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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 185(20): 3689-3704.e21, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179666

RESUMEN

Regulatory landscapes drive complex developmental gene expression, but it remains unclear how their integrity is maintained when incorporating novel genes and functions during evolution. Here, we investigated how a placental mammal-specific gene, Zfp42, emerged in an ancient vertebrate topologically associated domain (TAD) without adopting or disrupting the conserved expression of its gene, Fat1. In ESCs, physical TAD partitioning separates Zfp42 and Fat1 with distinct local enhancers that drive their independent expression. This separation is driven by chromatin activity and not CTCF/cohesin. In contrast, in embryonic limbs, inactive Zfp42 shares Fat1's intact TAD without responding to active Fat1 enhancers. However, neither Fat1 enhancer-incompatibility nor nuclear envelope-attachment account for Zfp42's unresponsiveness. Rather, Zfp42's promoter is rendered inert to enhancers by context-dependent DNA methylation. Thus, diverse mechanisms enabled the integration of independent Zfp42 regulation in the Fat1 locus. Critically, such regulatory complexity appears common in evolution as, genome wide, most TADs contain multiple independently expressed genes.


Asunto(s)
Cromatina , Placenta , Animales , Factor de Unión a CCCTC/metabolismo , Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Evolución Molecular , Femenino , Genoma , Mamíferos/metabolismo , Placenta/metabolismo , Embarazo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Cell ; 161(5): 1012-1025, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25959774

RESUMEN

Mammalian genomes are organized into megabase-scale topologically associated domains (TADs). We demonstrate that disruption of TADs can rewire long-range regulatory architecture and result in pathogenic phenotypes. We show that distinct human limb malformations are caused by deletions, inversions, or duplications altering the structure of the TAD-spanning WNT6/IHH/EPHA4/PAX3 locus. Using CRISPR/Cas genome editing, we generated mice with corresponding rearrangements. Both in mouse limb tissue and patient-derived fibroblasts, disease-relevant structural changes cause ectopic interactions between promoters and non-coding DNA, and a cluster of limb enhancers normally associated with Epha4 is misplaced relative to TAD boundaries and drives ectopic limb expression of another gene in the locus. This rewiring occurred only if the variant disrupted a CTCF-associated boundary domain. Our results demonstrate the functional importance of TADs for orchestrating gene expression via genome architecture and indicate criteria for predicting the pathogenicity of human structural variants, particularly in non-coding regions of the human genome.


Asunto(s)
Modelos Animales de Enfermedad , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Animales , Extremidades/anatomía & histología , Extremidades/crecimiento & desarrollo , Humanos , Deformidades Congénitas de las Extremidades/genética , Ratones , Regiones Promotoras Genéticas , ARN no Traducido/genética , ARN no Traducido/metabolismo , Receptor EphA4/genética
3.
Development ; 150(17)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37519269

RESUMEN

Changes in gene expression represent an important source of phenotypic innovation. Yet how such changes emerge and impact the evolution of traits remains elusive. Here, we explore the molecular mechanisms associated with the development of masculinizing ovotestes in female moles. By performing integrative analyses of epigenetic and transcriptional data in mole and mouse, we identified the co-option of SALL1 expression in mole ovotestes formation. Chromosome conformation capture analyses highlight a striking conservation of the 3D organization at the SALL1 locus, but an evolutionary divergence of enhancer activity. Interspecies reporter assays support the capability of mole-specific enhancers to activate transcription in urogenital tissues. Through overexpression experiments in transgenic mice, we further demonstrate the capability of SALL1 to induce kidney-related gene programs, which are a signature of mole ovotestes. Our results highlight the co-option of gene expression, through changes in enhancer activity, as a plausible mechanism for the evolution of traits.


Asunto(s)
Riñón , Topos , Animales , Femenino , Ratones , Riñón/metabolismo , Ratones Transgénicos , Topos/genética
4.
Mol Psychiatry ; 24(11): 1748-1768, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29728705

RESUMEN

RLIM, also known as RNF12, is an X-linked E3 ubiquitin ligase acting as a negative regulator of LIM-domain containing transcription factors and participates in X-chromosome inactivation (XCI) in mice. We report the genetic and clinical findings of 84 individuals from nine unrelated families, eight of whom who have pathogenic variants in RLIM (RING finger LIM domain-interacting protein). A total of 40 affected males have X-linked intellectual disability (XLID) and variable behavioral anomalies with or without congenital malformations. In contrast, 44 heterozygous female carriers have normal cognition and behavior, but eight showed mild physical features. All RLIM variants identified are missense changes co-segregating with the phenotype and predicted to affect protein function. Eight of the nine altered amino acids are conserved and lie either within a domain essential for binding interacting proteins or in the C-terminal RING finger catalytic domain. In vitro experiments revealed that these amino acid changes in the RLIM RING finger impaired RLIM ubiquitin ligase activity. In vivo experiments in rlim mutant zebrafish showed that wild type RLIM rescued the zebrafish rlim phenotype, whereas the patient-specific missense RLIM variants failed to rescue the phenotype and thus represent likely severe loss-of-function mutations. In summary, we identified a spectrum of RLIM missense variants causing syndromic XLID and affecting the ubiquitin ligase activity of RLIM, suggesting that enzymatic activity of RLIM is required for normal development, cognition and behavior.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Adolescente , Adulto , Animales , Niño , Preescolar , Trastorno de la Conducta/genética , Femenino , Genes Ligados a X , Células HEK293 , Humanos , Recién Nacido , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Ratones , Persona de Mediana Edad , Mutación , Linaje , Factores de Transcripción/genética , Ubiquitinación , Inactivación del Cromosoma X , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Am J Hum Genet ; 97(2): 302-10, 2015 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-26166480

RESUMEN

Export of mRNA from the cell nucleus to the cytoplasm is essential for protein synthesis, a process vital to all living eukaryotic cells. mRNA export is highly conserved and ubiquitous. Mutations affecting mRNA and mRNA processing or export factors, which cause aberrant retention of mRNAs in the nucleus, are thus emerging as contributors to an important class of human genetic disorders. Here, we report that variants in THOC2, which encodes a subunit of the highly conserved TREX mRNA-export complex, cause syndromic intellectual disability (ID). Affected individuals presented with variable degrees of ID and commonly observed features included speech delay, elevated BMI, short stature, seizure disorders, gait disturbance, and tremors. X chromosome exome sequencing revealed four missense variants in THOC2 in four families, including family MRX12, first ascertained in 1971. We show that two variants lead to decreased stability of THOC2 and its TREX-complex partners in cells derived from the affected individuals. Protein structural modeling showed that the altered amino acids are located in the RNA-binding domains of two complex THOC2 structures, potentially representing two different intermediate RNA-binding states of THOC2 during RNA transport. Our results show that disturbance of the canonical molecular pathway of mRNA export is compatible with life but results in altered neuronal development with other comorbidities.


Asunto(s)
Transporte Activo de Núcleo Celular/genética , Cromosomas Humanos X/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Modelos Moleculares , Mutación Missense/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Secuencia de Aminoácidos , Secuencia de Bases , Humanos , Discapacidad Intelectual Ligada al Cromosoma X/patología , Datos de Secuencia Molecular , Linaje , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/química , Análisis de Secuencia de ADN , Síndrome
6.
Brain ; 140(11): 2879-2894, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29053855

RESUMEN

Genetic epilepsies are caused by mutations in a range of different genes, many of them encoding ion channels, receptors or transporters. While the number of detected variants and genes increased dramatically in the recent years, pleiotropic effects have also been recognized, revealing that clinical syndromes with various degrees of severity arise from a single gene, a single mutation, or from different mutations showing similar functional defects. Accordingly, several genes coding for GABAA receptor subunits have been linked to a spectrum of benign to severe epileptic disorders and it was shown that a loss of function presents the major correlated pathomechanism. Here, we identified six variants in GABRA3 encoding the α3-subunit of the GABAA receptor. This gene is located on chromosome Xq28 and has not been previously associated with human disease. Five missense variants and one microduplication were detected in four families and two sporadic cases presenting with a range of epileptic seizure types, a varying degree of intellectual disability and developmental delay, sometimes with dysmorphic features or nystagmus. The variants co-segregated mostly but not completely with the phenotype in the families, indicating in some cases incomplete penetrance, involvement of other genes, or presence of phenocopies. Overall, males were more severely affected and there were three asymptomatic female mutation carriers compared to only one male without a clinical phenotype. X-chromosome inactivation studies could not explain the phenotypic variability in females. Three detected missense variants are localized in the extracellular GABA-binding NH2-terminus, one in the M2-M3 linker and one in the M4 transmembrane segment of the α3-subunit. Functional studies in Xenopus laevis oocytes revealed a variable but significant reduction of GABA-evoked anion currents for all mutants compared to wild-type receptors. The degree of current reduction correlated partially with the phenotype. The microduplication disrupted GABRA3 expression in fibroblasts of the affected patient. In summary, our results reveal that rare loss-of-function variants in GABRA3 increase the risk for a varying combination of epilepsy, intellectual disability/developmental delay and dysmorphic features, presenting in some pedigrees with an X-linked inheritance pattern.


Asunto(s)
Encefalopatías/genética , Fisura del Paladar/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Facies , Discapacidad Intelectual/genética , Nistagmo Patológico/genética , Receptores de GABA-A/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Femenino , Variación Genética , Humanos , Masculino , Microcefalia/genética , Mutagénesis Sitio-Dirigida , Oocitos/metabolismo , Técnicas de Placa-Clamp , Linaje , Receptores de GABA-A/metabolismo , Síndrome , Xenopus laevis , Adulto Joven , Ácido gamma-Aminobutírico/metabolismo
7.
Hum Mutat ; 38(4): 409-425, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28055140

RESUMEN

Impairment of translation initiation and its regulation within the integrated stress response (ISR) and related unfolded-protein response has been identified as a cause of several multisystemic syndromes. Here, we link MEHMO syndrome, whose genetic etiology was unknown, to this group of disorders. MEHMO is a rare X-linked syndrome characterized by profound intellectual disability, epilepsy, hypogonadism and hypogenitalism, microcephaly, and obesity. We have identified a C-terminal frameshift mutation (Ile465Serfs) in the EIF2S3 gene in three families with MEHMO syndrome and a novel maternally inherited missense EIF2S3 variant (c.324T>A; p.Ser108Arg) in another male patient with less severe clinical symptoms. The EIF2S3 gene encodes the γ subunit of eukaryotic translation initiation factor 2 (eIF2), crucial for initiation of protein synthesis and regulation of the ISR. Studies in patient fibroblasts confirm increased ISR activation due to the Ile465Serfs mutation and functional assays in yeast demonstrate that the Ile465Serfs mutation impairs eIF2γ function to a greater extent than tested missense mutations, consistent with the more severe clinical phenotype of the Ile465Serfs male mutation carriers. Thus, we propose that more severe EIF2S3 mutations cause the full MEHMO phenotype, while less deleterious mutations cause a milder form of the syndrome with only a subset of the symptoms.


Asunto(s)
Epilepsia , Factor 2 Eucariótico de Iniciación/genética , Hipogonadismo , Discapacidad Intelectual/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Microcefalia , Mutación , Secuencia de Aminoácidos , Salud de la Familia , Femenino , Genitales/anomalías , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/patología , Obesidad , Linaje , Análisis de Secuencia de ADN/métodos , Homología de Secuencia de Aminoácido , Síndrome
8.
Hum Mol Genet ; 24(25): 7171-81, 2015 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-26443594

RESUMEN

Next generation genomic technologies have made a significant contribution to the understanding of the genetic architecture of human neurodevelopmental disorders. Copy number variants (CNVs) play an important role in the genetics of intellectual disability (ID). For many CNVs, and copy number gains in particular, the responsible dosage-sensitive gene(s) have been hard to identify. We have collected 18 different interstitial microduplications and 1 microtriplication of Xq25. There were 15 affected individuals from 6 different families and 13 singleton cases, 28 affected males in total. The critical overlapping region involved the STAG2 gene, which codes for a subunit of the cohesin complex that regulates cohesion of sister chromatids and gene transcription. We demonstrate that STAG2 is the dosage-sensitive gene within these CNVs, as gains of STAG2 mRNA and protein dysregulate disease-relevant neuronal gene networks in cells derived from affected individuals. We also show that STAG2 gains result in increased expression of OPHN1, a known X-chromosome ID gene. Overall, we define a novel cohesinopathy due to copy number gain of Xq25 and STAG2 in particular.


Asunto(s)
Antígenos Nucleares/genética , Discapacidad Intelectual/genética , Proteínas de Ciclo Celular , Cromosomas Humanos X/genética , Variaciones en el Número de Copia de ADN/genética , Humanos , Masculino , Problema de Conducta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
9.
Am J Hum Genet ; 95(6): 729-35, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25434005

RESUMEN

Advances in understanding the etiology of Parkinson disease have been driven by the identification of causative mutations in families. Genetic analysis of an Australian family with three males displaying clinical features of early-onset parkinsonism and intellectual disability identified a ∼45 kb deletion resulting in the complete loss of RAB39B. We subsequently identified a missense mutation (c.503C>A [p.Thr168Lys]) in RAB39B in an unrelated Wisconsin kindred affected by a similar clinical phenotype. In silico and in vitro studies demonstrated that the mutation destabilized the protein, consistent with loss of function. In vitro small-hairpin-RNA-mediated knockdown of Rab39b resulted in a reduction in the density of α-synuclein immunoreactive puncta in dendritic processes of cultured neurons. In addition, in multiple cell models, we demonstrated that knockdown of Rab39b was associated with reduced steady-state levels of α-synuclein. Post mortem studies demonstrated that loss of RAB39B resulted in pathologically confirmed Parkinson disease. There was extensive dopaminergic neuron loss in the substantia nigra and widespread classic Lewy body pathology. Additional pathological features included cortical Lewy bodies, brain iron accumulation, tau immunoreactivity, and axonal spheroids. Overall, we have shown that loss-of-function mutations in RAB39B cause intellectual disability and pathologically confirmed early-onset Parkinson disease. The loss of RAB39B results in dysregulation of α-synuclein homeostasis and a spectrum of neuropathological features that implicate RAB39B in the pathogenesis of Parkinson disease and potentially other neurodegenerative disorders.


Asunto(s)
Genes Ligados a X , Discapacidad Intelectual/genética , Degeneración Nerviosa/genética , Enfermedad de Parkinson/genética , alfa-Sinucleína/metabolismo , Proteínas de Unión al GTP rab/genética , Sustitución de Aminoácidos , Australia , Secuencia de Bases , Dopamina/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Discapacidad Intelectual/fisiopatología , Cuerpos de Lewy/metabolismo , Masculino , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Mutación Missense , Degeneración Nerviosa/fisiopatología , Enfermedad de Parkinson/fisiopatología , Linaje , Análisis de Secuencia de ADN , Eliminación de Secuencia , Sustancia Negra/fisiopatología , Proteínas de Unión al GTP rab/metabolismo
10.
Am J Hum Genet ; 92(5): 681-95, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23623388

RESUMEN

Arthrogryposis multiplex congenita (AMC) is caused by heterogeneous pathologies leading to multiple antenatal joint contractures through fetal akinesia. Understanding the pathophysiology of this disorder is important for clinical care of the affected individuals and genetic counseling of the families. We thus aimed to establish the genetic basis of an AMC subtype that is associated with multiple dysmorphic features and intellectual disability (ID). We used haplotype analysis, next-generation sequencing, array comparative genomic hybridization, and chromosome breakpoint mapping to identify the pathogenic mutations in families and simplex cases. Suspected disease variants were verified by cosegregation analysis. We identified disease-causing mutations in the zinc-finger gene ZC4H2 in four families affected by X-linked AMC plus ID and one family affected by cerebral palsy. Several heterozygous females were also affected, but to a lesser degree. Furthermore, we found two ZC4H2 deletions and one rearrangement in two female and one male unrelated simplex cases, respectively. In mouse primary hippocampal neurons, transiently produced ZC4H2 localized to the postsynaptic compartment of excitatory synapses, and the altered protein influenced dendritic spine density. In zebrafish, antisense-morpholino-mediated zc4h2 knockdown caused abnormal swimming and impaired α-motoneuron development. All missense mutations identified herein failed to rescue the swimming defect of zebrafish morphants. We conclude that ZC4H2 point mutations, rearrangements, and small deletions cause a clinically variable broad-spectrum neurodevelopmental disorder of the central and peripheral nervous systems in both familial and simplex cases of both sexes. Our results highlight the importance of ZC4H2 for genetic testing of individuals presenting with ID plus muscle weakness and minor or major forms of AMC.


Asunto(s)
Anomalías Múltiples/genética , Artrogriposis/genética , Proteínas Portadoras/genética , Predisposición Genética a la Enfermedad/genética , Discapacidad Intelectual/genética , Plasticidad Neuronal/genética , Dedos de Zinc/genética , Anomalías Múltiples/patología , Animales , Artrogriposis/patología , Células Cultivadas , Puntos de Rotura del Cromosoma , Hibridación Genómica Comparativa , Femenino , Haplotipos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Immunoblotting , Hibridación in Situ , Discapacidad Intelectual/patología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Mutación/genética , Proteínas Nucleares , Linaje , Sinapsis/genética , Pez Cebra
11.
Am J Med Genet A ; 170A(1): 94-102, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26358559

RESUMEN

The clinical diagnosis of Lujan-Fryns syndrome (LFS) comprises X-linked intellectual disability (XLID) with marfanoid habitus, distinct combination of minor facial anomalies and nasal speech. However the definition of syndrome was significantly broadened since the original report and implies ID with marfanoid habitus. Mutations of three genes (MED12, UPF3B, and ZDHHC9) have been reported in "broadly defined" LFS. We examined these genes in 28 individuals with a tentative clinical diagnosis of LFS but we did not identify any causative mutation. By molecular karyotyping we detected other disorders, i.e., Phelan-McDermid syndrome and 16p11.2 microduplication, each in one patient. One affected individual was carrier of a different recurrent duplication on 16p11.2 that has been reported several times to the DECIPHER and ISCA databases in individuals with autism, intellectual disability (ID), and developmental delay. It may represent a new duplication syndrome. We also identified previously unreported de novo duplication on chromosome 12p13.31 which we considered to be disease-causing. X-exome sequencing of four individuals revealed private or non-recurrent mutations in NKAP and LAS1L in one patient each. While LFS is defined as a form of XLID, there seem to be various conditions that have rather similar phenotypes. Therefore, the combination of ID and marfanoid habitus in a male patient is not sufficient for the diagnosis of LFS. We suggest that the diagnosis of LFS in patients with ID and marfanoid habitus should be made only in presence of specific facial features, nasal speech and obvious X-linked segregation of the disorder or an unambiguously pathogenic mutation in the MED12.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Craneofaciales/diagnóstico , Genes Ligados a X/genética , Discapacidad Intelectual/diagnóstico , Síndrome de Marfan/diagnóstico , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Mutación/genética , Anomalías Múltiples/genética , Aciltransferasas/genética , Anomalías Craneofaciales/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/genética , Masculino , Síndrome de Marfan/genética , Complejo Mediador/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Linaje , Proteínas de Unión al ARN/genética
12.
Hum Mutat ; 36(12): 1155-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26290468

RESUMEN

RPL10 encodes ribosomal protein L10 (uL16), a highly conserved multifunctional component of the large ribosomal subunit, involved in ribosome biogenesis and function. Using X-exome resequencing, we identified a novel missense mutation (c.191C>T; p.(A64V)) in the N-terminal domain of the protein, in a family with two affected cousins presenting with X-linked intellectual disability, cerebellar hypoplasia, and spondylo-epiphyseal dysplasia (SED). We assessed the impact of the mutation on the translational capacity of the cell using yeast as model system. The mutation generates a functional ribosomal protein, able to complement the translational defects of a conditional lethal mutation of yeast rpl10. However, unlike previously reported mutations, this novel RPL10 missense mutation results in an increase in the actively translating ribosome population. Our results expand the mutational and clinical spectrum of RPL10 identifying a new genetic cause of SED and highlight the emerging role of ribosomal proteins in the pathogenesis of neurodevelopmental disorders.


Asunto(s)
Cerebelo/anomalías , Genes Ligados a X , Discapacidad Intelectual/genética , Mutación , Malformaciones del Sistema Nervioso/genética , Osteocondrodisplasias/genética , Proteínas Ribosómicas/genética , Preescolar , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Femenino , Estudios de Asociación Genética , Heterocigoto , Humanos , Discapacidad Intelectual/diagnóstico , Imagen por Resonancia Magnética , Masculino , Malformaciones del Sistema Nervioso/diagnóstico , Neuroimagen , Osteocondrodisplasias/diagnóstico , Fenotipo , Proteína Ribosómica L10 , Proteínas Ribosómicas/metabolismo , Análisis de Secuencia de ADN , Inactivación del Cromosoma X
13.
Hum Mutat ; 36(1): 106-17, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25385192

RESUMEN

Variants in cullin 4B (CUL4B) are a known cause of syndromic X-linked intellectual disability. Here, we describe an additional 25 patients from 11 families with variants in CUL4B. We identified nine different novel variants in these families and confirmed the pathogenicity of all nontruncating variants. Neuroimaging data, available for 15 patients, showed the presence of cerebral malformations in ten patients. The cerebral anomalies comprised malformations of cortical development (MCD), ventriculomegaly, and diminished white matter volume. The phenotypic heterogeneity of the cerebral malformations might result from the involvement of CUL-4B in various cellular pathways essential for normal brain development. Accordingly, we show that CUL-4B interacts with WDR62, a protein in which variants were previously identified in patients with microcephaly and a wide range of MCD. This interaction might contribute to the development of cerebral malformations in patients with variants in CUL4B.


Asunto(s)
Encéfalo/patología , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Malformaciones del Desarrollo Cortical/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteínas del Tejido Nervioso/metabolismo , Adolescente , Adulto , Proteínas de Ciclo Celular , Células Cultivadas , Niño , Preescolar , Estudios de Asociación Genética , Células HEK293 , Humanos , Lactante , Masculino , Malformaciones del Desarrollo Cortical/metabolismo , Malformaciones del Desarrollo Cortical/patología , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Discapacidad Intelectual Ligada al Cromosoma X/patología , Persona de Mediana Edad , Linaje , Análisis de Secuencia de ADN , Adulto Joven
14.
Hum Mol Genet ; 22(16): 3306-14, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23615299

RESUMEN

Existence of a discrete new X-linked intellectual disability (XLID) syndrome due to KIAA2022 deficiency was questioned by disruption of KIAA2022 by an X-chromosome pericentric inversion in a XLID family we reported in 2004. Three additional families with likely pathogenic KIAA2022 mutations were discovered within the frame of systematic parallel sequencing of familial cases of XLID or in the context of routine array-CGH evaluation of sporadic intellectual deficiency (ID) cases. The c.186delC and c.3597dupA KIAA2022 truncating mutations were identified by X-chromosome exome sequencing, while array CGH discovered a 70 kb microduplication encompassing KIAA2022 exon 1 in the third family. This duplication decreased KIAA2022 mRNA level in patients' lymphocytes by 60%. Detailed clinical examination of all patients, including the two initially reported, indicated moderate-to-severe ID with autistic features, strabismus in all patients, with no specific dysmorphic features other than a round face in infancy and no structural brain abnormalities on magnetic resonance imaging (MRI). Interestingly, the patient with decreased KIAA2022 expression had only mild ID with severe language delay and repetitive behaviors falling in the range of an autism spectrum disorder (ASD). Since little is known about KIAA2022 function, we conducted morphometric studies in cultured rat hippocampal neurons. We found that siRNA-mediated KIAA2022 knockdown resulted in marked impairment in neurite outgrowth including both the dendrites and the axons, suggesting a major role for KIAA2022 in neuron development and brain function.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Genes Ligados a X , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Neuritas/fisiología , Adolescente , Adulto , Animales , Encéfalo/metabolismo , Células Cultivadas , Niño , Preescolar , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Variación Genética , Humanos , Masculino , Neuritas/metabolismo , Ratas , Análisis de Secuencia de ARN , Adulto Joven
15.
Am J Hum Genet ; 91(4): 694-702, 2012 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23000143

RESUMEN

The discovery of mutations causing human disease has so far been biased toward protein-coding regions. Having excluded all annotated coding regions, we performed targeted massively parallel resequencing of the nonrepetitive genomic linkage interval at Xq28 of family MRX3. We identified in the binding site of transcription factor YY1 a regulatory mutation that leads to overexpression of the chromatin-associated transcriptional regulator HCFC1. When tested on embryonic murine neural stem cells and embryonic hippocampal neurons, HCFC1 overexpression led to a significant increase of the production of astrocytes and a considerable reduction in neurite growth. Two other nonsynonymous, potentially deleterious changes have been identified by X-exome sequencing in individuals with intellectual disability, implicating HCFC1 in normal brain function.


Asunto(s)
Factor C1 de la Célula Huésped/genética , Discapacidad Intelectual/genética , Mutación , ARN no Traducido/genética , Secuencia de Aminoácidos , Animales , Astrocitos/metabolismo , Sitios de Unión , Cromatina/genética , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/genética , Ratones , Datos de Secuencia Molecular , Factores de Transcripción/genética , Cromosoma X/genética , Factor de Transcripción YY1/genética
16.
J Med Genet ; 51(7): 487-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24812067

RESUMEN

INTRODUCTION: Kinesin superfamily (KIF) genes encode motor proteins that have fundamental roles in brain functioning, development, survival and plasticity by regulating the transport of cargo along microtubules within axons, dendrites and synapses. Mouse knockout studies support these important functions in the nervous system. The role of KIF genes in intellectual disability (ID) has so far received limited attention, although previous studies have suggested that many ID genes impinge on synaptic function. METHODS: By applying next-generation sequencing (NGS) in ID patients, we identified likely pathogenic mutations in KIF4A and KIF5C. To further confirm the pathogenicity of these mutations, we performed functional studies at the level of synaptic function in primary rat hippocampal neurons. RESULTS AND CONCLUSIONS: Four males from a single family with a disruptive mutation in the X-linked KIF4A (c.1489-8_1490delins10; p.?- exon skipping) showed mild to moderate ID and epilepsy. A female patient with a de novo missense mutation in KIF5C (c.11465A>C; p.(Glu237Lys)) presented with severe ID, epilepsy, microcephaly and cortical malformation. Knock-down of Kif4a in rat primary hippocampal neurons altered the balance between excitatory and inhibitory synaptic transmission, whereas the mutation in Kif5c affected its protein function at excitatory synapses. Our results suggest that mutations in KIF4A and KIF5C cause ID by tipping the balance between excitatory and inhibitory synaptic excitability.


Asunto(s)
Discapacidad Intelectual/genética , Cinesinas/genética , Adolescente , Animales , Secuencia de Bases , Células Cultivadas , Niño , Análisis Mutacional de ADN , Exones , Femenino , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación Missense , Neuronas/fisiología , Linaje , Cultivo Primario de Células , Ratas , Sinapsis/fisiología , Transmisión Sináptica
17.
Am J Hum Genet ; 88(5): 628-34, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21549342

RESUMEN

In a Dutch family with an X-linked postlingual progressive hearing impairment, a critical linkage interval was determined to span a region of 12.9 Mb flanked by the markers DXS7108 and DXS7110. This interval overlaps with the previously described DFNX4 locus and contains 75 annotated genes. Subsequent next-generation sequencing (NGS) detected one variant within the linkage interval, a nonsense mutation in SMPX. SMPX encodes the small muscle protein, X-linked (SMPX). Further screening was performed on 26 index patients from small families for which X-linked inheritance of nonsyndromic hearing impairment (NSHI) was not excluded. We detected a frameshift mutation in SMPX in one of the patients. Segregation analysis of both mutations in the families in whom they were found revealed that the mutations cosegregated with hearing impairment. Although we show that SMPX is expressed in many different organs, including the human inner ear, no obvious symptoms other than hearing impairment were observed in the patients. SMPX had previously been demonstrated to be specifically expressed in striated muscle and, therefore, seemed an unlikely candidate gene for hearing impairment. We hypothesize that SMPX functions in inner ear development and/or maintenance in the IGF-1 pathway, the integrin pathway through Rac1, or both.


Asunto(s)
Codón sin Sentido , Genes Ligados a X , Pérdida Auditiva/genética , Proteínas Musculares/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Mutación del Sistema de Lectura , Pérdida Auditiva/patología , Humanos , Factor I del Crecimiento Similar a la Insulina/genética , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia , Adulto Joven
18.
Bioinformatics ; 28(7): 1024-5, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22302574

RESUMEN

SUMMARY: We developed Breakpointer, a fast algorithm to locate breakpoints of structural variants (SVs) from single-end reads produced by next-generation sequencing. By taking advantage of local non-uniform read distribution and misalignments created by SVs, Breakpointer scans the alignment of single-end reads to identify regions containing potential breakpoints. The detection of such breakpoints can indicate insertions longer than the read length and SVs located in repetitve regions which might be missd by other methods. Thus, Breakpointer complements existing methods to locate SVs from single-end reads. AVAILABILITY: https://github.com/ruping/Breakpointer CONTACT: ruping@molgen.mpg.de SUPPLEMENTARY INFORMATION: Supplementary material is available at Bioinformatics online.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Variación Estructural del Genoma , Análisis de Secuencia de ADN/métodos , Artefactos , Humanos
19.
Bioinformatics ; 28(5): 619-27, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22238266

RESUMEN

MOTIVATION: The reliable detection of genomic variation in resequencing data is still a major challenge, especially for variants larger than a few base pairs. Sequencing reads crossing boundaries of structural variation carry the potential for their identification, but are difficult to map. RESULTS: Here we present a method for 'split' read mapping, where prefix and suffix match of a read may be interrupted by a longer gap in the read-to-reference alignment. We use this method to accurately detect medium-sized insertions and long deletions with precise breakpoints in genomic resequencing data. Compared with alternative split mapping methods, SplazerS significantly improves sensitivity for detecting large indel events, especially in variant-rich regions. Our method is robust in the presence of sequencing errors as well as alignment errors due to genomic mutations/divergence, and can be used on reads of variable lengths. Our analysis shows that SplazerS is a versatile tool applicable to unanchored or single-end as well as anchored paired-end reads. In addition, application of SplazerS to targeted resequencing data led to the interesting discovery of a complete, possibly functional gene retrocopy variant. AVAILABILITY: SplazerS is available from http://www.seqan.de/projects/ splazers. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Genómica/métodos , Mutación INDEL , Análisis de Secuencia de ADN , Algoritmos , Humanos
20.
Am J Med Genet A ; 161A(12): 3063-71, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24039113

RESUMEN

FG syndrome, Lujan syndrome, and Ohdo syndrome, the Maat-Kievit-Brunner type, have been described as distinct syndromes with overlapping non-specific features and different missense mutations of the MED12 gene have been reported in all of them. We report a family including 10 males and 1 female affected with profound non-specific intellectual disability (ID) which was linked to a 30-cM region extending from Xp11.21 (ALAS2) to Xq22.3 (COL4A5). Parallel sequencing of all X-chromosome exons identified a frameshift mutation (c.5898dupC) of MED12. Mutated mRNA was not affected by non-sense mediated RNA decay and induced an additional abnormal isoform due to activation of cryptic splice-sites in exon 41. Dysmorphic features common to most affected males were long narrow face, high forehead, flat malar area, high nasal bridge, and short philtrum. Language was absent or very limited. Most patients had a friendly personality. Cognitive impairment, varying from borderline to profound ID was similarly observed in seven heterozygous females. There was no correlation between cognitive function and X-chromosome inactivation profiles in blood cells. The severe degree of ID in male patients, as well as variable cognitive impairment in heterozygous females suggests that the duplication observed in the present family may have a more severe effect on MED12 function than missense mutations. In a cognitively impaired male from this family, who also presented with tall stature and dysmorphism and did not have the MED12 mutation, a 600-kb duplication at 17p13.3 including the YWHAE gene, was found in a mosaic state.


Asunto(s)
Anomalías Múltiples/genética , Agenesia del Cuerpo Calloso/genética , Ano Imperforado/genética , Blefarofimosis/genética , Blefaroptosis/genética , Estreñimiento/genética , Enfermedades Genéticas Ligadas al Cromosoma X , Cardiopatías Congénitas/genética , Discapacidad Intelectual/genética , Complejo Mediador/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Hipotonía Muscular/congénito , Proteínas 14-3-3/genética , Anomalías Múltiples/fisiopatología , Adulto , Anciano , Agenesia del Cuerpo Calloso/fisiopatología , Ano Imperforado/fisiopatología , Blefarofimosis/fisiopatología , Blefaroptosis/fisiopatología , Cromosomas Humanos X/genética , Estreñimiento/fisiopatología , Exones , Femenino , Mutación del Sistema de Lectura , Cardiopatías Congénitas/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Persona de Mediana Edad , Hipotonía Muscular/genética , Hipotonía Muscular/fisiopatología , Mutación , Inactivación del Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA