Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Med Genet ; 59(4): 346-350, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-33782093

RESUMEN

DICER1 syndrome is a rare paediatric autosomal dominant inherited disorder predisposing to various benign and malignant tumours. It is caused by a germline pathogenic variant in DICER1, and the second hit for tumour development is usually a missense hotspot pathogenic variant in the DICER1 ribonuclease IIIb domain. While DICER1 predisposing variants account for about 60% of ovarian Sertoli-Leydig cell tumours, no DICER1-related testicular stromal tumours have been described. Here we report the first two cases of testicular stromal tumours in children carrying a DICER1 germline pathogenic variant: a case of Sertoli cell tumour and a case of Leydig cell tumour diagnosed at 2 and 12 years of age, respectively. A somatic DICER1 hotspot pathogenic variant was detected in the Sertoli cell tumour. This report extends the spectrum of DICER1-related tumours to include testicular Sertoli cell tumour and potentially testicular Leydig cell tumour. Diagnosis of a testicular Sertoli cell tumour should prompt DICER1 genetic testing so that patients with a DICER1 germline pathogenic variant can benefit from established surveillance guidelines. DICER1 genetic evaluation may be considered for testicular Leydig cell tumour. Our findings suggest that miRNA dysregulation underlies the aetiology of some testicular stromal tumours.


Asunto(s)
Tumor de Células de Leydig , Síndromes Neoplásicos Hereditarios , Neoplasias Ováricas , Tumor de Células de Sertoli , Tumor de Células de Sertoli-Leydig , Neoplasias Testiculares , Niño , ARN Helicasas DEAD-box/genética , Femenino , Humanos , Tumor de Células de Leydig/diagnóstico , Tumor de Células de Leydig/genética , Masculino , Neoplasias Ováricas/genética , Ribonucleasa III/genética , Tumor de Células de Sertoli/genética , Tumor de Células de Sertoli-Leydig/genética , Tumor de Células de Sertoli-Leydig/patología , Neoplasias Testiculares/genética
2.
Clin Chem ; 68(2): 322-331, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34928333

RESUMEN

BACKGROUND: DICER1 syndrome is an autosomal dominant inherited syndrome predisposing to various benign and malignant tumors, mainly occurring in children and young adults, requiring broad surveillance starting at birth with repeated irradiating imaging exams and sedations for young patients. It is caused by monoallelic germline pathogenic variants in the DICER1 gene. More than 90% of tumors bear an additional somatic DICER1 missense hotspot mutation, as a second hit, involving 1 of 6 codons clustered in exons 24 and 25. We designed and in vitro validated a drop-off droplet digital PCR (ddPCR) system to scan all DICER1 hotspot codons, allowing for a liquid biopsy test, an alternative to sedation and radiation exposure. METHODS: Three drop-off ddPCR assays were designed, with 2 TaqMan probes per assay, 1 complementary to the wild-type sequence of the region containing hotspots and another 1 used as a reference. Eight tumor-derived DNAs and 5 synthetic oligonucleotides bearing DICER1 hotspot mutations were tested. RESULTS: All tested mutations were detected, with a limit of detection ranging from 0.07% to 0.31% for codons p. E1705, p. D1709, and p. D1713 in exon 24 and from 0.06% to 0.15% for codons p. G1809, p. D1810, and p. E1813 in exon 25. CONCLUSIONS: The high sensitivity of this method is compatible with its use for plasma circulating tumor DNA (ctDNA) analysis for early tumor detection in DICER1 syndrome patients. It may reduce the need for radiation exposure and sedation in surveillance protocols and may also improve patient prognosis. Clinical trials are needed to evaluate ctDNA analysis in these patients.


Asunto(s)
ADN Tumoral Circulante , Neoplasias , Niño , Codón , ARN Helicasas DEAD-box/genética , Humanos , Recién Nacido , Mutación , Neoplasias/genética , Reacción en Cadena de la Polimerasa/métodos , Ribonucleasa III/genética , Adulto Joven
3.
Hum Mutat ; 41(3): 608-618, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31729086

RESUMEN

Nijmegen breakage syndrome caused by biallelic pathogenic variants of the DNA-damage response gene NBN, is characterized by severe microcephaly, cancer proneness, infertility, and karyotype abnormalities. We previously reported NBN variants in siblings suffering from fertility defects. Here, we identify a new founder NBN variant (c.442A>G, p.(Thr148Ala)) in Lebanese patients associated with isolated infertility. Functional analyses explored preserved or altered functions correlated with their remarkably mild phenotype. Transcript and protein analyses supported the use of an alternative transcript with in-frame skipping of exons 4-5, leading to p84-NBN protein with a preserved forkhead-associated (FHA) domain. The level of NBN was dramatically reduced and the MRN complex delocalized to the cytoplasm. Interestingly, ataxia-elangiectasia mutated (ATM) also shifted from the nucleus to the cytoplasm, suggesting some interaction between ATM and the MRN complex at a steady state. The ATM pathway activation, attenuated in typical patients with NBS, appeared normal under camptothecin treatment in these new NBN-related infertile patients. Cell cycle checkpoint defect was present in these atypical patients, although to a lesser extent than in typical patients with NBS. In conclusion, we report three new NBN-related infertile patients and we suggest that preserved FHA domain could be responsible for the mild phenotype and intermediate DNA-damage response defects.


Asunto(s)
Proteínas de Ciclo Celular/genética , Reparación del ADN , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Infertilidad/diagnóstico , Infertilidad/genética , Proteínas Nucleares/genética , Adulto , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/metabolismo , Análisis Mutacional de ADN , Femenino , Citometría de Flujo , Regulación de la Expresión Génica , Estudios de Asociación Genética/métodos , Humanos , Infertilidad/metabolismo , Masculino , Síndrome de Nijmegen/diagnóstico , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Transducción de Señal
4.
J Allergy Clin Immunol ; 143(1): 325-334.e2, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29906526

RESUMEN

BACKGROUND: V(D)J recombination ensures the diversity of the adaptive immune system. Although its complete defect causes severe combined immunodeficiency (ie, T-B- severe combined immunodeficiency), its suboptimal activity is associated with a broad spectrum of immune manifestations, such as late-onset combined immunodeficiency and autoimmunity. The earliest molecular diagnosis of these patients is required to adopt the best therapy strategy, particularly when it involves a myeloablative conditioning regimen for hematopoietic stem cell transplantation. OBJECTIVE: We aimed at developing biomarkers based on analysis of the T-cell receptor (TCR) α repertoire to assist in the diagnosis of patients with primary immunodeficiencies with V(D)J recombination and DNA repair deficiencies. METHODS: We used flow cytometric (fluorescence-activated cell sorting) analysis to quantify TCR-Vα7.2-expressing T lymphocytes in peripheral blood and developed PROMIDISα, a multiplex RT-PCR/next-generation sequencing assay, to evaluate a subset of the TCRα repertoire in T lymphocytes. RESULTS: The combined fluorescence-activated cell sorting and PROMIDISα analyses revealed specific signatures in patients with V(D)J recombination-defective primary immunodeficiencies or ataxia telangiectasia/Nijmegen breakage syndromes. CONCLUSION: Analysis of the TCRα repertoire is particularly appropriate in a prospective way to identify patients with partial immune defects caused by suboptimal V(D)J recombination activity, a DNA repair defect, or both. It also constitutes a valuable tool for the retrospective in vivo functional validation of variants identified through exome or panel sequencing. Its broader implementation might be of interest to assist early diagnosis of patients presenting with hypomorphic DNA repair defects inclined to experience acute toxicity during prehematopoietic stem cell transplantation conditioning.


Asunto(s)
Síndromes de Inmunodeficiencia , Receptores de Antígenos de Linfocitos T alfa-beta , Recombinación V(D)J/inmunología , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Síndromes de Inmunodeficiencia/patología , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Estudios Retrospectivos
5.
Hum Mutat ; 40(10): 1690-1699, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31033087

RESUMEN

Ataxia-telangiectasia-like disorder (ATLD) is a rare genomic instability syndrome caused by biallelic variants of MRE11 (meiotic recombination 11) characterized by progressive cerebellar ataxia and typical karyotype abnormalities. These symptoms are common to those of ataxia-telangiectasia, which is consistent with the key role of MRE11 in ataxia-telangiectasia mutated (ATM) activation after DNA double-strand breaks. Three unrelated French patients were referred with ataxia. Only one had typical karyotype abnormalities. Unreported biallelic MRE11 variants were found in these three cases. Interestingly, one variant (c.424G>A) was present in two cases and haplotype analysis strongly suggested a French founder variant. Variants c.544G>A and c.314+4_314+7del lead to splice defects. The level of MRE11 in lymphoblastoid cell lines was consistently and dramatically reduced. Functional consequences were evaluated on activation of the ATM pathway via phosphorylation of ATM targets (KAP1 and CHK2), but no consistent defect was observed. However, an S-phase checkpoint activation defect after camptothecin was observed in these patients with ATLD. In conclusion, we report the first three French ATLD patients and a French founder variant, and propose an S-phase checkpoint activation study to evaluate the pathogenicity of MRE11 variants.


Asunto(s)
Ataxia Telangiectasia/diagnóstico , Ataxia Telangiectasia/etiología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Niño , Susceptibilidad a Enfermedades , Femenino , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Lactante , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Imagen por Resonancia Magnética , Mutación , Fenotipo , Empalme del ARN , Puntos de Control de la Fase S del Ciclo Celular/genética , Transducción de Señal , Transcriptoma
6.
Hum Mutat ; 40(10): 1713-1730, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31050087

RESUMEN

Ataxia-telangiectasia (A-T) is a recessive disorder caused by biallelic pathogenic variants of ataxia-telangiectasia mutated (ATM). This disease is characterized by progressive ataxia, telangiectasia, immune deficiency, predisposition to malignancies, and radiosensitivity. However, hypomorphic variants may be discovered associated with very atypical phenotypes, raising the importance of evaluating their pathogenic effects. In this study, multiple functional analyses were performed on lymphoblastoid cell lines from 36 patients, comprising 49 ATM variants, 24 being of uncertain significance. Thirteen patients with atypical phenotype and presumably hypomorphic variants were of particular interest to test strength of functional analyses and to highlight discrepancies with typical patients. Western-blot combined with transcript analyses allowed the identification of one missing variant, confirmed suspected splice defects and revealed unsuspected minor transcripts. Subcellular localization analyses confirmed the low level and abnormal cytoplasmic localization of ATM for most A-T cell lines. Interestingly, atypical patients had lower kinase defect and less altered cell-cycle distribution after genotoxic stress than typical patients. In conclusion, this study demonstrated the pathogenic effects of the 49 variants, highlighted the strength of KAP1 phosphorylation test for pathogenicity assessment and allowed the establishment of the Ataxia-TeLangiectasia Atypical Score to predict atypical phenotype. Altogether, we propose strategies for ATM variant detection and classification.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Ataxia Telangiectasia/diagnóstico , Ataxia Telangiectasia/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Empalme Alternativo , Ciclo Celular , Línea Celular , Análisis Mutacional de ADN , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Mutación , Fenotipo
7.
Pediatr Blood Cancer ; 65(6): e27005, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29469200

RESUMEN

Germline DICER1 pathogenic variants predispose to numerous benign and malignant tumors. In this report, we describe DICER1 gene analysis in an adolescent diagnosed with multinodular goiter, ovarian Sertoli-Leydig cell tumor, and lung cyst. DICER1 mutational screening at the DNA level failed to detect any pathogenic variant. Subsequent messenger RNA (mRNA) analysis revealed a 132 nucleotide intronic sequence exonization. This truncating event was caused by a deep intronic mutation generating a de novo acceptor splice site. This study demonstrates that some undetected DICER1 mutations should be investigated at the mRNA level.


Asunto(s)
ARN Helicasas DEAD-box/genética , Mutación de Línea Germinal , Bocio Nodular/genética , Intrones/genética , Enfermedades Pulmonares/genética , Neoplasias Ováricas/genética , Ribonucleasa III/genética , Tumor de Células de Sertoli-Leydig/genética , Adolescente , Anciano , Niño , Femenino , Bocio Nodular/patología , Humanos , Enfermedades Pulmonares/patología , Masculino , Neoplasias Ováricas/patología , Linaje , Pronóstico , Tumor de Células de Sertoli-Leydig/patología
8.
Am J Hum Genet ; 92(6): 974-80, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23684012

RESUMEN

The genetic cause of some familial nonsyndromic renal cell carcinomas (RCC) defined by at least two affected first-degree relatives is unknown. By combining whole-exome sequencing and tumor profiling in a family prone to cases of RCC, we identified a germline BAP1 mutation c.277A>G (p.Thr93Ala) as the probable genetic basis of RCC predisposition. This mutation segregated with all four RCC-affected relatives. Furthermore, BAP1 was found to be inactivated in RCC-affected individuals from this family. No BAP1 mutations were identified in 32 familial cases presenting with only RCC. We then screened for germline BAP1 deleterious mutations in familial aggregations of cancers within the spectrum of the recently described BAP1-associated tumor predisposition syndrome, including uveal melanoma, malignant pleural mesothelioma, and cutaneous melanoma. Among the 11 families that included individuals identified as carrying germline deleterious BAP1 mutations, 6 families presented with 9 RCC-affected individuals, demonstrating a significantly increased risk for RCC. This strongly argues that RCC belongs to the BAP1 syndrome and that BAP1 is a RCC-predisposition gene.


Asunto(s)
Carcinoma de Células Renales/genética , Mutación de Línea Germinal , Neoplasias Renales/genética , Mutación Missense , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Adulto , Secuencia de Bases , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Exoma , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
9.
Tunis Med ; 93(8-9): 511-5, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26815515

RESUMEN

BACKGROUND: Ataxia-telangiectasia (A-T) is a multisystem disorder characterized by progressive neurologic impairment, variable immunodeficiency, impaired organ maturation, X-ray hypersensitivity, oculocutaneous telangiectasia, and a predisposition to malignancy. AIM: We performed this study in order to describe clinical, immunological and molecular features of patients with AT followed in the south of Tunisia Methods: we performed a retrospective study (1996-2012) in the south of Tunisia about all cases of A-T in order to describe their clinical, immunological and molecular features. RESULTS: 11 cases of AT were found. The mean age at onset of symptoms was 20 months with extremes varying from 3 months to 4 years. The median time to diagnosis was 3.6 years (range: 0-12 years).The main clinical feature of cerebellar syndrome, ataxia, was present at diagnosis in 8 patients and occurred at mean ages of 2.8 years. Ocular telangiectasia occurred at a mean age of 3.9 years (extremes: 3 months and 7 years). Recurrent sino-pulmonary infections that affected 7 children occurred at the mean age of 4.3 years. The most common humoral immune abnormality was serum IgA deficiency. Lymphopenia was found in 7 cases and lack of CD4 T in 6 cases. Cytogenetic analyses showed chromosomal instability in all children and a translocation (7-14) in two patients. A molecular diagnosis established in 6 patients from 4 families showed 5 different mutations of ATM gene. After an average decline of 5 years and 6 months, 7 patients died of severe pulmonary infection. Among them, 3 were ATM mutated. CONCLUSION: Morbidity and mortality among patients with A- T are associated with ATM genotype.


Asunto(s)
Ataxia Telangiectasia/diagnóstico , Proteínas de la Ataxia Telangiectasia Mutada/genética , Preescolar , Femenino , Humanos , Lactante , Masculino , Mutación , Estudios Retrospectivos , Túnez
10.
J Natl Cancer Inst ; 116(4): 580-587, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38060262

RESUMEN

BACKGROUND: Monoallelic germline MBD4 pathogenic variants were recently reported to cause a predisposition to uveal melanoma, associated with a specific tumor mutational signature and good response to immunotherapy. Monoallelic tumor pathogenic variants have also been described in brain tumors, breast cancers, and myxofibrosarcomas, whereas biallelic germline MBD4 pathogenic variants have been involved in a recessive hereditary adenomatous polyposis and a specific type of acute myeloid leukemia. METHODS: We analyzed MBD4 for all patients with a diagnosis of uveal melanoma at Institut Curie since July 2021 and in the 3240 consecutive female probands explored at the Institut Curie for suspicion of predisposition to breast cancer between July 2021 and February 2023. RESULTS: We describe 25 families whose probands carry a monoallelic germline pathogenic variant in MBD4. Eighteen of these families presented with uveal melanoma (including a case patient with multiple uveal melanoma), and 7 families presented with breast cancer. Family histories showed the first familial case of uveal melanoma in monoallelic MBD4 pathogenic variant carriers and other various types of cancers in relatives, especially breast, renal, and colorectal tumors. CONCLUSIONS: Monoallelic MBD4 pathogenic variant may explain some cases of familial and multiple uveal melanoma as well as various cancer types, expanding the tumor spectrum of this predisposition. Further genetic testing in relatives combined with molecular tumor analyses will help define the tumor spectrum and estimate each tumor's risk.


Asunto(s)
Neoplasias de la Mama , Melanoma , Neoplasias Cutáneas , Neoplasias de la Úvea , Humanos , Adulto , Femenino , Predisposición Genética a la Enfermedad , Melanoma/epidemiología , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/genética , Mutación de Línea Germinal , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Endodesoxirribonucleasas/genética
11.
Blood ; 117(15): e161-70, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21325596

RESUMEN

Fanconi anemia (FA) is a genetic condition associated with bone marrow (BM) failure, myelodysplasia (MDS), and acute myeloid leukemia (AML). We studied 57 FA patients with hypoplastic or aplastic anemia (n = 20), MDS (n = 18), AML (n = 11), or no BM abnormality (n = 8). BM samples were analyzed by karyotype, high-density DNA arrays with respect to paired fibroblasts, and by selected oncogene sequencing. A specific pattern of chromosomal abnormalities was found in MDS/AML, which included 1q+ (44.8%), 3q+ (41.4%), -7/7q (17.2%), and 11q- (13.8%). Moreover, cryptic RUNX1/AML1 lesions (translocations, deletions, or mutations) were observed for the first time in FA (20.7%). Rare mutations of NRAS, FLT3-ITD, MLL-PTD, ERG amplification, and ZFP36L2-PRDM16 translocation, but no TP53, TET2, CBL, NPM1, and CEBPα mutations were found. Frequent homozygosity regions were related not to somatic copy-neutral loss of heterozygosity but to consanguinity, suggesting that homologous recombination is not a common progression mechanism in FA. Importantly, the RUNX1 and other chromosomal/genomic lesions were found at the MDS/AML stages, except for 1q+, which was found at all stages. These data have implications for staging and therapeutic managing in FA patients, and also to analyze the mechanisms of clonal evolution and oncogenesis in a background of genomic instability and BM failure.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Adolescente , Adulto , Médula Ósea/fisiología , Niño , Preescolar , Anemia de Fanconi/complicaciones , Femenino , Dosificación de Gen/genética , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Genes Supresores de Tumor , Homocigoto , Humanos , Leucemia Mieloide Aguda/etiología , Masculino , Persona de Mediana Edad , Síndromes Mielodisplásicos/etiología , Nucleofosmina , Polimorfismo de Nucleótido Simple , Adulto Joven
12.
J Allergy Clin Immunol ; 128(2): 382-9.e1, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21665257

RESUMEN

BACKGROUND: Ataxia-telangiectasia (A-T) is a rare genetic disease caused by germline biallelic mutations in the ataxia-telangiectasia mutated gene (ATM) that result in partial or complete loss of ATM expression or activity. The course of the disease is characterized by neurologic manifestations, infections, and cancers. OBJECTIVE: We studied A-T progression and investigated whether manifestations were associated with the ATM genotype. METHODS: We performed a retrospective cohort study in France of 240 patients with A-T born from 1954 to 2005 and analyzed ATM mutations in 184 patients, along with neurologic manifestations, infections, and cancers. RESULTS: Among patients with A-T, the Kaplan-Meier 20-year survival rate was 53.4%; the prognosis for these patients has not changed since 1954. Life expectancy was lower among patients with mutations in ATM that caused total loss of expression or function of the gene product (null mutations) compared with that seen in patients with hypomorphic mutations because of earlier onset of cancer (mainly hematologic malignancies). Cancer (hazard ratio, 2.7; 95% CI, 1.6-4.5) and respiratory tract infections (hazard ratio, 2.3; 95% CI, 1.4-3.8) were independently associated with mortality. Cancer (hazard ratio, 5.8; 95% CI, 2.9-11.6) was a major risk factor for mortality among patients with null mutations, whereas respiratory tract infections (hazard ratio, 4.1; 95% CI, 1.8-9.1) were the leading cause of death among patients with hypomorphic mutations. CONCLUSION: Morbidity and mortality among patients with A-T are associated with ATM genotype. This information could improve our prognostic ability and lead to adapted therapeutic strategies.


Asunto(s)
Ataxia Telangiectasia/genética , Ataxia Telangiectasia/mortalidad , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Adolescente , Adulto , Ataxia Telangiectasia/epidemiología , Ataxia Telangiectasia/fisiopatología , Proteínas de la Ataxia Telangiectasia Mutada , Niño , Preescolar , Estudios de Cohortes , Femenino , Francia/epidemiología , Genotipo , Humanos , Lactante , Recién Nacido , Leucemia/genética , Linfoma/genética , Masculino , Morbilidad , Mutación , Infecciones del Sistema Respiratorio/genética , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
13.
Hum Mutat ; 30(3): 424-30, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19105185

RESUMEN

Biallelic mutations in the NBN/NBS1 gene are the cause of Nijmegen breakage syndrome (NBS), a severe pediatric disease characterized by dysmorphy with a bird-like face, microcephaly, growth retardation, immune deficiency, and proneness to cancer. We here report two adult siblings that are compound heterozygotes for two previously unreported NBN nonsense mutations. These patients presented with the unique clinical symptom of fertility defects. Contrasting with the absence of any developmental abnormality, biological analyses revealed defects similar to those observed in NBS patients, including chromosomal instability, cellular hyperradiosensitivity and checkpoint defects as measured by radioresistant DNA synthesis (RDS). NBN mutations should thus be considered a new cause of infertility, and should be searched for if associated with the biological abnormalities of NBS.


Asunto(s)
Proteínas de Ciclo Celular/genética , Codón sin Sentido , Mutación de Línea Germinal , Infertilidad/genética , Proteínas Nucleares/genética , Adulto , Secuencia de Bases , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular Transformada , Análisis Mutacional de ADN , Femenino , Heterocigoto , Humanos , Infertilidad/metabolismo , Infertilidad/patología , Masculino , Síndrome de Nijmegen/genética , Síndrome de Nijmegen/patología , Proteínas Nucleares/metabolismo , Hermanos
14.
Genet Test ; 12(2): 257-61, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18471088

RESUMEN

Bloom's syndrome (BS) is a rare autosomal recessive disease predisposing patients to all types of cancers affecting the general population. BS cells display a high level of genetic instability, including a 10-fold increase in the rate of sister chromatid exchanges, currently the only objective criterion for BS diagnosis. We have developed a method for screening the BLM gene for mutations based on direct genomic DNA sequencing. A questionnaire based on clinical information, cytogenetic features, and family history was addressed to physicians prescribing BS genetic screening, with the aim of confirming or guiding diagnosis. We report here four BLM gene mutations, three of which have not been described before. Three of the mutations are frameshift mutations, and the fourth is a nonsense mutation. All these mutations introduce a stop codon, and may therefore be considered to have deleterious biological effect. This approach should make it possible to identify new mutations and to correlate them with clinical information.


Asunto(s)
Síndrome de Bloom/diagnóstico , Síndrome de Bloom/genética , ADN Helicasas/genética , Análisis Mutacional de ADN/métodos , Mutación , Adulto , Síndrome de Bloom/fisiopatología , Niño , Preescolar , Codón sin Sentido , Femenino , Mutación del Sistema de Lectura , Pruebas Genéticas , Genoma , Humanos , Lactante , Masculino , RecQ Helicasas , Análisis de Secuencia de ADN
16.
J Clin Invest ; 126(9): 3580-4, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27500492

RESUMEN

Fanconi anemia (FA) is a recessive genetic disease characterized by congenital abnormalities, chromosome instability, progressive bone marrow failure (BMF), and a strong predisposition to cancer. Twenty FA genes have been identified, and the FANC proteins they encode cooperate in a common pathway that regulates DNA crosslink repair and replication fork stability. We identified a child with severe BMF who harbored biallelic inactivating mutations of the translesion DNA synthesis (TLS) gene REV7 (also known as MAD2L2), which encodes the mutant REV7 protein REV7-V85E. Patient-derived cells demonstrated an extended FA phenotype, which included increased chromosome breaks and G2/M accumulation upon exposure to DNA crosslinking agents, γH2AX and 53BP1 foci accumulation, and enhanced p53/p21 activation relative to cells derived from healthy patients. Expression of WT REV7 restored normal cellular and functional phenotypes in the patient's cells, and CRISPR/Cas9 inactivation of REV7 in a non-FA human cell line produced an FA phenotype. Finally, silencing Rev7 in primary hematopoietic cells impaired progenitor function, suggesting that the DNA repair defect underlies the development of BMF in FA. Taken together, our genetic and functional analyses identified REV7 as a previously undescribed FA gene, which we term FANCV.


Asunto(s)
Anemia de Fanconi/genética , Proteínas Mad2/genética , Mutación , Alelos , Animales , Ciclo Celular , Línea Celular Tumoral , Niño , Inestabilidad Cromosómica , Rotura Cromosómica , Estudios de Cohortes , Reactivos de Enlaces Cruzados/química , Daño del ADN , Reparación del ADN , Femenino , Fibroblastos/metabolismo , Silenciador del Gen , Prueba de Complementación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Células Madre Hematopoyéticas/citología , Humanos , Lentivirus , Proteínas Mad2/metabolismo , Masculino , Ratones , Ratones Noqueados , Mitosis , Fenotipo
17.
J Clin Oncol ; 33(2): 202-8, 2015 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-25488969

RESUMEN

PURPOSE: Biallelic mutations in ATM cause ataxia-telangiectasia (AT), a rare inherited disease with a high incidence of cancer. Precise estimates of the risk, presentation, and outcomes of cancer in patients with AT need to be addressed in large series. PATIENTS AND METHODS: In this large retrospective cohort, 69 patients with cancers (24.5%) were identified among 279 patients with AT. Centralized review was performed on 60% of the lymphomas. Incidence rates were compared with the French population, and risk factors were analyzed. RESULTS: Eight patients developed acute leukemias (including four T-cell acute lymphoblastic leukemias), 12 developed Hodgkin lymphoma (HL), 38 developed non-Hodgkin lymphoma (NHL), three developed T-cell prolymphocytic leukemia (T-PLL), and eight developed carcinoma at a median age of 8.3, 10.6, 9.7, 24.2, and 31.4 years, respectively (P < .001). The majority of NHLs were aggressive B-cell NHL. Epstein-Barr virus was associated with all of the HLs and 50% of the NHLs. Overall survival was shorter in patients with AT who developed cancer compared with those who did not develop cancer (15 v 24 years, respectively; P < .001). Survival was improved in patients who achieved a major response to treatment (3.46 v 0.87 years for major v minor responses, respectively; P = .011). Immunodeficiency was associated with increased risk of cancer. ATM mutation type was associated with a difference in survival in the entire cohort but not with cancer incidence or cancer survival. CONCLUSION: B-cell NHL, HL, and acute lymphoblastic leukemia occur at a high rate and earlier age than carcinomas in AT. T-PLLs are rarer than initially reported. Prognosis is poor, but patients may benefit from treatment with an improved survival.


Asunto(s)
Ataxia Telangiectasia/complicaciones , Ataxia Telangiectasia/mortalidad , Leucemia/epidemiología , Linfoma/epidemiología , Neoplasias/diagnóstico , Neoplasias/epidemiología , Enfermedad Aguda , Adulto , Factores de Edad , Anciano , Femenino , Francia/epidemiología , Enfermedad de Hodgkin/epidemiología , Humanos , Síndromes de Inmunodeficiencia , Incidencia , Linfoma no Hodgkin/epidemiología , Masculino , Persona de Mediana Edad , Mutación , Neoplasias/genética , Neoplasias/mortalidad , Neoplasias/terapia , Pronóstico , Sistema de Registros , Estudios Retrospectivos
18.
Neurology ; 83(12): 1087-95, 2014 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-25122203

RESUMEN

OBJECTIVE: To assess the clinical spectrum of ataxia-telangiectasia (A-T) in adults, with a focus on movement disorders. METHODS: A total of 14 consecutive adults with A-T were included at 2 tertiary adult movement disorders centers and compared to 53 typical patients with A-T. Clinical evaluation, neurophysiologic and video-oculographic recording, imaging, laboratory investigations, and ATM analysis were performed. RESULTS: In comparison with typical A-T cases, our patients demonstrated later mean age at onset (6.1 vs 2.5 years, p < 0.0001), later loss of walking ability (p = 0.003), and longer survival (p = 0.0039). The presenting feature was ataxia in 71% and dysarthria and dystonia in 14% each. All patients displayed movement disorders, among which dystonia and subcortical myoclonus were the most common (86%), followed by tremor (43%). Video-oculographic recordings revealed mostly dysmetric saccades and 46% of patients had normal latencies (i.e., no oculomotor apraxia) and velocities. The α-fetoprotein (AFP) level was normal in 7%, chromosomal instability was found in 29% (vs 100% of typical patients, p = 0.0006), and immunoglobulin deficiency was found in 29% (vs 69%, p = 0.057). All patients exhibited 2 ATM mutations, including at least 1 missense mutation in 79% of them (vs 36%, p = 0.0067). CONCLUSION: There is great variability of phenotype and severity in A-T, including a wide spectrum of movement disorders. Karyotype and repeated AFP level assessments should be performed in adults with unexplained movement disorders as valuable clues towards the diagnosis. In case of a compatible phenotype, A-T should be considered even if age at onset is late and progression is slow.


Asunto(s)
Ataxia Telangiectasia/fisiopatología , Disartria/fisiopatología , Distonía/fisiopatología , Adulto , Edad de Inicio , Ataxia Telangiectasia/epidemiología , Ataxia Telangiectasia/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Inestabilidad Cromosómica/genética , Estudios de Cohortes , Progresión de la Enfermedad , Disartria/genética , Distonía/genética , Medidas del Movimiento Ocular , Femenino , Pleiotropía Genética , Humanos , Inmunoglobulinas/deficiencia , Masculino , Limitación de la Movilidad , Trastornos del Movimiento/genética , Trastornos del Movimiento/fisiopatología , Mutación Missense , Mioclonía/genética , Mioclonía/fisiopatología , Trastornos de la Motilidad Ocular/genética , Trastornos de la Motilidad Ocular/fisiopatología , Fenotipo , Índice de Severidad de la Enfermedad , Adulto Joven , alfa-Fetoproteínas
19.
J Clin Invest ; 121(1): 184-94, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21183791

RESUMEN

DNA damage checkpoints in the cell cycle may be important barriers against cancer progression in human cells. Fanconi anemia (FA) is an inherited DNA instability disorder that is associated with bone marrow failure and a strong predisposition to cancer. Although FA cells experience constitutive chromosomal breaks, cell cycle arrest at the G2 DNA damage checkpoint, and an excess of cell death, some patients do become clinically stable, and the mechanisms underlying this, other than spontaneous reversion of the disease-causing mutation, are not well understood. Here we have defined a clonal phenotype, termed attenuation, in which FA patients acquire an abrogation of the G2 checkpoint arrest. Attenuated cells expressed lower levels of CHK1 (also known as CHEK1) and p53. The attenuation could be recapitulated by modulating the ATR/CHK1 pathway, and CHK1 inhibition protected FA cells from cell death. FA patients who expressed the attenuated phenotype had mild bone marrow deficiency and reached adulthood, but several of them eventually developed myelodysplasia or leukemia. Better understanding of attenuation might help predict a patient's clinical course and guide choice of treatment. Our results also highlight the importance of evaluating the cellular DNA damage checkpoint and repair pathways in cancer therapies in general.


Asunto(s)
Daño del ADN , Anemia de Fanconi/complicaciones , Anemia de Fanconi/genética , Fase G2/genética , Leucemia Mieloide Aguda/etiología , Adolescente , Adulto , Secuencia de Bases , Médula Ósea/metabolismo , Médula Ósea/patología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Quinasa de Punto de Control 2 , Niño , Preescolar , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Femenino , Humanos , Lactante , Leucemia Mieloide Aguda/genética , Masculino , Metaloproteinasa 1 de la Matriz/genética , Persona de Mediana Edad , Modelos Biológicos , Mutación , Síndromes Mielodisplásicos/etiología , Síndromes Mielodisplásicos/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA