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.
Am J Hum Genet ; 111(3): 509-528, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38412861

RESUMEN

Neurodevelopmental disorders (NDDs) result from impaired development and functioning of the brain. Here, we identify loss-of-function (LoF) variation in ZFHX3 as a cause for syndromic intellectual disability (ID). ZFHX3 is a zinc-finger homeodomain transcription factor involved in various biological processes, including cell differentiation and tumorigenesis. We describe 42 individuals with protein-truncating variants (PTVs) or (partial) deletions of ZFHX3, exhibiting variable intellectual disability and autism spectrum disorder, recurrent facial features, relative short stature, brachydactyly, and, rarely, cleft palate. ZFHX3 LoF associates with a specific methylation profile in whole blood extracted DNA. Nuclear abundance of ZFHX3 increases during human brain development and neuronal differentiation. ZFHX3 was found to interact with the chromatin remodeling BRG1/Brm-associated factor complex and the cleavage and polyadenylation complex, suggesting a function in chromatin remodeling and mRNA processing. Furthermore, ChIP-seq for ZFHX3 revealed that it predominantly binds promoters of genes involved in nervous system development. We conclude that loss-of-function variants in ZFHX3 are a cause of syndromic ID associating with a specific DNA methylation profile.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/complicaciones , Haploinsuficiencia/genética , Trastornos del Neurodesarrollo/genética , Encéfalo/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
2.
Genet Med ; 26(5): 101087, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38288683

RESUMEN

PURPOSE: Interneuronopathies are a group of neurodevelopmental disorders characterized by deficient migration and differentiation of gamma-aminobutyric acidergic interneurons resulting in a broad clinical spectrum, including autism spectrum disorders, early-onset epileptic encephalopathy, intellectual disability, and schizophrenic disorders. SP9 is a transcription factor belonging to the Krüppel-like factor and specificity protein family, the members of which harbor highly conserved DNA-binding domains. SP9 plays a central role in interneuron development and tangential migration, but it has not yet been implicated in a human neurodevelopmental disorder. METHODS: Cases with SP9 variants were collected through international data-sharing networks. To address the specific impact of SP9 variants, in silico and in vitro assays were carried out. RESULTS: De novo heterozygous variants in SP9 cause a novel form of interneuronopathy. SP9 missense variants affecting the glutamate 378 amino acid result in severe epileptic encephalopathy because of hypomorphic and neomorphic DNA-binding effects, whereas SP9 loss-of-function variants result in a milder phenotype with epilepsy, developmental delay, and autism spectrum disorder. CONCLUSION: De novo heterozygous SP9 variants are responsible for a neurodevelopmental disease. Interestingly, variants located in conserved DNA-binding domains of KLF/SP family transcription factors may lead to neomorphic DNA-binding functions resulting in a combination of loss- and gain-of-function effects.


Asunto(s)
Trastorno del Espectro Autista , Epilepsia , Discapacidad Intelectual , Interneuronas , Factores de Transcripción Sp , Factores de Transcripción , Adolescente , Niño , Preescolar , Femenino , Humanos , Masculino , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Epilepsia/genética , Epilepsia/patología , Heterocigoto , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Interneuronas/metabolismo , Interneuronas/patología , Mutación Missense/genética , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción Sp/genética
3.
Hum Genet ; 141(1): 65-80, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34748075

RESUMEN

Pathogenic variants of the myelin transcription factor-1 like (MYT1L) gene include heterozygous missense, truncating variants and 2p25.3 microdeletions and cause a syndromic neurodevelopmental disorder (OMIM#616,521). Despite enrichment in de novo mutations in several developmental disorders and autism studies, the data on clinical characteristics and genotype-phenotype correlations are scarce, with only 22 patients with single nucleotide pathogenic variants reported. We aimed to further characterize this disorder at both the clinical and molecular levels by gathering a large series of patients with MYT1L-associated neurodevelopmental disorder. We collected genetic information on 40 unreported patients with likely pathogenic/pathogenic MYT1L variants and performed a comprehensive review of published data (total = 62 patients). We confirm that the main phenotypic features of the MYT1L-related disorder are developmental delay with language delay (95%), intellectual disability (ID, 70%), overweight or obesity (58%), behavioral disorders (98%) and epilepsy (23%). We highlight novel clinical characteristics, such as learning disabilities without ID (30%) and feeding difficulties during infancy (18%). We further describe the varied dysmorphic features (67%) and present the changes in weight over time of 27 patients. We show that patients harboring highly clustered missense variants in the 2-3-ZNF domains are not clinically distinguishable from patients with truncating variants. We provide an updated overview of clinical and genetic data of the MYT1L-associated neurodevelopmental disorder, hence improving diagnosis and clinical management of these patients.


Asunto(s)
Variación Genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Preescolar , Epilepsia/genética , Trastornos de Alimentación y de la Ingestión de Alimentos/genética , Femenino , Estudios de Asociación Genética , Heterocigoto , Humanos , Lactante , Trastornos del Desarrollo del Lenguaje/genética , Masculino , Obesidad/genética , Fenotipo , Adulto Joven
4.
Cleft Palate Craniofac J ; 59(11): 1346-1351, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-34714179

RESUMEN

OBJECTIVE: Facial dysostosis is a group of rare craniofacial congenital disabilities requiring multidisciplinary long-term care. This report presents the phenotypic and genotypic information from South India. DESIGN: The study is a case series. SETTING: This was an international collaborative study involving a tertiary craniofacial clinic and medical genetics unit. PATIENTS, PARTICIPANTS: The participants were 9 families with 17 affected individuals of facial dysostosis. INTERVENTION: Exome analysis focused on known genes associated with acrofacial and mandibulofacial syndromes. MAIN OUTCOME MEASURE: The outcome measure was to report phenotyptic and genetic heterogeneity in affected individuals. RESULTS: A Tessier cleft was seen in 7 (41%), lower eyelid coloboma in 12 (65%), ear anomalies in 10 (59%), uniolateral or bilateral aural atresia in 4 (24%), and deafness in 6 (35%). The facial gestalt of Treacher Collins syndrome (TCS) showed extensive phenotypic variations. Pathogenic variants in TCOF1 (Treacher Collins syndrome) were seen in six families, POLR1A (acrofacial dysostosis, Cincinnati type) and EFTUD2 (mandibulofacial dysostosis with microcephaly) in one each. One family (11.1%) had no detectable variation. Five out of six probands with Treacher Collins syndrome had other affected family members (83.3%), including a non-penetrant mother, identified after sequencing. CONCLUSION: Our report illustrates the molecular heterogeneity of mandibulofacial dysostosis in India.


Asunto(s)
Disostosis Mandibulofacial , Microcefalia , Cara , Genotipo , Humanos , Disostosis Mandibulofacial/genética , Microcefalia/genética , Factores de Elongación de Péptidos/genética , Ribonucleoproteína Nuclear Pequeña U5/genética , Síndrome
5.
Hum Genet ; 140(7): 1061-1076, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33811546

RESUMEN

Teebi hypertelorism syndrome (THS; OMIM 145420) is a rare craniofacial disorder characterized by hypertelorism, prominent forehead, short nose with broad or depressed nasal root. Some cases of THS have been attributed to SPECC1L variants. Homozygous variants in CDH11 truncating the transmembrane and intracellular domains have been implicated in Elsahy-Waters syndrome (EWS; OMIM 211380) with hypertelorism. We report THS due to CDH11 heterozygous missense variants on 19 subjects from 9 families. All affected residues in the extracellular region of Cadherin-11 (CHD11) are highly conserved across vertebrate species and classical cadherins. Six of the variants that cluster around the EC2-EC3 and EC3-EC4 linker regions are predicted to affect Ca2+ binding that is required for cadherin stability. Two of the additional variants [c.164G > C, p.(Trp55Ser) and c.418G > A, p.(Glu140Lys)] are also notable as they are predicted to directly affect trans-homodimer formation. Immunohistochemical study demonstrates that CDH11 is strongly expressed in human facial mesenchyme. Using multiple functional assays, we show that five variants from the EC1, EC2-EC3 linker, and EC3 regions significantly reduced the cell-substrate trans adhesion activity and one variant from EC3-EC4 linker results in changes in cell morphology, focal adhesion, and migration, suggesting dominant negative effect. Characteristic features in this cohort included depressed nasal root, cardiac and umbilical defects. These features distinguished this phenotype from that seen in SPECC1L-related hypertelorism syndrome and CDH11-related EWS. Our results demonstrate heterozygous variants in CDH11, which decrease cell-cell adhesion and increase cell migratory behavior, cause a form of THS, as termed CDH11-related THS.


Asunto(s)
Anomalías Múltiples/genética , Cadherinas/genética , Adhesión Celular/genética , Anomalías Craneofaciales/genética , Deformidades Congénitas del Pie/genética , Variación Genética/genética , Deformidades Congénitas de la Mano/genética , Hipertelorismo/genética , Secuencia de Aminoácidos , Movimiento Celular/genética , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Linaje , Fenotipo
6.
Genet Med ; 23(6): 1137-1142, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33564150

RESUMEN

PURPOSE: Noninvasive prenatal screening (NIPS) using cell-free DNA has transformed prenatal care. Belgium was the first country to implement and fully reimburse NIPS as a first-tier screening test offered to all pregnant women. A consortium consisting of all Belgian genetic centers report the outcome of two years genome-wide NIPS implementation. METHODS: The performance for the common trisomies and for secondary findings was evaluated based on 153,575 genome-wide NIP tests. Furthermore, the evolution of the number of invasive tests and the incidence of Down syndrome live births was registered. RESULTS: Trisomies 21, 18, and 13 were detected in respectively 0.32%, 0.07%, and 0.06% of cases, with overall positive predictive values (PPVs) of 92.4%, 84.6%, and 43.9%. Rare autosomal trisomies and fetal segmental imbalances were detected in respectively 0.23% and 0.07% of cases with PPVs of 4.1% and 47%. The number of invasive obstetric procedures decreased by 52%. The number of trisomy 21 live births dropped to 0.04%. CONCLUSION: Expanding the scope of NIPS beyond trisomy 21 fetal screening allows the implementation of personalized genomic medicine for the obstetric population. This genome-wide NIPS approach has been embedded successfully in prenatal genetic care in Belgium and might serve as a framework for other countries offering NIPS.


Asunto(s)
Trastornos de los Cromosomas , Síndrome de Down , Pruebas Prenatales no Invasivas , Aneuploidia , Trastornos de los Cromosomas/diagnóstico , Trastornos de los Cromosomas/epidemiología , Trastornos de los Cromosomas/genética , Síndrome de Down/diagnóstico , Síndrome de Down/epidemiología , Síndrome de Down/genética , Femenino , Humanos , Embarazo , Diagnóstico Prenatal , Trisomía
7.
Clin Genet ; 99(2): 259-268, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33131045

RESUMEN

The CAMTA1-associated phenotype was initially defined in patients with intragenic deletions and duplications who showed nonprogressive congenital ataxia, with or without intellectual disability. Here, we describe 10 individuals with CAMTA1 variants: nine previously unreported (likely) pathogenic variants comprising one missense, four frameshift and four nonsense variants, and one missense variant of unknown significance. Six patients were diagnosed following whole exome sequencing and four individuals with exome-based targeted panel analysis. Most of them present with developmental delay, manifesting in speech and motor delay. Other frequent findings are hypotonia, cognitive impairment, cerebellar dysfunction, oculomotor abnormalities, and behavioral problems. Feeding problems occur more frequently than previously observed. In addition, we present a systematic review of 19 previously published individuals with causal variants, including copy number, truncating, and missense variants. We note a tendency of more severe cognitive impairment and recurrent dysmorphic features in individuals with a copy number variant. Pathogenic variants are predominantly observed in and near the N- and C- terminal functional domains. Clinical heterogeneity is observed, but 3'-terminal variants seem to associate with less pronounced cerebellar dysfunction.


Asunto(s)
Proteínas de Unión al Calcio/genética , Enfermedades del Sistema Nervioso/genética , Transactivadores/genética , Adolescente , Niño , Preescolar , Trastornos del Conocimiento/genética , Análisis Mutacional de ADN , Discapacidades del Desarrollo/genética , Femenino , Humanos , Masculino , Fenotipo
8.
Nucleic Acids Res ; 47(4): 1605-1614, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30566647

RESUMEN

Shallow whole-genome sequencing to infer copy number alterations (CNAs) in the human genome is rapidly becoming the method par excellence for routine diagnostic use. Numerous tools exist to deduce aberrations from massive parallel sequencing data, yet most are optimized for research and often fail to redeem paramount needs in a clinical setting. Optimally, a read depth-based analytical software should be able to deal with single-end and low-coverage data-this to make sequencing costs feasible. Other important factors include runtime, applicability to a variety of analyses and overall performance. We compared the most important aspect, being normalization, across six different CNA tools, selected for their assumed ability to satisfy the latter needs. In conclusion, WISECONDOR, which uses a within-sample normalization technique, undoubtedly produced the best results concerning variance, distributional assumptions and basic ability to detect true variations. Nonetheless, as is the case with every tool, WISECONDOR has limitations, which arise through its exclusiveness for non-invasive prenatal testing. Therefore, this work presents WisecondorX in addition, an improved WISECONDOR that enables its use for varying types of applications. WisecondorX is freely available at https://github.com/CenterForMedicalGeneticsGhent/WisecondorX.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Femenino , Genoma Humano/genética , Humanos , Embarazo , Diagnóstico Prenatal , Secuenciación Completa del Genoma/métodos
9.
Prenat Diagn ; 40(10): 1272-1283, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32436253

RESUMEN

OBJECTIVE: Belgian genetic centers established a database containing data on all chromosomal microarrays performed in a prenatal context. A study was initiated to evaluate postnatal development in children diagnosed prenatally with a non-benign copy number variant (CNV). METHODS: All children diagnosed with a prenatally detected non-benign CNV in a Belgian genetic center between May 2013 and February 2015 were included in the patient population. The control population consisted of children who had undergone an invasive procedure during pregnancy, with no or only benign CNVs. Child development was evaluated at 36 months using three (3) questionnaires: Ages and Stages Questionnaire Third edition, Ages and Stages Questionnaire Social-Emotional Second Edition and a general questionnaire. RESULTS: A significant difference in communication and personal-social development was detected between children with a reported susceptibility CNV and both children with an unreported susceptibility CNV and the control population. The outcome of children with a particular CNV is discussed in a case-by-case manner. CONCLUSION: Our postnatal follow-up project of children with a prenatally detected non-benign CNV is the first nationwide project of its kind. A higher number of cases for each CNV category is however needed to confirm our findings.


Asunto(s)
Variaciones en el Número de Copia de ADN , Resultado del Embarazo/epidemiología , Diagnóstico Prenatal/estadística & datos numéricos , Bélgica/epidemiología , Estudios de Casos y Controles , Preescolar , Aberraciones Cromosómicas/estadística & datos numéricos , Estudios de Cohortes , Anomalías Congénitas/diagnóstico , Anomalías Congénitas/epidemiología , Anomalías Congénitas/genética , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Análisis por Micromatrices/métodos , Embarazo , Diagnóstico Prenatal/métodos
10.
Prenat Diagn ; 39(10): 925-933, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31219182

RESUMEN

OBJECTIVE: During routine noninvasive prenatal testing (NIPT), cell-free fetal DNA fraction is ideally derived from shallow-depth whole-genome sequencing data, preventing the need for additional experimental assays. The fraction of aligned reads to chromosome Y enables proper quantification for male fetuses, unlike for females, where advanced predictive procedures are required. This study introduces PREdict FetAl ComponEnt (PREFACE), a novel bioinformatics pipeline to establish fetal fraction in a gender-independent manner. METHODS: PREFACE combines the strengths of principal component analysis and neural networks to model copy number profiles. RESULTS: For sets of roughly 1100 male NIPT samples, a cross-validated Pearson correlation of 0.9 between predictions and fetal fractions according to Y chromosomal read counts was noted. PREFACE enables training with both male and unlabeled female fetuses. Using our complete cohort (nfemale = 2468, nmale = 2723), the correlation metric reached 0.94. CONCLUSIONS: Allowing individual institutions to generate optimized models sidelines between-laboratory bias, as PREFACE enables user-friendly training with a limited amount of retrospective data. In addition, our software provides the fetal fraction based on the copy number state of chromosome X. We show that these measures can predict mixed multiple pregnancies, sex chromosomal aneuploidies, and the source of observed aberrations.


Asunto(s)
Ácidos Nucleicos Libres de Células/análisis , Trastornos de los Cromosomas/diagnóstico , Feto/metabolismo , Pruebas Prenatales no Invasivas , Análisis de Componente Principal/métodos , Programas Informáticos , Trastornos de los Cromosomas/epidemiología , Estudios de Cohortes , Biología Computacional/métodos , Simulación por Computador , Femenino , Feto/fisiología , Pruebas Genéticas/métodos , Pruebas Genéticas/estadística & datos numéricos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Humanos , Recién Nacido , Masculino , Pruebas Prenatales no Invasivas/métodos , Pruebas Prenatales no Invasivas/estadística & datos numéricos , Embarazo , Pronóstico , Reproducibilidad de los Resultados , Estudios Retrospectivos , Análisis de Secuencia de ADN/métodos , Análisis de Secuencia de ADN/estadística & datos numéricos , Factores Sexuales
11.
Hemoglobin ; 43(2): 112-115, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31223040

RESUMEN

α-Thalassemia (α-thal) is a common hemoglobinopathy mainly caused by deletion of one or both α-globin genes. We describe an autochthonous Belgian family diagnosed with α-thal trait. Molecular analysis revealed a novel large deletion of at least 170 kb between 226.68 kb (0.2 Mb) and 402.68 kb (0.4 Mb) from the telomere of 16p, leaving the subtelomeric region intact. The deletion includes both α-globin genes (HBA1 and HBA2) but also flanking genes possibly related to non hematological effects: HBQ1, LUC7L, ITFG3, RGS11, ARHGDIG, PDIA2 and AXIN1. These genes are not contained in the region (0.9 and 1.7 Mb from the telomere of 16p) associated with α-thal intellectual disability (ATR-16) syndrome. However, further research is necessary to exclude other potential effects than α-thal in patients with a large deletion at 0.2-0.4 Mb from the telomere of 16p. Genetic counseling is important for carriers of this deletion as homozygosity for the α-globin (- -/) haplotype may lead to Hb Bart's (γ4) hydrops fetalis syndrome.


Asunto(s)
Eliminación de Secuencia/genética , Talasemia alfa/genética , Bélgica , Familia , Hemoglobinas Anormales , Heterocigoto , Humanos , Hidropesía Fetal , Telómero/genética , Globinas alfa/genética
12.
Hum Mutat ; 39(12): 1916-1925, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30084155

RESUMEN

Transposable elements modify human genome by inserting into new loci or by mediating homology-, microhomology-, or homeology-driven DNA recombination or repair, resulting in genomic structural variation. Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a rare lethal neonatal developmental lung disorder caused by point mutations or copy-number variant (CNV) deletions of FOXF1 or its distant tissue-specific enhancer. Eighty-five percent of 45 ACDMPV-causative CNV deletions, of which junctions have been sequenced, had at least one of their two breakpoints located in a retrotransposon, with more than half of them being Alu elements. We describe a novel ∼35 kb-large genomic instability hotspot at 16q24.1, involving two evolutionarily young LINE-1 (L1) elements, L1PA2 and L1PA3, flanking AluY, two AluSx, AluSx1, and AluJr elements. The occurrence of L1s at this location coincided with the branching out of the Homo-Pan-Gorilla clade, and was preceded by the insertion of AluSx, AluSx1, and AluJr. Our data show that, in addition to mediating recurrent CNVs, L1 and Alu retrotransposons can predispose the human genome to formation of variably sized CNVs, both of clinical and evolutionary relevance. Nonetheless, epigenetic or other genomic features of this locus might also contribute to its increased instability.


Asunto(s)
Cromosomas Humanos Par 16/genética , Variaciones en el Número de Copia de ADN , Inestabilidad Genómica , Síndrome de Circulación Fetal Persistente/genética , Elementos Alu , Evolución Molecular , Factores de Transcripción Forkhead/genética , Predisposición Genética a la Enfermedad , Humanos , Elementos de Nucleótido Esparcido Largo , Linaje , Mutación Puntual
13.
Prenat Diagn ; 38(13): 1120-1128, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30334587

RESUMEN

OBJECTIVE: With the replacement of karyotyping by chromosomal microarray (CMA) in invasive prenatal diagnosis, new challenges have arisen. By building a national database, we standardize the classification and reporting of prenatally detected copy number variants (CNVs) across Belgian genetic centers. This database, which will link genetic and ultrasound findings with postnatal development, forms a unique resource to investigate the pathogenicity of variants of uncertain significance and to refine the phenotypic spectrum of pathogenic and susceptibility CNVs. METHODS: The Belgian MicroArray Prenatal (BEMAPRE) consortium is a collaboration of all genetic centers in Belgium. We collected data from all invasive prenatal procedures performed between May 2013 and July 2016. RESULTS: In this three-year period, 13 266 prenatal CMAs were performed. By national agreement, a limited number of susceptibility CNVs and no variants of uncertain significance were reported. Added values for using CMA versus conventional karyotyping were 1.8% in the general invasive population and 2.7% in cases with an ultrasound anomaly. Of the reported CNVs, 31.5% would have remained undetected with non-invasive prenatal test as the first-tier test. CONCLUSION: The establishment of a national database for prenatal CNV data allows for a uniform reporting policy and the investigation of the prenatal and postnatal genotype-phenotype correlation.


Asunto(s)
Aberraciones Cromosómicas , Anomalías Congénitas/genética , Variaciones en el Número de Copia de ADN/genética , Haploinsuficiencia/genética , Análisis por Micromatrices/métodos , Adulto , Artrogriposis/diagnóstico , Artrogriposis/genética , Bélgica , Enfermedad de Charcot-Marie-Tooth/diagnóstico , Enfermedad de Charcot-Marie-Tooth/genética , Hibridación Genómica Comparativa , Anomalías Congénitas/diagnóstico , Bases de Datos Genéticas , Síndrome de DiGeorge/diagnóstico , Síndrome de DiGeorge/genética , Femenino , Predisposición Genética a la Enfermedad , Neuropatía Hereditaria Motora y Sensorial/diagnóstico , Neuropatía Hereditaria Motora y Sensorial/genética , Humanos , Ictiosis Ligada al Cromosoma X/diagnóstico , Ictiosis Ligada al Cromosoma X/genética , Cariotipificación , Embarazo , Diagnóstico Prenatal
14.
J Med Genet ; 54(9): 613-623, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28735298

RESUMEN

BACKGROUND: Mutations in forkhead box protein P1 (FOXP1) cause intellectual disability (ID) and specific language impairment (SLI), with or without autistic features (MIM: 613670). Despite multiple case reports no specific phenotype emerged so far. METHODS: We correlate clinical and molecular data of 25 novel and 23 previously reported patients with FOXP1 defects. We evaluated FOXP1 activity by an in vitro luciferase model and assessed protein stability in vitro by western blotting. RESULTS: Patients show ID, SLI, neuromotor delay (NMD) and recurrent facial features including a high broad forehead, bent downslanting palpebral fissures, ptosis and/or blepharophimosis and a bulbous nasal tip. Behavioural problems and autistic features are common. Brain, cardiac and urogenital malformations can be associated. More severe ID and NMD, sensorineural hearing loss and feeding difficulties are more common in patients with interstitial 3p deletions (14 patients) versus patients with monogenic FOXP1 defects (34 patients). Mutations result in impaired transcriptional repression and/or reduced protein stability. CONCLUSIONS: FOXP1-related ID syndrome is a recognisable entity with a wide clinical spectrum and frequent systemic involvement. Our data will be helpful to evaluate genotype-phenotype correlations when interpreting next-generation sequencing data obtained in patients with ID and/or SLI and will guide clinical management.


Asunto(s)
Factores de Transcripción Forkhead/genética , Discapacidad Intelectual/genética , Proteínas Represoras/genética , Trastorno del Espectro Autista/genética , Cara/anomalías , Femenino , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/metabolismo , Humanos , Trastornos del Lenguaje/genética , Masculino , Trastornos de la Destreza Motora/genética , Mutación , Mutación Missense , Trastornos del Neurodesarrollo/genética , Fenotipo , Estabilidad Proteica , Proteínas Represoras/química , Proteínas Represoras/metabolismo , Síndrome , Transcripción Genética
15.
Hum Mol Genet ; 24(11): 3038-49, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25669657

RESUMEN

Inherited dental malformations constitute a clinically and genetically heterogeneous group of disorders. Here, we report on four families, three of them consanguineous, with an identical phenotype, characterized by significant short stature with brachyolmia and hypoplastic amelogenesis imperfecta (AI) with almost absent enamel. This phenotype was first described in 1996 by Verloes et al. as an autosomal recessive form of brachyolmia associated with AI. Whole-exome sequencing resulted in the identification of recessive hypomorphic mutations including deletion, nonsense and splice mutations, in the LTBP3 gene, which is involved in the TGF-beta signaling pathway. We further investigated gene expression during mouse development and tooth formation. Differentiated ameloblasts synthesizing enamel matrix proteins and odontoblasts expressed the gene. Study of an available knockout mouse model showed that the mutant mice displayed very thin to absent enamel in both incisors and molars, hereby recapitulating the AI phenotype in the human disorder.


Asunto(s)
Amelogénesis Imperfecta/genética , Proteínas de Unión a TGF-beta Latente/genética , Osteocondrodisplasias/genética , Adolescente , Amelogénesis Imperfecta/diagnóstico por imagen , Animales , Secuencia de Bases , Niño , Consanguinidad , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Estudios de Asociación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Missense , Osteocondrodisplasias/diagnóstico por imagen , Linaje , Radiografía , Eliminación de Secuencia
16.
Am J Med Genet A ; 173(11): 3104-3108, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28884947

RESUMEN

We present a 4-year-old girl with delayed neuromotor development, short stature of prenatal onset, and specific behavioral and craniofacial features harboring an intragenic deletion in the ARID2 gene. The phenotype confirmed the major features of the recently described ARID2-related intellectual disability syndrome. However, our patient showed overlapping features with Nicolaides-Baraitser syndrome and Coffin-Siris syndrome, providing further arguments to reclassify these disorders as "SWI/SNF-related intellectual disability syndromes."


Asunto(s)
Proteínas Cromosómicas no Histona/genética , Discapacidad Intelectual/genética , Trastornos Motores/genética , Factores de Transcripción/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Preescolar , Cara/anomalías , Cara/patología , Facies , Femenino , Deformidades Congénitas del Pie/diagnóstico , Deformidades Congénitas del Pie/genética , Deformidades Congénitas del Pie/patología , Predisposición Genética a la Enfermedad , Deformidades Congénitas de la Mano/diagnóstico , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/patología , Humanos , Hipotricosis/diagnóstico , Hipotricosis/genética , Hipotricosis/patología , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Micrognatismo/diagnóstico , Micrognatismo/genética , Micrognatismo/patología , Trastornos Motores/fisiopatología , Cuello/anomalías , Cuello/patología
17.
Nat Methods ; 10(11): 1063-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24173381

RESUMEN

Two surveys of over 1,700 publications whose authors use quantitative real-time PCR (qPCR) reveal a lack of transparent and comprehensive reporting of essential technical information. Reporting standards are significantly improved in publications that cite the Minimum Information for Publication of Quantitative Real-Time PCR Experiments (MIQE) guidelines, although such publications are still vastly outnumbered by those that do not.


Asunto(s)
Servicios de Información , Reacción en Cadena de la Polimerasa/métodos , Recolección de Datos
18.
Prenat Diagn ; 36(8): 699-707, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27176606

RESUMEN

OBJECTIVES: To implement non-invasive prenatal testing (NIPT) for fetal aneuploidies with semiconductor sequencing in an academic cytogenomic laboratory and to evaluate the first 15-month experience on clinical samples. METHODS: We validated a NIPT protocol for cell-free fetal DNA sequencing from maternal plasma for the detection of trisomy 13, 18 and 21 on a semiconductor sequencing instrument. Fetal DNA fraction calculation for all samples and several quality parameters were implemented in the workflow. One thousand eighty-one clinical NIPT samples were analysed, following the described protocol. RESULTS: Non-invasive prenatal testing was successfully implemented and validated on 201 normal and 74 aneuploid samples. From 1081 clinical samples, 17 samples showed an abnormal result: 14 trisomy 21 samples, one trisomy 18 and one trisomy 16 were detected. Also a maternal copy number variation on chromosome 13 was observed, which could potentially lead to a false positive trisomy 13 result. One sex discordant result was reported, possibly attributable to a vanishing twin. Moreover, our combined fetal fraction calculation enabled a more reliable risk estimate for trisomy 13, 18 and 21. CONCLUSIONS: Non-invasive prenatal testing for trisomy 21, 18 and 13 has a very high specificity and sensitivity. Because of several biological phenomena, diagnostic invasive confirmation of abnormal results remains required. © 2016 The Authors. Prenatal Diagnosis published by John Wiley & Sons, Ltd.


Asunto(s)
Trastornos de los Cromosomas/diagnóstico , ADN/sangre , Diagnóstico Prenatal/métodos , Semiconductores , Análisis de Secuencia de ADN/métodos , Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 13/genética , Cromosomas Humanos Par 18/genética , ADN/análisis , Variaciones en el Número de Copia de ADN , Síndrome de Down/diagnóstico , Síndrome de Down/genética , Reacciones Falso Positivas , Femenino , Pruebas Genéticas/métodos , Humanos , Embarazo , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Trisomía/diagnóstico , Trisomía/genética , Síndrome de la Trisomía 13 , Síndrome de la Trisomía 18
19.
Am J Hum Genet ; 91(4): 636-45, 2012 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-22981119

RESUMEN

CaBPs are a family of Ca(2+)-binding proteins related to calmodulin and are localized in the brain and sensory organs, including the retina and cochlea. Although their physiological roles are not yet fully elucidated, CaBPs modulate Ca(2+) signaling through effectors such as voltage-gated Ca(v) Ca(2+) channels. In this study, we identified a splice-site mutation (c.637+1G>T) in Ca(2+)-binding protein 2 (CABP2) in three consanguineous Iranian families affected by moderate-to-severe hearing loss. This mutation, most likely a founder mutation, probably leads to skipping of exon 6 and premature truncation of the protein (p.Phe164Serfs(∗)4). Compared with wild-type CaBP2, the truncated CaBP2 showed altered Ca(2+) binding in isothermal titration calorimetry and less potent regulation of Ca(v)1.3 Ca(2+) channels. We show that genetic defects in CABP2 cause moderate-to-severe sensorineural hearing impairment. The mutation might cause a hypofunctional CaBP2 defective in Ca(2+) sensing and effector regulation in the inner ear.


Asunto(s)
Proteínas de Unión al Calcio/genética , Trastornos de los Cromosomas/genética , Cóclea/fisiopatología , Células Ciliadas Auditivas/patología , Células Ciliadas Auditivas/fisiología , Pérdida Auditiva/genética , Mutación , Calcio/metabolismo , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Cóclea/metabolismo , Consanguinidad , Exones/genética , Femenino , Genes Recesivos , Predisposición Genética a la Enfermedad , Células HEK293 , Células Ciliadas Auditivas/metabolismo , Pérdida Auditiva/metabolismo , Pérdida Auditiva/fisiopatología , Humanos , Masculino , Linaje
20.
medRxiv ; 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37292950

RESUMEN

Neurodevelopmental disorders (NDDs) result from impaired development and functioning of the brain. Here, we identify loss-of-function variation in ZFHX3 as a novel cause for syndromic intellectual disability (ID). ZFHX3, previously known as ATBF1, is a zinc-finger homeodomain transcription factor involved in multiple biological processes including cell differentiation and tumorigenesis. Through international collaboration, we collected clinical and morphometric data (Face2Gene) of 41 individuals with protein truncating variants (PTVs) or (partial) deletions of ZFHX3 . We used data mining, RNA and protein analysis to identify the subcellular localization and spatiotemporal expression of ZFHX3 in multiple in vitro models. We identified the DNA targets of ZFHX3 using ChIP seq. Immunoprecipitation followed by mass spectrometry indicated potential binding partners of endogenous ZFHX3 in neural stem cells that were subsequently confirmed by reversed co-immunoprecipitation and western blot. We evaluated a DNA methylation profile associated with ZFHX3 haploinsufficiency using DNA methylation analysis on whole blood extracted DNA of six individuals with ZFHX3 PTVs and four with a (partial) deletion of ZFHX3 . A reversed genetic approach characterized the ZFHX3 orthologue in Drosophila melanogaster . Loss-of-function variation of ZFHX3 consistently associates with (mild) ID and/or behavioural problems, postnatal growth retardation, feeding difficulties, and recognizable facial characteristics, including the rare occurrence of cleft palate. Nuclear abundance of ZFHX3 increases during human brain development and neuronal differentiation in neural stem cells and SH-SY5Y cells, ZFHX3 interacts with the chromatin remodelling BRG1/Brm-associated factor complex and the cleavage and polyadenylation complex. In line with a role for chromatin remodelling, ZFHX3 haploinsufficiency associates with a specific DNA methylation profile in leukocyte-derived DNA. The target genes of ZFHX3 are implicated in neuron and axon development. In Drosophila melanogaster , z fh2, considered to be the ZFHX3 orthologue, is expressed in the third instar larval brain. Ubiquitous and neuron-specific knockdown of zfh2 results in adult lethality underscoring a key role for zfh2 in development and neurodevelopment. Interestingly, ectopic expression of zfh2 as well as ZFHX3 in the developing wing disc results in a thoracic cleft phenotype. Collectively, our data shows that loss-of-function variants in ZFHX3 are a cause of syndromic ID, that associates with a specific DNA methylation profile. Furthermore, we show that ZFHX3 participates in chromatin remodelling and mRNA processing.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA