Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Hum Mutat ; 43(12): 1816-1823, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36317458

RESUMEN

Advanced bioinformatics algorithms allow detection of multiple-exon copy-number variations (CNVs) from exome sequencing (ES) data, while detection of single-exon CNVs remains challenging. A retrospective review of Baylor Genetics' clinical ES patient cohort identified four individuals with homozygous single-exon deletions of TBCK (exon 23, NM_001163435.2), a gene associated with an autosomal recessive neurodevelopmental phenotype. To evaluate the prevalence of this deletion and its contribution to disease, we retrospectively analyzed single nucleotide polymorphism (SNP) array data for 8194 individuals undergoing ES, followed by PCR confirmation and RT-PCR on individuals carrying homozygous or heterozygous exon 23 TBCK deletions. A fifth individual was diagnosed with the TBCK-related disorder due to a heterozygous exon 23 deletion in trans with a c.1860+1G>A (NM_001163435.2) pathogenic variant, and three additional heterozygous carriers were identified. Affected individuals and carriers were from diverse ethnicities including European Caucasian, South Asian, Middle Eastern, Hispanic American and African American, with only one family reporting consanguinity. RT-PCR revealed two out-of-frame transcripts related to the exon 23 deletion. Our results highlight the importance of identifying single-exon deletions in clinical ES, especially for genes carrying recurrent deletions. For patients with early-onset hypotonia and psychomotor delay, this single-exon TBCK deletion might be under-recognized due to technical limitations of ES.


Asunto(s)
Hipotonía Muscular , Enfermedades Musculares , Proteínas Serina-Treonina Quinasas , Humanos , Variaciones en el Número de Copia de ADN , Exoma , Secuenciación del Exoma , Exones/genética , Hipotonía Muscular/genética , Enfermedades Musculares/genética , Proteínas Serina-Treonina Quinasas/genética , Estudios Retrospectivos , Lactante
2.
Genome Med ; 14(1): 113, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36180924

RESUMEN

BACKGROUND: In medical genetics, discovery and characterization of disease trait contributory genes and alleles depends on genetic reasoning, study design, and patient ascertainment; we suggest a segmental haploid genetics approach to enhance gene discovery and molecular diagnostics. METHODS: We constructed a genome-wide map for nonallelic homologous recombination (NAHR)-mediated recurrent genomic deletions and used this map to estimate population frequencies of NAHR deletions based on large-scale population cohorts and region-specific studies. We calculated recessive disease carrier burden using high-quality pathogenic or likely pathogenic variants from ClinVar and gnomAD. We developed a NIRD (NAHR deletion Impact to Recessive Disease) score for recessive disorders by quantifying the contribution of NAHR deletion to the overall allele load that enumerated all pairwise combinations of disease-causing alleles; we used a Punnett square approach based on an assumption of random mating. Literature mining was conducted to identify all reported patients with defects in a gene with a high NIRD score; meta-analysis was performed on these patients to estimate the representation of NAHR deletions in recessive traits from contemporary human genomics studies. Retrospective analyses of extant clinical exome sequencing (cES) were performed for novel rare recessive disease trait gene and allele discovery from individuals with NAHR deletions. RESULTS: We present novel genomic insights regarding the genome-wide impact of NAHR recurrent segmental variants on recessive disease burden; we demonstrate the utility of NAHR recurrent deletions to enhance discovery in the challenging context of autosomal recessive (AR) traits and biallelic variation. Computational results demonstrate new mutations mediated by NAHR, involving recurrent deletions at 30 genomic regions, likely drive recessive disease burden for over 74% of loci within these segmental deletions or at least 2% of loci genome-wide. Meta-analyses on 170 literature-reported patients implicate that NAHR deletions are depleted from the ascertained pool of AR trait alleles. Exome reanalysis of personal genomes from subjects harboring recurrent deletions uncovered new disease-contributing variants in genes including COX10, ERCC6, PRRT2, and OTUD7A. CONCLUSIONS: Our results demonstrate that genomic sequencing of personal genomes with NAHR deletions could dramatically improve allele and gene discovery and enhance clinical molecular diagnosis. Moreover, results suggest NAHR events could potentially enable human haploid genetic screens as an approach to experimental inquiry into disease biology.


Asunto(s)
Genómica , Enfermedades Raras , Secuencia de Bases , Recombinación Homóloga , Humanos , Enfermedades Raras/genética , Estudios Retrospectivos
3.
Genet Med ; 24(2): 364-373, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906496

RESUMEN

PURPOSE: BRG1/BRM-associated factor (BAF) complex is a chromatin remodeling complex that plays a critical role in gene regulation. Defects in the genes encoding BAF subunits lead to BAFopathies, a group of neurodevelopmental disorders with extensive locus and phenotypic heterogeneity. METHODS: We retrospectively analyzed data from 16,243 patients referred for clinical exome sequencing (ES) with a focus on the BAF complex. We applied a genotype-first approach, combining predicted genic constraints to propose candidate BAFopathy genes. RESULTS: We identified 127 patients carrying pathogenic variants, likely pathogenic variants, or de novo variants of unknown clinical significance in 11 known BAFopathy genes. Those include 34 patients molecularly diagnosed using ES reanalysis with new gene-disease evidence (n = 21) or variant reclassifications in known BAFopathy genes (n = 13). We also identified de novo or predicted loss-of-function variants in 4 candidate BAFopathy genes, including ACTL6A, BICRA (implicated in Coffin-Siris syndrome during this study), PBRM1, and SMARCC1. CONCLUSION: We report the mutational spectrum of BAFopathies in an ES cohort. A genotype-driven and pathway-based reanalysis of ES data identified new evidence for candidate genes involved in BAFopathies. Further mechanistic and phenotypic characterization of additional patients are warranted to confirm their roles in human disease and to delineate their associated phenotypic spectrums.


Asunto(s)
Anomalías Múltiples , Deformidades Congénitas de la Mano , Micrognatismo , Anomalías Múltiples/genética , Actinas/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Exoma/genética , Deformidades Congénitas de la Mano/genética , Humanos , Micrognatismo/genética , Estudios Retrospectivos
4.
Clin Genet ; 100(2): 227-233, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33963760

RESUMEN

PPP3CA encodes the catalytic subunit of calcineurin, a calcium-calmodulin-regulated serine-threonine phosphatase. Loss-of-function (LoF) variants in the catalytic domain have been associated with epilepsy, while gain-of-function (GoF) variants in the auto-inhibitory domain cause multiple congenital abnormalities. We herein report five new patients with de novo PPP3CA variants. Interestingly, the two frameshift variants in this study and the six truncating variants reported previously are all located within a 26-amino acid region in the regulatory domain (RD). Patients with a truncating variant had more severe earlier onset seizures compared to patients with a LoF missense variant, while autism spectrum disorder was a more frequent feature in the latter. Expression studies of a truncating variant showed apparent RNA expression from the mutant allele, but no detectable mutant protein. Our data suggest that PPP3CA truncating variants clustered in the RD, causing more severe early-onset refractory epilepsy and representing a type of variants distinct from LoF or GoF missense variants.


Asunto(s)
Calcineurina/genética , Epilepsia/genética , Mutación , Adolescente , Calcineurina/metabolismo , Niño , Preescolar , Epilepsia Refractaria/etiología , Epilepsia Refractaria/genética , Epilepsia/etiología , Femenino , Expresión Génica , Humanos , Masculino , Análisis de Secuencia de ARN
5.
Pediatr Neurol ; 114: 16-20, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33189025

RESUMEN

BACKGROUND: Pathogenic variants in the IGHMBP2 gene cause recessive spinal motor neuropathies of variable phenotype, including a predominantly distal motor impairment of Charcot-Marie-Tooth type 2S and the more severe condition of spinal muscular atrophy with respiratory distress type 1 in which infantile respiratory failure predominates. METHODS: We describe the first reported case of spinal muscular atrophy with respiratory distress type 1 caused by a novel deep intronic variant in IGHMBP2 (NM_002180c.712-610A>G). RESULTS: The variant was detected by whole genome sequencing. Reverse transcription-polymerase chain reaction and complimentary DNA sequencing were used to characterize the impact of the novel variant. CONCLUSIONS: This report illustrates the utility in clinical practice of genome sequencing and RNA analysis, compared with exome sequencing alone.


Asunto(s)
Proteínas de Unión al ADN/genética , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/fisiopatología , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/fisiopatología , Factores de Transcripción/genética , Humanos , Lactante , Masculino , Atrofia Muscular Espinal/diagnóstico , Atrofia Muscular Espinal/patología , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Análisis de Secuencia de ARN , Secuenciación Completa del Genoma
6.
Genet Med ; 22(10): 1633-1641, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32576985

RESUMEN

PURPOSE: Improved resolution of molecular diagnostic technologies enabled detection of smaller sized exonic level copy-number variants (CNVs). The contribution of CNVs to autosomal recessive (AR) conditions may be better recognized using a large clinical cohort. METHODS: We retrospectively investigated the CNVs' contribution to AR conditions in cases subjected to chromosomal microarray analysis (CMA, N = ~70,000) and/or clinical exome sequencing (ES, N = ~12,000) at Baylor Genetics; most had pediatric onset neurodevelopmental disorders. RESULTS: CNVs contributed to biallelic variations in 87 cases, including 81 singletons and three affected sibling pairs. Seventy cases had CNVs affecting both alleles, and 17 had a CNV and a single-nucleotide variant (SNV)/indel in trans. In total, 94.3% of AR-CNVs affected one gene; among these 41.4% were single-exon and 35.0% were multiexon partial-gene events. Sixty-nine percent of homozygous AR-CNVs were embedded in homozygous genomic intervals. Five cases had large deletions unmasking an SNV/indel on the intact allele for a recessive condition, resulting in multiple molecular diagnoses. CONCLUSIONS: AR-CNVs are often smaller in size, transmitted through generations, and underrecognized due to limitations in clinical CNV detection methods. Our findings from a large clinical cohort emphasized integrated CNV and SNV/indel analyses for precise clinical and molecular diagnosis especially in the context of genomic disorders.


Asunto(s)
Variaciones en el Número de Copia de ADN , Mutación INDEL , Niño , Variaciones en el Número de Copia de ADN/genética , Exones , Humanos , Estudios Retrospectivos , Secuenciación del Exoma
7.
Am J Med Genet A ; 182(1): 189-194, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31633297

RESUMEN

Hennekam lymphangiectasia-lymphedema syndrome is an autosomal recessive disorder characterized by congenital lymphedema, intestinal lymphangiectasia, facial dysmorphism, and variable intellectual disability. Known disease genes include CCBE1, FAT4, and ADAMTS3. In a patient with clinically diagnosed Hennekam syndrome but without mutations or copy-number changes in the three known disease genes, we identified a homozygous single-exon deletion affecting FBXL7. Specifically, exon 3, which encodes the F-box domain and several leucine-rich repeats of FBXL7, is eliminated. Our analyses of databases representing >100,000 control individuals failed to identify biallelic loss-of-function variants in FBXL7. Published studies in Drosophila indicate Fbxl7 interacts with Fat, of which human FAT4 is an ortholog, and mutation of either gene yields similar morphological consequences. These data suggest that FBXL7 may be the fourth gene for Hennekam syndrome, acting via a shared pathway with FAT4.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas F-Box/genética , Predisposición Genética a la Enfermedad , Linfangiectasia Intestinal/genética , Linfedema/genética , Proteínas ADAMTS/genética , Alelos , Animales , Preescolar , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/patología , Drosophila melanogaster/genética , Genotipo , Homocigoto , Humanos , Linfangiectasia Intestinal/complicaciones , Linfangiectasia Intestinal/patología , Linfedema/complicaciones , Linfedema/patología , Masculino , Técnicas de Diagnóstico Molecular/métodos , Mutación/genética , Linaje , Fenotipo , Procolágeno N-Endopeptidasa/genética
9.
Genome Med ; 11(1): 12, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30819258

RESUMEN

BACKGROUND: Neurodevelopmental disorders are genetically and phenotypically heterogeneous encompassing developmental delay (DD), intellectual disability (ID), autism spectrum disorders (ASDs), structural brain abnormalities, and neurological manifestations with variants in a large number of genes (hundreds) associated. To date, a few de novo mutations potentially disrupting TCF20 function in patients with ID, ASD, and hypotonia have been reported. TCF20 encodes a transcriptional co-regulator structurally related to RAI1, the dosage-sensitive gene responsible for Smith-Magenis syndrome (deletion/haploinsufficiency) and Potocki-Lupski syndrome (duplication/triplosensitivity). METHODS: Genome-wide analyses by exome sequencing (ES) and chromosomal microarray analysis (CMA) identified individuals with heterozygous, likely damaging, loss-of-function alleles in TCF20. We implemented further molecular and clinical analyses to determine the inheritance of the pathogenic variant alleles and studied the spectrum of phenotypes. RESULTS: We report 25 unique inactivating single nucleotide variants/indels (1 missense, 1 canonical splice-site variant, 18 frameshift, and 5 nonsense) and 4 deletions of TCF20. The pathogenic variants were detected in 32 patients and 4 affected parents from 31 unrelated families. Among cases with available parental samples, the variants were de novo in 20 instances and inherited from 4 symptomatic parents in 5, including in one set of monozygotic twins. Two pathogenic loss-of-function variants were recurrent in unrelated families. Patients presented with a phenotype characterized by developmental delay, intellectual disability, hypotonia, variable dysmorphic features, movement disorders, and sleep disturbances. CONCLUSIONS: TCF20 pathogenic variants are associated with a novel syndrome manifesting clinical characteristics similar to those observed in Smith-Magenis syndrome. Together with previously described cases, the clinical entity of TCF20-associated neurodevelopmental disorders (TAND) emerges from a genotype-driven perspective.


Asunto(s)
Anomalías Craneofaciales/genética , Discapacidades del Desarrollo/genética , Mutación INDEL , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Síndrome de Smith-Magenis/genética , Factores de Transcripción/genética , Adolescente , Niño , Preescolar , Anomalías Craneofaciales/patología , Discapacidades del Desarrollo/patología , Femenino , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Hipotonía Muscular/patología , Síndrome de Smith-Magenis/patología , Factores de Transcripción/metabolismo , Adulto Joven
10.
Am J Hum Genet ; 103(1): 154-162, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29961569

RESUMEN

TRAF7 is a multi-functional protein involved in diverse signaling pathways and cellular processes. The phenotypic consequence of germline TRAF7 variants remains unclear. Here we report missense variants in TRAF7 in seven unrelated individuals referred for clinical exome sequencing. The seven individuals share substantial phenotypic overlap, with developmental delay, congenital heart defects, limb and digital anomalies, and dysmorphic features emerging as key unifying features. The identified variants are de novo in six individuals and comprise four distinct missense changes, including a c.1964G>A (p.Arg655Gln) variant that is recurrent in four individuals. These variants affect evolutionarily conserved amino acids and are located in key functional domains. Gene-specific mutation rate analysis showed that the occurrence of the de novo variants in TRAF7 (p = 2.6 × 10-3) and the recurrent de novo c.1964G>A (p.Arg655Gln) variant (p = 1.9 × 10-8) in our exome cohort was unlikely to have occurred by chance. In vitro analyses of the observed TRAF7 mutations showed reduced ERK1/2 phosphorylation. Our findings suggest that missense mutations in TRAF7 are associated with a multisystem disorder and provide evidence of a role for TRAF7 in human development.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Mutación Missense/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Adulto , Aminoácidos/genética , Niño , Preescolar , Exoma/genética , Femenino , Cardiopatías Congénitas/genética , Humanos , Lactante , Recién Nacido , Sistema de Señalización de MAP Quinasas/genética , Masculino , Anomalías Musculoesqueléticas/genética , Fenotipo
11.
Am J Med Genet A ; 176(4): 973-979, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29423971

RESUMEN

SHANK3 encodes for a scaffolding protein that links neurotransmitter receptors to the cytoskeleton and is enriched in postsynaptic densities of excitatory synapses. Deletions or mutations in one copy of the SHANK3 gene cause Phelan-McDermid syndrome, also called 22q13.3 deletion syndrome, a neurodevelopmental disorder with common features including global developmental delay, absent to severely impaired language, autistic behavior, and minor dysmorphic features. By whole exome sequencing, we identified two de novo novel variants including one frameshift pathogenic variant and one missense variant of unknown significance in a 14-year-old boy with delayed motor milestones, delayed language acquisition, autism, intellectual disability, ataxia, progressively worsening spasticity of the lower extremities, dysmorphic features, short stature, microcephaly, failure to thrive, chronic constipation, intrauterine growth restriction, and bilateral inguinal hernias. Both changes are within the CpG island in exon 21, separated by a 375 bp sequence. Next generation sequencing of PCR products revealed that the two variants are most frequently associated with each other. Sanger sequencing of the cloned PCR products further confirmed that both changes were on a single allele. The clinical presentation in this individual is consistent with other patients with a truncating mutation in exon 21, suggesting that the missense change contributes none or minimally to the phenotypes. This is the first report of two de novo mutations in one SHANK3 allele.


Asunto(s)
Alelos , Trastorno Autístico/diagnóstico , Trastorno Autístico/genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Mutación , Proteínas del Tejido Nervioso/genética , Adolescente , Análisis Mutacional de ADN , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Fenotipo , Secuenciación del Exoma
13.
Hum Genet ; 136(4): 377-386, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28251352

RESUMEN

Impairment of ubiquitin-proteasome system activity involving ubiquitin ligase genes UBE3A, UBE3B, and HUWE1 and deubiquitinating enzyme genes USP7 and USP9X has been reported in patients with neurodevelopmental delays. To date, only a handful of single-nucleotide variants (SNVs) and copy-number variants (CNVs) involving TRIP12, encoding a member of the HECT domain E3 ubiquitin ligases family on chromosome 2q36.3 have been reported. Using chromosomal microarray analysis and whole-exome sequencing (WES), we have identified, respectively, five deletion CNVs and four inactivating SNVs (two frameshifts, one missense, and one splicing) in TRIP12. Seven of these variants were found to be de novo; parental studies could not be completed in two families. Quantitative PCR analyses of the splicing mutation showed a dramatically decreased level of TRIP12 mRNA in the proband compared to the family controls, indicating a loss-of-function mechanism. The shared clinical features include intellectual disability with or without autistic spectrum disorders, speech delay, and facial dysmorphism. Our findings demonstrate that E3 ubiquitin ligase TRIP12 plays an important role in nervous system development and function. The nine presented pathogenic variants further document that TRIP12 haploinsufficiency causes a childhood-onset neurodevelopmental disorder. Finally, our data enable expansion of the phenotypic spectrum of ubiquitin-proteasome dependent disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas Portadoras/genética , Facies , Haploinsuficiencia , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Trastorno del Espectro Autista/complicaciones , Niño , Preescolar , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Femenino , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Trastornos del Desarrollo del Lenguaje/complicaciones , Masculino
14.
Am J Med Genet A ; 170(8): 2206-11, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27250922

RESUMEN

Mutations in CRIPT encoding cysteine-rich PDZ domain-binding protein are rare, and to date have been reported in only two patients with autosomal recessive primordial dwarfism and distinctive facies. Here, we describe a female with biallelic mutations in CRIPT presenting with postnatal growth retardation, global developmental delay, and dysmorphic features including frontal bossing, high forehead, and sparse hair and eyebrows. Additional clinical features included high myopia, admixed hyper- and hypopigmented macules primarily on the face, arms, and legs, and syndactyly of 4-5 toes bilaterally. Using whole exome sequencing (WES) and chromosomal microarray analysis (CMA), we detected a c.8G>A (p.C3Y) missense variant in exon 1 of the CRIPT gene inherited from the mother and a 1,331 bp deletion encompassing exon 1, inherited from the father. The c.8G>A (p.C3Y) missense variant in CRIPT was apparently homozygous in the proband due to the exon 1 deletion. Our findings illustrate the clinical utility of combining WES with copy number variant (CNV) analysis to provide a molecular diagnosis to patients with rare Mendelian disorders. Our findings also illustrate the clinical spectrum of CRIPT related mutations. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Exones , Estudios de Asociación Genética , Mutación Missense , Fenotipo , Eliminación de Secuencia , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Alelos , Sustitución de Aminoácidos , Preescolar , Análisis Mutacional de ADN , Enanismo/diagnóstico , Enanismo/genética , Facies , Femenino , Humanos , Microcefalia/diagnóstico , Microcefalia/genética , Linaje
15.
Am J Hum Genet ; 98(2): 347-57, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26805781

RESUMEN

The underlying genetic etiology of rhabdomyolysis remains elusive in a significant fraction of individuals presenting with recurrent metabolic crises and muscle weakness. Using exome sequencing, we identified bi-allelic mutations in TANGO2 encoding transport and Golgi organization 2 homolog (Drosophila) in 12 subjects with episodic rhabdomyolysis, hypoglycemia, hyperammonemia, and susceptibility to life-threatening cardiac tachyarrhythmias. A recurrent homozygous c.460G>A (p.Gly154Arg) mutation was found in four unrelated individuals of Hispanic/Latino origin, and a homozygous ∼34 kb deletion affecting exons 3-9 was observed in two families of European ancestry. One individual of mixed Hispanic/European descent was found to be compound heterozygous for c.460G>A (p.Gly154Arg) and the deletion of exons 3-9. Additionally, a homozygous exons 4-6 deletion was identified in a consanguineous Middle Eastern Arab family. No homozygotes have been reported for these changes in control databases. Fibroblasts derived from a subject with the recurrent c.460G>A (p.Gly154Arg) mutation showed evidence of increased endoplasmic reticulum stress and a reduction in Golgi volume density in comparison to control. Our results show that the c.460G>A (p.Gly154Arg) mutation and the exons 3-9 heterozygous deletion in TANGO2 are recurrent pathogenic alleles present in the Latino/Hispanic and European populations, respectively, causing considerable morbidity in the homozygotes in these populations.


Asunto(s)
Arritmias Cardíacas/genética , Debilidad Muscular/genética , Rabdomiólisis/genética , Alelos , Árabes/genética , Arritmias Cardíacas/diagnóstico , Secuencia de Bases , Niño , Preescolar , Estrés del Retículo Endoplásmico/genética , Exoma , Exones , Femenino , Eliminación de Gen , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Hispánicos o Latinos/genética , Homocigoto , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Debilidad Muscular/diagnóstico , Linaje , Rabdomiólisis/diagnóstico , Población Blanca/genética
16.
Am J Hum Genet ; 97(6): 904-13, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26637980

RESUMEN

Meier-Gorlin syndrome (MGS) is a genetically heterogeneous primordial dwarfism syndrome known to be caused by biallelic loss-of-function mutations in one of five genes encoding pre-replication complex proteins: ORC1, ORC4, ORC6, CDT1, and CDC6. Mutations in these genes cause disruption of the origin of DNA replication initiation. To date, only an autosomal-recessive inheritance pattern has been described in individuals with this disorder, with a molecular etiology established in about three-fourths of cases. Here, we report three subjects with MGS and de novo heterozygous mutations in the 5' end of GMNN, encoding the DNA replication inhibitor geminin. We identified two truncating mutations in exon 2 (the 1(st) coding exon), c.16A>T (p.Lys6(∗)) and c.35_38delTCAA (p.Ile12Lysfs(∗)4), and one missense mutation, c.50A>G (p.Lys17Arg), affecting the second-to-last nucleotide of exon 2 and possibly RNA splicing. Geminin is present during the S, G2, and M phases of the cell cycle and is degraded during the metaphase-anaphase transition by the anaphase-promoting complex (APC), which recognizes the destruction box sequence near the 5' end of the geminin protein. All three GMNN mutations identified alter sites 5' to residue Met28 of the protein, which is located within the destruction box. We present data supporting a gain-of-function mechanism, in which the GMNN mutations result in proteins lacking the destruction box and hence increased protein stability and prolonged inhibition of replication leading to autosomal-dominant MGS.


Asunto(s)
Microtia Congénita/genética , Enanismo/genética , Geminina/genética , Trastornos del Crecimiento/genética , Micrognatismo/genética , Mutación , Rótula/anomalías , Adolescente , Secuencia de Aminoácidos , Secuencia de Bases , Ciclo Celular/genética , Preescolar , Microtia Congénita/metabolismo , Enanismo/metabolismo , Enanismo/patología , Exones , Femenino , Geminina/metabolismo , Expresión Génica , Genes Dominantes , Trastornos del Crecimiento/metabolismo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Patrón de Herencia , Masculino , Micrognatismo/metabolismo , Datos de Secuencia Molecular , Rótula/metabolismo , Linaje , Estabilidad Proteica , Proteolisis , Empalme del ARN , Alineación de Secuencia
17.
Am J Hum Genet ; 95(5): 579-83, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439098

RESUMEN

5q31.3 microdeletion syndrome is characterized by neonatal hypotonia, encephalopathy with or without epilepsy, and severe developmental delay, and the minimal critical deletion interval harbors three genes. We describe 11 individuals with clinical features of 5q31.3 microdeletion syndrome and de novo mutations in PURA, encoding transcriptional activator protein Pur-α, within the critical region. These data implicate causative PURA mutations responsible for the severe neurological phenotypes observed in this syndrome.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Proteínas de Unión al ADN/genética , Hipotonía Muscular/genética , Convulsiones/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caenorhabditis elegans/genética , Mapeo Cromosómico , Humanos , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN , Síndrome
18.
Am J Hum Genet ; 94(5): 784-9, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24791903

RESUMEN

Clinical whole-exome sequencing (WES) for identification of mutations leading to Mendelian disease has been offered to the medical community since 2011. Clinically undiagnosed neurological disorders are the most frequent basis for test referral, and currently, approximately 25% of such cases are diagnosed at the molecular level. To date, there are approximately 4,000 "known" disease-associated loci, and many are associated with striking dysmorphic features, making genotype-phenotype correlations relatively straightforward. A significant fraction of cases, however, lack characteristic dysmorphism or clinical pathognomonic traits and are dependent upon molecular tests for definitive diagnoses. Further, many molecular diagnoses are guided by recent gene-disease association discoveries. Hence, there is a critical interplay between clinical testing and research leading to gene-disease association discovery. Here, we describe four probands, all of whom presented with hypotonia, intellectual disability, global developmental delay, and mildly dysmorphic facial features. Three of the four also had sleep apnea. Each was a simplex case without a remarkable family history. Using WES, we identified AHDC1 de novo truncating mutations that most likely cause this genetic syndrome.


Asunto(s)
Proteínas de Unión al ADN/genética , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Hipotonía Muscular/genética , Síndromes de la Apnea del Sueño/genética , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Lactante , Masculino , Mutación , Síndrome
19.
J Cell Biol ; 182(1): 89-101, 2008 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-18625844

RESUMEN

Deadenylation is the major step triggering mammalian mRNA decay. One consequence of deadenylation is the formation of nontranslatable messenger RNA (mRNA) protein complexes (messenger ribonucleoproteins [mRNPs]). Nontranslatable mRNPs may accumulate in P-bodies, which contain factors involved in translation repression, decapping, and 5'-to-3' degradation. We demonstrate that deadenylation is required for mammalian P-body formation and mRNA decay. We identify Pan2, Pan3, and Caf1 deadenylases as new P-body components and show that Pan3 helps recruit Pan2, Ccr4, and Caf1 to P-bodies. Pan3 knockdown causes a reduction of P-bodies and has differential effects on mRNA decay. Knocking down Caf1 or overexpressing a Caf1 catalytically inactive mutant impairs deadenylation and mRNA decay. P-bodies are not detected when deadenylation is blocked and are restored when the blockage is released. When deadenylation is impaired, P-body formation is not restorable, even when mRNAs exit the translating pool. These results support a dynamic interplay among deadenylation, mRNP remodeling, and P-body formation in selective decay of mammalian mRNA.


Asunto(s)
Estructuras Citoplasmáticas/metabolismo , Poliadenilación , Estabilidad del ARN , Animales , Estructuras Citoplasmáticas/efectos de los fármacos , Exorribonucleasas , Humanos , Ratones , Modelos Biológicos , Complejos Multiproteicos/metabolismo , Células 3T3 NIH , Poli A/metabolismo , Poliadenilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas/metabolismo , Puromicina/farmacología , Estabilidad del ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR4/metabolismo , Proteínas Represoras , Ribonucleasas
20.
Mol Cell Biol ; 27(22): 7791-801, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17785442

RESUMEN

In mammalian cells, mRNA decay begins with deadenylation, which involves two consecutive phases mediated by the PAN2-PAN3 and the CCR4-CAF1 complexes, respectively. The regulation of the critical deadenylation step and its relationship with RNA-processing bodies (P-bodies), which are thought to be a site where poly(A)-shortened mRNAs get degraded, are poorly understood. Using the Tet-Off transcriptional pulsing approach to investigate mRNA decay in mouse NIH 3T3 fibroblasts, we found that TOB, an antiproliferative transcription factor, enhances mRNA deadenylation in vivo. Results from glutathione S-transferase pull-down and coimmunoprecipitation experiments indicate that TOB can simultaneously interact with the poly(A) nuclease complex CCR4-CAF1 and the cytoplasmic poly(A)-binding protein, PABPC1. Combining these findings with those from mutagenesis studies, we further identified the protein motifs on TOB and PABPC1 that are necessary for their interaction and found that interaction with PABPC1 is necessary for TOB's deadenylation-enhancing effect. Moreover, our immunofluorescence microscopy results revealed that TOB colocalizes with P-bodies, suggesting a role of TOB in linking deadenylation to the P-bodies. Our findings reveal a new mechanism by which the fate of mammalian mRNA is modulated at the deadenylation step by a protein that recruits poly(A) nuclease(s) to the 3' poly(A) tail-PABP complex.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína I de Unión a Poli(A)/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células COS , Chlorocebus aethiops , Exorribonucleasas/genética , Exorribonucleasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Complejos Multiproteicos/metabolismo , Células 3T3 NIH , Fenilalanina/metabolismo , Proteína I de Unión a Poli(A)/genética , Poliadenilación , Estructura Terciaria de Proteína , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...