Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Allergy Clin Immunol ; 143(2): 726-735, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-29772310

RESUMO

BACKGROUND: Mutations in recombination-activating gene (RAG) 1 and RAG2 are associated with a broad range of clinical and immunologic phenotypes in human subjects. OBJECTIVE: Using a flow cytometry-based assay, we aimed to measure the recombinase activity of naturally occurring RAG2 mutant proteins and to correlate our results with the severity of the clinical and immunologic phenotype. METHODS: Abelson virus-transformed Rag2-/- pro-B cells engineered to contain an inverted green fluorescent protein (GFP) cassette flanked by recombination signal sequences were transduced with retroviruses encoding either wild-type or 41 naturally occurring RAG2 variants. Bicistronic vectors were used to introduce compound heterozygous RAG2 variants. The percentage of GFP-expressing cells was evaluated by using flow cytometry, and high-throughput sequencing was used to analyze rearrangements at the endogenous immunoglobulin heavy chain (Igh) locus. RESULTS: The RAG2 variants showed a wide range of recombination activity. Mutations associated with severe combined immunodeficiency and Omenn syndrome had significantly lower activity than those detected in patients with less severe clinical presentations. Four variants (P253R, F386L, N474S, and M502V) previously thought to be pathogenic were found to have wild-type levels of activity. Use of bicistronic vectors permitted us to assess more carefully the effect of compound heterozygous mutations, with good correlation between GFP expression and the number and diversity of Igh rearrangements. CONCLUSIONS: Our data support genotype-phenotype correlation in the setting of RAG2 deficiency. The assay described can be used to define the possible disease-causing role of novel RAG2 variants and might help predict the severity of the clinical phenotype.


Assuntos
Linfócitos B/fisiologia , Proteínas de Ligação a DNA/genética , Cadeias Pesadas de Imunoglobulinas/genética , Mutação/genética , Proteínas Nucleares/genética , Receptores de Antígenos de Linfócitos B/genética , Imunodeficiência Combinada Severa/genética , Adolescente , Linhagem Celular Transformada , Criança , Pré-Escolar , Progressão da Doença , Feminino , Técnicas de Silenciamento de Genes , Estudos de Associação Genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lactente , Recém-Nascido , Masculino , Polimorfismo Genético
2.
Arthritis Rheumatol ; 68(7): 1758-68, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26815131

RESUMO

OBJECTIVE: Treg cell-mediated suppression of Teff cells is impaired in juvenile idiopathic arthritis (JIA); however, the basis for this dysfunction is incompletely understood. Animal models of autoimmunity and immunodeficiency demonstrate that a diverse Treg cell repertoire is essential to maintain Treg cell function. The present study was undertaken to investigate the Treg and Teff cell repertoires in JIA. METHODS: Treg cells (CD4+CD25+CD127(low) ) and Teff cells (CD4+CD25-) were isolated from peripheral blood and synovial fluid obtained from JIA patients, healthy controls, and children with Lyme arthritis. Treg cell function was measured in suppressive assays. The T cell receptor ß chain (TRB) was amplified by multiplex polymerase chain reaction and next-generation sequencing was performed, with amplicons sequenced using an Illumina HiSeq platform. Data were analyzed using ImmunoSEQ, International ImMunoGeneTics system, and the Immunoglobulin Analysis Tools. RESULTS: Compared to findings in controls, the JIA peripheral blood Treg cell repertoire was restricted, and clonotypic expansions were found in both blood and synovial fluid Treg cells. Skewed usage and pairing of TRB variable and joining genes, including overuse of gene segments that have been associated with other autoimmune conditions, was observed. JIA patients shared a substantial portion of synovial fluid Treg cell clonotypes that were private to JIA and not identified in Lyme arthritis. CONCLUSION: We identified restriction and clonotypic expansions in the JIA Treg cell repertoire with sharing of Treg cell clonotypes across patients. These findings suggest that abnormalities in the Treg cell repertoire, possibly engendered by shared antigenic triggers, may contribute to disease pathogenesis in JIA.


Assuntos
Artrite Juvenil/imunologia , Linfócitos T Reguladores/fisiologia , Adolescente , Artrite Juvenil/sangue , Células Cultivadas , Criança , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Masculino , Líquido Sinovial/citologia , Subpopulações de Linfócitos T/fisiologia
4.
Sci Immunol ; 1(6)2016 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-28783691

RESUMO

Recombination-activating genes 1 and 2 (RAG1 and RAG2) play a critical role in T and B cell development by initiating the recombination process that controls the expression of T cell receptor (TCR) and immunoglobulin genes. Mutations in the RAG1 and RAG2 genes in humans cause a broad spectrum of phenotypes, including severe combined immunodeficiency (SCID) with lack of T and B cells, Omenn syndrome, leaky SCID, and combined immunodeficiency with granulomas or autoimmunity (CID-G/AI). Using next-generation sequencing, we analyzed the TCR and B cell receptor (BCR) repertoire in 12 patients with RAG mutations presenting with Omenn syndrome (n = 5), leaky SCID (n = 3), or CID-G/AI (n = 4). Restriction of repertoire diversity skewed usage of variable (V), diversity (D), and joining (J) segment genes, and abnormalities of CDR3 length distribution were progressively more prominent in patients with a more severe phenotype. Skewed usage of V, D, and J segment genes was present also within unique sequences, indicating a primary restriction of repertoire. Patients with Omenn syndrome had a high proportion of class-switched immunoglobulin heavy chain transcripts and increased somatic hypermutation rate, suggesting in vivo activation of these B cells. These data provide a framework to better understand the phenotypic heterogeneity of RAG deficiency.

5.
J Allergy Clin Immunol ; 136(1): 140-150.e7, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25917813

RESUMO

BACKGROUND: The endonuclease ARTEMIS, which is encoded by the DCLRE1C gene, is a component of the nonhomologous end-joining pathway and participates in hairpin opening during the V(D)J recombination process and repair of a subset of DNA double-strand breaks. Patients with ARTEMIS deficiency usually present with severe combined immunodeficiency (SCID) and cellular radiosensitivity, but hypomorphic mutations can cause milder phenotypes (leaky SCID). OBJECTIVE: We sought to correlate the functional effect of human DCLRE1C mutations on phenotypic presentation in patients with ARTEMIS deficiency. METHODS: We studied the recombination and DNA repair activity of 41 human DCLRE1C mutations in Dclre1c(-/-) v-abl kinase-transformed pro-B cells retrovirally engineered with a construct that allows quantification of recombination activity by means of flow cytometry. For assessment of DNA repair efficacy, resolution of γH2AX accumulation was studied after ionizing radiation. RESULTS: Low or absent activity was detected for mutations causing a typical SCID phenotype. Most of the patients with leaky SCID were compound heterozygous for 1 loss-of-function and 1 hypomorphic allele, with significant residual levels of recombination and DNA repair activity. Deletions disrupting the C-terminus result in truncated but partially functional proteins and are often associated with leaky SCID. Overexpression of hypomorphic mutants might improve the functional defect. CONCLUSIONS: Correlation between the nature and location of DCLRE1C mutations, functional activity, and the clinical phenotype has been observed. Hypomorphic variants that have been reported in the general population can be disease causing if combined in trans with a loss-of-function allele. Therapeutic strategies aimed at inducing overexpression of hypomorphic alleles might be beneficial.


Assuntos
Linfócitos B/fisiologia , Mutação/genética , Proteínas Nucleares/genética , Imunodeficiência Combinada Severa/genética , Adolescente , Adulto , Alelos , Linfócitos B/efeitos da radiação , Linhagem Celular Transformada , Criança , Pré-Escolar , Análise Mutacional de DNA , Reparo do DNA/genética , Proteínas de Ligação a DNA , Endonucleases , Heterozigoto , Histonas/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Proteínas Oncogênicas v-abl/genética , Proteínas Oncogênicas v-abl/metabolismo , Fenótipo , Tolerância a Radiação/genética , Radiação Ionizante , Recombinação V(D)J/genética , Adulto Jovem
7.
J Allergy Clin Immunol ; 134(6): 1375-1380, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24996264

RESUMO

BACKGROUND: Recombination-activating gene 1 (RAG1) deficiency presents with a varied spectrum of combined immunodeficiency, ranging from a T(-)B(-)NK(+) type of disease to a T(+)B(+)NK(+) phenotype. OBJECTIVE: We sought to assess the genetic background of patients with common variable immunodeficiency (CVID). METHODS: A patient given a diagnosis of CVID, who was born to a consanguineous family and thus would be expected to show an autosomal recessive inheritance, was subjected to clinical evaluation, immunologic assays, homozygosity gene mapping, exome sequencing, Sanger sequencing, and functional analysis. RESULTS: The 14-year-old patient, who had liver granuloma, extranodal marginal zone B-cell lymphoma, and autoimmune neutropenia, presented with a clinical picture resembling CVID. Genetic analysis of this patient showed a homozygous hypomorphic RAG1 mutation (c.1073 G>A, p.C358Y) with a residual functional capacity of 48% of wild-type protein. CONCLUSION: Our finding broadens the range of disorders associated with RAG1 mutations and might have important therapeutic implications.


Assuntos
Imunodeficiência de Variável Comum/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Adolescente , Imunodeficiência de Variável Comum/imunologia , Granuloma/genética , Granuloma/imunologia , Humanos , Hepatopatias/genética , Hepatopatias/imunologia , Linfoma de Células B/genética , Linfoma de Células B/imunologia , Masculino , Mutação , Neutropenia/genética , Neutropenia/imunologia , Fenótipo
8.
J Clin Immunol ; 34(5): 551-4, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24817258

RESUMO

PURPOSE: To identify mechanisms of disease in a child born to consanguineous parents, who presented with Omenn syndrome (OS) and was found to carry a heterozygous RAG1 mutation in peripheral blood DNA. METHODS: Mutation analysis was performed on whole blood and buccal swab DNA. Recombination activity of the mutant RAG1 protein and diversity of T cell repertoire were tested. RESULTS: Apparent heterozygosity for a novel, functionally null RAG1 mutation in peripheral blood DNA from a patient with OS was shown to be secondary to true somatic reversion. Analysis of T cell repertoire demonstrated expression of various TCRBV families, but an overall restricted pattern. CONCLUSIONS: This is the first case of true somatic reversion of a RAG1 mutation in a patient with OS. The reversion event likely occurred at a stage where only a limited pool of T cell progenitors capable of performing V(D)J recombination could be generated. This work emphasizes the importance of performing functional studies to investigate the significance of novel genetic variants, and to consider somatic reversion as a possible disease modifier in SCID.


Assuntos
Proteínas de Homeodomínio/genética , Mosaicismo , Imunodeficiência Combinada Severa/genética , Transposases/genética , Linfócitos B/imunologia , Linfócitos B/patologia , Análise Mutacional de DNA , Genótipo , Transplante de Células-Tronco Hematopoéticas , Heterozigoto , Proteínas de Homeodomínio/sangue , Humanos , Lactente , Masculino , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/patologia , Imunodeficiência Combinada Severa/terapia , Linfócitos T/imunologia , Linfócitos T/patologia , Transposases/sangue
9.
J Allergy Clin Immunol ; 133(4): 1099-108, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24290284

RESUMO

BACKGROUND: The recombination-activating gene (RAG) 1/2 proteins play a critical role in the development of T and B cells by initiating the VDJ recombination process that leads to generation of a broad T-cell receptor (TCR) and B-cell receptor repertoire. Pathogenic mutations in the RAG1/2 genes result in various forms of primary immunodeficiency, ranging from T(-)B(-) severe combined immune deficiency to delayed-onset disease with granuloma formation, autoimmunity, or both. It is not clear what contributes to such heterogeneity of phenotypes. OBJECTIVE: We sought to investigate the molecular basis for phenotypic diversity presented in patients with various RAG1 mutations. METHODS: We have developed a flow cytometry-based assay that allows analysis of RAG recombination activity based on green fluorescent protein expression and have assessed the induction of the Ighc locus rearrangements in mouse Rag1(-/-) pro-B cells reconstituted with wild-type or mutant human RAG1 (hRAG1) using deep sequencing technology. RESULTS: Here we demonstrate correlation between defective recombination activity of hRAG1 mutant proteins and severity of the clinical and immunologic phenotype and provide insights on the molecular mechanisms accounting for such phenotypic diversity. CONCLUSIONS: Using a sensitive assay to measure the RAG1 activity level of 79 mutations in a physiologic setting, we demonstrate correlation between recombination activity of RAG1 mutants and the severity of clinical presentation and show that RAG1 mutants can induce specific abnormalities of the VDJ recombination process.


Assuntos
Estudos de Associação Genética , Proteínas de Homeodomínio/genética , Imunodeficiência Combinada Severa/genética , Recombinação V(D)J , Alelos , Linfócitos B/imunologia , Linfócitos B/metabolismo , Criança , Pré-Escolar , Ordem dos Genes , Rearranjo Gênico , Proteínas de Homeodomínio/metabolismo , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Lactente , Recém-Nascido , Mutação , Fenótipo , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
11.
Clin Immunol ; 146(2): 84-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23280491

RESUMO

X-linked lymphoproliferative (XLP) disease is a primary immunodeficiency syndrome associated with the inability to control Epstein-Barr virus (EBV), lymphoma, and hypogammaglobulinemia. XLP is caused by mutations in the SH2D1A gene, which encodes the SLAM-associated protein (SAP), or in the BIRC4 gene, which encodes the X-linked inhibitor of apoptosis protein (XIAP). Here we report a patient with recurrent respiratory tract infections and early onset agammaglobulinemia who carried a unique disease-causing intronic loss-of-function mutation in SH2D1A. The intronic mutation affected SH2D1A gene transcription but not mRNA splicing, and led to markedly reduced level of SAP protein. Despite undetectable serum immunoglobulins, the patient's B cells replicated and differentiated into antibody producing cells normally in vitro.


Assuntos
Agamaglobulinemia/genética , Agamaglobulinemia/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Íntrons/genética , Agamaglobulinemia/tratamento farmacológico , Animais , Pré-Escolar , Feminino , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Lactente , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Íntrons/imunologia , Masculino , Camundongos , Proteína Associada à Molécula de Sinalização da Ativação Linfocitária , Domínios de Homologia de src/genética , Domínios de Homologia de src/imunologia
13.
J Clin Immunol ; 33(3): 513-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23143406

RESUMO

Major Histocompatibility Complex (MHC) class II deficiency is a combined primary immunodeficiency disease that leads to overwhelming and recurrent infections. It was found to account for 19 % of combined immune deficiency cases in the National Primary Immunodeficiency Disorders Registry in Kuwait, a country with high prevalence of consanguinity. We present the clinical, immunologic and molecular features of 11 Kuwaiti patients who presented with MHC class II deficiency between 2004 and 2011.


Assuntos
Antígenos de Histocompatibilidade Classe II/genética , Imunodeficiência Combinada Severa/epidemiologia , Imunodeficiência Combinada Severa/genética , Adolescente , Adulto , Criança , Pré-Escolar , Consanguinidade , Proteínas de Ligação a DNA , Feminino , Ordem dos Genes , Transplante de Células-Tronco Hematopoéticas , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Imunoglobulinas Intravenosas , Kuweit/epidemiologia , Masculino , Pessoa de Meia-Idade , Mutação , Sistema de Registros , Imunodeficiência Combinada Severa/diagnóstico , Imunodeficiência Combinada Severa/terapia , Fatores de Transcrição/genética , Resultado do Tratamento , Adulto Jovem
15.
Blood ; 119(12): 2819-28, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22302739

RESUMO

Wiskott Aldrich syndrome (WAS) is caused by mutations in the WAS gene that encodes for a protein (WASp) involved in cytoskeleton organization in hematopoietic cells. Several distinctive abnormalities of T, B, and natural killer lymphocytes; dendritic cells; and phagocytes have been found in WASp-deficient patients and mice; however, the in vivo consequence of WASp deficiency within individual blood cell lineages has not been definitively evaluated. By conditional gene deletion we have generated mice with selective deficiency of WASp in the B-cell lineage (B/WcKO mice). We show that this is sufficient to cause a severe reduction of marginal zone B cells and inability to respond to type II T-independent Ags, thereby recapitulating phenotypic features of complete WASp deficiency. In addition, B/WcKO mice showed prominent signs of B-cell dysregulation, as indicated by an increase in serum IgM levels, expansion of germinal center B cells and plasma cells, and elevated autoantibody production. These findings are accompanied by hyperproliferation of WASp-deficient follicular and germinal center B cells in heterozygous B/WcKO mice in vivo and excessive differentiation of WASp-deficient B cells into class-switched plasmablasts in vitro, suggesting that WASp-dependent B cell-intrinsic mechanisms critically contribute to WAS-associated autoimmunity.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Proteína da Síndrome de Wiskott-Aldrich/imunologia , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Autoantígenos/imunologia , Contagem de Células , Modelos Animais de Doenças , Citometria de Fluxo , Imunofluorescência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína da Síndrome de Wiskott-Aldrich/deficiência , Proteína da Síndrome de Wiskott-Aldrich/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...