deaminase 2 (DADA2) is caused by biallelic deleterious mutations in CECR1 DADA2 results in variable autoinflammation and vasculopathy (recurrent fevers, livedo reticularis, polyarteritis nodosa, lacunar ischemic strokes, and intracranial hemorrhages), immunodeficiency and bone marrow failure. Tumor necrosis factor-α blockade is the treatment of choice for the autoinflammation and vascular manifestations. Hematopoietic stem cell transplantation (HSCT) represents a potential definitive treatment. We present a cohort of 14 patients from 6 countries who received HSCT for DADA2. Indication for HSCT was bone marrow dysfunction or immunodeficiency. Six of 14 patients had vasculitis pre-HSCT. The median age at HSCT was 7.5 years. Conditioning regimens were myeloablative (9) and reduced intensity (5). Donors were HLA-matched sibling (n = 1), HLA-matched unrelated (n = 9), HLA-mismatched unrelated (n = 3), and HLA haploidentical sibling (n = 1). All patients are alive and well with no new vascular events and resolution of hematological and immunological phenotype at a median follow-up of 18 months (range, 5 months to 13 years). Plasma ADA2 enzyme activity normalized in those tested post-HSCT (7/7), as early as day +14 (myeloid engraftment). Post-HSCT hematological autoimmunity (cytopenias) was reported in 4 patients, acute graft-versus-host disease grade 1 in 2, grade 2 in 3, and grade 3-4 in 1, and moderate chronic graft-versus-host disease in 1 patient. In conclusion, in 14 patients, HSCT was an effective and definitive treatment of DADA2.


Asunto(s)
Adenosina Desaminasa/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Péptidos y Proteínas de Señalización Intercelular/genética , Mutación , Adenosina Desaminasa/sangre , Adenosina Desaminasa/metabolismo , Adolescente , Niño , Preescolar , Estudios de Cohortes , Femenino , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Síndromes de Inmunodeficiencia/enzimología , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular/sangre , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Masculino , Fenotipo , Acondicionamiento Pretrasplante/métodos
9.
J Immunol Res ; 2017: 1514294, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28900629

RESUMEN

Sialic acid acetylesterase (SIAE) deficiency was suggested to lower the levels of ligands for sialic acid-binding immunoglobulin-like receptors, decreasing the threshold for B-cell activation. In humans, studies of rare heterozygous loss-of-function mutations in SIAE gene in common autoimmune diseases, including juvenile idiopathic arthritis (JIA), yielded inconsistent results. Considering the distinct pathogenesis of the two main subtypes of JIA, autoinflammatory systemic (sJIA) and autoimmune oligo/polyarticular (aJIA), and a predisposition to autoimmunity displayed by patients and families with primary antibody deficiencies (PADs), the aim of our study was to analyze whether SIAE rare variants are associated with both the phenotype of JIA and the autoimmunity risk in families with PADs. A cohort of 69 patients with JIA, 117 healthy children, 54 patients, and family members with PADs were enrolled in the study. Three novel SIAE variants (p.Q343P, p.Y495X, and c.1320+33T>C) were found only in patients with aJIA but interestingly also in their healthy relatives without autoimmunity, while none of PAD patients or their relatives carried SIAE defects. Our results show that SIAE rare variants are not causative of autoimmunity as single defects.


Asunto(s)
Acetilesterasa/genética , Artritis Juvenil/genética , Variación Genética , Síndromes de Inmunodeficiencia/genética , Acetilesterasa/metabolismo , Adolescente , Alelos , Artritis Juvenil/enzimología , Artritis Juvenil/inmunología , Autoinmunidad , Estudios de Casos y Controles , Niño , Preescolar , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Síndromes de Inmunodeficiencia/enzimología , Masculino , Mutación , Polimorfismo de Nucleótido Simple
10.
Cell Signal ; 28(9): 1237-1251, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27196803

RESUMEN

Deletions in the gene encoding signal-transducing inositol phospholipid-specific phospholipase C-γ2 (PLCγ2) are associated with the novel human hereditary disease PLAID (PLCγ2-associated antibody deficiency and immune dysregulation). PLAID is characterized by a rather puzzling concurrence of augmented and diminished functions of the immune system, such as cold urticaria triggered by only minimal decreases in temperature, autoimmunity, and immunodeficiency. Understanding of the functional effects of the genomic alterations at the level of the affected enzyme, PLCγ2, is currently lacking. PLCγ2 is critically involved in coupling various cell surface receptors to regulation of important functions of immune cells such as mast cells, B cells, monocytes/macrophages, and neutrophils. PLCγ2 is unique by carrying three Src (SH) and one split pleckstrin homology domain (spPH) between the two catalytic subdomains (spPHn-SH2n-SH2c-SH3-spPHc). Prevailing evidence suggests that activation of PLCγ2 is primarily due to loss of SH-region-mediated autoinhibition and/or enhanced plasma membrane translocation. Here, we show that the two PLAID PLCγ2 mutants lacking portions of the SH region are strongly (>100-fold), rapidly, and reversibly activated by cooling by only a few degrees. We found that the mechanism(s) underlying PLCγ2 PLAID mutant activation by cool temperatures is distinct from a mere loss of SH-region-mediated autoinhibition and dependent on both the integrity and the pliability of the spPH domain. The results suggest a new mechanism of PLCγ activation with unique thermodynamic features and assign a novel regulatory role to its spPH domain. Involvement of this mechanism in other human disease states associated with cooling such as exertional asthma and certain acute coronary events appears an intriguing possibility.


Asunto(s)
Frío , Síndromes de Inmunodeficiencia/enzimología , Fosfolipasa C gamma/metabolismo , Animales , Células COS , Chlorocebus aethiops , Activación Enzimática , Receptores ErbB/metabolismo , Exones/genética , Eliminación de Gen , Humanos , Síndromes de Inmunodeficiencia/patología , Isoenzimas/metabolismo , Biosíntesis de Proteínas , Dominios Proteicos
11.
Biochimie ; 126: 27-30, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26853819

RESUMEN

Disruptions in folate-mediated one-carbon metabolism (FOCM) are associated with risk for several pathologies including developmental anomalies such as neural tube defects and congenital heart defects, diseases of aging including cognitive decline, neurodegeneration and epithelial cancers, and hematopoietic disorders including megaloblastic anemia. However, the causal pathways and mechanisms that underlie these pathologies remain unresolved. Because folate-dependent anabolic pathways are tightly interconnected and best described as a metabolic network, the identification of causal pathways and associated mechanisms of pathophysiology remains a major challenge in identifying the contribution of individual pathways to disease phenotypes. Investigations of genetic mouse models and human inborn errors of metabolism enable a more precise dissection of the pathways that constitute the FOCM network and enable elucidation of causal pathways associated with NTDs. In this overview, we summarize recent evidence that the enzyme MTHFD1 plays an essential role in FOCM in humans and in mice, and that it determines the partitioning of folate-activated one carbon units between the folate-dependent de novo thymidylate and homocysteine remethylation pathways through its regulated nuclear localization. We demonstrate that impairments in MTHFD1 activity compromise both homocysteine remethylation and de novo thymidylate biosynthesis, and provide evidence that MTHFD1-associated disruptions in de novo thymidylate biosynthesis lead to genome instability that may underlie folate-associated immunodeficiency and birth defects.


Asunto(s)
Inestabilidad Genómica , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Timidina Monofosfato/biosíntesis , Animales , Anomalías Congénitas/enzimología , Anomalías Congénitas/genética , Ácido Fólico/biosíntesis , Ácido Fólico/genética , Homocisteína/biosíntesis , Homocisteína/genética , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Errores Innatos del Metabolismo/enzimología , Errores Innatos del Metabolismo/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Ratones , Antígenos de Histocompatibilidad Menor/genética
13.
Oncotarget ; 6(42): 44151-60, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26683521

RESUMEN

Genetic deficiencies provide insights into gene function in humans. Here we describe a patient with a very rare genetic deficiency of ADAM17. We show that the patient's PBMCs had impaired cytokine secretion in response to LPS stimulation, correlating with the clinical picture of severe bacteremia from which the patient suffered. ADAM17 was shown to cleave CD16, a major NK killer receptor. Functional analysis of patient's NK cells demonstrated that his NK cells express normal levels of activating receptors and maintain high surface levels of CD16 following mAb stimulation. Activation of individual NK cell receptors showed that the patient's NK cells are more potent when activated directly by CD16, albeit no difference was observed in Antibody Depedent Cytotoxicity (ADCC) assays. Our data suggest that ADAM17 inhibitors currently considered for clinical use to boost CD16 activity should be cautiously applied, as they might have severe side effects resulting from impaired cytokine secretion.


Asunto(s)
Proteínas ADAM/deficiencia , Citocinas/metabolismo , Síndromes de Inmunodeficiencia/enzimología , Células Asesinas Naturales/enzimología , Leucocitos Mononucleares/enzimología , Activación de Linfocitos , Proteínas ADAM/genética , Proteínas ADAM/inmunología , Proteína ADAM17 , Citotoxicidad Celular Dependiente de Anticuerpos , Línea Celular Tumoral , Preescolar , Citocinas/inmunología , Resultado Fatal , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Inmunidad Innata , Síndromes de Inmunodeficiencia/diagnóstico , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Lipopolisacáridos/farmacología , Masculino , Fenotipo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo
14.
J Pediatr Hematol Oncol ; 37(8): 616-22, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26479985

RESUMEN

Glucose-6-phosphatase catalytic subunit 3 (G6PC3) deficiency was recently defined as a new severe congenital neutropenia subgroup remarkable with congenital heart defects, urogenital malformations, endocrine abnormalities, and prominent superficial veins. Here, we report 3 patients with G6PC3 deficiency presenting with recurrent diarrhea, failure to thrive, and sinopulmonary infections leading to bronchiectasis. In patient I and II, a combined immune deficiency was suspected due to early-onset disease with lymphopenia, neutropenia, and thrombocytopenia, along with variable reductions in lymphocyte subpopulations and favorable response to intravenous γ-globulin therapy. Apart from neutropenia, all 3 patients had intermittent thrombocytopenia, anemia, and lymphopenia. All patients had failure to thrive and some of the classic syndromic features of G6PC3 deficiency, including cardiac abnormalities and visibility of superficial veins in all, endocrinologic problems in PI and PIII, and urogenital abnormalities in PII. Our experience suggests that a diagnosis of congenital neutropenia due to G6PC3 may not be as straightforward in such patients with combined lymphopenia and thrombocytopenia. A high index of suspicion and the other syndromic features of G6PC3 were clues to diagnosis. Screening of all combined immune deficiencies with neutropenia may help to uncover the whole spectra of G6PC3 deficiency.


Asunto(s)
Anomalías Múltiples/genética , Glucosa-6-Fosfatasa/genética , Enfermedad del Almacenamiento de Glucógeno Tipo I/genética , Síndromes de Inmunodeficiencia/genética , Subgrupos Linfocitarios/patología , Neutropenia/genética , Anomalías Múltiples/enzimología , Adolescente , Bronquiectasia/etiología , Dominio Catalítico , Linaje de la Célula , Niño , Codón sin Sentido , Colitis/enzimología , Colitis/genética , Consanguinidad , Diarrea/enzimología , Diarrea/genética , Exones/genética , Insuficiencia de Crecimiento/enzimología , Insuficiencia de Crecimiento/genética , Femenino , Mutación del Sistema de Lectura , Enfermedad del Almacenamiento de Glucógeno Tipo I/inmunología , Humanos , Síndromes de Inmunodeficiencia/enzimología , Linfopenia/congénito , Linfopenia/enzimología , Linfopenia/genética , Masculino , Mutagénesis Insercional , Neutropenia/enzimología , Linaje , Fenotipo , Sitios de Empalme de ARN/genética , Infecciones del Sistema Respiratorio/complicaciones , Trombocitopenia/congénito , Trombocitopenia/enzimología , Trombocitopenia/genética , Turquía
15.
J Clin Immunol ; 35(1): 11-4, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25352054

RESUMEN

Gain of function (GOF) mutation in the p110δ catalytic subunit of the phosphatidylinositol-3-OH kinase (PIK3CD) is the cause of a primary immunodeficiency (PID) characterized by recurrent sinopulmonary infections and lymphoproliferation. We describe a family of two adults and three children with GOF mutation in PIK3CD, all with recurrent sinopulmonary infections and varied infectious and non-infectious complications. The two adults have Primary Sclerosing Cholangitis (PSC) without evidence of Cryptosporidium parvum infection and have required liver transplantation. PSC is a novel phenotype of GOF mutation in PIK3CD.


Asunto(s)
Colangitis Esclerosante/enzimología , Colangitis Esclerosante/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Mutación Missense , Adulto , Sustitución de Aminoácidos , Niño , Colangitis Esclerosante/inmunología , Fosfatidilinositol 3-Quinasa Clase I/inmunología , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Femenino , Heterocigoto , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Hígado/patología , Masculino , Linaje
16.
J Exp Med ; 211(13): 2537-47, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25488983

RESUMEN

Class IA phosphatidylinositol 3-kinases (PI3K), which generate PIP3 as a signal for cell growth and proliferation, exist as an intracellular complex of a catalytic subunit bound to a regulatory subunit. We and others have previously reported that heterozygous mutations in PIK3CD encoding the p110δ catalytic PI3K subunit cause a unique disorder termed p110δ-activating mutations causing senescent T cells, lymphadenopathy, and immunodeficiency (PASLI) disease. We report four patients from three families with a similar disease who harbor a recently reported heterozygous splice site mutation in PIK3R1, which encodes the p85α, p55α, and p50α regulatory PI3K subunits. These patients suffer from recurrent sinopulmonary infections and lymphoproliferation, exhibit hyperactive PI3K signaling, and have prominent expansion and skewing of peripheral blood CD8(+) T cells toward terminally differentiated senescent effector cells with short telomeres. The PIK3R1 splice site mutation causes skipping of an exon, corresponding to loss of amino acid residues 434-475 in the inter-SH2 domain. The mutant p85α protein is expressed at low levels in patient cells and activates PI3K signaling when overexpressed in T cells from healthy subjects due to qualitative and quantitative binding changes in the p85α-p110δ complex and failure of the C-terminal region to properly inhibit p110δ catalytic activity.


Asunto(s)
Empalme Alternativo/genética , Genes Dominantes , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Trastornos Linfoproliferativos/genética , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Adolescente , Adulto , Formación de Anticuerpos , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Dominio Catalítico , Diferenciación Celular , Preescolar , Fosfatidilinositol 3-Quinasa Clase Ia , Activación Enzimática , Exones/genética , Femenino , Heterocigoto , Humanos , Síndromes de Inmunodeficiencia/inmunología , Trastornos Linfoproliferativos/enzimología , Trastornos Linfoproliferativos/inmunología , Masculino , Datos de Secuencia Molecular , Linaje , Fosfatidilinositol 3-Quinasas/química , Estructura Terciaria de Proteína , Eliminación de Secuencia , Transducción de Señal , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Telómero/metabolismo
17.
Hum Mol Genet ; 23(24): 6448-57, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25027325

RESUMEN

Immunodeficiency, centromeric instability and facial anomalies type I (ICF1) syndrome is a rare genetic disease caused by mutations in DNA methyltransferase (DNMT) 3B, a de novo DNA methyltransferase. However, the molecular basis of how DNMT3B deficiency leads to ICF1 pathogenesis is unclear. Induced pluripotent stem cell (iPSC) technology facilitates the study of early human developmental diseases via facile in vitro paradigms. Here, we generate iPSCs from ICF Type 1 syndrome patient fibroblasts followed by directed differentiation of ICF1-iPSCs to mesenchymal stem cells (MSCs). By performing genome-scale bisulfite sequencing, we find that DNMT3B-deficient iPSCs exhibit global loss of non-CG methylation and select CG hypomethylation at gene promoters and enhancers. Further unbiased scanning of ICF1-iPSC methylomes also identifies large megabase regions of CG hypomethylation typically localized in centromeric and subtelomeric regions. RNA sequencing of ICF1 and control iPSCs reveals abnormal gene expression in ICF1-iPSCs relevant to ICF syndrome phenotypes, some directly associated with promoter or enhancer hypomethylation. Upon differentiation of ICF1 iPSCs to MSCs, we find virtually all CG hypomethylated regions remained hypomethylated when compared with either wild-type iPSC-derived MSCs or primary bone-marrow MSCs. Collectively, our results show specific methylome and transcriptome defects in both ICF1-iPSCs and differentiated somatic cell lineages, providing a valuable stem cell system for further in vitro study of the molecular pathogenesis of ICF1 syndrome. GEO accession number: GSE46030.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Epigénesis Genética , Genoma Humano , Síndromes de Inmunodeficiencia/genética , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Mesenquimatosas/enzimología , Diferenciación Celular , ADN (Citosina-5-)-Metiltransferasas/deficiencia , Metilación de ADN , Elementos de Facilitación Genéticos , Fibroblastos/enzimología , Fibroblastos/patología , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/patología , Células Madre Pluripotentes Inducidas/patología , Células Madre Mesenquimatosas/patología , Regiones Promotoras Genéticas , ADN Metiltransferasa 3B
18.
Nature ; 510(7504): 288-92, 2014 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-24870241

RESUMEN

Lymphocyte functions triggered by antigen recognition and co-stimulation signals are associated with a rapid and intense cell division, and hence with metabolism adaptation. The nucleotide cytidine 5' triphosphate (CTP) is a precursor required for the metabolism of DNA, RNA and phospholipids. CTP originates from two sources: a salvage pathway and a de novo synthesis pathway that depends on two enzymes, the CTP synthases (or synthetases) 1 and 2 (CTPS1 with CTPS2); the respective roles of these two enzymes are not known. CTP synthase activity is a potentially important step for DNA synthesis in lymphocytes. Here we report the identification of a loss-of-function homozygous mutation (rs145092287) in CTPS1 in humans that causes a novel and life-threatening immunodeficiency, characterized by an impaired capacity of activated T and B cells to proliferate in response to antigen receptor-mediated activation. In contrast, proximal and distal T-cell receptor (TCR) signalling events and responses were only weakly affected by the absence of CTPS1. Activated CTPS1-deficient cells had decreased levels of CTP. Normal T-cell proliferation was restored in CTPS1-deficient cells by expressing wild-type CTPS1 or by addition of exogenous CTP or its nucleoside precursor, cytidine. CTPS1 expression was found to be low in resting T cells, but rapidly upregulated following TCR activation. These results highlight a key and specific role of CTPS1 in the immune system by its capacity to sustain the proliferation of activated lymphocytes during the immune response. CTPS1 may therefore represent a therapeutic target of immunosuppressive drugs that could specifically dampen lymphocyte activation.


Asunto(s)
Ligasas de Carbono-Nitrógeno/deficiencia , Ligasas de Carbono-Nitrógeno/metabolismo , Activación de Linfocitos , Linfocitos/citología , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Complejo CD3/inmunología , Ligasas de Carbono-Nitrógeno/genética , Proliferación Celular , Preescolar , Citidina Trifosfato/metabolismo , Femenino , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Lactante , Recién Nacido , Activación de Linfocitos/genética , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Mutación/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo
19.
J Leukoc Biol ; 94(6): 1243-51, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23935208

RESUMEN

Chemokine receptor interactions coordinate leukocyte migration in inflammation. Chemokine receptors are GPCRs that when activated, are phosphorylated by GRKs to turn off G protein-mediated signaling yet recruit additional signaling machinery. Recently, GRK3 was identified as a negative regulator of CXCL12/CXCR4 signaling that is defective in human WHIM syndrome. Here, we report that GRK3-/- mice exhibit numerous features of human WHIM, such as impaired CXCL12-mediated desensitization, enhanced CXCR4 signaling to ERK activation, altered granulocyte migration, and a mild myelokathexis. Moreover, GRK3-/- protects mice from two acute models of inflammatory arthritis (K/BxN serum transfer and CAIA). In these granulocyte-dependent disease models, protection of GRK3-/- mice is mediated by retention of cells in the marrow, fewer circulating granulocytes in the peripheral blood, and reduced granulocytes in the joints during active inflammation. In contrast to WHIM, GRK3-/- mice have minimal hypogammaglobulinemia and a peripheral leukocytosis with increased lymphocytes and absent neutropenia. Thus, we conclude that the loss of GRK3-mediated regulation of CXCL12/CXCR4 signaling contributes to some, but not all, of the complete WHIM phenotype and that GRK3 inhibition may be beneficial in the treatment of inflammatory arthritis.


Asunto(s)
Quinasa 3 del Receptor Acoplado a Proteína-G/inmunología , Síndromes de Inmunodeficiencia/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Verrugas/inmunología , Animales , Línea Celular Transformada , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Quinasa 3 del Receptor Acoplado a Proteína-G/genética , Quinasa 3 del Receptor Acoplado a Proteína-G/metabolismo , Granulocitos/enzimología , Granulocitos/inmunología , Granulocitos/patología , Humanos , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Enfermedades de Inmunodeficiencia Primaria , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Verrugas/enzimología , Verrugas/genética , Verrugas/patología
20.
J Immunol ; 191(4): 1765-74, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23842753

RESUMEN

The IFN-inducible protein Irgm1 (LRG-47) belongs to the family of immunity-related GTPases that function in cell-autonomous resistance against intracellular pathogens in mice. Irgm1 deficiency is associated with a severe immunodeficiency syndrome. The protein has been variously interpreted as a direct effector molecule on bacterial phagosomes or on other organelles or as an inducer of autophagy. In this study, we re-examined one of these claims, namely that Irgm1 targets mycobacterial and listerial phagosomes. We found no colocalization of endogenous Irgm1, using two immunofluorescent staining techniques, either in fibroblasts or in macrophages. We demonstrated the predicted existence of two protein isoforms of Irgm1 derived from differential splicing and described immunological reagents for their detection. Both Irgm1 isoforms localize to the Golgi apparatus and weakly to mitochondria; however, only the long Irgm1 isoforms can be detected on endolysosomal membranes. Together with the previous observation that the general immunodeficiency phenotype of Irgm1(-/-) mice is reversed in Irgm1/Irgm3 double-deficient mice, our results argue against a direct effector function of Irgm1 at the bacterial phagosome. We discuss these findings in the context of evidence that Irgm1 functions as a negative regulator of other members of the immunity-related GTPase protein family.


Asunto(s)
Proteínas de Unión al GTP/inmunología , Interferón gamma/farmacología , Fagosomas/inmunología , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Especificidad de Anticuerpos , Línea Celular , Fibroblastos/química , Técnica del Anticuerpo Fluorescente Directa , GTP Fosfohidrolasas/clasificación , Proteínas de Unión al GTP/análisis , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Aparato de Golgi/química , Humanos , Inmunización , Síndromes de Inmunodeficiencia/enzimología , Síndromes de Inmunodeficiencia/genética , Listeria monocytogenes/inmunología , Macrófagos/química , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/química , Datos de Secuencia Molecular , Mycobacterium bovis/inmunología , Fragmentos de Péptidos/inmunología , Fagosomas/microbiología , Isoformas de Proteínas/análisis , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...