Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genet Med ; 23(11): 2150-2159, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34345024

RESUMEN

PURPOSE: DYRK1A syndrome is among the most frequent monogenic forms of intellectual disability (ID). We refined the molecular and clinical description of this disorder and developed tools to improve interpretation of missense variants, which remains a major challenge in human genetics. METHODS: We reported clinical and molecular data for 50 individuals with ID harboring DYRK1A variants and developed (1) a specific DYRK1A clinical score; (2) amino acid conservation data generated from 100 DYRK1A sequences across different taxa; (3) in vitro overexpression assays to study level, cellular localization, and kinase activity of DYRK1A mutant proteins; and (4) a specific blood DNA methylation signature. RESULTS: This integrative approach was successful to reclassify several variants as pathogenic. However, we questioned the involvement of some others, such as p.Thr588Asn, still reported as likely pathogenic, and showed it does not cause an obvious phenotype in mice. CONCLUSION: Our study demonstrated the need for caution when interpreting variants in DYRK1A, even those occurring de novo. The tools developed will be useful to interpret accurately the variants identified in the future in this gene.


Asunto(s)
Discapacidad Intelectual , Microcefalia , Proteínas Serina-Treonina Quinasas , Proteínas Tirosina Quinasas , Animales , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Ratones , Fenotipo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Quinasas DyrK
2.
Clin Genet ; 99(2): 318-324, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33169370

RESUMEN

Bardet-Biedl syndrome (BBS) is a ciliopathy characterized by retinitis pigmentosa, obesity, polydactyly, cognitive impairment and renal failure. Pathogenic variants in 24 genes account for the molecular basis of >80% of cases. Toward saturated discovery of the mutational basis of the disorder, we carefully explored our cohorts and identified a hominid-specific SINE-R/VNTR/Alu type F (SVA-F) insertion in exon 13 of BBS1 in eight families. In six families, the repeat insertion was found in trans with c.1169 T > G, p.Met390Arg and in two families the insertion was found in addition to other recessive BBS loci. Whole genome sequencing, de novo assembly and SNP array analysis were performed to characterize the genomic event. This insertion is extremely rare in the general population (found in 8 alleles of 8 BBS cases but not in >10 800 control individuals from gnomAD-SV) and due to a founder effect. Its 2435 bp sequence contains hallmarks of LINE1 mediated retrotransposition. Functional studies with patient-derived cell lines confirmed that the BBS1 SVA-F is deleterious as evidenced by a significant depletion of both mRNA and protein levels. Such findings highlight the importance of dedicated bioinformatics pipelines to identify all types of variation.


Asunto(s)
Síndrome de Bardet-Biedl/genética , Proteínas Asociadas a Microtúbulos/genética , Retroelementos , Estudios de Cohortes , Femenino , Efecto Fundador , Frecuencia de los Genes , Humanos , Masculino , Mutagénesis Insercional , Linaje , Secuenciación Completa del Genoma
3.
Clin Genet ; 99(5): 650-661, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33415748

RESUMEN

Megalencephaly-CApillary malformation-Polymicrogyria (MCAP) syndrome results from somatic mosaic gain-of-function variants in PIK3CA. Main features are macrocephaly, somatic overgrowth, cutaneous vascular malformations, connective tissue dysplasia, neurodevelopmental delay, and brain anomalies. The objectives of this study were to describe the clinical and radiological features of MCAP, to suggest relevant clinical endpoints applicable in future trials of targeted drug therapy. Based on a French collaboration, we collected clinical features of 33 patients (21 females, 12 males, median age of 9.9 years) with MCAP carrying mosaic PIK3CA pathogenic variants. MRI images were reviewed for 21 patients. The main clinical features reported were macrocephaly at birth (20/31), postnatal macrocephaly (31/32), body/facial asymmetry (21/33), cutaneous capillary malformations (naevus flammeus 28/33, cutis marmorata 17/33). Intellectual disability was present in 15 patients. Among the MRI images reviewed, the neuroimaging findings were megalencephaly (20/21), thickening of corpus callosum (16/21), Chiari malformation (12/21), ventriculomegaly/hydrocephaly (10/21), cerebral asymmetry (6/21) and polymicrogyria (2/21). This study confirms the main known clinical features that defines MCAP syndrome. Taking into account the phenotypic heterogeneity in MCAP patients, in the context of emerging clinical trials, we suggest that patients should be evaluated based on the main neurocognitive expression on each patient.


Asunto(s)
Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/fisiopatología , Ensayos Clínicos como Asunto , Megalencefalia/diagnóstico por imagen , Megalencefalia/fisiopatología , Neuroimagen , Enfermedades Cutáneas Vasculares/diagnóstico por imagen , Enfermedades Cutáneas Vasculares/fisiopatología , Telangiectasia/congénito , Anomalías Múltiples/tratamiento farmacológico , Adolescente , Adulto , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I/genética , Estudios de Cohortes , Femenino , Predicción , Humanos , Imagen por Resonancia Magnética , Masculino , Megalencefalia/tratamiento farmacológico , Enfermedades Cutáneas Vasculares/tratamiento farmacológico , Telangiectasia/diagnóstico por imagen , Telangiectasia/tratamiento farmacológico , Telangiectasia/fisiopatología , Adulto Joven
4.
Am J Med Genet A ; 185(12): 3877-3883, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34357686

RESUMEN

Kleefstra syndrome (KS) is a rare autosomic dominant genetic disorder caused by euchromatic histone methyltransferase 1 (EHMT1) alterations. Patients mainly present with moderate to severe intellectual disability, a severe delay in/or absence of speech, autism spectrum disorder, childhood hypotonia, neuropsychiatric anomalies, and distinctive dysmorphic features. Here, we report the cases of a male and a female, two younger siblings of three, with asymptomatic parents. An EHMT1 new mutation was identified. Both presented with a typical core phenotype. Some specific features were noted, such as macrocephaly (previously reported) and enuresis (not yet described). Parental analysis identified the mutation in the mosaic state in the father. Reverse phenotyping enabled us to highlight the pauci phenotype features of inguinal hernia, azoospermia, and possible behavioral disorders. This allowed us to adapt his follow-up and genetic counseling for the family. Our three reported cases provide a new description of KS with an intragenic EHMT1 mutation, whereas in the literature most reported cases have EHMT1 deletions. Moreover, in the areas of next-generation sequencing and trio techniques with parental segregation, it is important to remain cautious about disregarding variants based on an autosomal recessive hypothesis.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Megalencefalia/genética , Anomalías Múltiples/patología , Adolescente , Adulto , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Hibridación Genómica Comparativa , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/patología , Femenino , Asesoramiento Genético , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Masculino , Megalencefalia/patología , Mosaicismo , Mutación , Fenotipo , Adulto Joven
5.
J Med Genet ; 57(5): 301-307, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-30287593

RESUMEN

BACKGROUND: The clinical significance of 16p13.11 duplications remains controversial while frequently detected in patients with developmental delay (DD), intellectual deficiency (ID) or autism spectrum disorder (ASD). Previously reported patients were not or poorly characterised. The absence of consensual recommendations leads to interpretation discrepancy and makes genetic counselling challenging. This study aims to decipher the genotype-phenotype correlations to improve genetic counselling and patients' medical care. METHODS: We retrospectively analysed data from 16 013 patients referred to 12 genetic centers for DD, ID or ASD, and who had a chromosomal microarray analysis. The referring geneticists of patients for whom a 16p13.11 duplication was detected were asked to complete a questionnaire for detailed clinical and genetic data for the patients and their parents. RESULTS: Clinical features are mainly speech delay and learning disabilities followed by ASD. A significant risk of cardiovascular disease was noted. About 90% of the patients inherited the duplication from a parent. At least one out of four parents carrying the duplication displayed a similar phenotype to the propositus. Genotype-phenotype correlations show no impact of the size of the duplicated segment on the severity of the phenotype. However, NDE1 and miR-484 seem to have an essential role in the neurocognitive phenotype. CONCLUSION: Our study shows that 16p13.11 microduplications are likely pathogenic when detected in the context of DD/ID/ASD and supports an essential role of NDE1 and miR-484 in the neurocognitive phenotype. Moreover, it suggests the need for cardiac evaluation and follow-up and a large study to evaluate the aortic disease risk.


Asunto(s)
Trastorno del Espectro Autista/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , MicroARNs/genética , Proteínas Asociadas a Microtúbulos/genética , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Adolescente , Adulto , Trastorno del Espectro Autista/patología , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Niño , Preescolar , Cromosomas Humanos Par 16/genética , Discapacidades del Desarrollo/patología , Femenino , Duplicación de Gen/genética , Estudios de Asociación Genética , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Fenotipo , Factores de Riesgo , Adulto Joven
6.
Genet Med ; 22(3): 538-546, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31723249

RESUMEN

PURPOSE: Intellectual disability (ID) and autism spectrum disorder (ASD) are genetically heterogeneous neurodevelopmental disorders. We sought to delineate the clinical, molecular, and neuroimaging spectrum of a novel neurodevelopmental disorder caused by variants in the zinc finger protein 292 gene (ZNF292). METHODS: We ascertained a cohort of 28 families with ID due to putatively pathogenic ZNF292 variants that were identified via targeted and exome sequencing. Available data were analyzed to characterize the canonical phenotype and examine genotype-phenotype relationships. RESULTS: Probands presented with ID as well as a spectrum of neurodevelopmental features including ASD, among others. All ZNF292 variants were de novo, except in one family with dominant inheritance. ZNF292 encodes a highly conserved zinc finger protein that acts as a transcription factor and is highly expressed in the developing human brain supporting its critical role in neurodevelopment. CONCLUSION: De novo and dominantly inherited variants in ZNF292 are associated with a range of neurodevelopmental features including ID and ASD. The clinical spectrum is broad, and most individuals present with mild to moderate ID with or without other syndromic features. Our results suggest that variants in ZNF292 are likely a recurrent cause of a neurodevelopmental disorder manifesting as ID with or without ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/patología , Niño , Preescolar , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/diagnóstico por imagen , Trastornos del Neurodesarrollo/patología , Neuroimagen/métodos , Secuenciación del Exoma/métodos
7.
Clin Genet ; 98(1): 10-18, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32233106

RESUMEN

Overlapping syndromes such as Noonan, Cardio-Facio-Cutaneous, Noonan syndrome (NS) with multiple lentigines and Costello syndromes are genetically heterogeneous conditions sharing a dysregulation of the RAS/mitogen-activated protein kinase (MAPK) pathway and are known collectively as the RASopathies. PTPN11 was the first disease-causing gene identified in NS and remains the more prevalent. We report seven patients from three families presenting heterozygous missense variants in PTPN11 probably responsible for a disease phenotype distinct from the classical Noonan syndrome. The clinical presentation and common features of these seven cases overlap with the SHORT syndrome. The latter is the consequence of PI3K/AKT signaling deregulation with the predominant disease-causing gene being PIK3R1. Our data suggest that the phenotypic spectrum associated with pathogenic variants of PTPN11 could be wider than previously described, and this could be due to the dual activity of SHP2 (ie, PTPN11 gene product) on the RAS/MAPK and PI3K/AKT signaling.


Asunto(s)
Variación Genética/genética , Trastornos del Crecimiento/genética , Hipercalcemia/genética , Enfermedades Metabólicas/genética , Nefrocalcinosis/genética , Síndrome de Noonan/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteínas Quinasas Activadas por Mitógenos/genética , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/genética
8.
J Med Genet ; 56(8): 526-535, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30923172

RESUMEN

BACKGROUND: Balanced chromosomal rearrangements associated with abnormal phenotype are rare events, but may be challenging for genetic counselling, since molecular characterisation of breakpoints is not performed routinely. We used next-generation sequencing to characterise breakpoints of balanced chromosomal rearrangements at the molecular level in patients with intellectual disability and/or congenital anomalies. METHODS: Breakpoints were characterised by a paired-end low depth whole genome sequencing (WGS) strategy and validated by Sanger sequencing. Expression study of disrupted and neighbouring genes was performed by RT-qPCR from blood or lymphoblastoid cell line RNA. RESULTS: Among the 55 patients included (41 reciprocal translocations, 4 inversions, 2 insertions and 8 complex chromosomal rearrangements), we were able to detect 89% of chromosomal rearrangements (49/55). Molecular signatures at the breakpoints suggested that DNA breaks arose randomly and that there was no major influence of repeated elements. Non-homologous end-joining appeared as the main mechanism of repair (55% of rearrangements). A diagnosis could be established in 22/49 patients (44.8%), 15 by gene disruption (KANSL1, FOXP1, SPRED1, TLK2, MBD5, DMD, AUTS2, MEIS2, MEF2C, NRXN1, NFIX, SYNGAP1, GHR, ZMIZ1) and 7 by position effect (DLX5, MEF2C, BCL11B, SATB2, ZMIZ1). In addition, 16 new candidate genes were identified. Systematic gene expression studies further supported these results. We also showed the contribution of topologically associated domain maps to WGS data interpretation. CONCLUSION: Paired-end WGS is a valid strategy and may be used for structural variation characterisation in a clinical setting.


Asunto(s)
Aberraciones Cromosómicas , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Reordenamiento Génico , Estudios de Asociación Genética , Fenotipo , Secuenciación Completa del Genoma , Adolescente , Adulto , Biomarcadores , Niño , Preescolar , Puntos de Rotura del Cromosoma , Variaciones en el Número de Copia de ADN , Femenino , Estudios de Asociación Genética/métodos , Humanos , Lactante , Masculino , Relación Estructura-Actividad , Translocación Genética , Adulto Joven
9.
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
10.
Clin Genet ; 96(4): 317-329, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31245841

RESUMEN

Pathogenic variants in FLNC encoding filamin C have been firstly reported to cause myopathies, and were recently linked to isolated cardiac phenotypes. Our aim was to estimate the prevalence of FLNC pathogenic variants in subtypes of cardiomyopathies and to study the relations between phenotype and genotype. DNAs from a cohort of 1150 unrelated index-patients with isolated cardiomyopathy (700 hypertrophic, 300 dilated, 50 restrictive cardiomyopathies, and 100 left ventricle non-compactions) have been sequenced on a custom panel of 51 cardiomyopathy disease-causing genes. An FLNC pathogenic variant was identified in 28 patients corresponding to a prevalence ranging from 1% to 8% depending on the cardiomyopathy subtype. Truncating variants were always identified in patients with dilated cardiomyopathy, while missense or in-frame indel variants were found in other phenotypes. A personal or family history of sudden cardiac death (SCD) was significantly higher in patients with truncating variants than in patients carrying missense variants (P = .01). This work reported the first observation of a left ventricular non-compaction associated with a unique probably causal variant in FLNC which highlights the role of FLNC in cardiomyopathies. A correlation between the nature of the variant and the cardiomyopathy subtype was observed as well as with SCD risk.


Asunto(s)
Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Filaminas/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Alelos , Cardiomiopatías/epidemiología , Ecocardiografía , Electrocardiografía , Femenino , Pruebas Genéticas , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Imagen por Resonancia Magnética , Masculino , Mutación , Linaje , Fenotipo , Prevalencia , Análisis de Secuencia de ADN
11.
Hum Mutat ; 39(3): 319-332, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29243349

RESUMEN

Autosomal recessive microcephaly or microcephaly primary hereditary (MCPH) is a genetically heterogeneous neurodevelopmental disorder characterized by a reduction in brain volume, indirectly measured by an occipitofrontal circumference (OFC) 2 standard deviations or more below the age- and sex-matched mean (-2SD) at birth and -3SD after 6 months, and leading to intellectual disability of variable severity. The abnormal spindle-like microcephaly gene (ASPM), the human ortholog of the Drosophila melanogaster "abnormal spindle" gene (asp), encodes ASPM, a protein localized at the centrosome of apical neuroprogenitor cells and involved in spindle pole positioning during neurogenesis. Loss-of-function mutations in ASPM cause MCPH5, which affects the majority of all MCPH patients worldwide. Here, we report 47 unpublished patients from 39 families carrying 28 new ASPM mutations, and conduct an exhaustive review of the molecular, clinical, neuroradiological, and neuropsychological features of the 282 families previously reported (with 161 distinct ASPM mutations). Furthermore, we show that ASPM-related microcephaly is not systematically associated with intellectual deficiency and discuss the association between the structural brain defects (strong reduction in cortical volume and surface area) that modify the cortical map of these patients and their cognitive abilities.


Asunto(s)
Microcefalia/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Preescolar , Cognición , Estudios de Cohortes , Familia , Femenino , Estudios de Asociación Genética , Geografía , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Microcefalia/epidemiología
12.
Am J Med Genet C Semin Med Genet ; 175(4): 417-430, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29178447

RESUMEN

CHARGE syndrome (CS) is a genetic disorder whose first description included Coloboma, Heart disease, Atresia of choanae, Retarded growth and development, Genital hypoplasia, and Ear anomalies and deafness, most often caused by a genetic mutation in the CHD7 gene. Two features were then added: semicircular canal anomalies and arhinencephaly/olfactory bulb agenesis, with classification of typical, partial, or atypical forms on the basis of major and minor clinical criteria. The detection rate of a pathogenic variant in the CHD7 gene varies from 67% to 90%. To try to have an overview of this heterogenous clinical condition and specify a genotype-phenotype relation, we conducted a national study of phenotype and genotype in 119 patients with CS. Selected clinical diagnostic criteria were from Verloes (2005), updated by Blake & Prasad (). Besides obtaining a detailed clinical description, when possible, patients underwent a full ophthalmologic examination, audiometry, temporal bone CT scan, gonadotropin analysis, and olfactory-bulb MRI. All patients underwent CHD7 sequencing and MLPA analysis. We found a pathogenic CHD7 variant in 83% of typical CS cases and 58% of atypical cases. Pathogenic variants in the CHD7 gene were classified by the expected impact on the protein. In all, 90% of patients had a typical form of CS and 10% an atypical form. The most frequent features were deafness/semicircular canal hypoplasia (94%), pituitary defect/hypogonadism (89%), external ear anomalies (87%), square-shaped face (81%), and arhinencephaly/anosmia (80%). Coloboma (73%), heart defects (65%), and choanal atresia (43%) were less frequent.


Asunto(s)
Síndrome CHARGE/diagnóstico , Síndrome CHARGE/genética , Estudios de Asociación Genética , Genotipo , Fenotipo , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adolescente , Adulto , Alelos , Sustitución de Aminoácidos , Sistema Nervioso Central/anomalías , Niño , Preescolar , Estudios de Cohortes , Nervios Craneales/anomalías , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Femenino , Francia , Pruebas Genéticas , Humanos , Lactante , Masculino , Técnicas de Diagnóstico Molecular , Adulto Joven
13.
Genet Med ; 19(9): 989-997, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28151489

RESUMEN

PURPOSE: Postzygotic activating mutations of PIK3CA cause a wide range of mosaic disorders collectively referred to as PIK3CA-related overgrowth spectrum (PROS). We describe the diagnostic yield and characteristics of PIK3CA sequencing in PROS. METHODS: We performed ultradeep next-generation sequencing (NGS) of PIK3CA in various tissues from 162 patients referred to our clinical laboratory and assessed diagnostic yield by phenotype and tissue tested. RESULTS: We identified disease-causing mutations in 66.7% (108/162) of patients, with mutant allele levels as low as 1%. The diagnostic rate was higher (74%) in syndromic than in isolated cases (35.5%; P = 9.03 × 10-5). We identified 40 different mutations and found strong oncogenic mutations more frequently in patients without brain overgrowth (50.6%) than in those with brain overgrowth (15.2%; P = 0.00055). Mutant allele levels were higher in skin and overgrown tissues than in blood and buccal samples (P = 3.9 × 10-25), regardless of the phenotype. CONCLUSION: Our data demonstrate the value of ultradeep NGS for molecular diagnosis of PROS, highlight its substantial allelic heterogeneity, and confirm that optimal diagnosis requires fresh skin or surgical samples from affected regions. Our findings may be of value in guiding future recommendations for genetic testing in PROS and other mosaic conditions.Genet Med advance online publication 02 February 2017.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Estudios de Asociación Genética , Pruebas Genéticas , Trastornos del Crecimiento/diagnóstico , Trastornos del Crecimiento/genética , Mutación , Adolescente , Adulto , Alelos , Sustitución de Aminoácidos , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Manejo de la Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Mosaicismo , Fenotipo , Diagnóstico Prenatal , Análisis de Secuencia de ADN , Adulto Joven
14.
Am J Med Genet A ; 173(10): 2782-2788, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28815877

RESUMEN

Prenatal growth is a complex dynamic process controlled by various genetic and environmental factors. Among genetic syndromes characterized by growth restriction, MULIBREY nanism represents a rare autosomal recessive condition presenting with severe pre- and post-natal growth failure, characteristic dysmorphic features but normal neurological development. The phenotype of MULIBREY nanism is variable and overlaps with others such as the Silver-Russell syndrome. We report here three patients in two distinct non-Finnish families from North France who were first suspected to have Silver-Russell syndrome which failed to be confirmed on molecular analyses. Clinical features in the three patients led us to also consider the diagnosis of MULIBREY nanism. Sequencing of the TRIM37 gene showed the three patients shared a novel nonsense mutation (c.181 C>T p.Arg61*) in a heterozygous state. Quantitative fluorescent multiplex PCR identified a new deletion of exons 15 and 16 in TRIM37 in one isolated patient and another deletion of exon 9 in two siblings. Breakpoints of both the deletions were localized in Alu sequences. Given the high number of Alu repeats, which predispose to gene rearrangements, one should always consider such genetic rearrangements in the molecular diagnosis of non-Finnish MULIBREY nanism patients. Early diagnosis of the disease would prompt careful cardiac follow up of such patients as cardiological complication is a characteristic feature of the MULIBREY nanism as described in this report.


Asunto(s)
Reordenamiento Génico , Enanismo Mulibrey/genética , Mutación , Proteínas Nucleares/genética , Adolescente , Niño , Preescolar , Femenino , Francia , Humanos , Lactante , Masculino , Enanismo Mulibrey/patología , Pronóstico , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
15.
Am J Med Genet A ; 173(11): 2923-2946, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28948695

RESUMEN

Facial femoral syndrome (FFS) is a rare congenital abnormality, also known as femoral hypoplasia-unusual facies syndrome, characterized by variable degrees of femoral hypoplasia, associated with specific facial features. Other organ malformations are sometimes present. Most cases are sporadic, but rare family observations suggest genetic origin. However, no chromosomal or genetic abnormalities have ever been incriminated. We conducted a comprehensive literature review and added three new unreported observations. Through these 92 cases, authors aimed to determine sonographic signs that should direct towards diagnosis, and discuss potential genetic etiology. Diagnosis was suspected prenatally in 27.2% of cases, and maternal diabetes was found in 42.4% of patients. When fetal karyotype was available, it was normal in 97.1% of cases, but genomic variations of unknown significance were discovered in all three cases in which array comparative genomic hybridization (CGH) techniques were applied. Femoral affection defining FFS was hypoplasia in 78.3% of cases, agenesis in 12%, and both in 9.8%. Affection was bilateral in 84.8% of cases. Retrognathia was present in 65.2% of cases, cleft lip and/or palate in 63%, and other organ malformations in 53.3%. Intellectual development was normal in 79.2% of cases. Better prenatal recognition of this pathology, notably frequently associated malformations, should lead to a more precise estimation of functional prognosis. It seems likely that today's tendency to systematically employ array-CGH and exome/genome sequencing methods to investigate malformative sequences will allow the identification of a causal genetic abnormality in the near future.


Asunto(s)
Anomalías Múltiples/diagnóstico , Fémur/anomalías , Síndrome de Pierre Robin/diagnóstico , Diagnóstico Prenatal , Ultrasonografía Prenatal/métodos , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/genética , Anomalías Múltiples/fisiopatología , Adulto , Labio Leporino/diagnóstico , Labio Leporino/diagnóstico por imagen , Labio Leporino/genética , Labio Leporino/fisiopatología , Hibridación Genómica Comparativa , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/diagnóstico por imagen , Diabetes Gestacional/fisiopatología , Femenino , Fémur/diagnóstico por imagen , Fémur/fisiopatología , Feto , Humanos , Recién Nacido , Masculino , Persona de Mediana Edad , Síndrome de Pierre Robin/diagnóstico por imagen , Síndrome de Pierre Robin/genética , Síndrome de Pierre Robin/fisiopatología , Embarazo
16.
Am J Hum Genet ; 93(1): 141-9, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23810378

RESUMEN

Short stature, hyperextensibility of joints and/or inguinal hernia, ocular depression, Rieger anomaly, and teething delay (SHORT) syndrome is a developmental disorder with an unknown genetic cause and hallmarks that include insulin resistance and lack of subcutaneous fat. We ascertained two unrelated individuals with SHORT syndrome, hypothesized that the observed phenotype was most likely due to de novo mutations in the same gene, and performed whole-exome sequencing in the two probands and their unaffected parents. We then confirmed our initial observations in four other subjects with SHORT syndrome from three families, as well as 14 unrelated subjects presenting with syndromic insulin resistance and/or generalized lipoatrophy associated with dysmorphic features and growth retardation. Overall, we identified in nine affected individuals from eight families de novo or inherited PIK3R1 mutations, including a mutational hotspot (c.1945C>T [p.Arg649Trp]) present in four families. PIK3R1 encodes the p85α, p55α, and p50α regulatory subunits of class IA phosphatidylinositol 3 kinases (PI3Ks), which are known to play a key role in insulin signaling. Functional data from fibroblasts derived from individuals with PIK3R1 mutations showed severe insulin resistance for both proximal and distal PI3K-dependent signaling. Our findings extend the genetic causes of severe insulin-resistance syndromes and provide important information with respect to the function of PIK3R1 in normal development and its role in human diseases, including growth delay, Rieger anomaly and other ocular affections, insulin resistance, diabetes, paucity of fat, and ovarian cysts.


Asunto(s)
Trastornos del Crecimiento/genética , Hipercalcemia/genética , Resistencia a la Insulina/genética , Enfermedades Metabólicas/genética , Nefrocalcinosis/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Análisis Mutacional de ADN , Exoma , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Edad Gestacional , Glucosa/metabolismo , Glucosa/farmacología , Humanos , Insulina/metabolismo , Insulina/farmacología , Masculino , Mutación , Linaje , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
17.
Gastroenterology ; 149(4): 1017-29.e3, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26116798

RESUMEN

BACKGROUND & AIMS: Patients with bi-allelic germline mutations in mismatch repair (MMR) genes (MLH1, MSH2, MSH6, or PMS2) develop a rare but severe variant of Lynch syndrome called constitutional MMR deficiency (CMMRD). This syndrome is characterized by early-onset colorectal cancers, lymphomas or leukemias, and brain tumors. There is no satisfactory method for diagnosis of CMMRD because screens for mutations in MMR genes are noninformative for 30% of patients. MMR-deficient cancer cells are resistant to genotoxic agents and have microsatellite instability (MSI), due to accumulation of errors in repetitive DNA sequences. We investigated whether these features could be used to identify patients with CMMRD. METHODS: We examined MSI by PCR analysis and tolerance to methylating or thiopurine agents (functional characteristics of MMR-deficient tumor cells) in lymphoblastoid cells (LCs) from 3 patients with CMMRD and 5 individuals with MMR-proficient LCs (controls). Using these assays, we defined experimental parameters that allowed discrimination of a series of 14 patients with CMMRD from 52 controls (training set). We then used the same parameters to assess 23 patients with clinical but not genetic features of CMMRD. RESULTS: In the training set, we identified parameters, based on MSI and LC tolerance to methylation, that detected patients with CMMRD vs controls with 100% sensitivity and 100% specificity. Among 23 patients suspected of having CMMRD, 6 had MSI and LC tolerance to methylation (CMMRD highly probable), 15 had neither MSI nor LC tolerance to methylation (unlikely to have CMMRD), and 2 were considered doubtful for CMMRD based on having only 1 of the 2 features. CONCLUSION: The presence of MSI and tolerance to methylation in LCs identified patients with CMMRD with 100% sensitivity and specificity. These features could be used in diagnosis of patients.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Neoplasias Colorrectales/diagnóstico , Resistencia a Antineoplásicos , Pruebas Genéticas , Mutación de Línea Germinal , Linfocitos/efectos de los fármacos , Inestabilidad de Microsatélites , Síndromes Neoplásicos Hereditarios/diagnóstico , Proteínas Adaptadoras Transductoras de Señales/genética , Adenosina Trifosfatasas/genética , Adulto , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Células CACO-2 , Estudios de Casos y Controles , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Colorrectales Hereditarias sin Poliposis/tratamiento farmacológico , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/metabolismo , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Análisis Mutacional de ADN , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Células HCT116 , Herencia , Humanos , Linfocitos/metabolismo , Masculino , Metilación , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Reacción en Cadena de la Polimerasa Multiplex , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS/genética , Síndromes Neoplásicos Hereditarios/tratamiento farmacológico , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/metabolismo , Síndromes Neoplásicos Hereditarios/patología , Proteínas Nucleares/genética , Fenotipo , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Transfección , Adulto Joven
18.
Hum Mol Genet ; 22(25): 5121-35, 2013 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-23906836

RESUMEN

Chromatin remodeling complexes are known to modify chemical marks on histones or to induce conformational changes in the chromatin in order to regulate transcription. De novo dominant mutations in different members of the SWI/SNF chromatin remodeling complex have recently been described in individuals with Coffin-Siris (CSS) and Nicolaides-Baraitser (NCBRS) syndromes. Using a combination of whole-exome sequencing, NGS-based sequencing of 23 SWI/SNF complex genes, and molecular karyotyping in 46 previously undescribed individuals with CSS and NCBRS, we identified a de novo 1-bp deletion (c.677delG, p.Gly226Glufs*53) and a de novo missense mutation (c.914G>T, p.Cys305Phe) in PHF6 in two individuals diagnosed with CSS. PHF6 interacts with the nucleosome remodeling and deacetylation (NuRD) complex implicating dysfunction of a second chromatin remodeling complex in the pathogenesis of CSS-like phenotypes. Altogether, we identified mutations in 60% of the studied individuals (28/46), located in the genes ARID1A, ARID1B, SMARCB1, SMARCE1, SMARCA2, and PHF6. We show that mutations in ARID1B are the main cause of CSS, accounting for 76% of identified mutations. ARID1B and SMARCB1 mutations were also found in individuals with the initial diagnosis of NCBRS. These individuals apparently belong to a small subset who display an intermediate CSS/NCBRS phenotype. Our proposed genotype-phenotype correlations are important for molecular screening strategies.


Asunto(s)
Anomalías Múltiples/genética , Ensamble y Desensamble de Cromatina/genética , Cara/anomalías , Deformidades Congénitas del Pie/genética , Deformidades Congénitas de la Mano/genética , Hipotricosis/genética , Discapacidad Intelectual/genética , Micrognatismo/genética , Cuello/anomalías , Eliminación de Secuencia/genética , Anomalías Múltiples/patología , Adolescente , Adulto , Proteínas Portadoras/genética , Niño , Preescolar , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Exoma/genética , Cara/patología , Facies , Femenino , Deformidades Congénitas del Pie/patología , Deformidades Congénitas de la Mano/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hipotricosis/patología , Lactante , Recién Nacido , Discapacidad Intelectual/patología , Cariotipificación , Masculino , Micrognatismo/patología , Mutación Missense , Cuello/patología , Proteínas Represoras , Proteína SMARCB1 , Factores de Transcripción/genética
19.
Am J Med Genet A ; 167A(3): 504-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25572454

RESUMEN

Interstitial microdeletions of 20q chromosome are rare, only 17 patients have been reported in the literature to date. Among them, only six carried a proximal 20q11.21-q11.23 deletion, with a size ranging from 2.6 to 6.8 Mb. The existence of a 20q11.2 microdeletion syndrome has been proposed, based on five previously reported cases that displayed anomalies of the extremities, intellectual disability, feeding difficulties, craniofacial dysmorphism and variable malformations. To further characterize this syndrome, we report on six new patients with 20q11.2 microdeletions diagnosed by whole-genome array-based comparative genomic hybridization. These patient reports more precisely refined the phenotype and narrowed the minimal critical region involved in this syndrome. Careful clinical assessment confirms the distinctive clinical phenotype. The craniofacial dysmorphism consists of high forehead, frontal bossing, enophthalmos, and midface hypoplasia. We have identified a 1.62 megabase minimal critical region involved in this syndrome encompassing three genes­GDF5, EPB41L1, andSAMHD1­which are strong candidates for different aspects of the phenotype. These results support that 20q11.2 microdeletion syndrome is a new contiguous gene deletion syndrome with a recognizable phenotype.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 20 , Fenotipo , Adolescente , Niño , Preescolar , Aberraciones Cromosómicas , Puntos de Rotura del Cromosoma , Hibridación Genómica Comparativa , Facies , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Cariotipificación , Masculino , Síndrome , Adulto Joven
20.
Am J Med Genet A ; 167A(1): 111-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25425167

RESUMEN

Array comparative genomic hybridization (array CGH) has proven its utility in uncovering cryptic rearrangements in patients with X-linked intellectual disability. In 2009, Giorda et al. identified inherited and de novo recurrent Xp11.23p11.22 microduplications in two males and six females from a wide cohort of patients presenting with syndromic intellectual disability. To date, 14 females and 5 males with an overlapping microduplication have been reported in the literature. To further characterize this emerging syndrome, we collected clinical and microarray data from 17 new patients, 10 females, and 7 males. The Xp11.23p11.2 microduplications detected by array CGH ranged in size from 331 Kb to 8.9 Mb. Five patients harbored 4.5 Mb recurrent duplications mediated by non-allelic homologous recombination between segmental duplications and 12 harbored atypical duplications. The chromosomal rearrangement occurred de novo in eight patients and was inherited in six affected males from three families. Patients shared several common major characteristics including moderate to severe intellectual disability, early onset of puberty, language impairment, and age related epileptic syndromes such as West syndrome and focal epilepsy with activation during sleep evolving in some patients to continuous spikes-and-waves during slow sleep. Atypical microduplications allowed us to identify minimal critical regions that might be responsible for specific clinical findings of the syndrome and to suggest possible candidate genes: FTSJ1 and SHROOM4 for intellectual disability along with PQBP1 and SLC35A2 for epilepsy. Xp11.23p11.22 microduplication is a recently-recognized syndrome associated with intellectual disability, epilepsy, and early onset of puberty in females. In this study, we propose several genes that could contribute to the phenotype.


Asunto(s)
Cromosomas Humanos X/genética , Estudios de Asociación Genética , Duplicaciones Segmentarias en el Genoma/genética , Adolescente , Adulto , Niño , Preescolar , Mapeo Cromosómico , Hibridación Genómica Comparativa , Electroencefalografía , Epilepsia/genética , Femenino , Humanos , Masculino , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA