Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Cell ; 146(6): 889-903, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21925314

RESUMEN

Complex genomic rearrangements (CGRs) consisting of two or more breakpoint junctions have been observed in genomic disorders. Recently, a chromosome catastrophe phenomenon termed chromothripsis, in which numerous genomic rearrangements are apparently acquired in one single catastrophic event, was described in multiple cancers. Here, we show that constitutionally acquired CGRs share similarities with cancer chromothripsis. In the 17 CGR cases investigated, we observed localization and multiple copy number changes including deletions, duplications, and/or triplications, as well as extensive translocations and inversions. Genomic rearrangements involved varied in size and complexities; in one case, array comparative genomic hybridization revealed 18 copy number changes. Breakpoint sequencing identified characteristic features, including small templated insertions at breakpoints and microhomology at breakpoint junctions, which have been attributed to replicative processes. The resemblance between CGR and chromothripsis suggests similar mechanistic underpinnings. Such chromosome catastrophic events appear to reflect basic DNA metabolism operative throughout an organism's life cycle.


Asunto(s)
Aberraciones Cromosómicas , Reparación del ADN , Discapacidades del Desarrollo/genética , Neoplasias/genética , Secuencia de Bases , Niño , Preescolar , Rotura Cromosómica , Hibridación Genómica Comparativa , Replicación del ADN , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Datos de Secuencia Molecular
2.
Am J Hum Genet ; 108(5): 929-941, 2021 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-33811806

RESUMEN

Proteins involved in transcriptional regulation harbor a demonstrated enrichment of mutations in neurodevelopmental disorders. The Sin3 (Swi-independent 3)/histone deacetylase (HDAC) complex plays a central role in histone deacetylation and transcriptional repression. Among the two vertebrate paralogs encoding the Sin3 complex, SIN3A variants cause syndromic intellectual disability, but the clinical consequences of SIN3B haploinsufficiency in humans are uncharacterized. Here, we describe a syndrome hallmarked by intellectual disability, developmental delay, and dysmorphic facial features with variably penetrant autism spectrum disorder, congenital malformations, corpus callosum defects, and impaired growth caused by disruptive SIN3B variants. Using chromosomal microarray or exome sequencing, and through international data sharing efforts, we identified nine individuals with heterozygous SIN3B deletion or single-nucleotide variants. Five individuals harbor heterozygous deletions encompassing SIN3B that reside within a ∼230 kb minimal region of overlap on 19p13.11, two individuals have a rare nonsynonymous substitution, and two individuals have a single-nucleotide deletion that results in a frameshift and predicted premature termination codon. To test the relevance of SIN3B impairment to measurable aspects of the human phenotype, we disrupted the orthologous zebrafish locus by genome editing and transient suppression. The mutant and morphant larvae display altered craniofacial patterning, commissural axon defects, and reduced body length supportive of an essential role for Sin3 function in growth and patterning of anterior structures. To investigate further the molecular consequences of SIN3B variants, we quantified genome-wide enhancer and promoter activity states by using H3K27ac ChIP-seq. We show that, similar to SIN3A mutations, SIN3B disruption causes hyperacetylation of a subset of enhancers and promoters in peripheral blood mononuclear cells. Together, these data demonstrate that SIN3B haploinsufficiency leads to a hitherto unknown intellectual disability/autism syndrome, uncover a crucial role of SIN3B in the central nervous system, and define the epigenetic landscape associated with Sin3 complex impairment.


Asunto(s)
Trastorno del Espectro Autista/genética , Haploinsuficiencia/genética , Histona Desacetilasas/metabolismo , Discapacidad Intelectual/genética , Proteínas Represoras/genética , Acetilación , Adolescente , Animales , Niño , Preescolar , Variaciones en el Número de Copia de ADN/genética , Femenino , Histonas/química , Histonas/metabolismo , Humanos , Lactante , Larva/genética , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutación , Proteínas Represoras/deficiencia , Proteínas Represoras/metabolismo , Síndrome , Adulto Joven , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética
3.
Am J Med Genet A ; 188(9): 2750-2759, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35543142

RESUMEN

The pre-mRNA-processing factor 8, encoded by PRPF8, is a scaffolding component of a spliceosome complex involved in the removal of introns from mRNA precursors. Previously, heterozygous pathogenic variants in PRPF8 have been associated with autosomal dominant retinitis pigmentosa. More recently, PRPF8 was suggested as a candidate gene for autism spectrum disorder due to the enrichment of sequence variants in this gene in individuals with neurodevelopmental disorders. We report 14 individuals with various forms of neurodevelopmental conditions, found to have heterozygous, predominantly de novo, missense, and loss-of-function variants in PRPF8. These individuals have clinical features that may represent a new neurodevelopmental syndrome.


Asunto(s)
Trastorno del Espectro Autista , Trastornos del Neurodesarrollo , Retinitis Pigmentosa , Trastorno del Espectro Autista/genética , Heterocigoto , Humanos , Trastornos del Neurodesarrollo/genética , Proteínas de Unión al ARN/genética , Retinitis Pigmentosa/genética
4.
Am J Hum Genet ; 102(1): 69-87, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29290338

RESUMEN

Neurofibromatosis type 1 (NF1), a common genetic disorder with a birth incidence of 1:2,000-3,000, is characterized by a highly variable clinical presentation. To date, only two clinically relevant intragenic genotype-phenotype correlations have been reported for NF1 missense mutations affecting p.Arg1809 and a single amino acid deletion p.Met922del. Both variants predispose to a distinct mild NF1 phenotype with neither externally visible cutaneous/plexiform neurofibromas nor other tumors. Here, we report 162 individuals (129 unrelated probands and 33 affected relatives) heterozygous for a constitutional missense mutation affecting one of five neighboring NF1 codons-Leu844, Cys845, Ala846, Leu847, and Gly848-located in the cysteine-serine-rich domain (CSRD). Collectively, these recurrent missense mutations affect ∼0.8% of unrelated NF1 mutation-positive probands in the University of Alabama at Birmingham (UAB) cohort. Major superficial plexiform neurofibromas and symptomatic spinal neurofibromas were more prevalent in these individuals compared with classic NF1-affected cohorts (both p < 0.0001). Nearly half of the individuals had symptomatic or asymptomatic optic pathway gliomas and/or skeletal abnormalities. Additionally, variants in this region seem to confer a high predisposition to develop malignancies compared with the general NF1-affected population (p = 0.0061). Our results demonstrate that these NF1 missense mutations, although located outside the GAP-related domain, may be an important risk factor for a severe presentation. A genotype-phenotype correlation at the NF1 region 844-848 exists and will be valuable in the management and genetic counseling of a significant number of individuals.


Asunto(s)
Codón/genética , Estudios de Asociación Genética , Mutación Missense/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Adolescente , Secuencia de Aminoácidos , Niño , Estudios de Cohortes , Simulación por Computador , Demografía , Femenino , Heterocigoto , Humanos , Masculino , Neurofibromina 1/química , Fenotipo , Adulto Joven
5.
Genet Med ; 23(6): 1028-1040, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33658631

RESUMEN

PURPOSE: We describe a novel neurobehavioral phenotype of autism spectrum disorder (ASD), intellectual disability, and/or attention-deficit/hyperactivity disorder (ADHD) associated with de novo or inherited deleterious variants in members of the RFX family of genes. RFX genes are evolutionarily conserved transcription factors that act as master regulators of central nervous system development and ciliogenesis. METHODS: We assembled a cohort of 38 individuals (from 33 unrelated families) with de novo variants in RFX3, RFX4, and RFX7. We describe their common clinical phenotypes and present bioinformatic analyses of expression patterns and downstream targets of these genes as they relate to other neurodevelopmental risk genes. RESULTS: These individuals share neurobehavioral features including ASD, intellectual disability, and/or ADHD; other frequent features include hypersensitivity to sensory stimuli and sleep problems. RFX3, RFX4, and RFX7 are strongly expressed in developing and adult human brain, and X-box binding motifs as well as RFX ChIP-seq peaks are enriched in the cis-regulatory regions of known ASD risk genes. CONCLUSION: These results establish a likely role of deleterious variation in RFX3, RFX4, and RFX7 in cases of monogenic intellectual disability, ADHD and ASD, and position these genes as potentially critical transcriptional regulators of neurobiological pathways associated with neurodevelopmental disease pathogenesis.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad , Trastorno del Espectro Autista , Trastorno Autístico , Discapacidad Intelectual , Adulto , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Humanos , Discapacidad Intelectual/genética , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/genética
6.
Am J Hum Genet ; 101(6): 995-1005, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29198722

RESUMEN

A recurrent de novo missense variant within the C-terminal Sin3-like domain of ZSWIM6 was previously reported to cause acromelic frontonasal dysostosis (AFND), an autosomal-dominant severe frontonasal and limb malformation syndrome, associated with neurocognitive and motor delay, via a proposed gain-of-function effect. We present detailed phenotypic information on seven unrelated individuals with a recurrent de novo nonsense variant (c.2737C>T [p.Arg913Ter]) in the penultimate exon of ZSWIM6 who have severe-profound intellectual disability and additional central and peripheral nervous system symptoms but an absence of frontonasal or limb malformations. We show that the c.2737C>T variant does not trigger nonsense-mediated decay of the ZSWIM6 mRNA in affected individual-derived cells. This finding supports the existence of a truncated ZSWIM6 protein lacking the Sin3-like domain, which could have a dominant-negative effect. This study builds support for a key role for ZSWIM6 in neuronal development and function, in addition to its putative roles in limb and craniofacial development, and provides a striking example of different variants in the same gene leading to distinct phenotypes.


Asunto(s)
Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Trastornos Neurocognitivos/genética , Sistema Nervioso Central/anomalías , Sistema Nervioso Central/embriología , Codón sin Sentido/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Deformidades Congénitas de las Extremidades/genética , Disostosis Mandibulofacial/genética , Sistema Nervioso Periférico/anomalías , Sistema Nervioso Periférico/enzimología
7.
Am J Hum Genet ; 101(5): 716-724, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100085

RESUMEN

DHX30 is a member of the family of DExH-box helicases, which use ATP hydrolysis to unwind RNA secondary structures. Here we identified six different de novo missense mutations in DHX30 in twelve unrelated individuals affected by global developmental delay (GDD), intellectual disability (ID), severe speech impairment and gait abnormalities. While four mutations are recurrent, two are unique with one affecting the codon of one recurrent mutation. All amino acid changes are located within highly conserved helicase motifs and were found to either impair ATPase activity or RNA recognition in different in vitro assays. Moreover, protein variants exhibit an increased propensity to trigger stress granule (SG) formation resulting in global translation inhibition. Thus, our findings highlight the prominent role of translation control in development and function of the central nervous system and also provide molecular insight into how DHX30 dysfunction might cause a neurodevelopmental disorder.


Asunto(s)
Discapacidades del Desarrollo/genética , Mutación Missense/genética , ARN Helicasas/genética , Adenosina Trifosfatasas/genética , Adolescente , Aminoácidos/genética , Línea Celular , Línea Celular Tumoral , Sistema Nervioso Central/patología , Niño , Preescolar , Femenino , Células HEK293 , Humanos , Discapacidad Intelectual/genética , Masculino , ARN/genética
8.
Am J Hum Genet ; 100(4): 676-688, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28343629

RESUMEN

Ubiquitination is a posttranslational modification that regulates many cellular processes including protein degradation, intracellular trafficking, cell signaling, and protein-protein interactions. Deubiquitinating enzymes (DUBs), which reverse the process of ubiquitination, are important regulators of the ubiquitin system. OTUD6B encodes a member of the ovarian tumor domain (OTU)-containing subfamily of deubiquitinating enzymes. Herein, we report biallelic pathogenic variants in OTUD6B in 12 individuals from 6 independent families with an intellectual disability syndrome associated with seizures and dysmorphic features. In subjects with predicted loss-of-function alleles, additional features include global developmental delay, microcephaly, absent speech, hypotonia, growth retardation with prenatal onset, feeding difficulties, structural brain abnormalities, congenital malformations including congenital heart disease, and musculoskeletal features. Homozygous Otud6b knockout mice were subviable, smaller in size, and had congenital heart defects, consistent with the severity of loss-of-function variants in humans. Analysis of peripheral blood mononuclear cells from an affected subject showed reduced incorporation of 19S subunits into 26S proteasomes, decreased chymotrypsin-like activity, and accumulation of ubiquitin-protein conjugates. Our findings suggest a role for OTUD6B in proteasome function, establish that defective OTUD6B function underlies a multisystemic human disorder, and provide additional evidence for the emerging relationship between the ubiquitin system and human disease.


Asunto(s)
Anomalías Múltiples/genética , Endopeptidasas/genética , Discapacidad Intelectual/genética , Adolescente , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Humanos , Masculino , Ratones , Linaje , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Convulsiones/genética
9.
Genet Med ; 21(7): 1652-1656, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30568308

RESUMEN

PURPOSE: Brain malformations caused by 17p13.3 deletions include lissencephaly with deletions of the larger Miller-Dieker syndrome region or smaller deletions of only PAFAH1B1, white matter changes, and a distinct syndrome due to deletions including YWHAE and CRK but sparing PAFAH1B1. We sought to understand the significance of 17p13.3 deletions between the YWHAE/CRK and PAFAH1B1 loci. METHODS: We analyzed the clinical features of six individuals from five families with 17p13.3 deletions between and not including YWHAE/CRK and PAFAH1B1 identified among individuals undergoing clinical chromosomal microarray testing or research genome sequencing. RESULTS: Five individuals from four families had multifocal white matter lesions while a sixth had a normal magnetic resonance image. A combination of our individuals and a review of those in the literature with white matter changes and deletions in this chromosomal region narrows the overlapping region for this brain phenotype to ~345 kb, including 11 RefSeq genes, with RTN4RL1 haploinsufficiency as the best candidate for causing this phenotype. CONCLUSION: While previous literature has hypothesized dysmorphic features and white matter changes related to YWHAE, our cohort contributes evidence to the presence of additional genetic changes within 17p13.3 required for proper brain development.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 17 , Leucoencefalopatías/genética , 1-Alquil-2-acetilglicerofosfocolina Esterasa/genética , Proteínas 14-3-3/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Recién Nacido , Leucoencefalopatías/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Proteínas Asociadas a Microtúbulos/genética
10.
Genet Med ; 21(3): 663-675, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30158690

RESUMEN

PURPOSE: Defects in the cohesin pathway are associated with cohesinopathies, notably Cornelia de Lange syndrome (CdLS). We aimed to delineate pathogenic variants in known and candidate cohesinopathy genes from a clinical exome perspective. METHODS: We retrospectively studied patients referred for clinical exome sequencing (CES, N = 10,698). Patients with causative variants in novel or recently described cohesinopathy genes were enrolled for phenotypic characterization. RESULTS: Pathogenic or likely pathogenic single-nucleotide and insertion/deletion variants (SNVs/indels) were identified in established disease genes including NIPBL (N = 5), SMC1A (N = 14), SMC3 (N = 4), RAD21 (N = 2), and HDAC8 (N = 8). The phenotypes in this genetically defined cohort skew towards the mild end of CdLS spectrum as compared with phenotype-driven cohorts. Candidate or recently reported cohesinopathy genes were supported by de novo SNVs/indels in STAG1 (N = 3), STAG2 (N = 5), PDS5A (N = 1), and WAPL (N = 1), and one inherited SNV in PDS5A. We also identified copy-number deletions affecting STAG1 (two de novo, one of unknown inheritance) and STAG2 (one of unknown inheritance). Patients with STAG1 and STAG2 variants presented with overlapping features yet without characteristic facial features of CdLS. CONCLUSION: CES effectively identified disease-causing alleles at the mild end of the cohensinopathy spectrum and enabled characterization of candidate disease genes.


Asunto(s)
Variación Biológica Poblacional/genética , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Adolescente , Alelos , Antígenos Nucleares/genética , Proteínas Portadoras/genética , Niño , Preescolar , Estudios de Cohortes , Síndrome de Cornelia de Lange/diagnóstico , Síndrome de Cornelia de Lange/genética , Exoma/genética , Femenino , Frecuencia de los Genes/genética , Heterogeneidad Genética , Humanos , Mutación INDEL/genética , Masculino , Mutación , Proteínas Nucleares/genética , Fenotipo , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas/genética , Estudios Retrospectivos , Secuenciación del Exoma/métodos , Cohesinas
11.
Am J Hum Genet ; 95(5): 565-78, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439725

RESUMEN

The 17p13.1 microdeletion syndrome is a recently described genomic disorder with a core clinical phenotype of intellectual disability, poor to absent speech, dysmorphic features, and a constellation of more variable clinical features, most prominently microcephaly. We identified five subjects with copy-number variants (CNVs) on 17p13.1 for whom we performed detailed clinical and molecular studies. Breakpoint mapping and retrospective analysis of published cases refined the smallest region of overlap (SRO) for microcephaly to a genomic interval containing nine genes. Dissection of this phenotype in zebrafish embryos revealed a complex genetic architecture: dosage perturbation of four genes (ASGR1, ACADVL, DVL2, and GABARAP) impeded neurodevelopment and decreased dosage of the same loci caused a reduced mitotic index in vitro. Moreover, epistatic analyses in vivo showed that dosage perturbations of discrete gene pairings induce microcephaly. Taken together, these studies support a model in which concomitant dosage perturbation of multiple genes within the CNV drive the microcephaly and possibly other neurodevelopmental phenotypes associated with rearrangements in the 17p13.1 SRO.


Asunto(s)
Anomalías Múltiples/genética , Dosificación de Gen/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis , Receptor de Asialoglicoproteína/genética , Secuencia de Bases , Línea Celular , Puntos de Rotura del Cromosoma , Deleción Cromosómica , Cromosomas Humanos Par 17/genética , Proteínas Dishevelled , Citometría de Flujo , Humanos , Inmunohistoquímica , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , Fosfoproteínas/genética , Estudios Retrospectivos , Análisis de Secuencia de ADN , Síndrome de Smith-Magenis , Síndrome , Pez Cebra
12.
Am J Med Genet A ; 173(9): 2415-2421, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28742282

RESUMEN

Multiple Epiphyseal Dysplasia (MED) is a relatively mild skeletal dysplasia characterized by mild short stature, joint pain, and early-onset osteoarthropathy. Dominantly inherited mutations in COMP, MATN3, COL9A1, COL9A2, and COL9A3, and recessively inherited mutations in SLC26A2, account for the molecular basis of disease in about 80-85% of the cases. In two families with recurrent MED of an unknown molecular basis, we used exome sequencing and candidate gene analysis to identify homozygosity for recessively inherited missense mutations in CANT1, which encodes calcium-activated nucleotidase 1. The MED phenotype is thus allelic to the more severe Desbuquois dysplasia phenotype and the results identify CANT1 as a second locus for recessively inherited MED.


Asunto(s)
Genes Recesivos , Nucleotidasas/genética , Osteocondrodisplasias/genética , Adulto , Secuencia de Bases , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Masculino , Mutación Missense/genética , Osteocondrodisplasias/diagnóstico por imagen , Osteocondrodisplasias/fisiopatología , Linaje , Radiografía
14.
Am J Hum Genet ; 93(2): 197-210, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-23810381

RESUMEN

White matter hyperintensities (WMHs) of the brain are important markers of aging and small-vessel disease. WMHs are rare in healthy children and, when observed, often occur with comorbid neuroinflammatory or vasculitic processes. Here, we describe a complex 4 kb deletion in 2q36.3 that segregates with early childhood communication disorders and WMH in 15 unrelated families predominantly from Southeast Asia. The premature brain aging phenotype with punctate and multifocal WMHs was observed in ~70% of young carrier parents who underwent brain MRI. The complex deletion removes the penultimate exon 3 of TM4SF20, a gene encoding a transmembrane protein of unknown function. Minigene analysis showed that the resultant net loss of an exon introduces a premature stop codon, which, in turn, leads to the generation of a stable protein that fails to target to the plasma membrane and accumulates in the cytoplasm. Finally, we report this deletion to be enriched in individuals of Vietnamese Kinh descent, with an allele frequency of about 1%, embedded in an ancestral haplotype. Our data point to a constellation of early language delay and WMH phenotypes, driven by a likely toxic mechanism of TM4SF20 truncation, and highlight the importance of understanding and managing population-specific low-frequency pathogenic alleles.


Asunto(s)
Envejecimiento Prematuro/genética , Secuencia de Bases , Predisposición Genética a la Enfermedad , Trastornos del Desarrollo del Lenguaje/genética , Leucoencefalopatías/genética , Eliminación de Secuencia , Tetraspaninas/genética , Edad de Inicio , Envejecimiento Prematuro/complicaciones , Envejecimiento Prematuro/etnología , Envejecimiento Prematuro/patología , Pueblo Asiatico , Encéfalo/metabolismo , Encéfalo/patología , Niño , Preescolar , Cromosomas Humanos Par 2 , Exones , Femenino , Humanos , Trastornos del Desarrollo del Lenguaje/complicaciones , Trastornos del Desarrollo del Lenguaje/etnología , Trastornos del Desarrollo del Lenguaje/patología , Leucoencefalopatías/complicaciones , Leucoencefalopatías/etnología , Leucoencefalopatías/patología , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Linaje , Análisis de Secuencia de ADN
15.
Am J Med Genet A ; 170(7): 1791-8, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27133397

RESUMEN

Neurodevelopmental disorders (NDD) are common, with 1-3% of general population being affected, but the etiology is unknown in most individuals. Clinical whole-exome sequencing (WES) has proven to be a powerful tool for the identification of pathogenic variants leading to Mendelian disorders, among which NDD represent a significant percentage. Performing WES with a trio-approach has proven to be extremely effective in identifying de novo pathogenic variants as a common cause of NDD. Here we report six unrelated individuals with a common phenotype consisting of NDD with severe speech delay, hypotonia, and facial dysmorphism. These patients underwent WES with a trio approach and de novo heterozygous predicted pathogenic novel variants in the KAT6A gene were identified. The KAT6A gene encodes a histone acetyltransfrease protein and it has long been known for its structural involvement in acute myeloid leukemia; however, it has not previously been associated with any congenital disorder. In animal models the KAT6A ortholog is involved in transcriptional regulation during development. Given the similar findings in animal models and our patient's phenotypes, we hypothesize that KAT6A could play a role in development of the brain, face, and heart in humans. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Exoma/genética , Histona Acetiltransferasas/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Adulto , Niño , Preescolar , Femenino , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Mutación , Trastornos del Neurodesarrollo/fisiopatología , Análisis de Secuencia de ADN
16.
Hum Mutat ; 34(2): 385-94, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23161826

RESUMEN

De novo mutations in SYNGAP1, which codes for a RAS/RAP GTP-activating protein, cause nonsyndromic intellectual disability (NSID). All disease-causing point mutations identified until now in SYNGAP1 are truncating, raising the possibility of an association between this type of mutations and NSID. Here, we report the identification of the first pathogenic missense mutations (c.1084T>C [p.W362R], c.1685C>T [p.P562L]) and three novel truncating mutations (c.283dupC [p.H95PfsX5], c.2212_2213del [p.S738X], and (c.2184del [p.N729TfsX31]) in SYNGAP1 in patients with NSID. A subset of these patients also showed ataxia, autism, and a specific form of generalized epilepsy that can be refractory to treatment. All of these mutations occurred de novo, except c.283dupC, which was inherited from a father who is a mosaic. Biolistic transfection of wild-type SYNGAP1 in pyramidal cells from cortical organotypic cultures significantly reduced activity-dependent phosphorylated extracellular signal-regulated kinase (pERK) levels. In contrast, constructs expressing p.W362R, p.P562L, or the previously described p.R579X had no significant effect on pERK levels. These experiments suggest that the de novo missense mutations, p.R579X, and possibly all the other truncating mutations in SYNGAP1 result in a loss of its function. Moreover, our study confirms the involvement of SYNGAP1 in autism while providing novel insight into the epileptic manifestations associated with its disruption.


Asunto(s)
Trastorno Autístico/genética , Epilepsia/genética , Haploinsuficiencia , Discapacidad Intelectual/genética , Proteínas Activadoras de ras GTPasa/genética , Adolescente , Secuencia de Aminoácidos , Trastorno Autístico/fisiopatología , Western Blotting , Niño , Preescolar , Clonación Molecular , Epilepsia/fisiopatología , Exoma , Quinasas MAP Reguladas por Señal Extracelular/genética , Femenino , Células HEK293 , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Fosforilación , Conformación Proteica , Análisis de Secuencia de ADN , Transfección , Proteínas Activadoras de ras GTPasa/metabolismo
17.
Am J Med Genet A ; 161A(5): 1110-6, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23610052

RESUMEN

We present three patients with overlapping interstitial deletions of 19p13.3 identified by high resolution SNP microarray analysis. All three had a similar phenotype characterized by intellectual disability or developmental delay, structural heart abnormalities, large head relative to height and weight or macrocephaly, and minor facial anomalies. Deletion sizes ranged from 792 Kb to 1.0 Mb and included a common region arr [hg19] 19p13.3 (3,814,392-4,136,989), containing eight genes: ZFR2, ATCAY, NMRK2, DAPK3, EEF2, PIAS4, ZBTB7A, MAP2K2, and two non-coding RNA's MIR637 and SNORDU37. The patient phenotypes were compared with three previous single patient reports with similar interstitial 19p13.3 deletions and six additional patients from the DECIPHER and ISCA databases to determine if a common haploinsufficient phenotype for the region can be established.


Asunto(s)
Anomalías Múltiples/genética , Cromosomas Humanos Par 19/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Polimorfismo de Nucleótido Simple/genética , Eliminación de Secuencia/genética , Niño , Preescolar , Femenino , Humanos , Hibridación Fluorescente in Situ , Masculino , Análisis por Micromatrices , Adulto Joven
18.
Hum Mutat ; 31(12): 1326-42, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20848651

RESUMEN

Array comparative genomic hybridization (aCGH) is a powerful tool for the molecular elucidation and diagnosis of disorders resulting from genomic copy-number variation (CNV). However, intragenic deletions or duplications--those including genomic intervals of a size smaller than a gene--have remained beyond the detection limit of most clinical aCGH analyses. Increasing array probe number improves genomic resolution, although higher cost may limit implementation, and enhanced detection of benign CNV can confound clinical interpretation. We designed an array with exonic coverage of selected disease and candidate genes and used it clinically to identify losses or gains throughout the genome involving at least one exon and as small as several hundred base pairs in size. In some patients, the detected copy-number change occurs within a gene known to be causative of the observed clinical phenotype, demonstrating the ability of this array to detect clinically relevant CNVs with subkilobase resolution. In summary, we demonstrate the utility of a custom-designed, exon-targeted oligonucleotide array to detect intragenic copy-number changes in patients with various clinical phenotypes.


Asunto(s)
Hibridación Genómica Comparativa/métodos , Variaciones en el Número de Copia de ADN/genética , Exones/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Puntos de Rotura del Cromosoma , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética , Adulto Joven
19.
Hum Genet ; 126(4): 589-602, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19557438

RESUMEN

We report four new patients with a submicroscopic deletion in 15q24 manifesting developmental delay, short stature, hypotonia, digital abnormalities, joint laxity, genital abnormalities, and characteristic facial features. These clinical features are shared with six recently reported patients with a 15q24 microdeletion, supporting the notion that this is a recognizable syndrome. We describe a case of an ~2.6 Mb microduplication involving a portion of the minimal deletion critical region in a 15-year-old male with short stature, mild mental retardation, attention deficit hyperactivity disorder, Asperger syndrome, decreased joint mobility, digital abnormalities, and characteristic facial features. Some of these features are shared with a recently reported case with a 15q24 microduplication involving the minimal deletion critical region. We also report two siblings and their mother with duplication adjacent and distal to this region exhibiting mild developmental delay, hypotonia, tapering fingers, characteristic facial features, and prominent ears. The deletion and duplication breakpoints were mapped by array comparative genomic hybridization and the genomic structure in 15q24 was analyzed further. Surprisingly, in addition to the previously recognized three low-copy repeat clusters (BP1, BP2, and BP3), we identified two other paralogous low-copy repeat clusters that likely mediated the formation of alternative sized 15q24 genomic rearrangements via non-allelic homologous recombination.


Asunto(s)
Rotura Cromosómica , Deleción Cromosómica , Mapeo Cromosómico , Cromosomas Humanos Par 15/genética , Duplicación de Gen , Adolescente , Niño , Preescolar , Hibridación Genómica Comparativa , Femenino , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Lactante , Masculino , Fenotipo
20.
Clin Case Rep ; 6(7): 1208-1213, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29988648

RESUMEN

Clinical diagnostic exome sequencing (DES) is currently infrequently used for detecting uniparental disomy (UPD). We present a patient with a dual diagnosis of GLI2 haploinsufficiency as well as UPD of chromosome 20, both identified through DES. We therefore recommend routine UPD analysis during DES to identify this genetic aberration.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA