Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 61(5): 469-476, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38458756

RESUMEN

BACKGROUND: Marfan syndrome (MFS) is a multisystem disease with a unique combination of skeletal, cardiovascular and ocular features. Geleophysic/acromicric dysplasias (GPHYSD/ACMICD), characterised by short stature and extremities, are described as 'the mirror image' of MFS. The numerous FBN1 pathogenic variants identified in MFS are located all along the gene and lead to the same final pathogenic sequence. Conversely, in GPHYSD/ACMICD, the 28 known heterozygous FBN1 pathogenic variants all affect exons 41-42 encoding TGFß-binding protein-like domain 5 (TB5). METHODS: Since 1996, more than 5000 consecutive probands have been referred nationwide to our laboratory for molecular diagnosis of suspected MFS. RESULTS: We identified five MFS probands carrying distinct heterozygous pathogenic in-frame variants affecting the TB5 domain of FBN1. The clinical data showed that the probands displayed a classical form of MFS. Strikingly, one missense variant affects an amino acid that was previously involved in GPHYSD. CONCLUSION: Surprisingly, pathogenic variants in the TB5 domain of FBN1 can lead to two opposite phenotypes: GPHYSD/ACMICD and MFS, suggesting the existence of different pathogenic sequences with the involvement of tissue specificity. Further functional studies are ongoing to determine the precise role of this domain in the physiopathology of each disease.


Asunto(s)
Enfermedades del Desarrollo Óseo , Deformidades Congénitas de las Extremidades , Síndrome de Marfan , Humanos , Enfermedades del Desarrollo Óseo/genética , Fibrilina-1/genética , Síndrome de Marfan/genética , Síndrome de Marfan/patología , Mutación
2.
Am J Hum Genet ; 106(2): 153-169, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31978331

RESUMEN

Cilia and flagella are evolutionarily conserved organelles whose motility relies on the outer and inner dynein arm complexes (ODAs and IDAs). Defects in ODAs and IDAs result in primary ciliary dyskinesia (PCD), a disease characterized by recurrent airway infections and male infertility. PCD mutations in assembly factors have been shown to cause a combined ODA-IDA defect, affecting both cilia and flagella. We identified four loss-of-function mutations in TTC12, which encodes a cytoplasmic protein, in four independent families in which affected individuals displayed a peculiar PCD phenotype characterized by the absence of ODAs and IDAs in sperm flagella, contrasting with the absence of only IDAs in respiratory cilia. Analyses of both primary cells from individuals carrying TTC12 mutations and human differentiated airway cells invalidated for TTC12 by a CRISPR-Cas9 approach revealed an IDA defect restricted to a subset of single-headed IDAs that are different in flagella and cilia, whereas TTC12 depletion in the ciliate Paramecium tetraurelia recapitulated the sperm phenotype. Overall, our study, which identifies TTC12 as a gene involved in PCD, unveils distinct dynein assembly mechanisms in human motile cilia versus flagella.


Asunto(s)
Cilios/patología , Trastornos de la Motilidad Ciliar/etiología , Dineínas/metabolismo , Flagelos/patología , Mutación , Proteínas/genética , Cola del Espermatozoide/patología , Adulto , Axonema , Niño , Cilios/metabolismo , Trastornos de la Motilidad Ciliar/patología , Dineínas/genética , Femenino , Flagelos/metabolismo , Homocigoto , Humanos , Infertilidad Masculina/etiología , Infertilidad Masculina/patología , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Motilidad Espermática , Cola del Espermatozoide/metabolismo , Adulto Joven
3.
Electrophoresis ; 39(24): 3123-3132, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29869806

RESUMEN

Congenital disorders of glycosylation (CDG) are rare autosomal genetic diseases affecting the glycosylation of proteins and lipids. Since CDG-related clinical symptoms are classically extremely variable and nonspecific, a combination of electrophoretic, mass spectrometric, and gene sequencing techniques is often mandatory for obtaining a definitive CDG diagnosis, as well as identifying causative gene mutations and deciphering the underlying biochemical mechanisms. Here, we illustrate the potential of integrating data from capillary electrophoresis of transferrin, two-dimensional electrophoresis of N- and O-glycoproteins, mass spectrometry analyses of total serum N-linked glycans and mucin core1 O-glycosylated apolipoprotein C-III for the determination of various culprit CDG gene mutations. "Step-by-step" diagnosis pathways of four particular and new CDG cases, including MGAT2-CDG, ATP6V0A2-CDG, SLC35A2-CDG, and SLC35A3-CDG, are described as illustrative examples.


Asunto(s)
Trastornos Congénitos de Glicosilación , Electroforesis/métodos , Espectrometría de Masas/métodos , Análisis de Secuencia de ADN/métodos , Adolescente , Niño , Preescolar , Trastornos Congénitos de Glicosilación/sangre , Trastornos Congénitos de Glicosilación/diagnóstico , Femenino , Glicómica , Glicoproteínas/sangre , Glicoproteínas/química , Humanos , Lactante , Masculino , Polisacáridos/análisis , Polisacáridos/química
4.
J Med Genet ; 54(12): 843-851, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28954837

RESUMEN

BACKGROUND: Phosphomannomutase 2-congenital disorder of glycosylation (PMM2-CDG) is a multisystem inborn error of metabolism. OBJECTIVES: To better characterise the natural history of PMM2-CDG. METHODS: Medical charts of 96 patients with PMM2-CDG (86 families, 41 males, 55 females) were retrospectively reviewed. Data on clinical, laboratory and molecular parameters at diagnosis were analysed. Follow-up data at last examination were reported for 25 patients. RESULTS: The patients were born between 1963 and 2011. Diagnosis of PMM2-CDG was made at a mean (SD) age of 6.8 (8.5) years. The presenting signs were mostly neurological (hypotonia, intellectual disability, cerebellar syndrome) and observed in almost all the patients. A total of 38 patients (14 males, 24 females) exhibited, in addition to neurological signs, visceral features including at least one of these: feeding difficulty requiring a nutritional support (n=23), cardiac features (n=20; pericarditis: 14, cardiac malformation: 9, cardiomyopathy: 2), hepato-gastrointestinal features (n=12; chronic diarrhoea: 7, protein-losing enteropathy: 1, ascites: 3, liver failure: 1, portal hypertension: 1), kidney features (n=4; nephrotic syndrome: 2, tubulopathy: 2) and hydrops fetalis (n=1). Twelve patients died at a mean age of 3.8 years (especially from pericarditis and other cardiac issues). Laboratory abnormalities mostly included elevated transaminases and abnormal coagulation parameters. High thyreostimulin levels, hypocholesterolemia, hypoalbuminemia and elevated transaminases were associated with the visceral phenotype. Besides the common Arg141His PMM2 variant harboured by half of the patients, 45 different variants were observed. CONCLUSIONS: PMM2-CDG clinical phenotype is heterogeneous in terms of clinical course, with no clear division between neurological and visceral presentations.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/genética , Estudios de Asociación Genética , Fosfotransferasas (Fosfomutasas)/genética , Adolescente , Alelos , Sustitución de Aminoácidos , Niño , Preescolar , Trastornos Congénitos de Glicosilación/mortalidad , Femenino , Estudios de Seguimiento , Humanos , Lactante , Masculino , Mutación , Fenotipo , Fosfotransferasas (Fosfomutasas)/metabolismo
5.
Hum Mol Genet ; 23(9): 2279-89, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24319099

RESUMEN

Non-syndromic arthrogryposis multiplex congenita (AMC) is characterized by multiple congenital contractures resulting from reduced fetal mobility. Genetic mapping and whole exome sequencing (WES) were performed in 31 multiplex and/or consanguineous undiagnosed AMC families. Although this approach identified known AMC genes, we here report pathogenic mutations in two new genes. Homozygous frameshift mutations in CNTNAP1 were found in four unrelated families. Patients showed a marked reduction in motor nerve conduction velocity (<10 m/s) and transmission electron microscopy (TEM) of sciatic nerve in the index cases revealed severe abnormalities of both nodes of Ranvier width and myelinated axons. CNTNAP1 encodes CASPR, an essential component of node of Ranvier domains which underlies saltatory conduction of action potentials along the myelinated axons, an important process for neuronal function. A homozygous missense mutation in adenylate cyclase 6 gene (ADCY6) was found in another family characterized by a lack of myelin in the peripheral nervous system (PNS) as determined by TEM. Morpholino knockdown of the zebrafish orthologs led to severe and specific defects in peripheral myelin in spite of the presence of Schwann cells. ADCY6 encodes a protein that belongs to the adenylate cyclase family responsible for the synthesis of cAMP. Elevation of cAMP can mimic axonal contact in vitro and upregulates myelinating signals. Our data indicate an essential and so far unknown role of ADCY6 in PNS myelination likely through the cAMP pathway. Mutations of genes encoding proteins of Ranvier domains or involved in myelination of Schwann cells are responsible for novel and severe human axoglial diseases.


Asunto(s)
Adenilil Ciclasas/genética , Artrogriposis/genética , Artrogriposis/patología , Moléculas de Adhesión Celular Neuronal/genética , Axones/patología , Axones/ultraestructura , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Microscopía Electrónica de Transmisión , Mutación/genética , Vaina de Mielina/patología , Sistema Nervioso Periférico/patología , Sistema Nervioso Periférico/ultraestructura , Embarazo , Células de Schwann/metabolismo
6.
Genet Med ; 17(8): 651-9, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25394172

RESUMEN

PURPOSE: Congenital hypogonadotropic hypogonadism (CHH) and split hand/foot malformation (SHFM) are two rare genetic conditions. Here we report a clinical entity comprising the two. METHODS: We identified patients with CHH and SHFM through international collaboration. Probands and available family members underwent phenotyping and screening for FGFR1 mutations. The impact of identified mutations was assessed by sequence- and structure-based predictions and/or functional assays. RESULTS: We identified eight probands with CHH with (n = 3; Kallmann syndrome) or without anosmia (n = 5) and SHFM, seven of whom (88%) harbor FGFR1 mutations. Of these seven, one individual is homozygous for p.V429E and six individuals are heterozygous for p.G348R, p.G485R, p.Q594*, p.E670A, p.V688L, or p.L712P. All mutations were predicted by in silico analysis to cause loss of function. Probands with FGFR1 mutations have severe gonadotropin-releasing hormone deficiency (absent puberty and/or cryptorchidism and/or micropenis). SHFM in both hands and feet was observed only in the patient with the homozygous p.V429E mutation; V429 maps to the fibroblast growth factor receptor substrate 2α binding domain of FGFR1, and functional studies of the p.V429E mutation demonstrated that it decreased recruitment and phosphorylation of fibroblast growth factor receptor substrate 2α to FGFR1, thereby resulting in reduced mitogen-activated protein kinase signaling. CONCLUSION: FGFR1 should be prioritized for genetic testing in patients with CHH and SHFM because the likelihood of a mutation increases from 10% in the general CHH population to 88% in these patients.


Asunto(s)
Hipogonadismo/congénito , Hipogonadismo/genética , Deformidades Congénitas de las Extremidades/genética , Mutación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia Conservada , Femenino , Estudios de Asociación Genética , Humanos , Hipogonadismo/metabolismo , Deformidades Congénitas de las Extremidades/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Linaje , Fosforilación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo
7.
J Med Genet ; 51(11): 724-36, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25167861

RESUMEN

BACKGROUND: Intellectual disability (ID) is characterised by an extreme genetic heterogeneity. Several hundred genes have been associated to monogenic forms of ID, considerably complicating molecular diagnostics. Trio-exome sequencing was recently proposed as a diagnostic approach, yet remains costly for a general implementation. METHODS: We report the alternative strategy of targeted high-throughput sequencing of 217 genes in which mutations had been reported in patients with ID or autism as the major clinical concern. We analysed 106 patients with ID of unknown aetiology following array-CGH analysis and other genetic investigations. Ninety per cent of these patients were males, and 75% sporadic cases. RESULTS: We identified 26 causative mutations: 16 in X-linked genes (ATRX, CUL4B, DMD, FMR1, HCFC1, IL1RAPL1, IQSEC2, KDM5C, MAOA, MECP2, SLC9A6, SLC16A2, PHF8) and 10 de novo in autosomal-dominant genes (DYRK1A, GRIN1, MED13L, TCF4, RAI1, SHANK3, SLC2A1, SYNGAP1). We also detected four possibly causative mutations (eg, in NLGN3) requiring further investigations. We present detailed reasoning for assigning causality for each mutation, and associated patients' clinical information. Some genes were hit more than once in our cohort, suggesting they correspond to more frequent ID-associated conditions (KDM5C, MECP2, DYRK1A, TCF4). We highlight some unexpected genotype to phenotype correlations, with causative mutations being identified in genes associated to defined syndromes in patients deviating from the classic phenotype (DMD, TCF4, MECP2). We also bring additional supportive (HCFC1, MED13L) or unsupportive (SHROOM4, SRPX2) evidences for the implication of previous candidate genes or mutations in cognitive disorders. CONCLUSIONS: With a diagnostic yield of 25% targeted sequencing appears relevant as a first intention test for the diagnosis of ID, but importantly will also contribute to a better understanding regarding the specific contribution of the many genes implicated in ID and autism.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Técnicas de Diagnóstico Molecular/métodos , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN/métodos , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Análisis de Secuencia de ADN/métodos , Adulto Joven
9.
J Pediatr ; 162(3): 593-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23022111

RESUMEN

OBJECTIVE: To estimate the prevalence of congenital cytomegalovirus (cCMV) among causes of bilateral hearing loss in young French children. STUDY DESIGN: Children <3 years old with hearing loss were prospectively included at their first visit to a referral center. Cytomegalovirus polymerase chain reaction was performed on dried blood spots from Guthrie cards. Medical records were reviewed. RESULTS: One hundred children with bilateral hearing loss were included at a median age of 15 months; the prevalence of cCMV was 8% (8/100) (95% CI, 2.7%-13.3%) in this population and 15.4% (8/52) in the subpopulation of children with profound bilateral hearing loss. Delayed neurodevelopment and brain abnormalities on computed tomography scan were found more often in children with cCMV than in children with hearing loss without cCMV (P = .027, P = .005). In 6 of 8 cCMV cases, cCMV infection had not been diagnosed before the study. CONCLUSIONS: In a comprehensive study of the causes of bilateral hearing loss in young French children, cCMV is the second most frequent cause of hearing loss after connexin mutations. It underlines that a majority of French children with hearing loss and cCMV are not diagnosed early and therefore may not benefit from early intervention including the possibility of neonatal antiviral treatment. These results make the case for promoting systematic cytomegalovirus screening in neonates with confirmed hearing loss identified through neonatal hearing screening.


Asunto(s)
Infecciones por Citomegalovirus/complicaciones , Citomegalovirus/aislamiento & purificación , Pérdida Auditiva Bilateral/epidemiología , Pérdida Auditiva Bilateral/virología , Preescolar , Femenino , Francia/epidemiología , Humanos , Lactante , Masculino , Reacción en Cadena de la Polimerasa , Prevalencia , Estudios Prospectivos
10.
Hum Mutat ; 33(6): 949-59, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22396310

RESUMEN

Centronuclear myopathy (CNM) is a genetically heterogeneous disorder associated with general skeletal muscle weakness, type I fiber predominance and atrophy, and abnormally centralized nuclei. Autosomal dominant CNM is due to mutations in the large GTPase dynamin 2 (DNM2), a mechanochemical enzyme regulating cytoskeleton and membrane trafficking in cells. To date, 40 families with CNM-related DNM2 mutations have been described, and here we report 60 additional families encompassing a broad genotypic and phenotypic spectrum. In total, 18 different mutations are reported in 100 families and our cohort harbors nine known and four new mutations, including the first splice-site mutation. Genotype-phenotype correlation hypotheses are drawn from the published and new data, and allow an efficient screening strategy for molecular diagnosis. In addition to CNM, dissimilar DNM2 mutations are associated with Charcot-Marie-Tooth (CMT) peripheral neuropathy (CMTD1B and CMT2M), suggesting a tissue-specific impact of the mutations. In this study, we discuss the possible clinical overlap of CNM and CMT, and the biological significance of the respective mutations based on the known functions of dynamin 2 and its protein structure. Defects in membrane trafficking due to DNM2 mutations potentially represent a common pathological mechanism in CNM and CMT.


Asunto(s)
Dinamina II/genética , Genes Dominantes , Estudios de Asociación Genética , Mutación , Miopatías Estructurales Congénitas/genética , Secuencia de Aminoácidos , Dinamina II/química , Humanos , Datos de Secuencia Molecular , Miopatías Estructurales Congénitas/diagnóstico , Polimorfismo Genético , Alineación de Secuencia
11.
J Med Genet ; 48(6): 417-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21415077

RESUMEN

BACKGROUND: Geleophysic dysplasia (GD, OMIM 231050) is an autosomal recessive disorder characterised by short stature, small hands and feet, stiff joints, and thick skin. Patients often present with a progressive cardiac valvular disease which can lead to an early death. In a previous study including six GD families, we have mapped the disease gene on chromosome 9q34.2 and identified mutations in the A Disintegrin And Metalloproteinase with Thrombospondin repeats-like 2 gene (ADAMTSL2). METHODS: Following this study, we have collected the samples of 30 additional GD families, including 33 patients and identified ADAMTSL2 mutations in 14/33 patients, comprising 13 novel mutations. The absence of mutation in 19 patients prompted us to compare the two groups of GD patients, namely group 1, patients with ADAMTSL2 mutations (n=20, also including the 6 patients from our previous study), and group 2, patients without ADAMTSL2 mutations (n=19). RESULTS: The main discriminating features were facial dysmorphism and tip-toe walking, which were almost constantly observed in group 1. No differences were found concerning heart involvement, skin thickness, recurrent respiratory and ear infections, bronchopulmonary insufficiency, laryngo-tracheal stenosis, deafness, and radiographic features. CONCLUSIONS: It is concluded that GD is a genetically heterogeneous condition. Ongoing studies will hopefully lead to the identification of another disease gene.


Asunto(s)
Enanismo/genética , Proteínas de la Matriz Extracelular , Anomalías del Ojo/genética , Anomalías Cutáneas/genética , Adolescente , Adulto , Enfermedades del Desarrollo Óseo , Niño , Preescolar , Tejido Conectivo/anomalías , Tejido Conectivo/patología , Tejido Conectivo/fisiopatología , Enanismo/etnología , Enanismo/fisiopatología , Europa (Continente)/epidemiología , Proteínas de la Matriz Extracelular/genética , Anomalías del Ojo/etnología , Anomalías del Ojo/fisiopatología , Femenino , Heterogeneidad Genética , Humanos , Cuerpos de Inclusión/genética , Lactante , Japón/epidemiología , Deformidades Congénitas de las Extremidades , Masculino , Medio Oriente/epidemiología , Mutación , Linaje
12.
Am J Respir Crit Care Med ; 181(8): 851-61, 2010 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-20056902

RESUMEN

RATIONALE: Activin A receptor type II-like kinase-1 (ACVRL1, also known as ALK1) mutation is a cause of hereditary hemorrhagic telangiectasia (HHT) and/or heritable pulmonary arterial hypertension (PAH). OBJECTIVES: To describe the characteristics of patients with PAH carrying an ACVRL1 mutation. METHODS: We reviewed clinical, functional, and hemodynamic characteristics of 32 patients with PAH carrying an ACVRL1 mutation, corresponding to 9 patients from the French PAH Network and 23 from literature analysis. These cases were compared with 370 patients from the French PAH Network (93 with a bone morphogenetic protein receptor type 2 [BMPR2] mutation and 277 considered as idiopathic cases without identified mutation). Distribution of mutations in the ACVRL1 gene in patients with PAH was compared with the HHT Mutation Database. MEASUREMENTS AND MAIN RESULTS: At diagnosis, ACVRL1 mutation carriers were significantly younger (21.8 +/- 16.7 yr) than BMPR2 mutation carriers and noncarriers (35.7 +/- 14.9 and 47.6 +/- 16.3 yr, respectively; P < 0.0001). In seven of the nine patients from the French PAH Network, PAH diagnosis preceded manifestations of HHT. ACVRL1 mutation carriers had better hemodynamic status at diagnosis, but none responded to acute vasodilator challenge and they had shorter survival when compared with other patients with PAH despite similar use of specific therapies. ACVRL1 mutations in exon 10 were more frequently observed in patients with PAH, as compared with what was observed in the HHT Mutation Database (33.3 vs. 5%; P < 0.0001). CONCLUSIONS: ACVRL1 mutation carriers were characterized by a younger age at PAH diagnosis. Despite less severe initial hemodynamics and similar management, these patients had worse prognosis compared with other patients with PAH, suggesting more rapid disease progression.


Asunto(s)
Receptores de Activinas Tipo II/genética , Hipertensión Pulmonar/genética , Adolescente , Adulto , Distribución por Edad , Presión Sanguínea , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Niño , Preescolar , Prueba de Esfuerzo/métodos , Prueba de Esfuerzo/estadística & datos numéricos , Femenino , Francia , Humanos , Hipertensión Pulmonar/fisiopatología , Lactante , Masculino , Persona de Mediana Edad , Oxígeno/sangre , Mutación Puntual/genética , Arteria Pulmonar , Presión Esfenoidal Pulmonar , Sistema de Registros , Eliminación de Secuencia/genética , Análisis de Supervivencia , Resistencia Vascular , Adulto Joven
13.
Eur J Med Genet ; 64(4): 104166, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33571694

RESUMEN

CHD3-related syndrome, also known as Snijders Blok-Campeau syndrome, is a rare developmental disorder described in 2018, caused by de novo pathogenic variants in the CHD3 gene. This syndrome is characterized by global developmental delay, speech delay, intellectual disability, hypotonia and behavioral disorders including autism spectrum disorder (ASD). Typical dysmorphic features include macrocephaly, hypertelorism, enophthalmia, sparse eyebrows, bulging forehead, midface hypoplasia, prominent nose and pointed chin. To our knowledge, there have been no other clinical descriptions of patients since the initial publication. We report the clinical description of a 21-year-old patient harboring a pathogenic de novo variant in CHD3. We reviewed the clinical features of the 35 previously reported patients. Main features were severe intellectual disability, dysmorphic facies, macrocephaly, cryptorchidism, pectus carinatum, severe ophthalmologic abnormalities and behavioral disorders including ASD, and a frank happy demeanor. Hypersociability, which was a noticeable clinical feature in our case, despite ASD, is an uncommon behavioral feature in syndromic intellectual disabilities. Our report supports hypersociability as a suggestive feature of CHD3-related syndrome along with developmental delay, macrocephaly and a dysmorphic facies.


Asunto(s)
Anomalías Craneofaciales/genética , ADN Helicasas/genética , Discapacidades del Desarrollo/genética , Megalencefalia/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Conducta Social , Anomalías Craneofaciales/patología , Discapacidades del Desarrollo/patología , Humanos , Masculino , Megalencefalia/patología , Mutación , Síndrome , Adulto Joven
14.
Hum Mutat ; 31(1): E1021-42, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19802897

RESUMEN

In six index cases/families referred for Marfan syndrome (MFS) molecular diagnosis, we identified six novel mutations in the FBN1 gene: c.1753G>C (p.Gly585Arg), c.2456G>A (p.Gly819Glu), c.4981G>A (p.Gly1661Arg), c.5339G>A (p.Gly1780Glu), c.6418G>A (p.Gly2140Arg) and c.6419G>A (p.Gly2140Glu). These variants, predicted to result in Glycine substitutions are located at the third position of a 4 amino acids loop-region of calcium-binding Epidermal Growth Factor-like (cb-EGF) fibrillin-1 domains 5, 9, 24, 25 and 32. Familial segregation studies showing cosegregation with MFS manifestations or de novo inheritance in addition to in silico analyses (conservation, 3D modeling) suggest evidence for a crucial role of the respective Glycine positions. Extending these analyses to all Glycine residue at position 3 of this 4 residues loop in fibrillin-1 cb-EGF with the UMD predictor tool and alignment of 2038 available related sequences strongly support a steric strain that only allows Glycine or even Alanine residues for domain structure maintenance and for the fibrillin functions. Our data compared with those of the literature strongly suggest the existence of a cb-EGF domain subtype with implications for related diseases.


Asunto(s)
Calcio/metabolismo , Factor de Crecimiento Epidérmico , Síndrome de Marfan/genética , Proteínas de Microfilamentos/genética , Mutación Missense , Adolescente , Adulto , Anciano , Niño , Factor de Crecimiento Epidérmico/química , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Fibrilina-1 , Fibrilinas , Glicina/química , Glicina/genética , Humanos , Masculino , Síndrome de Marfan/diagnóstico , Proteínas de Microfilamentos/química , Persona de Mediana Edad , Modelos Moleculares , Linaje , Análisis de Secuencia de ADN , Adulto Joven
15.
Hum Mutat ; 31(8): 961-5, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20524212

RESUMEN

Type I hyperprolinemia (HPI) is an autosomal recessive disorder associated with cognitive and psychiatric troubles, caused by alterations of the Proline Dehydrogenase gene (PRODH) at 22q11. HPI results from PRODH deletion and/or missense mutations reducing proline oxidase (POX) activity. The goals of this study were first to measure in controls the frequency of PRODH variations described in HPI patients, second to assess the functional effect of PRODH mutations on POX activity, and finally to establish genotype/enzymatic activity correlations in a new series of HPI patients. Eight of 14 variants occurred at polymorphic frequency in 114 controls. POX activity was determined for six novel mutations and two haplotypes. The c.1331G>A, p.G444D allele has a drastic effect, whereas the c.23C>T, p.P8L allele and the c.[56C>A; 172G>A], p.[Q19P; A58T] haplotype result in a moderate decrease in activity. Among the 19 HPI patients, 10 had a predicted residual activity <50%. Eight out of nine subjects with a predicted residual activity > or = 50% bore at least one c.824C>A, p.T275N allele, which has no detrimental effect on activity but whose frequency in controls is only 3%. Our results suggest that PRODH mutations lead to a decreased POX activity or affect other biological parameters causing hyperprolinemia.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Estudios de Asociación Genética , Prolina/metabolismo , Adolescente , Adulto , Alelos , Errores Innatos del Metabolismo de los Aminoácidos/enzimología , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Mutación Missense/genética , Prolina Oxidasa/genética
16.
Transl Psychiatry ; 10(1): 77, 2020 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-32094338

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder with a strong genetic component whose knowledge evolves quickly. Next-generation sequencing is the only effective technology to deal with the high genetic heterogeneity of ASD in a clinical setting. However, rigorous criteria to classify rare genetic variants conferring ASD susceptibility are currently lacking. We have performed whole-exome sequencing to identify both nucleotide variants and copy number variants (CNVs) in 253 ASD patients, including 68 patients with intellectual disability (ID) and 90 diagnosed as Asperger syndrome. Using explicit criteria to classify both susceptibility genes and susceptibility variants we prioritized 217 genes belonging to the following categories: syndromic genes, genes with an excess of de novo protein truncating variants and genes targeted by rare CNVs. We obtained a susceptibility variant detection rate of 19.7% (95% CI: [15-25.2%]). The rate for CNVs was 7.1% (95% CI: [4.3-11%]) and 12.6% (95% CI: [8.8-17.4%]) for nucleotide variants. The highest rate (30.1%, 95% CI: [20.2-43.2%]) was obtained in the ASD + ID subgroup. A strong contributor for at risk nucleotide variants was the recently identified set of genes (n = 81) harboring an excess of de novo protein truncating variants. Since there is currently no evidence that the genes targeted here are necessary and sufficient to cause ASD, we recommend to avoid the term "causative of ASD" when delivering the information about a variant to a family and to use instead the term "genetic susceptibility factor contributing to ASD".


Asunto(s)
Trastorno del Espectro Autista , Trastorno del Espectro Autista/genética , Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Secuenciación del Exoma
17.
Am J Med Genet A ; 149A(6): 1108-15, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19449422

RESUMEN

An hypothalamic hamartoma is an abnormal mass of mature glio-neuronal tissue present in the hypothalamic area. It usually measures <2 cm of diameter. Most of the time, this hamartoma occurs in Pallister-Hall syndrome (PHS), due to heterozygous GLI3 mutations. We report on five fetuses with giant diencephalic hamartoma and other midline brain and facial malformations, without mutation in the GLI3 gene or genomic rearrangements in three of them. The fetuses showed facial asymmetry, unilateral ear and eye anomalies, and facial cleft. Extracephalic malformations consisted of vertebral anomalies and short nails, without polydactyly and cardiac malformation. The diencephalon was replaced by an encephaloid mass protruding into the facial cleft. Normal cerebral structures were not detectable. In one patient, holoprosencephaly of the syntelencephalic type was noted. Arhinencephaly was present in all patients. Histologically, the ill-defined, multilobulated lesion was made of neuroblastic and neurocytic cell foci, lying in a fibrillar network, elaborating sometimes perivascular pseudorosettes, with a maturation gradient in accordance with the fetal age. Owing to their location, the tumors could be described as diencephalic, rather than hypothalamic hamartomas. The striking asymmetry of the facial anomalies and the diencephalic malformations are not in the spectrum observed with PHS and related syndromes, suggesting a distinct entity involving abnormal morphogenetic developmental fields at around 5 weeks of gestation.


Asunto(s)
Anomalías Múltiples/genética , Diencéfalo/patología , Hamartoma/genética , Síndrome de Pallister-Hall , Aborto Inducido , Adulto , Facies , Femenino , Edad Gestacional , Humanos , Embarazo , Síndrome , Ultrasonografía Prenatal
18.
Hum Mutat ; 28(11): 1098-107, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17565729

RESUMEN

Sotos syndrome is an overgrowth syndrome characterized by distinctive facial features, learning difficulties, and macrocephaly with frequent pre- and postnatal overgrowth with advanced bone age. Here, we report on our experience in the molecular diagnostic of Sotos syndrome on 116 patients. Using direct sequencing and a quantitative multiplex PCR of short fluorescent fragments (QMPSF)-based assay allowing accurate detection of both total and partial NSD1 deletions, we identified NSD1 abnormalities in 104 patients corresponding to 102 Sotos families (90%). NSD1 point mutations were detected in 80% of the index cases, large deletions removing the NSD1 gene entirely in 14%, and intragenic NSD1 rearrangements in 6%. Among the 69 detected distinct point mutations, 48 were novel. The QMPSF assay detected an exonic duplication and a mosaic partial deletion. QMPSF mapping of the 15 large deletions revealed the heterogeneity of the deletions, which vary in size from 1 to 4.5 Mb. Clinical features of NSD1-positive Sotos patients revealed that the phenotype in patients with nontruncating mutations was less severe that in patients with truncating mutations. This study confirms the heterogeneity of NSD1 alterations in Sotos syndrome and therefore the need to complete sequencing analysis by screening for partial deletions and duplications to ensure an accurate molecular diagnosis of this syndrome.


Asunto(s)
Anomalías Múltiples/genética , Heterogeneidad Genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Recién Nacido , Masculino , Reacción en Cadena de la Polimerasa , Síndrome
19.
Eur J Hum Genet ; 15(11): 1145-55, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17637808

RESUMEN

Mitochondrial DNA (mtDNA) mutations have been implicated in non-syndromic hearing loss either as primary or as predisposing factors. As only a part of the mitochondrial genome is usually explored in deafness, its prevalence is probably under-estimated. Among 1350 families with non-syndromic sensorineural hearing loss collected through a French collaborative network, we selected 29 large families with a clear maternal lineage and screened them for known mtDNA mutations in 12S rRNA, tRNASer(UCN) and tRNALeu(UUR) genes. When no mutation could be identified, a whole mitochondrial genome screening was performed, using a microarray resequencing chip: the MitoChip version 2.0 developed by Affymetrix Inc. Known mtDNA mutations was found in nine of the 29 families, which are described in the article: five with A1555G, two with the T7511C, one with 7472insC and one with A3243G mutation. In the remaining 20 families, the resequencing Mitochip detected 258 mitochondrial homoplasmic variants and 107 potentially heteroplasmic variants. Controls were made by direct sequencing on selected fragments and showed a high sensibility of the MitoChip but a low specificity, especially for heteroplasmic variations. An original analysis on the basis of species conservation, frequency and phylogenetic investigation was performed to select the more probably pathogenic variants. The entire genome analysis allowed us to identify five additional families with a putatively pathogenic mitochondrial variant: T669C, C1537T, G8078A, G12236A and G15077A. These results indicate that the new MitoChip platform is a rapid and valuable tool for identification of new mtDNA mutations in deafness.


Asunto(s)
ADN Mitocondrial/genética , Genoma Mitocondrial , Pérdida Auditiva Sensorineural/genética , Mitocondrias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , Adulto , Animales , Bovinos , Niño , Perros , Electroforesis en Gel Bidimensional/instrumentación , Electroforesis en Gel Bidimensional/métodos , Femenino , Humanos , Masculino , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/instrumentación , Linaje , Mutación Puntual , Ratas , Análisis de Secuencia de ADN/instrumentación
20.
Eur J Hum Genet ; 14(9): 1009-17, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16773131

RESUMEN

In contrast to the numerous well-known microdeletion syndromes, only a few microduplications have been described, and this discrepancy may be due in part to methodological bias. In order to facilitate the detection of genomic microdeletions and microduplications, we developed a new assay based on QMPSF (Quantitative Multiplex PCR of Short fluorescent Fragments) able to explore simultaneously 12 candidate loci involved in mental retardation (MR) and known to be the target of genomic rearrangements. We first screened 153 patients with MR and facial dysmorphism associated with malformations, or growth anomalies, or familial history, with cytogenetically normal chromosomes, and the absence of FRAXA mutation and subtelomeric rearrangements. In this series, we found a 5q35 deletion removing the NSD1 gene in a patient with severe epilepsy, profound MR and, retrospectively, craniofacial features of Sotos syndrome. In a second series, we screened 140 patients with MR and behaviour disturbance who did not fulfil the de Vries criteria for subtelomeric rearrangements and who had a normal karyotype and no detectable FRAXA mutation. We detected a 22q11 deletion in a patient with moderate MR, obesity, and facial dysmorphism and a 4 Mb 17p11 duplication in a patient with moderate MR, behaviour disturbance, strabismus, and aspecific facial features. This new QMPSF assay can be gradually upgraded to include additional loci involved in newly recognised microduplication/microdeletion syndromes, and should facilitate wide screenings of patients with idiopathic MR and provide better estimates of the microduplication frequency in the MR population.


Asunto(s)
Deleción Cromosómica , Duplicación de Gen , Discapacidad Intelectual/genética , Reacción en Cadena de la Polimerasa/métodos , Preescolar , Aberraciones Cromosómicas , Cromosomas Humanos Par 17 , Femenino , Síndrome del Cromosoma X Frágil/genética , Reordenamiento Génico , Genoma Humano , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Proteínas Nucleares/genética , Reproducibilidad de los Resultados , Telómero/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA