Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 159
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 49(16): 9310-9326, 2021 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-34387696

RESUMO

Artemis (SNM1C/DCLRE1C) is an endonuclease that plays a key role in development of B- and T-lymphocytes and in dsDNA break repair by non-homologous end-joining (NHEJ). Artemis is phosphorylated by DNA-PKcs and acts to open DNA hairpin intermediates generated during V(D)J and class-switch recombination. Artemis deficiency leads to congenital radiosensitive severe acquired immune deficiency (RS-SCID). Artemis belongs to a superfamily of nucleases containing metallo-ß-lactamase (MBL) and ß-CASP (CPSF-Artemis-SNM1-Pso2) domains. We present crystal structures of the catalytic domain of wildtype and variant forms of Artemis, including one causing RS-SCID Omenn syndrome. The catalytic domain of the Artemis has similar endonuclease activity to the phosphorylated full-length protein. Our structures help explain the predominantly endonucleolytic activity of Artemis, which contrasts with the predominantly exonuclease activity of the closely related SNM1A and SNM1B MBL fold nucleases. The structures reveal a second metal binding site in its ß-CASP domain unique to Artemis, which is amenable to inhibition by compounds including ebselen. By combining our structural data with that from a recently reported Artemis structure, we were able model the interaction of Artemis with DNA substrates. The structures, including one of Artemis with the cephalosporin ceftriaxone, will help enable the rational development of selective SNM1 nuclease inhibitors.


Assuntos
Proteínas de Ciclo Celular/ultraestrutura , Proteínas de Ligação a DNA/ultraestrutura , Endonucleases/ultraestrutura , Exodesoxirribonucleases/ultraestrutura , Imunodeficiência Combinada Severa/genética , Linfócitos B/enzimologia , Domínio Catalítico/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cristalografia por Raios X , Reparo do DNA por Junção de Extremidades/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Endonucleases/antagonistas & inibidores , Endonucleases/química , Endonucleases/genética , Inibidores Enzimáticos/química , Exodesoxirribonucleases/química , Exodesoxirribonucleases/genética , Humanos , Fosforilação/genética , Dobramento de Proteína , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/patologia , Linfócitos T/enzimologia
2.
Eur J Immunol ; 51(1): 206-219, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32707604

RESUMO

Adenosine deaminase 2 deficiency (DADA2) is an autoinflammatory disease characterized by inflammatory vasculopathy, early strokes associated often with hypogammaglobulinemia. Pure red cell aplasia, thrombocytopenia, and neutropenia have been reported. The defect is due to biallelic loss of function of ADA2 gene, coding for a protein known to regulate the catabolism of extracellular adenosine. We therefore investigated immune phenotype and B- and T-cell responses in 14 DADA2 patients to address if ADA2 mutation affects B- and T-cell function. Here, we show a significant decrease in memory B cells, in particular class switch memory, and an expansion of CD21low B cells in DADA2 patients. In vitro stimulated B lymphocytes were able to secrete nonfunctional ADA2 protein, suggesting a cell intrinsic defect resulting in an impairment of B-cell proliferation and differentiation. Moreover, CD4+ and CD8+ T cells were diminished; however, the frequency of circulating T follicular helper cells was significantly increased but they had an impairment in IL-21 production possibly contributing to an impaired B cell help. Our findings suggest that ADA2 mutation could lead to a B-cell intrinsic defect but also to a defective Tfh cell function, which could contribute to the immunodeficient phenotype reported in DADA2 patients.


Assuntos
Adenosina Desaminase/deficiência , Agamaglobulinemia/imunologia , Linfócitos B/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Imunodeficiência Combinada Severa/imunologia , Células T Auxiliares Foliculares/imunologia , Adenosina Desaminase/genética , Adenosina Desaminase/imunologia , Adolescente , Adulto , Agamaglobulinemia/enzimologia , Agamaglobulinemia/genética , Linfócitos B/enzimologia , Linfócitos B/patologia , Estudos de Casos e Controles , Diferenciação Celular , Proliferação de Células , Criança , Pré-Escolar , Feminino , Humanos , Memória Imunológica , Imunofenotipagem , Técnicas In Vitro , Lactente , Recém-Nascido , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interleucinas/biossíntese , Ativação Linfocitária , Masculino , Mutação , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Células T Auxiliares Foliculares/patologia
3.
J Clin Immunol ; 41(7): 1633-1647, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34324127

RESUMO

PURPOSE: Deficiency of adenosine deaminase 2 (DADA2) is an inherited inborn error of immunity, characterized by autoinflammation (recurrent fever), vasculopathy (livedo racemosa, polyarteritis nodosa, lacunar ischemic strokes, and intracranial hemorrhages), immunodeficiency, lymphoproliferation, immune cytopenias, and bone marrow failure (BMF). Tumor necrosis factor (TNF-α) blockade is the treatment of choice for the vasculopathy, but often fails to reverse refractory cytopenia. We aimed to study the outcome of hematopoietic cell transplantation (HCT) in patients with DADA2. METHODS: We conducted a retrospective study on the outcome of HCT in patients with DADA2. The primary outcome was overall survival (OS). RESULTS: Thirty DADA2 patients from 12 countries received a total of 38 HCTs. The indications for HCT were BMF, immune cytopenia, malignancy, or immunodeficiency. Median age at HCT was 9 years (range: 2-28 years). The conditioning regimens for the final transplants were myeloablative (n = 20), reduced intensity (n = 8), or non-myeloablative (n = 2). Donors were HLA-matched related (n = 4), HLA-matched unrelated (n = 16), HLA-haploidentical (n = 2), or HLA-mismatched unrelated (n = 8). After a median follow-up of 2 years (range: 0.5-16 years), 2-year OS was 97%, and 2-year GvHD-free relapse-free survival was 73%. The hematological and immunological phenotypes resolved, and there were no new vascular events. Plasma ADA2 enzyme activity normalized in 16/17 patients tested. Six patients required more than one HCT. CONCLUSION: HCT was an effective treatment for DADA2, successfully reversing the refractory cytopenia, as well as the vasculopathy and immunodeficiency. CLINICAL IMPLICATIONS: HCT is a definitive cure for DADA2 with > 95% survival.


Assuntos
Agamaglobulinemia/terapia , Transtornos da Insuficiência da Medula Óssea/terapia , Transplante de Células-Tronco Hematopoéticas , Imunodeficiência Combinada Severa/terapia , Adenosina Desaminase/deficiência , Adolescente , Adulto , Agamaglobulinemia/enzimologia , Agamaglobulinemia/genética , Agamaglobulinemia/mortalidade , Transtornos da Insuficiência da Medula Óssea/enzimologia , Transtornos da Insuficiência da Medula Óssea/genética , Transtornos da Insuficiência da Medula Óssea/mortalidade , Criança , Pré-Escolar , Feminino , Doença Enxerto-Hospedeiro/etiologia , Doença Enxerto-Hospedeiro/mortalidade , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Estimativa de Kaplan-Meier , Masculino , Estudos Retrospectivos , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/mortalidade , Resultado do Tratamento , Adulto Jovem
4.
Blood ; 129(21): 2928-2938, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28331055

RESUMO

Reticular dysgenesis (RD) is a rare congenital disorder defined clinically by the combination of severe combined immunodeficiency (SCID), agranulocytosis, and sensorineural deafness. Mutations in the gene encoding adenylate kinase 2 were identified to cause the disorder. Hematopoietic stem cell transplantation (HSCT) is the only option to cure this otherwise fatal disease. Retrospective data on clinical presentation, genetics, and outcome of HSCT were collected from centers in Europe, Asia, and North America for a total of 32 patients born between 1982 and 2011. Age at presentation was <4 weeks in 30 of 32 patients (94%). Grafts originated from mismatched family donors in 17 patients (55%), from matched family donors in 6 patients (19%), and from unrelated marrow or umbilical cord blood donors in 8 patients (26%). Thirteen patients received secondary or tertiary transplants. After transplantation, 21 of 31 patients were reported alive at a mean follow-up of 7.9 years (range: 0.6-23.6 years). All patients who died beyond 6 months after HSCT had persistent or recurrent agranulocytosis due to failure of donor myeloid engraftment. In the absence of conditioning, HSCT was ineffective to overcome agranulocytosis, and inclusion of myeloablative components in the conditioning regimens was required to achieve stable lymphomyeloid engraftment. In comparison with other SCID entities, considerable differences were noted regarding age at presentation, onset, and type of infectious complications, as well as the requirement of conditioning prior to HSCT. Although long-term survival is possible in the presence of mixed chimerism, high-level donor myeloid engraftment should be targeted to avoid posttransplant neutropenia.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Transplante de Células-Tronco Hematopoéticas , Leucopenia/mortalidade , Leucopenia/terapia , Imunodeficiência Combinada Severa/mortalidade , Imunodeficiência Combinada Severa/terapia , Condicionamento Pré-Transplante , Doadores não Relacionados , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Adolescente , Adulto , Idade de Início , Aloenxertos , Criança , Intervalo Livre de Doença , Feminino , Humanos , Leucopenia/enzimologia , Leucopenia/genética , Masculino , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Taxa de Sobrevida
5.
Blood ; 125(10): 1643-52, 2015 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-25587035

RESUMO

Erythrocyte possesses high sphingosine kinase 1 (SphK1) activity and is the major cell type supplying plasma sphingosine-1-phosphate, a signaling lipid regulating multiple physiological and pathological functions. Recent studies revealed that erythrocyte SphK1 activity is upregulated in sickle cell disease (SCD) and contributes to sickling and disease progression. However, how erythrocyte SphK1 activity is regulated remains unknown. Here we report that adenosine induces SphK1 activity in human and mouse sickle and normal erythrocytes in vitro. Next, using 4 adenosine receptor-deficient mice and pharmacological approaches, we determined that the A2B adenosine receptor (ADORA2B) is essential for adenosine-induced SphK1 activity in human and mouse normal and sickle erythrocytes in vitro. Subsequently, we provide in vivo genetic evidence that adenosine deaminase (ADA) deficiency leads to excess plasma adenosine and elevated erythrocyte SphK1 activity. Lowering adenosine by ADA enzyme therapy or genetic deletion of ADORA2B significantly reduced excess adenosine-induced erythrocyte SphK1 activity in ADA-deficient mice. Finally, we revealed that protein kinase A-mediated extracellular signal-regulated kinase 1/2 activation functioning downstream of ADORA2B underlies adenosine-induced erythrocyte SphK1 activity. Overall, our findings reveal a novel signaling network regulating erythrocyte SphK1 and highlight innovative mechanisms regulating SphK1 activity in normal and SCD.


Assuntos
Adenosina/sangue , Anemia Falciforme/sangue , Anemia Falciforme/enzimologia , Eritrócitos Anormais/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/sangue , Receptor A2B de Adenosina/sangue , Adenosina Desaminase/sangue , Adenosina Desaminase/deficiência , Adenosina Desaminase/genética , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Agamaglobulinemia/sangue , Agamaglobulinemia/enzimologia , Agamaglobulinemia/genética , Anemia Falciforme/genética , Animais , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/sangue , Eritrócitos/efeitos dos fármacos , Eritrócitos/enzimologia , Eritrócitos/metabolismo , Eritrócitos Anormais/efeitos dos fármacos , Eritrócitos Anormais/enzimologia , Hemoglobina Falciforme/genética , Hemoglobina Falciforme/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Biológicos , Receptor A2B de Adenosina/deficiência , Receptor A2B de Adenosina/genética , Imunodeficiência Combinada Severa/sangue , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Transdução de Sinais
6.
Pediatr Int ; 58(10): 1076-1080, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27593409

RESUMO

Severe combined immunodeficiency (SCID) is the most severe form of primary immunodeficiency disease, and it is characterized by marked impairment in cellular and humoral immunity. Mutations in several genes cause SCID, one of which is Janus kinase 3 (JAK3), resulting in autosomal recessive T(-)B(+)NK(-) SCID. Only three patients with JAK3-deficient SCID have been reported in Japan. We herein describe the case of a 6-month-old girl with pneumocystis pneumonia, who was diagnosed with SCID with compound heterozygous JAK3 mutations (c.1568G>A + c.421-10G>A). One of the mutations was previously reported in another Japanese patient. The other mutation was a novel and de novo relatively deep intronic mutation causing aberrant RNA splicing. The patient was successfully treated with bone marrow transplantation from a haploidentical donor.


Assuntos
DNA/genética , Janus Quinase 3/genética , Mutação , Imunodeficiência Combinada Severa/genética , Análise Mutacional de DNA , Feminino , Heterozigoto , Humanos , Lactente , Janus Quinase 3/metabolismo , Japão , Imunodeficiência Combinada Severa/enzimologia
7.
Immunology ; 141(3): 377-87, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24164480

RESUMO

Loss of ζ-associated protein 70 (Zap70) results in severe immunodeficiency in humans and mice because of the critical role of Zap70 in T-cell receptor (TCR) signalling. Here we describe a novel mouse strain generated by N-ethyl-N-nitrosourea mutagenesis, with the reduced protein stability (rps) mutation in Zap70. The A243V rps mutation resulted in decreased Zap70 protein and a reduced duration of TCR-induced calcium responses, equivalent to that induced by a 50% decrease in catalytically active Zap70. The reduction of signalling through Zap70 was insufficient to substantially perturb thymic differentiation of conventional CD4 and CD8 T cells, although Foxp3(+) regulatory T cells demonstrated altered thymic production and peripheral homeostasis. Despite the mild phenotype, the Zap70(A243V) variant lies just above the functional threshold for TCR signalling competence, as T cells relying on only a single copy of the Zap70(rps) allele for TCR signalling demonstrated no intracellular calcium response to TCR stimulation. This addition to the Zap70 allelic series indicates that a rate-limiting threshold for Zap70 protein levels exists at which signalling capacity switches from nearly intact to effectively null.


Assuntos
Sinalização do Cálcio , Mutação , Receptores de Antígenos de Linfócitos T/metabolismo , Imunodeficiência Combinada Severa/enzimologia , Subpopulações de Linfócitos T/enzimologia , Proteína-Tirosina Quinase ZAP-70/deficiência , Sequência de Aminoácidos , Animais , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/metabolismo , Predisposição Genética para Doença , Heterozigoto , Cinética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Dados de Sequência Molecular , Fenótipo , Estabilidade Proteica , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Baço/enzimologia , Baço/imunologia , Subpopulações de Linfócitos T/imunologia , Timócitos/enzimologia , Timócitos/imunologia , Proteína-Tirosina Quinase ZAP-70/genética , Proteína-Tirosina Quinase ZAP-70/imunologia , Proteína-Tirosina Quinase ZAP-70/metabolismo
10.
J Biol Chem ; 286(6): 4081-9, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-20876536

RESUMO

The unfolded protein response (UPR) is a homeostatic signaling mechanism that balances the protein folding capacity of the endoplasmic reticulum (ER) with the secretory protein load of the cell. ER protein folding capacity is dependent on the abundance of chaperones, which is increased in response to UPR signaling, and on a sufficient ATP supply for their activity. An essential branch of the UPR entails the splicing of XBP1 mRNA to form the XBP1 transcription factor. XBP1 has been shown to be required during adipocyte differentiation, enabling mature adipocytes to secrete adiponectin, and during differentiation of B cells into antibody-secreting plasma cells. Here we find that adenylate kinase 2 (AK2), a mitochondrial enzyme that regulates adenine nucleotide interconversion within the intermembrane space, is markedly induced during adipocyte and B cell differentiation. Depletion of AK2 by RNAi impairs adiponectin secretion in 3T3-L1 adipocytes, IgM secretion in BCL1 cells, and the induction of the UPR during differentiation of both cell types. These results reveal a new mechanism by which mitochondria support ER function and suggest that specific mitochondrial defects may give rise to impaired UPR signaling. The requirement for AK2 for UPR induction may explain the pathogenesis of the profound hematopoietic defects of reticular dysgenesis, a disease associated with mutations of the AK2 gene in humans.


Assuntos
Trifosfato de Adenosina/metabolismo , Adenilato Quinase/metabolismo , Metabolismo Energético/fisiologia , Mitocôndrias/enzimologia , Proteínas Mitocondriais/metabolismo , Plasmócitos/metabolismo , Resposta a Proteínas não Dobradas/fisiologia , Células 3T3-L1 , Trifosfato de Adenosina/genética , Adenilato Quinase/genética , Adiponectina/genética , Adiponectina/metabolismo , Animais , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Hematopoese/genética , Humanos , Leucopenia/enzimologia , Leucopenia/genética , Camundongos , Mitocôndrias/genética , Proteínas Mitocondriais/genética , Mutação , Splicing de RNA/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Fatores de Transcrição de Fator Regulador X , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Proteína 1 de Ligação a X-Box
12.
Invest Clin ; 53(3): 315-24, 2012 Sep.
Artigo em Espanhol | MEDLINE | ID: mdl-23248974

RESUMO

Adenosine deaminase is an enzyme of the purine metabolism whose function is to convert adenosine to inosine and deoxyadenosine to deoxyinosine. The ecto-ADA1 binding to the cell surface through CD26 contributes to the regulation of cytokines and stimulates the proliferation of T cells by activating CD45. The deficiency of this enzyme generates the severe combined immunodeficiency syndrome, characterized by the accumulation of deoxyadenosine and adenine metabolites, which have toxic effects on lymphocytes, affecting DNA synthesis and consequently, clonal expansion. Early diagnosis of this immunodeficiency is essential, as it significantly reduces morbidity and mortality associated with recurrent infections. Recent advances in molecular biology and genetics have led to the identification of genetic defects of many primary immunodeficiencies and the development of promising diagnostic tools and treatment.


Assuntos
Adenosina Desaminase/deficiência , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/etiologia , Humanos
13.
Blood ; 114(17): 3524-32, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19638621

RESUMO

Adenosine deaminase deficiency is a disorder of purine metabolism leading to severe combined immunodeficiency (ADA-SCID). Without treatment, the condition is fatal and requires early intervention. Haematopoietic stem cell transplantation is the major treatment for ADA-SCID, although survival following different donor sources varies considerably. Unlike other SCID forms, 2 other options are available for ADA-SCID: enzyme replacement therapy (ERT) with pegylated bovine ADA, and autologous haematopoietic stem cell gene therapy (GT). Due to the rarity of the condition, the lack of large scale outcome studies, and availability of different treatments, guidance on treatment strategies is limited. We have reviewed the currently available evidence and together with our experience of managing this condition propose a consensus management strategy. Matched sibling donor transplants represent a successful treatment option with high survival rates and excellent immune recovery. Mismatched parental donor transplants have a poor survival outcome and should be avoided unless other treatments are unavailable. ERT and GT both show excellent survival, and therefore the choice between ERT, MUD transplant, or GT is difficult and dependent on several factors, including accessibility to the different modalities, response of patients to long-term ERT, and the attitudes of physicians and parents to the short- and potential long-term risks associated with different treatments.


Assuntos
Adenosina Desaminase/deficiência , Transplante de Células-Tronco Hematopoéticas , Imunodeficiência Combinada Severa/terapia , Animais , Bovinos , Humanos , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/etiologia
14.
Blood ; 114(17): 3546-56, 2009 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-19652199

RESUMO

Gene transfer into hematopoietic stem cells by gamma-retroviral vectors (RVs) is an effective treatment for inherited blood disorders, although potentially limited by the risk of insertional mutagenesis. We evaluated the genomic impact of RV integration in T lymphocytes from adenosine deaminase-deficient severe combined immunodeficiency (ADA-SCID) patients 10 to 30 months after infusion of autologous, genetically corrected CD34(+) cells. Expression profiling on ex vivo T-cell bulk population revealed no difference with respect to healthy controls. To assess the effect of vector integration on gene expression at the single-cell level, primary T-cell clones were isolated from 2 patients. T-cell clones harbored either 1 (89.8%) or 2 (10.2%) vector copies per cell and displayed partial to full correction of ADA expression, purine metabolism, and T-cell receptor-driven functions. Analysis of RV integration sites indicated a high diversity in T-cell origin, consistently with the polyclonal T-cell receptor-Vbeta repertoire. Quantitative transcript analysis of 120 genes within a 200-kb window around RV integration sites showed modest (2.8- to 5.2-fold) dysregulation of 5.8% genes in 18.6% of the T-cell clones compared with controls. Nonetheless, affected clones maintained a stable phenotype and normal in vitro functions. These results confirm that RV-mediated gene transfer for ADA-SCID is safe, and provide crucial information for the development of future gene therapy protocols. The trials described herein have been registered at http://www.clinicaltrials.gov as #NCT00598481 and #NCT00599781.


Assuntos
Adenosina Desaminase/genética , Terapia Genética , Vetores Genéticos/uso terapêutico , Retroviridae/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos T/patologia , Adenosina Desaminase/deficiência , Antígenos CD34 , Biomarcadores Tumorais/genética , Células Cultivadas , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Purinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Imunodeficiência Combinada Severa/enzimologia , Imunodeficiência Combinada Severa/genética , Linfócitos T/metabolismo , Transdução Genética , Integração Viral
15.
Nat Med ; 4(1): 58-64, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9427607

RESUMO

Janus kinase-3 (JAK3) deficiency has recently been identified as a cause of severe combined immunodeficiency (SCID) in humans. We used a mouse model of Jak3-deficient SCID to test a gene therapy approach for treatment of this disease. Transfer of a Jak3 retroviral vector to repopulating hematopoietic stem cells resulted in increased numbers of T and B lymphocytes, reversal of hypogammaglobulinemia, restoration of T-cell activation upon stimulation with mitogens, and development of an antigen-specific immune response after immunization. Analysis for vector copy number in lymphoid and myeloid populations showed a large in vivo selective advantage for Jak3-expressing lymphoid cells. These results show that gene replacement is a feasible treatment strategy for this disease and that naturally occurring in vivo selection of corrected cells is an important advantage of this approach.


Assuntos
Linfócitos B/imunologia , Terapia Genética/métodos , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Imunodeficiência Combinada Severa/imunologia , Imunodeficiência Combinada Severa/terapia , Linfócitos T/imunologia , Agamaglobulinemia/etiologia , Agamaglobulinemia/terapia , Animais , Formação de Anticorpos , Células da Medula Óssea/citologia , Transplante de Medula Óssea/métodos , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Janus Quinase 3 , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Reação em Cadeia da Polimerase , Proteínas Tirosina Quinases/biossíntese , Retroviridae , Imunodeficiência Combinada Severa/enzimologia , Baço/imunologia
16.
J Leukoc Biol ; 110(3): 409-424, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33988272

RESUMO

Deficiency of adenosine deaminase 2 (DADA2) is a rare autosomal recessive disease caused by loss-of-function variants in the ADA2 gene. DADA2 typically presents in childhood and is characterized by vasculopathy, stroke, inflammation, immunodeficiency, as well as hematologic manifestations. ADA2 protein is predominantly present in stimulated monocytes, dendritic cells, and macrophages. To elucidate molecular mechanisms in DADA2, CD14+ monocytes from 14 patients and 6 healthy donors were analyzed using single-cell RNA sequencing (scRNA-seq). Monocytes were purified by positive selection based on CD14 expression. Subpopulations were imputed from their transcriptomes. Based on scRNA-seq, monocytes could be classified as classical, intermediate, and nonclassical. Further, we used gene pathway analytics to interpret patterns of up- and down-regulated gene transcription. In DADA2, the frequency of nonclassical monocytes was higher compared with that of healthy donors, and M1 macrophage markers were up-regulated in patients. By comparing gene expression of each monocyte subtype between patients and healthy donors, we identified upregulated immune response pathways, including IFNα/ß and IFNγ signaling, in all monocyte subtypes. Distinctively, the TNFR2 noncanonical NF-κB pathway was up-regulated only in nonclassical monocytes. Patients' plasma showed increased IFNγ and TNFα levels. Our results suggest that elevated IFNγ activates cell signaling, leading to differentiation into M1 macrophages from monocytes and release of TNFα. Immune responses and more general response to stimuli pathways were up-regulated in DADA2 monocytes, and protein synthesis pathways were down-regulated, perhaps as stress responses. Our identification of novel aberrant immune pathways has implications for therapeutic approaches in DADA2 (registered at clinicaltrials.gov NCT00071045).


Assuntos
Agamaglobulinemia/genética , Agamaglobulinemia/patologia , Monócitos/patologia , Análise de Sequência de RNA , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/patologia , Análise de Célula Única , Adenosina Desaminase/genética , Adolescente , Adulto , Agamaglobulinemia/sangue , Agamaglobulinemia/enzimologia , Criança , Pré-Escolar , Citocinas/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Interferons/metabolismo , Masculino , Pessoa de Meia-Idade , Mutação/genética , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Imunodeficiência Combinada Severa/sangue , Imunodeficiência Combinada Severa/enzimologia , Transdução de Sinais , Doadores de Tecidos , Adulto Jovem
17.
DNA Repair (Amst) ; 8(2): 202-8, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19022407

RESUMO

Artemis is a key factor of the nonhomologous end-joining (NHEJ) pathway, which is critical for DNA double-strand break (DSB) repair in eukaryotic cells. It belongs to the beta-CASP family of nucleases, forming a distinct group within the metallo-beta-lactamase superfamily. Proteins of this group are specific for nucleic acids and contain an original domain, the beta-CASP domain, which serves as a cap covering the active site displayed by the metallo-beta-lactamase domain.Here, we have identified in the highly divergent sequences of the beta-CASP domains from DNA-specific nucleases two conserved residues (Artemis E213 and H254), which are not present in RNA-specific enzymes, and shown that H254 plays a key role in the Artemis function, as it is critical for its full activity in vitro. Moreover, inherited mutation of H254 results in radiosensitive severe combined immune deficiency (RS-SCID) in humans. This residue might play a key role in specificity towards DNA, if not directly in zinc binding.


Assuntos
Histidina/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Sequência de Bases , Sequência Conservada , DNA/metabolismo , Análise Mutacional de DNA , Proteínas de Ligação a DNA , Endonucleases , Células HeLa , Humanos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Proteínas Nucleares/genética , Estrutura Terciária de Proteína , Imunodeficiência Combinada Severa/enzimologia , Relação Estrutura-Atividade
18.
J Exp Med ; 192(9): 1223-36, 2000 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-11067872

RESUMO

Human, but not murine, adenosine deaminase (ADA) forms a complex with the cell membrane protein CD26/dipeptidyl peptidase IV. CD26-bound ADA has been postulated to regulate extracellular adenosine levels and to modulate the costimulatory function of CD26 on T lymphocytes. Absence of ADA-CD26 binding has been implicated in causing severe combined immunodeficiency due to ADA deficiency. Using human-mouse ADA hybrids and ADA point mutants, we have localized the amino acids critical for CD26 binding to the helical segment 126-143. Arg142 in human ADA and Gln142 in mouse ADA largely determine the capacity to bind CD26. Recombinant human ADA bearing the R142Q mutation had normal catalytic activity per molecule, but markedly impaired binding to a CD26(+) ADA-deficient human T cell line. Reduced CD26 binding was also found with ADA from red cells and T cells of a healthy individual whose only expressed ADA has the R142Q mutation. Conversely, ADA with the E217K active site mutation, the only ADA expressed by a severely immunodeficient patient, showed normal CD26 binding. These findings argue that ADA binding to CD26 is not essential for immune function in humans.


Assuntos
Adenosina Desaminase/metabolismo , Substituição de Aminoácidos/genética , Dipeptidil Peptidase 4/metabolismo , Mutação Puntual/genética , Imunodeficiência Combinada Severa/genética , Adenosina Desaminase/química , Adenosina Desaminase/genética , Adenosina Desaminase/imunologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Bovinos , Células Cultivadas , Citometria de Fluxo , Deleção de Genes , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Ligação Proteica , Coelhos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/metabolismo , Imunodeficiência Combinada Severa/enzimologia , Linfócitos T/enzimologia , Linfócitos T/metabolismo
19.
J Exp Med ; 183(6): 2687-92, 1996 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8676091

RESUMO

Mutations affecting the expression of the Janus family kinase JAK3 were recently shown to be responsible for autosomal recessive severe combined immunodeficiency (SCID). JAK3-deficient patients present with a clinical phenotype virtually indistinguishable from boys affected by X-linked SCID, a disease caused by genetic defects of the common gamma chain (gamma c) that is a shared component of the receptors for IL-2, IL-4, IL-7, IL-9, and IL-15. The specific interaction of JAK3 and gamma c represents the biochemical basis for the similarities between these two immunodeficiencies. Both forms of SCID are characterized by recurrent, severe infections leading to death in infancy unless successfully treated by allogeneic bone marrow transplantation. Because of the potentially lethal complications associated with allogeneic bone marrow transplantation and the frequent lack of suitable marrow donors, the development of alternative forms of therapy is highly desirable. To this end, we investigated a retroviral-mediated gene correction approach for JAK3-deficiency. A vector carrying a copy of JAK3 cDNA was constructed and used to transduce B cell lines derived from patients with JAK3-deficient SCID. We demonstrate restoration of JAK3 expression and phosphorylation upon IL-2 and IL-4 stimulation. Furthermore, patients' cells transduced with JAK3 acquired the ability to proliferate normally in response to IL-2. These data indicate that the biological defects of JAK3-deficient cells can be efficiently corrected in vitro by retroviral-mediated gene transfer, thus providing the basis for future investigation of gene therapy as treatment for JAK3-deficient SCID.


Assuntos
Terapia Genética , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Receptores de Interleucina/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos B/imunologia , Western Blotting , Células Cultivadas , Criança , Clonagem Molecular , Consanguinidade , Genes Recessivos , Vetores Genéticos , Humanos , Interleucina-2/farmacologia , Janus Quinase 3 , Ativação Linfocitária/efeitos dos fármacos , Substâncias Macromoleculares , Masculino , Proteínas Tirosina Quinases/biossíntese , Proteínas Recombinantes/biossíntese , Retroviridae , Imunodeficiência Combinada Severa/enzimologia , Transfecção , Cromossomo X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA