Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 51(W1): W39-W45, 2023 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-37216590

RESUMEN

Much of the human genetics variant repertoire is composed of single nucleotide variants (SNV) and small insertion/deletions (indel) but structural variants (SV) remain a major part of our modified DNA. SV detection has often been a complex question to answer either because of the necessity to use different technologies (array CGH, SNP array, Karyotype, Optical Genome Mapping…) to detect each category of SV or to get an appropriate resolution (Whole Genome Sequencing). Thanks to the deluge of pangenomic analysis, Human geneticists are accumulating SV and their interpretation remains time consuming and challenging. The AnnotSV webserver (https://www.lbgi.fr/AnnotSV/) aims at being an efficient tool to (i) annotate and interpret SV potential pathogenicity in the context of human diseases, (ii) recognize potential false positive variants from all the SV identified and (iii) visualize the patient variants repertoire. The most recent developments in the AnnotSV webserver are: (i) updated annotations sources and ranking, (ii) three novel output formats to allow diverse utilization (analysis, pipelines), as well as (iii) two novel user interfaces including an interactive circos view.


Asunto(s)
Mutación INDEL , Polimorfismo de Nucleótido Simple , Programas Informáticos , Humanos , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Mapeo Restrictivo , Análisis de Secuencia de ADN , Secuenciación Completa del Genoma , Enfermedad/genética
2.
Am J Med Genet A ; 194(4): e63476, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37974505

RESUMEN

Cat Eye Syndrome (CES) is a rare genetic disease caused by the presence of a small supernumerary marker chromosome derived from chromosome 22, which results in a partial tetrasomy of 22p-22q11.21. CES is classically defined by association of iris coloboma, anal atresia, and preauricular tags or pits, with high clinical and genetic heterogeneity. We conducted an international retrospective study of patients carrying genomic gain in the 22q11.21 chromosomal region upstream from LCR22-A identified using FISH, MLPA, and/or array-CGH. We report a cohort of 43 CES cases. We highlight that the clinical triad represents no more than 50% of cases. However, only 16% of CES patients presented with the three signs of the triad and 9% not present any of these three signs. We also highlight the importance of other impairments: cardiac anomalies are one of the major signs of CES (51% of cases), and high frequency of intellectual disability (47%). Ocular motility defects (45%), abdominal malformations (44%), ophthalmologic malformations (35%), and genitourinary tract defects (32%) are other frequent clinical features. We observed that sSMC is the most frequent chromosomal anomaly (91%) and we highlight the high prevalence of mosaic cases (40%) and the unexpectedly high prevalence of parental transmission of sSMC (23%). Most often, the transmitting parent has mild or absent features and carries the mosaic marker at a very low rate (<10%). These data allow us to better delineate the clinical phenotype associated with CES, which must be taken into account in the cytogenetic testing for this syndrome. These findings draw attention to the need for genetic counseling and the risk of recurrence.


Asunto(s)
Aneuploidia , Trastornos de los Cromosomas , Cromosomas Humanos Par 22 , Anomalías del Ojo , Cardiopatías Congénitas , Humanos , Estudios Retrospectivos , Hibridación Fluorescente in Situ , Cromosomas Humanos Par 22/genética , Cardiopatías Congénitas/genética
3.
Am J Med Genet A ; 191(1): 52-63, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36196855

RESUMEN

A small but growing body of scientific literature is emerging about clinical findings in patients with 19p13.3 microdeletion or duplication. Recently, a proximal 19p13.3 microduplication syndrome was described, associated with growth delay, microcephaly, psychomotor delay and dysmorphic features. The aim of our study was to better characterize the syndrome associated with duplications in the proximal 19p13.3 region (prox 19p13.3 dup), and to propose a comprehensive analysis of the underlying genomic mechanism. We report the largest cohort of patients with prox 19p13.3 dup through a collaborative study. We collected 24 new patients with terminal or interstitial 19p13.3 duplication characterized by array-based Comparative Genomic Hybridization (aCGH). We performed mapping, phenotype-genotype correlations analysis, critical region delineation and explored three-dimensional chromatin interactions by analyzing Topologically Associating Domains (TADs). We define a new 377 kb critical region (CR 1) in chr19: 3,116,922-3,494,377, GRCh37, different from the previously described critical region (CR 2). The new 377 kb CR 1 includes a TAD boundary and two enhancers whose common target is PIAS4. We hypothesize that duplications of CR 1 are responsible for tridimensional structural abnormalities by TAD disruption and misregulation of genes essentials for the control of head circumference during development, by breaking down the interactions between enhancers and the corresponding targeted gene.


Asunto(s)
Anomalías Múltiples , Microcefalia , Humanos , Hibridación Genómica Comparativa , Anomalías Múltiples/genética , Microcefalia/genética , Síndrome , Estudios de Asociación Genética
4.
Nucleic Acids Res ; 49(W1): W21-W28, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34023905

RESUMEN

With the dramatic increase of pangenomic analysis, Human geneticists have generated large amount of genomic data including millions of small variants (SNV/indel) but also thousands of structural variations (SV) mainly from next-generation sequencing and array-based techniques. While the identification of the complete SV repertoire of a patient is getting possible, the interpretation of each SV remains challenging. To help identifying human pathogenic SV, we have developed a web server dedicated to their annotation and ranking (AnnotSV) as well as their visualization and interpretation (knotAnnotSV) freely available at the following address: https://www.lbgi.fr/AnnotSV/. A large amount of annotations from >20 sources is integrated in our web server including among others genes, haploinsufficiency, triplosensitivity, regulatory elements, known pathogenic or benign genomic regions, phenotypic data. An ACMG/ClinGen compliant prioritization module allows the scoring and the ranking of SV into 5 SV classes from pathogenic to benign. Finally, the visualization interface displays the annotated SV in an interactive way including popups, search fields, filtering options, advanced colouring to highlight pathogenic SV and hyperlinks to the UCSC genome browser or other public databases. This web server is designed for diagnostic and research analysis by providing important resources to the user.


Asunto(s)
Variación Estructural del Genoma , Programas Informáticos , Genoma Humano , Genómica , Humanos , Internet , Anotación de Secuencia Molecular , Fenotipo , Polimorfismo de Nucleótido Simple
5.
Int J Mol Sci ; 24(10)2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37240074

RESUMEN

Bardet-Biedl syndrome (BBS) is an autosomal recessive ciliopathy that affects multiple organs, leading to retinitis pigmentosa, polydactyly, obesity, renal anomalies, cognitive impairment, and hypogonadism. Until now, biallelic pathogenic variants have been identified in at least 24 genes delineating the genetic heterogeneity of BBS. Among those, BBS5 is a minor contributor to the mutation load and is one of the eight subunits forming the BBSome, a protein complex implied in protein trafficking within the cilia. This study reports on a European BBS5 patient with a severe BBS phenotype. Genetic analysis was performed using multiple next-generation sequencing (NGS) tests (targeted exome, TES and whole exome, WES), and biallelic pathogenic variants could only be identified using whole-genome sequencing (WGS), including a previously missed large deletion of the first exons. Despite the absence of family samples, the biallelic status of the variants was confirmed. The BBS5 protein's impact was confirmed on the patient's cells (presence/absence and size of the cilium) and ciliary function (Sonic Hedgehog pathway). This study highlights the importance of WGS and the challenge of reliable structural variant detection in patients' genetic explorations as well as functional tests to assess a variant's pathogenicity.


Asunto(s)
Síndrome de Bardet-Biedl , Polidactilia , Humanos , Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/patología , Proteínas del Citoesqueleto/genética , Proteínas Hedgehog/genética , Mutación , Fenotipo , Proteínas de Unión a Fosfato/genética , Transporte de Proteínas , Masculino , Preescolar
6.
Hum Mutat ; 43(3): 347-361, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35005812

RESUMEN

We report the screening of a large panel of genes in a series of 100 fetuses (98 families) affected with severe renal defects. Causative variants were identified in 22% of cases, greatly improving genetic counseling. The percentage of variants explaining the phenotype was different according to the type of phenotype. The highest diagnostic yield was found in cases affected with the ciliopathy-like phenotype (11/15 families and, in addition, a single heterozygous or a homozygous Class 3 variant in PKHD1 in three unrelated cases with autosomal recessive polycystic kidney disease). The lowest diagnostic yield was observed in cases with congenital anomalies of the kidney and urinary tract (9/78 families and, in addition, Class 3 variants in GREB1L in three unrelated cases with bilateral renal agenesis). Inheritance was autosomal recessive in nine genes (PKHD1, NPHP3, CEP290, TMEM67, DNAJB11, FRAS1, ACE, AGT, and AGTR1), and autosomal dominant in six genes (PKD1, PKD2, PAX2, EYA1, BICC1, and MYOCD). Finally, we developed an original approach of next-generation sequencing targeted RNA sequencing using the custom capture panel used for the sequencing of DNA, to validate one MYOCD heterozygous splicing variant identified in two male siblings with megabladder and inherited from their healthy mother.


Asunto(s)
Enfermedades Renales , Riñón Poliquístico Autosómico Dominante , Antígenos de Neoplasias , Proteínas de Ciclo Celular/genética , Proteínas del Citoesqueleto/genética , Femenino , Feto/anomalías , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Riñón/anomalías , Enfermedades Renales/congénito , Enfermedades Renales/diagnóstico , Enfermedades Renales/genética , Masculino , Mutación , Riñón Poliquístico Autosómico Dominante/genética
7.
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
8.
Hum Mutat ; 41(1): 240-254, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31549751

RESUMEN

Polydactyly is one of the most frequent inherited defects of the limbs characterized by supernumerary digits and high-genetic heterogeneity. Among the many genes involved, either in isolated or syndromic forms, eight have been implicated in postaxial polydactyly (PAP). Among those, IQCE has been recently identified in a single consanguineous family. Using whole-exome sequencing in patients with uncharacterized ciliopathies, including PAP, we identified three families with biallelic pathogenic variations in IQCE. Interestingly, the c.895_904del (p.Val301Serfs*8) was found in all families without sharing a common haplotype, suggesting a recurrent mechanism. Moreover, in two families, the systemic phenotype could be explained by additional pathogenic variants in known genes (TULP1, ATP6V1B1). RNA expression analysis on patients' fibroblasts confirms that the dysfunction of IQCE leads to the dysregulation of genes associated with the hedgehog-signaling pathway, and zebrafish experiments demonstrate a full spectrum of phenotypes linked to defective cilia: Body curvature, kidney cysts, left-right asymmetry, misdirected cilia in the pronephric duct, and retinal defects. In conclusion, we identified three additional families confirming IQCE as a nonsyndromic PAP gene. Our data emphasize the importance of taking into account the complete set of variations of each individual, as each clinical presentation could finally be explained by multiple genes.


Asunto(s)
Ciliopatías/diagnóstico , Ciliopatías/genética , Dedos/anomalías , Predisposición Genética a la Enfermedad , Variación Genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Fenotipo , Polidactilia/diagnóstico , Polidactilia/genética , Dedos del Pie/anomalías , Animales , Consanguinidad , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Estudios de Asociación Genética/métodos , Homocigoto , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Linaje , Transducción de Señal , Transcriptoma , Secuenciación del Exoma , Pez Cebra
9.
Hum Mutat ; 41(12): 2167-2178, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131162

RESUMEN

Herein, we report the screening of a large panel of genes in a series of 80 fetuses with congenital heart defects (CHDs) and/or heterotaxy and no cytogenetic anomalies. There were 49 males (61%/39%), with a family history in 28 cases (35%) and no parental consanguinity in 77 cases (96%). All fetuses had complex CHD except one who had heterotaxy and midline anomalies while 52 cases (65%) had heterotaxy in addition to CHD. Altogether, 29 cases (36%) had extracardiac and extra-heterotaxy anomalies. A pathogenic variant was found in 10/80 (12.5%) cases with a higher percentage in the heterotaxy group (8/52 cases, 15%) compared with the non-heterotaxy group (2/28 cases, 7%), and in 3 cases with extracardiac and extra-heterotaxy anomalies (3/29, 10%). The inheritance was recessive in six genes (DNAI1, GDF1, MMP21, MYH6, NEK8, and ZIC3) and dominant in two genes (SHH and TAB2). A homozygous pathogenic variant was found in three cases including only one case with known consanguinity. In conclusion, after removing fetuses with cytogenetic anomalies, next-generation sequencing discovered a causal variant in 12.5% of fetal cases with CHD and/or heterotaxy. Genetic counseling for future pregnancies was greatly improved. Surprisingly, unexpected consanguinity accounts for 20% of cases with identified pathogenic variants.


Asunto(s)
Feto/anomalías , Cardiopatías Congénitas/genética , Síndrome de Heterotaxia/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Análisis Citogenético , Familia , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Mutación/genética , Linaje
10.
Clin Genet ; 98(2): 166-171, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32361989

RESUMEN

Bardet-Biedl syndrome (BBS) is a rare ciliopathy with variable retinal dystrophy, polydactyly, renal abnormalities, obesity, cognitive impairment, and hypogonadism. Biallelic pathogenic variants have been identified in 24 genes, leading to BBS in an autosomal recessive inheritance pattern. In this study, we investigated a cohort of 16 families (20 individuals) presenting with typical BBS originating from La Réunion Island using sequencing (Sanger and high-throughput methods) and SNP array. In eight families (12 individuals) we identified the same ARL6/BBS3 variation [c.535G > A, p.(Asp179Asn)]. Bioinformatics and functional analyses revealed an effect of this variant on the splicing of ARL6/BBS3. Owing to the relatively high frequency of this variant, a possible founder effect was suspected. Genotyping of six individuals revealed a common 3.8-Mb haplotype and estimated the most recent common ancestor to about eight generations confirmed by the known genealogy. Knowledge of this founder effect modifies our diagnostic strategy and enables a personalized genetic counseling for patients from La Réunion Island. Being the first description of BBS patients from La Réunion Island, we could estimate its prevalence between ~1/45000 and ~ 1/66000 individuals.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Síndrome de Bardet-Biedl/genética , Predisposición Genética a la Enfermedad , Polidactilia/genética , Adolescente , Alelos , Síndrome de Bardet-Biedl/fisiopatología , Niño , Preescolar , Femenino , Efecto Fundador , Genotipo , Haplotipos , Humanos , Masculino , Mutación , Linaje , Polidactilia/fisiopatología , Polimorfismo de Nucleótido Simple/genética
11.
Hum Mutat ; 40(10): 1826-1840, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31116475

RESUMEN

Mutations in genes encoding aminoacyl-tRNA synthetases have been reported in several neurological disorders. KARS is a dual localized lysyl-tRNA synthetase and its cytosolic isoform belongs to the multiple aminoacyl-tRNA synthetase complex (MSC). Biallelic mutations in the KARS gene were described in a wide phenotypic spectrum ranging from nonsyndromic deafness to complex impairments. Here, we report on a patient with severe neurological and neurosensory disease investigated by whole-exome sequencing and found to carry biallelic mutations c.683C>T (p.Pro228Leu) and c.871T>G (p.Phe291Val), the second one being novel, in the KARS gene. The patient presented with an atypical clinical presentation with an optic neuropathy not previously reported. At the cellular level, we show that cytoplasmic KARS was expressed at a lower level in patient cells and displayed decreased interaction with MSC. In vitro, these two KARS variants have a decreased aminoacylation activity compared with wild-type KARS, the p.Pro228Leu being the most affected. Our data suggest that dysfunction of cytoplasmic KARS resulted in a decreased level of translation of the nuclear-encoded lysine-rich proteins belonging to the respiratory chain complex, thus impairing mitochondria functions.


Asunto(s)
Aminoacil-ARNt Sintetasas/genética , Lisina-ARNt Ligasa/genética , Mutación , Enfermedades del Sistema Nervioso/complicaciones , Enfermedades del Sistema Nervioso/genética , Enfermedades del Nervio Óptico/complicaciones , Trastornos de la Sensación/complicaciones , Trastornos de la Sensación/genética , Alelos , Secuencia de Aminoácidos , Aminoacil-ARNt Sintetasas/química , Aminoacil-ARNt Sintetasas/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lisina-ARNt Ligasa/química , Lisina-ARNt Ligasa/metabolismo , Imagen por Resonancia Magnética , Modelos Moleculares , Enfermedades del Sistema Nervioso/diagnóstico , Enfermedades del Nervio Óptico/diagnóstico , Linaje , Unión Proteica , Conformación Proteica , Trastornos de la Sensación/diagnóstico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Brain ; 141(11): 3160-3178, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30351409

RESUMEN

Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control neuronal excitability and their dysfunction has been linked to epileptogenesis but few individuals with neurological disorders related to variants altering HCN channels have been reported so far. In 2014, we described five individuals with epileptic encephalopathy due to de novo HCN1 variants. To delineate HCN1-related disorders and investigate genotype-phenotype correlations further, we assembled a cohort of 33 unpublished patients with novel pathogenic or likely pathogenic variants: 19 probands carrying 14 different de novo mutations and four families with dominantly inherited variants segregating with epilepsy in 14 individuals, but not penetrant in six additional individuals. Sporadic patients had epilepsy with median onset at age 7 months and in 36% the first seizure occurred during a febrile illness. Overall, considering familial and sporadic patients, the predominant phenotypes were mild, including genetic generalized epilepsies and genetic epilepsy with febrile seizures plus (GEFS+) spectrum. About 20% manifested neonatal/infantile onset otherwise unclassified epileptic encephalopathy. The study also included eight patients with variants of unknown significance: one adopted patient had two HCN1 variants, four probands had intellectual disability without seizures, and three individuals had missense variants inherited from an asymptomatic parent. Of the 18 novel pathogenic missense variants identified, 12 were associated with severe phenotypes and clustered within or close to transmembrane domains, while variants segregating with milder phenotypes were located outside transmembrane domains, in the intracellular N- and C-terminal parts of the channel. Five recurrent variants were associated with similar phenotypes. Using whole-cell patch-clamp, we showed that the impact of 12 selected variants ranged from complete loss-of-function to significant shifts in activation kinetics and/or voltage dependence. Functional analysis of three different substitutions altering Gly391 revealed that these variants had different consequences on channel biophysical properties. The Gly391Asp variant, associated with the most severe, neonatal phenotype, also had the most severe impact on channel function. Molecular dynamics simulation on channel structure showed that homotetramers were not conducting ions because the permeation path was blocked by cation(s) strongly complexed to the Asp residue, whereas heterotetramers showed an instantaneous current component possibly linked to deformation of the channel pore. In conclusion, our results considerably expand the clinical spectrum related to HCN1 variants to include common generalized epilepsy phenotypes and further illustrate how HCN1 has a pivotal function in brain development and control of neuronal excitability.


Asunto(s)
Epilepsia Generalizada/genética , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Mutación/genética , Canales de Potasio/genética , Espasmos Infantiles/genética , Adolescente , Adulto , Anciano , Animales , Células CHO , Niño , Preescolar , Cricetulus , Estimulación Eléctrica , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Potenciales de la Membrana/genética , Persona de Mediana Edad , Modelos Moleculares , Mutagénesis Sitio-Dirigida/métodos , Adulto Joven
13.
Hum Mutat ; 39(7): 983-992, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29688594

RESUMEN

Ciliopathies represent a wide spectrum of rare diseases with overlapping phenotypes and a high genetic heterogeneity. Among those, IFT140 is implicated in a variety of phenotypes ranging from isolated retinis pigmentosa to more syndromic cases. Using whole-genome sequencing in patients with uncharacterized ciliopathies, we identified a novel recurrent tandem duplication of exon 27-30 (6.7 kb) in IFT140, c.3454-488_4182+2588dup p.(Tyr1152_Thr1394dup), missed by whole-exome sequencing. Pathogenicity of the mutation was assessed on the patients' skin fibroblasts. Several hundreds of patients with a ciliopathy phenotype were screened and biallelic mutations were identified in 11 families representing 12 pathogenic variants of which seven are novel. Among those unrelated families especially with a Mainzer-Saldino syndrome, eight carried the same tandem duplication (two at the homozygous state and six at the heterozygous state). In conclusion, we demonstrated the implication of structural variations in IFT140-related diseases expanding its mutation spectrum. We also provide evidences for a unique genomic event mediated by an Alu-Alu recombination occurring on a shared haplotype. We confirm that whole-genome sequencing can be instrumental in the ability to detect structural variants for genomic disorders.


Asunto(s)
Proteínas Portadoras/genética , Ataxia Cerebelosa/genética , Ciliopatías/genética , Retinitis Pigmentosa/genética , Secuenciación Completa del Genoma , Elementos Alu/genética , Ataxia Cerebelosa/patología , Ciliopatías/patología , Bases de Datos Genéticas , Exones/genética , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Mutación/genética , Linaje , Fenotipo , Retinitis Pigmentosa/patología
14.
Am J Hum Genet ; 96(4): 666-74, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25817018

RESUMEN

We have identified TUBGCP4 variants in individuals with autosomal-recessive microcephaly and chorioretinopathy. Whole-exome sequencing performed on one family with two affected siblings and independently on another family with one affected child revealed compound-heterozygous mutations in TUBGCP4. Subsequent Sanger sequencing was performed on a panel of individuals from 12 French families affected by microcephaly and ophthalmic manifestations, and one other individual was identified with compound-heterozygous mutations in TUBGCP4. One synonymous variant was common to all three families and was shown to induce exon skipping; the other mutations were frameshift mutations and a deletion. TUBGCP4 encodes γ-tubulin complex protein 4, a component belonging to the γ-tubulin ring complex (γ-TuRC) and known to regulate the nucleation and organization of microtubules. Functional analysis of individual fibroblasts disclosed reduced levels of the γ-TuRC, altered nucleation and organization of microtubules, abnormal nuclear shape, and aneuploidy. Moreover, zebrafish treated with morpholinos against tubgcp4 were found to have reduced head volume and eye developmental anomalies with chorioretinal dysplasia. In summary, the identification of TUBGCP4 mutations in individuals with microcephaly and a spectrum of anomalies in eye development, particularly photoreceptor anomalies, provides evidence of an important role for the γ-TuRC in brain and eye development.


Asunto(s)
Enfermedades de la Coroides/genética , Enfermedades Hereditarias del Ojo/genética , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/genética , Microtúbulos/genética , Enfermedades de la Retina/genética , Tubulina (Proteína)/metabolismo , Secuencia de Bases , Exoma/genética , Mutación del Sistema de Lectura/genética , Francia , Componentes del Gen , Humanos , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Linaje , Análisis de Secuencia de ADN
15.
Hum Genet ; 136(4): 463-479, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28283832

RESUMEN

Subtelomeric 1q43q44 microdeletions cause a syndrome associating intellectual disability, microcephaly, seizures and anomalies of the corpus callosum. Despite several previous studies assessing genotype-phenotype correlations, the contribution of genes located in this region to the specific features of this syndrome remains uncertain. Among those, three genes, AKT3, HNRNPU and ZBTB18 are highly expressed in the brain and point mutations in these genes have been recently identified in children with neurodevelopmental phenotypes. In this study, we report the clinical and molecular data from 17 patients with 1q43q44 microdeletions, four with ZBTB18 mutations and seven with HNRNPU mutations, and review additional data from 37 previously published patients with 1q43q44 microdeletions. We compare clinical data of patients with 1q43q44 microdeletions with those of patients with point mutations in HNRNPU and ZBTB18 to assess the contribution of each gene as well as the possibility of epistasis between genes. Our study demonstrates that AKT3 haploinsufficiency is the main driver for microcephaly, whereas HNRNPU alteration mostly drives epilepsy and determines the degree of intellectual disability. ZBTB18 deletions or mutations are associated with variable corpus callosum anomalies with an incomplete penetrance. ZBTB18 may also contribute to microcephaly and HNRNPU to thin corpus callosum, but with a lower penetrance. Co-deletion of contiguous genes has additive effects. Our results confirm and refine the complex genotype-phenotype correlations existing in the 1qter microdeletion syndrome and define more precisely the neurodevelopmental phenotypes associated with genetic alterations of AKT3, ZBTB18 and HNRNPU in humans.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 1 , Ribonucleoproteínas Nucleares Heterogéneas/genética , Mutación , Trastornos del Neurodesarrollo/genética , Fenotipo , Proteínas Represoras/genética , Humanos
16.
Acta Neuropathol ; 134(6): 889-904, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28685322

RESUMEN

X-linked myotubular myopathy (XLMTM), a severe congenital myopathy, is caused by mutations in the MTM1 gene located on the X chromosome. A majority of affected males die in the early postnatal period, whereas female carriers are believed to be usually asymptomatic. Nevertheless, several affected females have been reported. To assess the phenotypic and pathological spectra of carrier females and to delineate diagnostic clues, we characterized 17 new unrelated affected females and performed a detailed comparison with previously reported cases at the clinical, muscle imaging, histological, ultrastructural and molecular levels. Taken together, the analysis of this large cohort of 43 cases highlights a wide spectrum of clinical severity ranging from severe neonatal and generalized weakness, similar to XLMTM male, to milder adult forms. Several females show a decline in respiratory function. Asymmetric weakness is a noteworthy frequent specific feature potentially correlated to an increased prevalence of highly skewed X inactivation. Asymmetry of growth was also noted. Other diagnostic clues include facial weakness, ptosis and ophthalmoplegia, skeletal and joint abnormalities, and histopathological signs that are hallmarks of centronuclear myopathy such as centralized nuclei and necklace fibers. The histopathological findings also demonstrate a general disorganization of muscle structure in addition to these specific hallmarks. Thus, MTM1 mutations in carrier females define a specific myopathy, which may be independent of the presence of an XLMTM male in the family. As several of the reported affected females carry large heterozygous MTM1 deletions not detectable by Sanger sequencing, and as milder phenotypes present as adult-onset limb-girdle myopathy, the prevalence of this myopathy is likely to be greatly underestimated. This report should aid diagnosis and thus the clinical management and genetic counseling of MTM1 carrier females. Furthermore, the clinical and pathological history of this cohort may be useful for therapeutic projects in males with XLMTM, as it illustrates the spectrum of possible evolution of the disease in patients surviving long term.


Asunto(s)
Heterocigoto , Mutación , Miopatías Estructurales Congénitas/diagnóstico , Proteínas Tirosina Fosfatasas no Receptoras/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Diagnóstico Diferencial , Femenino , Humanos , Persona de Mediana Edad , Miopatías Estructurales Congénitas/genética , Miopatías Estructurales Congénitas/patología , Miopatías Estructurales Congénitas/fisiopatología , Fenotipo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Índice de Severidad de la Enfermedad
17.
Am J Med Genet A ; 173(2): 479-486, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27623003

RESUMEN

Focal dermal hypoplasia (FDH) is a rare syndrome characterized by pleiotropic features knowing to involve mostly skin and limbs. Although FDH has been described in children and adults, the cardinal signs of the fetal phenotype are not straightforward impacting the quality of the prenatal diagnosis. We describe in depth the ultrasound, radiological, macroscopical, and histological phenotype of three female fetuses with a severe form of FDH, propose a review of the literature and an attempt to delineate minimal and cardinal signs for FDH diagnosis. This report confirms the variability of FDH phenotype, highlights unreported FDH features, and allows delineating evocative clinical associations for prenatal diagnosis, namely intrauterine growth retardation, limbs malformations, anterior wall/diaphragm defects, and eye anomalies. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Hipoplasia Dérmica Focal/diagnóstico , Hipoplasia Dérmica Focal/genética , Aborto Inducido , Aciltransferasas/genética , Autopsia , Análisis Mutacional de ADN , Femenino , Feto/anomalías , Estudios de Asociación Genética , Pruebas Genéticas , Genotipo , Humanos , Masculino , Proteínas de la Membrana/genética , Mutación , Fenotipo , Diagnóstico Prenatal , Radiografía , Ultrasonografía Prenatal
18.
J Hum Genet ; 61(5): 447-50, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26763875

RESUMEN

Bardet-Biedl syndrome (BBS; MIM 209900) is a recessive heterogeneous ciliopathy characterized by retinitis pigmentosa (RP), postaxial polydactyly, obesity, hypogonadism, cognitive impairment and kidney dysfunction. So far, 20 BBS genes have been identified, with the last reported ones being found in one or very few families. Whole-exome sequencing was performed in a consanguineous family in which two affected children presented typical BBS features (retinitis pigmentosa, postaxial polydactyly, obesity, hypogonadism and cognitive impairment) without any mutation identified in known BBS genes at the time of the study. We identified a homozygous splice-site mutation (NM_015662.2: c.4428+3A>G) in both affected siblings in the last reported BBS gene, namely, Intraflagellar Transport 172 Homolog (IFT172). Familial mutation segregation was consistent with autosomal recessive inheritance. IFT172 mutations were initially reported in Jeune and Mainzer-Saldino syndromes. Recently, mutations have also been found in isolated RP and Bardet-Biedl-like ciliopathy. This is the second report of IFT172 mutations in BBS patients validating IFT172 as the twentieth BBS gene (BBS20). Moreover, another IFT gene, IFT27, was already associated with BBS, confirming the implication of IFT genes in the pathogenesis of BBS.


Asunto(s)
Síndrome de Bardet-Biedl/diagnóstico , Síndrome de Bardet-Biedl/genética , Proteínas Portadoras/genética , Mutación , Proteínas Adaptadoras Transductoras de Señales , Niño , Preescolar , Biología Computacional/métodos , Proteínas del Citoesqueleto , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Masculino , Linaje , Fenotipo , Esqueleto/diagnóstico por imagen , Esqueleto/patología
19.
J Med Genet ; 51(2): 132-6, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24026985

RESUMEN

BACKGROUND: Bardet-Biedl syndrome (BBS) is a recessive and genetically heterogeneous ciliopathy characterised by retinitis pigmentosa, obesity, kidney dysfunction, postaxial polydactyly, behavioural dysfunction and hypogonadism. 7 of the 17 BBS gene products identified to date assemble together with the protein BBIP1/BBIP10 into the BBSome, a protein complex that ferries signalling receptors to and from cilia. METHODS AND RESULTS: Exome sequencing performed on a sporadic BBS case revealed for the first time a homozygous stop mutation (NM_001195306: c.173T>G, p.Leu58*) in the BBIP1 gene. This mutation is pathogenic since no BBIP1 protein could be detected in fibroblasts from the patient, and BBIP1[Leu58*] is unable to associate with the BBSome subunit BBS4. CONCLUSIONS: These findings identify BBIP1 as the 18th BBS gene (BBS18) and suggest that BBSome assembly may represent a unifying pathomechanism for BBS.


Asunto(s)
Síndrome de Bardet-Biedl/genética , Proteínas Portadoras/genética , Codón sin Sentido , Exoma , Animales , Síndrome de Bardet-Biedl/metabolismo , Secuencia de Bases , Consanguinidad , Análisis Mutacional de ADN , Fibroblastos/metabolismo , Estudios de Asociación Genética , Ligamiento Genético , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA