Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 172(5): 924-936.e11, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29474920

RESUMEN

Certain mutations can cause proteins to accumulate in neurons, leading to neurodegeneration. We recently showed, however, that upregulation of a wild-type protein, Ataxin1, caused by haploinsufficiency of its repressor, the RNA-binding protein Pumilio1 (PUM1), also causes neurodegeneration in mice. We therefore searched for human patients with PUM1 mutations. We identified eleven individuals with either PUM1 deletions or de novo missense variants who suffer a developmental syndrome (Pumilio1-associated developmental disability, ataxia, and seizure; PADDAS). We also identified a milder missense mutation in a family with adult-onset ataxia with incomplete penetrance (Pumilio1-related cerebellar ataxia, PRCA). Studies in patient-derived cells revealed that the missense mutations reduced PUM1 protein levels by ∼25% in the adult-onset cases and by ∼50% in the infantile-onset cases; levels of known PUM1 targets increased accordingly. Changes in protein levels thus track with phenotypic severity, and identifying posttranscriptional modulators of protein expression should identify new candidate disease genes.


Asunto(s)
Discapacidades del Desarrollo/genética , Predisposición Genética a la Enfermedad , Haploinsuficiencia/genética , Mutación/genética , Proteínas de Unión al ARN/genética , Convulsiones/genética , Adolescente , Adulto , Edad de Inicio , Anciano de 80 o más Años , Animales , Secuencia de Bases , Niño , Preescolar , Discapacidades del Desarrollo/diagnóstico por imagen , Evolución Molecular , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Lactante , Masculino , Ratones , Persona de Mediana Edad , Mutación Missense/genética , Neuronas/metabolismo , Neuronas/patología , Linaje , Estabilidad Proteica , Convulsiones/diagnóstico por imagen
2.
Am J Hum Genet ; 108(1): 186-193, 2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33417887

RESUMEN

POLR3B encodes the second-largest catalytic subunit of RNA polymerase III, an enzyme involved in transcription. Bi-allelic pathogenic variants in POLR3B are a well-established cause of hypomyelinating leukodystrophy. We describe six unrelated individuals with de novo missense variants in POLR3B and a clinical presentation substantially different from POLR3-related leukodystrophy. These individuals had afferent ataxia, spasticity, variable intellectual disability and epilepsy, and predominantly demyelinating sensory motor peripheral neuropathy. Protein modeling and proteomic analysis revealed a distinct mechanism of pathogenicity; the de novo POLR3B variants caused aberrant association of individual enzyme subunits rather than affecting overall enzyme assembly or stability. We expand the spectrum of disorders associated with pathogenic variants in POLR3B to include a de novo heterozygous POLR3B-related disorder.


Asunto(s)
Ataxia/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , ARN Polimerasa III/genética , Adolescente , Adulto , Ataxia Cerebelosa/genética , Niño , Preescolar , Femenino , Genes Recesivos/genética , Heterocigoto , Humanos , Masculino , Mutación Missense/genética , Proteómica/métodos , Adulto Joven
3.
Am J Hum Genet ; 102(2): 309-320, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29394990

RESUMEN

Exome sequencing has markedly enhanced the discovery of genes implicated in Mendelian disorders, particularly for individuals in whom a known clinical entity could not be assigned. This has led to the recognition that phenotypic heterogeneity resulting from allelic mutations occurs more commonly than previously appreciated. Here, we report that missense variants in CDC42, a gene encoding a small GTPase functioning as an intracellular signaling node, underlie a clinically heterogeneous group of phenotypes characterized by variable growth dysregulation, facial dysmorphism, and neurodevelopmental, immunological, and hematological anomalies, including a phenotype resembling Noonan syndrome, a developmental disorder caused by dysregulated RAS signaling. In silico, in vitro, and in vivo analyses demonstrate that mutations variably perturb CDC42 function by altering the switch between the active and inactive states of the GTPase and/or affecting CDC42 interaction with effectors, and differentially disturb cellular and developmental processes. These findings reveal the remarkably variable impact that dominantly acting CDC42 mutations have on cell function and development, creating challenges in syndrome definition, and exemplify the importance of functional profiling for syndrome recognition and delineation.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Craneofaciales/genética , Heterogeneidad Genética , Atrofia Muscular/genética , Mutación Missense , Trastornos del Neurodesarrollo/genética , Síndrome de Noonan/genética , Proteína de Unión al GTP cdc42/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Adolescente , Adulto , Niño , Preescolar , Anomalías Craneofaciales/metabolismo , Anomalías Craneofaciales/patología , Femenino , Expresión Génica , Humanos , Lactante , Masculino , Modelos Moleculares , Atrofia Muscular/metabolismo , Atrofia Muscular/patología , Trastornos del Neurodesarrollo/metabolismo , Trastornos del Neurodesarrollo/patología , Síndrome de Noonan/metabolismo , Síndrome de Noonan/patología , Fenotipo , Estructura Secundaria de Proteína , Índice de Severidad de la Enfermedad , Proteína de Unión al GTP cdc42/química , Proteína de Unión al GTP cdc42/metabolismo
5.
Am J Hum Genet ; 101(2): 267-273, 2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28777933

RESUMEN

Ribosomal RNA (rRNA) is transcribed from rDNA by RNA polymerase I (Pol I) to produce the 45S precursor of the 28S, 5.8S, and 18S rRNA components of the ribosome. Two transcription factors have been defined for Pol I in mammals, the selectivity factor SL1, and the upstream binding transcription factor (UBF), which interacts with the upstream control element to facilitate the assembly of the transcription initiation complex including SL1 and Pol I. In seven unrelated affected individuals, all suffering from developmental regression starting at 2.5-7 years, we identified a heterozygous variant, c.628G>A in UBTF, encoding p.Glu210Lys in UBF, which occurred de novo in all cases. While the levels of UBF, Ser388 phosphorylated UBF, and other Pol I-related components (POLR1E, TAF1A, and TAF1C) remained unchanged in cells of an affected individual, the variant conferred gain of function to UBF, manifesting by markedly increased UBF binding to the rDNA promoter and to the 5'- external transcribed spacer. This was associated with significantly increased 18S expression, and enlarged nucleoli which were reduced in number per cell. The data link neurodegeneration in childhood with altered rDNA chromatin status and rRNA metabolism.


Asunto(s)
Encefalopatías/genética , Nucléolo Celular/patología , Enfermedades Neurodegenerativas/genética , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , ARN Ribosómico 18S/biosíntesis , Adolescente , Adulto , Atrofia/genética , Encéfalo/patología , Encefalopatías/patología , Niño , Cromatina/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Enfermedades Neurodegenerativas/patología , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , Adulto Joven
6.
Am J Hum Genet ; 100(1): 91-104, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-27939640

RESUMEN

Identification of over 500 epigenetic regulators in humans raises an interesting question regarding how chromatin dysregulation contributes to different diseases. Bromodomain and PHD finger-containing protein 1 (BRPF1) is a multivalent chromatin regulator possessing three histone-binding domains, one non-specific DNA-binding module, and several motifs for interacting with and activating three lysine acetyltransferases. Genetic analyses of fish brpf1 and mouse Brpf1 have uncovered an important role in skeletal, hematopoietic, and brain development, but it remains unclear how BRPF1 is linked to human development and disease. Here, we describe an intellectual disability disorder in ten individuals with inherited or de novo monoallelic BRPF1 mutations. Symptoms include infantile hypotonia, global developmental delay, intellectual disability, expressive language impairment, and facial dysmorphisms. Central nervous system and spinal abnormalities are also seen in some individuals. These clinical features overlap with but are not identical to those reported for persons with KAT6A or KAT6B mutations, suggesting that BRPF1 targets these two acetyltransferases and additional partners in humans. Functional assays showed that the resulting BRPF1 variants are pathogenic and impair acetylation of histone H3 at lysine 23, an abundant but poorly characterized epigenetic mark. We also found a similar deficiency in different lines of Brpf1-knockout mice. These data indicate that aberrations in the chromatin regulator gene BRPF1 cause histone H3 acetylation deficiency and a previously unrecognized intellectual disability syndrome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Cromatina/metabolismo , Histonas/metabolismo , Discapacidad Intelectual/genética , Mutación , Proteínas Nucleares/genética , Acetilación , Adolescente , Alelos , Animales , Proteínas Portadoras/genética , Niño , Cromatina/química , Proteínas de Unión al ADN , Discapacidades del Desarrollo/genética , Cara/anomalías , Femenino , Histona Acetiltransferasas/genética , Humanos , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hipotonía Muscular/genética , Síndrome
7.
Am J Hum Genet ; 99(3): 711-719, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27545680

RESUMEN

The overall understanding of the molecular etiologies of intellectual disability (ID) and developmental delay (DD) is increasing as next-generation sequencing technologies identify genetic variants in individuals with such disorders. However, detailed analyses conclusively confirming these variants, as well as the underlying molecular mechanisms explaining the diseases, are often lacking. Here, we report on an ID syndrome caused by de novo heterozygous loss-of-function (LoF) mutations in SON. The syndrome is characterized by ID and/or DD, malformations of the cerebral cortex, epilepsy, vision problems, musculoskeletal abnormalities, and congenital malformations. Knockdown of son in zebrafish resulted in severe malformation of the spine, brain, and eyes. Importantly, analyses of RNA from affected individuals revealed that genes critical for neuronal migration and cortex organization (TUBG1, FLNA, PNKP, WDR62, PSMD3, and HDAC6) and metabolism (PCK2, PFKL, IDH2, ACY1, and ADA) are significantly downregulated because of the accumulation of mis-spliced transcripts resulting from erroneous SON-mediated RNA splicing. Our data highlight SON as a master regulator governing neurodevelopment and demonstrate the importance of SON-mediated RNA splicing in human development.


Asunto(s)
Encéfalo/embriología , Encéfalo/metabolismo , Proteínas de Unión al ADN/genética , Genes Esenciales/genética , Discapacidad Intelectual/genética , Antígenos de Histocompatibilidad Menor/genética , Mutación/genética , Empalme del ARN/genética , Animales , Encéfalo/anomalías , Encéfalo/patología , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Discapacidades del Desarrollo/fisiopatología , Anomalías del Ojo/genética , Femenino , Haploinsuficiencia/genética , Cabeza/anomalías , Heterocigoto , Humanos , Discapacidad Intelectual/patología , Discapacidad Intelectual/fisiopatología , Masculino , Enfermedades Metabólicas/genética , Enfermedades Metabólicas/metabolismo , Antígenos de Histocompatibilidad Menor/análisis , Antígenos de Histocompatibilidad Menor/metabolismo , Linaje , ARN Mensajero/análisis , Columna Vertebral/anomalías , Síndrome , Pez Cebra/anomalías , Pez Cebra/embriología , Pez Cebra/genética
8.
J Med Genet ; 55(8): 561-566, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28866611

RESUMEN

BACKGROUND: The list of Mendelian disorders of the epigenetic machinery has expanded rapidly during the last 5 years. A few missense variants in the chromatin remodeler CHD1 have been found in several large-scale sequencing efforts focused on uncovering the genetic aetiology of autism. OBJECTIVES: To explore whether variants in CHD1 are associated with a human phenotype. METHODS: We used GeneMatcher to identify other physicians caring for patients with variants in CHD1. We also explored the epigenetic consequences of one of these variants in cultured fibroblasts. RESULTS: Here we describe six CHD1 heterozygous missense variants in a cohort of patients with autism, speech apraxia, developmental delay and facial dysmorphic features. Importantly, three of these variants occurred de novo. We also report on a subject with a de novo deletion covering a large fraction of the CHD1 gene without any obvious neurological phenotype. Finally, we demonstrate increased levels of the closed chromatin modification H3K27me3 in fibroblasts from a subject carrying a de novo variant in CHD1. CONCLUSIONS: Our results suggest that variants in CHD1 can lead to diverse phenotypic outcomes; however, the neurodevelopmental phenotype appears to be limited to patients with missense variants, which is compatible with a dominant negative mechanism of disease.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación Missense , Niño , Preescolar , ADN Helicasas/química , Proteínas de Unión al ADN/química , Discapacidades del Desarrollo/diagnóstico , Facies , Femenino , Fibroblastos/metabolismo , Estudios de Asociación Genética/métodos , Histonas/metabolismo , Humanos , Lactante , Modelos Moleculares , Fenotipo , Conformación Proteica , Relación Estructura-Actividad
9.
Am J Med Genet A ; 173(1): 213-216, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27865048

RESUMEN

Neuroligins are post-synaptic, cellular adhesion molecules implicated in synaptic formation and function. NLGN2 is strongly linked to inhibitory, GABAergic signaling and is crucial for maintaining the excitation-inhibition balance in the brain. Disruption of the excitation-inhibition balance is associated with neuropsychiatric disease. In animal models, altered NLGN2 expression causes anxiety, developmental delay, motor discoordination, social impairment, aggression, and sensory processing defects. In humans, mutations in NLGN3 and NLGN4 are linked to autism and schizophrenia; NLGN2 missense variants are implicated in schizophrenia. Copy number variants encompassing NLGN2 on 17p13.1 are associated with autism, intellectual disability, metabolic syndrome, diabetes, and dysmorphic features, but an isolated NLGN2 nonsense variant has not yet been described in humans. Here, we describe a 15-year-old male with severe anxiety, obsessive-compulsive behaviors, developmental delay, autism, obesity, macrocephaly, and some dysmorphic features. Exome sequencing identified a heterozygous, de novo, c.441C>A p.(Tyr147Ter) variant in NLGN2 that is predicted to cause loss of normal protein function. This is the first report of an NLGN2 nonsense variant in humans, adding to the accumulating evidence that links synaptic proteins with a spectrum of neurodevelopmental phenotypes. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Ansiedad/genética , Trastorno Autístico/genética , Moléculas de Adhesión Celular Neuronal/genética , Codón sin Sentido , Hiperfagia/genética , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Obesidad/genética , Adolescente , Alelos , Ansiedad/diagnóstico , Trastorno Autístico/diagnóstico , Biomarcadores , Exoma , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Estudios de Asociación Genética , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hiperfagia/diagnóstico , Hibridación Fluorescente in Situ , Discapacidad Intelectual/diagnóstico , Masculino , Pruebas Neuropsicológicas , Obesidad/diagnóstico , Síndrome
10.
Hum Genet ; 135(7): 699-705, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27048600

RESUMEN

Whole exome sequencing (WES) can be used to efficiently identify de novo genetic variants associated with genetically heterogeneous conditions including intellectual disabilities. We have performed WES for 4102 (1847 female; 2255 male) intellectual disability/developmental delay cases and we report five patients with a neurodevelopmental disorder associated with developmental delay, intellectual disability, behavioral problems, hypotonia, speech problems, microcephaly, pachygyria and dysmorphic features in whom we have identified de novo missense and canonical splice site mutations in CSNK2A1, the gene encoding CK2α, the catalytic subunit of protein kinase CK2, a ubiquitous serine/threonine kinase composed of two regulatory (ß) and two catalytic (α and/or α') subunits. Somatic mutations in CSNK2A1 have been implicated in various cancers; however, this is the first study to describe a human condition associated with germline mutations in any of the CK2 subunits.


Asunto(s)
Trastorno Dismórfico Corporal/genética , Discapacidad Intelectual/genética , Trastornos del Neurodesarrollo/genética , Adolescente , Trastorno Dismórfico Corporal/fisiopatología , Quinasa de la Caseína II/genética , Niño , Preescolar , Exoma/genética , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/patología , Mutación , Trastornos del Neurodesarrollo/fisiopatología
11.
Lancet Oncol ; 16(5): 569-82, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25882982

RESUMEN

BACKGROUND: Rhabdoid brain tumours, also called atypical teratoid rhabdoid tumours, are lethal childhood cancers with characteristic genetic alterations of SMARCB1/hSNF5. Lack of biological understanding of the substantial clinical heterogeneity of these tumours restricts therapeutic advances. We integrated genomic and clinicopathological analyses of a cohort of patients with atypical teratoid rhabdoid tumours to find out the molecular basis for clinical heterogeneity in these tumours. METHODS: We obtained 259 rhabdoid tumours from 37 international institutions and assessed transcriptional profiles in 43 primary tumours and copy number profiles in 38 primary tumours to discover molecular subgroups of atypical teratoid rhabdoid tumours. We used gene and pathway enrichment analyses to discover group-specific molecular markers and did immunohistochemical analyses on 125 primary tumours to evaluate clinicopathological significance of molecular subgroup and ASCL1-NOTCH signalling. FINDINGS: Transcriptional analyses identified two atypical teratoid rhabdoid tumour subgroups with differential enrichment of genetic pathways, and distinct clinicopathological and survival features. Expression of ASCL1, a regulator of NOTCH signalling, correlated with supratentorial location (p=0·004) and superior 5-year overall survival (35%, 95% CI 13-57, and 20%, 6-34, for ASCL1-positive and ASCL1-negative tumours, respectively; p=0·033) in 70 patients who received multimodal treatment. ASCL1 expression also correlated with superior 5-year overall survival (34%, 7-61, and 9%, 0-21, for ASCL1-positive and ASCL1-negative tumours, respectively; p=0·001) in 39 patients who received only chemotherapy without radiation. Cox hazard ratios for overall survival in patients with differential ASCL1 enrichment treated with chemotherapy with or without radiation were 2·02 (95% CI 1·04-3·85; p=0·038) and 3·98 (1·71-9·26; p=0·001). Integrated analyses of molecular subgroupings with clinical prognostic factors showed three distinct clinical risk groups of tumours with different therapeutic outcomes. INTERPRETATION: An integration of clinical risk factors and tumour molecular groups can be used to identify patients who are likely to have improved long-term radiation-free survival and might help therapeutic stratification of patients with atypical teratoid rhabdoid tumours. FUNDING: C17 Research Network, Genome Canada, b.r.a.i.n.child, Mitchell Duckman, Tal Doron and Suri Boon foundations.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Genómica , Receptores Notch/biosíntesis , Tumor Rabdoide/genética , Teratoma/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Niño , Preescolar , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Lactante , Masculino , Pronóstico , Receptores Notch/genética , Tumor Rabdoide/patología , Factores de Riesgo , Transducción de Señal/genética , Teratoma/patología
12.
Hum Genet ; 133(3): 321-30, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24154661

RESUMEN

Beckwith-Wiedemann syndrome (BWS), an overgrowth and tumor predisposition syndrome is clinically heterogeneous. Its variable presentation makes molecular diagnosis particularly important for appropriate counseling of patients with respect to embyronal tumor risk and recurrence risk. BWS is characterized by macrosomia, omphalocele, and macroglossia. Additional clinical features can include hemihyperplasia, embryonal tumors, umbilical hernia, and ear anomalies. BWS is etiologically heterogeneous arising from dysregulation of one or both of the chromosome 11p15.5 imprinting centers (IC) and/or imprinted growth regulatory genes on chromosome 11p15.5. Most BWS cases are sporadic and result from loss of maternal methylation at imprinting center 2 (IC2), gain of maternal methylation at imprinting center 1 (IC1) or paternal uniparental disomy (UPD). Heritable forms of BWS (15 %) have been attributed mainly to mutations in the growth suppressor gene CDKN1C, but have also infrequently been identified in patients with copy number variations (CNVs) in the chromosome 11p15.5 region. Four hundred and thirty-four unrelated BWS patients referred to the molecular diagnostic laboratory were tested by methylation-specific multiplex ligation-dependent probe amplification. Molecular alterations were detected in 167 patients, where 103 (62 %) showed loss of methylation at IC2, 23 (14 %) had gain of methylation at IC1, and 41 (25 %) showed changes at both ICs usually associated with paternal UPD. In each of the three groups, we identified patients in whom the abnormalities in the chromosome 11p15.5 region were due to CNVs. Surprisingly, 14 patients (9 %) demonstrated either deletions or duplications of the BWS critical region that were confirmed using comparative genomic hybridization array analysis. The majority of these CNVs were associated with a methylation change at IC1. Our results suggest that CNVs in the 11p15.5 region contribute significantly to the etiology of BWS. We highlight the importance of performing deletion/duplication testing in addition to methylation analysis in the molecular investigation of BWS to improve our understanding of the molecular basis of this disorder, and to provide accurate genetic counseling.


Asunto(s)
Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/genética , Cromosomas Humanos Par 11/genética , Variaciones en el Número de Copia de ADN/genética , Deleción Cromosómica , Cromosomas Humanos Par 4/genética , Hibridación Genómica Comparativa , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Metilación de ADN , Femenino , Reordenamiento Génico , Impresión Genómica , Genotipo , Humanos , Masculino , Linaje , Fenotipo
13.
Genet Med ; 16(8): 625-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24556927

RESUMEN

PURPOSE: The purpose of this study was to determine the molecular consequences of the variant c.3700 A>G in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, a variant that has been predicted to cause a missense mutation in the CFTR protein (p.Ile1234Val). METHODS: Clinical assays of CFTR function were performed, and genomic DNA from patients homozygous for c.3700 A>G and their family members was sequenced. Total RNA was extracted from epithelial cells of the patients, transcribed into complementary DNA, and sequenced. CFTR complementary DNA clones containing the missense mutation p.Ile1234Val or a truncated exon 19 (p.Ile1234_Arg1239del) were constructed and heterologously expressed to test CFTR protein synthesis and processing. RESULTS: In vivo functional measurements revealed that the individuals homozygous for the variant c.3700 A>G exhibited defective CFTR function. We show that this mutation in exon 19 activates a cryptic donor splice site 18 bp upstream of the original donor splice site, resulting in deletion of six amino acids (r.3700_3717del; p.Ile1234_Arg1239del). This deletion, similar to p.Phe508del, causes a primary defect in folding and processing. Importantly, Lumacaftor (VX-809), currently in clinical trial for cystic fibrosis patients with the major cystic fibrosis-causing mutation, p.Phe508del, partially ameliorated the processing defect caused by p.Ile1234_Arg1239del. CONCLUSION: These studies highlight the need to verify molecular and clinical consequences of CFTR variants to define possible therapeutic strategies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Isoleucina/metabolismo , Valina/metabolismo , Adolescente , Adulto , Aminopiridinas/farmacología , Animales , Benzodioxoles/farmacología , Línea Celular , Cricetinae , Fibrosis Quística/tratamiento farmacológico , Exones , Células HEK293 , Homocigoto , Humanos , Masculino , Mutación Missense , Qatar , Empalme del ARN
14.
Acta Neuropathol ; 128(6): 853-62, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25315281

RESUMEN

Although telomeres are maintained in most cancers by telomerase activation, a subset of tumors utilize alternative lengthening of telomeres (ALT) to sustain self-renewal capacity. In order to study the prevalence and significance of ALT in childhood brain tumors we screened 517 pediatric brain tumors using the novel C-circle assay. We examined the association of ALT with alterations in genes found to segregate with specific histological phenotypes and with clinical outcome. ALT was detected almost exclusively in malignant tumors (p = 0.001). ALT was highly enriched in primitive neuroectodermal tumors (12 %), choroid plexus carcinomas (23 %) and high-grade gliomas (22 %). Furthermore, in contrast to adult gliomas, pediatric low grade gliomas which progressed to high-grade tumors did not exhibit the ALT phenotype. Somatic but not germline TP53 mutations were highly associated with ALT (p = 1.01 × 10(-8)). Of the other alterations examined, only ATRX point mutations and reduced expression were associated with the ALT phenotype (p = 0.0005). Interestingly, ALT attenuated the poor outcome conferred by TP53 mutations in specific pediatric brain tumors. Due to very poor prognosis, one year overall survival was quantified in malignant gliomas, while in children with choroid plexus carcinoma, five year overall survival was investigated. For children with TP53 mutant malignant gliomas, one year overall survival was 63 ± 12 and 23 ± 10 % for ALT positive and negative tumors, respectively (p = 0.03), while for children with TP53 mutant choroid plexus carcinomas, 5 years overall survival was 67 ± 19 and 27 ± 13 % for ALT positive and negative tumors, respectively (p = 0.07). These observations suggest that the presence of ALT is limited to a specific group of childhood brain cancers which harbor somatic TP53 mutations and may influence the outcome of these patients. Analysis of ALT may contribute to risk stratification and targeted therapies to improve outcome for these children.


Asunto(s)
Neoplasias Encefálicas/genética , Carcinoma/genética , Neoplasias del Plexo Coroideo/genética , Glioma/genética , Tumores Neuroectodérmicos Primitivos/genética , Telómero , Proteína p53 Supresora de Tumor/genética , Adolescente , Neoplasias Encefálicas/fisiopatología , Carcinoma/fisiopatología , Neoplasias del Plexo Coroideo/fisiopatología , Estudios de Cohortes , ADN Helicasas/genética , Glioma/fisiopatología , Humanos , Estimación de Kaplan-Meier , Mutación , Clasificación del Tumor , Tumores Neuroectodérmicos Primitivos/fisiopatología , Proteínas Nucleares/genética , Fenotipo , Pronóstico , Telómero/metabolismo , Proteína Nuclear Ligada al Cromosoma X
15.
Hum Genet ; 132(11): 1245-52, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23812740

RESUMEN

Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a myocardial disease characterized by fibro-fatty replacement of right ventricular free wall myocardium and life-threatening ventricular arrhythmias. A missense mutation, c.1073C>T (p.S358L) in the transmembrane protein 43 (TMEM43) gene, has been genetically identified to cause ARVC type 5 in a founder population from Newfoundland. It is unclear whether this mutation occurs in other populations outside of this founder population or if other variants of TMEM43 are associated with ARVC disease. We sought to identify non-Newfoundland individuals with TMEM43 variants among patient samples sent for genetic assessment for possible ARVC. Of 195 unrelated individuals with suspected ARVC, mutation of desmosomal proteins was seen in 28 and the p.S358L TMEM43 mutation in six. We identified a de novo p.S358L mutation in a non-Newfoundland patient and five separate rare TMEM43 (four novel) sequence variants in non-Newfoundland patients, each occurring in an evolutionarily conserved amino acid. TMEM43 mutations occur outside of the founder population of the island of Newfoundland where it was originally described. TMEM43 sequencing should be incorporated into clinical genetic testing for ARVC patients.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica/genética , Proteínas de la Membrana/genética , Mutación Missense , Arritmias Cardíacas/genética , Displasia Ventricular Derecha Arritmogénica/fisiopatología , Desmosomas/genética , Desmosomas/metabolismo , Efecto Fundador , Predisposición Genética a la Enfermedad , Ventrículos Cardíacos/fisiopatología , Heterocigoto , Humanos , Proteínas de la Membrana/metabolismo , Terranova y Labrador , Linaje , Análisis de Secuencia de ADN
16.
Am J Hum Genet ; 87(5): 631-42, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-21056402

RESUMEN

DNA copy-number variations (CNVs) underlie many neuropsychiatric conditions, but they have been less studied in cancer. We report the association of a 17p13.1 CNV, childhood-onset developmental delay (DD), and cancer. Through a screen of over 4000 patients with diverse diagnoses, we identified eight probands harboring microdeletions at TP53 (17p13.1). We used a purpose-built high-resolution array with 93.75% breakpoint accuracy to fine map these microdeletions. Four patients were found to have a common phenotype including DD, hypotonia, and hand and foot abnormalities, constituting a unique syndrome. Notably, these patients were not affected with cancer. Moreover, none of the TP53-deletion patients affected with cancer (n = 4) had neurocognitive impairments. DD patients have larger deletions, which encompass but do not disrupt TP53, whereas cancer-affected patients harbor CNVs with at least one breakpoint within TP53. Most 17p13.1 deletions arise by Alu-mediated nonallelic homologous recombination. Furthermore, we identify a critical genomic region associated with DD and containing six underexpressed genes. We conclude that, although they overlap, 17p13.1 CNVs are associated with distinct phenotypes depending on the position of the breakpoint with respect to TP53. Further, detailed characterization of breakpoints revealed a common formation signature. Future studies should consider whether other loci in the genome also give rise to phenotypically distinct disorders by means of a common mechanism, resulting in a similar formation signature.


Asunto(s)
Neoplasias/genética , Adulto , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 17 , Variaciones en el Número de Copia de ADN , Discapacidades del Desarrollo/genética , Genes p53 , Humanos , Fenotipo , Síndrome
18.
Am J Med Genet A ; 158A(6): 1388-94, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22585446

RESUMEN

Beckwith-Wiedemann syndrome (BWS) is an overgrowth disorder with variability in clinical manifestations and molecular causes. In most cases, patients with BWS have normal development. Cases with developmental delay are usually attributed to neonatal hypoglycemia or chromosome abnormalities involving copy number variation for genes beyond the critical BWS region at 11p15.5. Brain abnormalities have not previously been recognized within the BWS phenotypic spectrum. We report on seven cases of BWS associated with posterior fossa abnormalities. Of these, two cases presented with Blake's pouch cyst, two with Dandy-Walker variant (DWV; hypoplasia of the inferior part of the vermis), one with Dandy-Walker malformation (DWM) and one with a complex of DWM, dysgenesis of the corpus callosum and brain stem abnormality. In all these cases, molecular findings involved the centromeric imprinted domain on chromosome locus 11p15.5, which includes imprinting center 2 (IC2) and the imprinted growth suppressor gene, CDKN1C. Three cases had loss of methylation at IC2, two had CDKN1C mutations, and one had loss of methylation at IC2 and a microdeletion. In one case no mutation/methylation abnormality was detected. These findings together with previously reported correlations suggest that genes in imprinted domain 2 at 11p15.5 are involved in normal midline development of several organs including the brain. Our data suggest that brain malformations may present as a finding within the BWS phenotype when the molecular etiology involves imprinted domain 2. Brain imaging may be useful in identifying such malformations in individuals with BWS and neurodevelopmental issues.


Asunto(s)
Síndrome de Beckwith-Wiedemann/diagnóstico , Encéfalo/anomalías , Síndrome de Beckwith-Wiedemann/complicaciones , Síndrome de Beckwith-Wiedemann/genética , Encéfalo/patología , Preescolar , Cromosomas Humanos Par 11 , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Metilación de ADN , Resultado Fatal , Femenino , Eliminación de Gen , Impresión Genómica , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Masculino , Mutación
19.
Muscle Nerve ; 45(5): 752-5, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22499106

RESUMEN

INTRODUCTION: Congenital muscular dystrophies (CMD) with hypoglycosylated α-dystroglycan due to POMT1 mutations are associated with clinical phenotypes that vary in severity. METHODS: We describe a patient with congenital hypotonia, generalized weakness, elevated creatine kinase (CK), and normal brain imaging. RESULTS: Histochemical analysis of the index case's muscle showed deficiency of glycosylated α-dystroglycan and secondary merosin deficiency. Genetic testing revealed a novel mutation in exon 20 of the POMT1 gene. CONCLUSIONS: Our patient's milder form of CMD adds to the emerging evidence of an expanding phenotype caused by POMT1 mutations. The histopathological findings of the muscle biopsy in this case support the need for careful clinical, genetic, and histochemical diagnostic interpretation.


Asunto(s)
Manosiltransferasas/genética , Distrofias Musculares/genética , Mutación , Encéfalo/patología , Creatina Quinasa/sangre , Distroglicanos/metabolismo , Electromiografía , Pruebas Genéticas , Glicosilación , Humanos , Lactante , Laminina/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Fenotipo
20.
Am J Med Genet A ; 152A(7): 1808-11, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20583174

RESUMEN

Long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency is an autosomal recessive disorder affecting mitochondrial fatty acid oxidation due to mutations in the HADHA gene. We report on a 22-month-old child who was identified on expanded newborn screening with an abnormal acylcarnitine pattern and increased C14OH. Molecular analysis showed that the child was homozygous for the common mutation, c.1526G > C (p.Glu510Gln) in the HADHA gene. Carrier testing on the parental samples revealed that the father was heterozygous for the mutation whereas the mother did not carry the mutation. Short tandem repeat testing with markers covering both short and long arms of chromosome 2 showed that the child has paternal uniparental isodisomy. We highlight the importance of parental testing in cases of homozygosity in autosomal recessive disorders and its impact on genetic counseling of the family.


Asunto(s)
3-Hidroxiacil-CoA Deshidrogenasas/deficiencia , Cromosomas Humanos Par 2/genética , Padre , Disomía Uniparental/genética , 3-Hidroxiacil-CoA Deshidrogenasas/genética , Secuencia de Bases , Segregación Cromosómica/genética , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , 3-Hidroxiacil-CoA Deshidrogenasa de Cadena Larga , Masculino , Datos de Secuencia Molecular , Linaje , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA