Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Front Pediatr ; 10: 844845, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35433545

RESUMEN

The role of lysine methyltransferases (KMTs) and demethylases (KDMs) in the regulation of chromatin modification is well-established. Recently, deleterious heterozygous variants in KMT5B were implicated in individuals with intellectual disability (ID) and/or autism spectrum disorder. We describe three unrelated patients with global developmental delay (GDD) or ID, macrocephaly and additional features. Using whole exome sequencing, each of the probands was found to harbor a distinct de novo heterozygous disease-causing variant in KMT5B: c.541C > G (p.His181Asp); c.833A > T (p.Asn278Ile); or c.391_394delAAAG (p.Lys131GlufsTer6). We discuss herein their clinical presentations, and compare them to those of previously reported patients. Furthermore, using a three-dimensional computational model of the KMT5B protein, we demonstrate the predicted structural effects of the two missense variants. Our findings support the role of de novo missense and nonsense variants in KMT5B-associated GDD/ID, and suggest that this gene should be considered in the differential diagnosis of neurodevelopmental disorders accompanied by macrocephaly and/or overgrowth.

2.
Clin Genet ; 98(4): 353-364, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33111345

RESUMEN

Mutations in more than 150 genes are responsible for inherited hearing loss, with thousands of different, severe causal alleles that vary among populations. The Israeli Jewish population includes communities of diverse geographic origins, revealing a wide range of deafness-associated variants and enabling clinical characterization of the associated phenotypes. Our goal was to identify the genetic causes of inherited hearing loss in this population, and to determine relationships among genotype, phenotype, and ethnicity. Genomic DNA samples from informative relatives of 88 multiplex families, all of self-identified Jewish ancestry, with either non-syndromic or syndromic hearing loss, were sequenced for known and candidate deafness genes using the HEar-Seq gene panel. The genetic causes of hearing loss were identified for 60% of the families. One gene was encountered for the first time in human hearing loss: ATOH1 (Atonal), a basic helix-loop-helix transcription factor responsible for autosomal dominant progressive hearing loss in a five-generation family. Our results show that genomic sequencing with a gene panel dedicated to hearing loss is effective for genetic diagnoses in a diverse population. Comprehensive sequencing enables well-informed genetic counseling and clinical management by medical geneticists, otolaryngologists, audiologists, and speech therapists and can be integrated into newborn screening for deafness.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Sordera/genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva/genética , Adolescente , Adulto , Niño , Preescolar , Sordera/epidemiología , Sordera/patología , Femenino , Estudios de Asociación Genética , Pérdida Auditiva/epidemiología , Pérdida Auditiva/patología , Humanos , Israel/epidemiología , Judíos/genética , Masculino , Linaje , Adulto Joven
3.
Genet Med ; 21(11): 2442-2452, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31160754

RESUMEN

PURPOSE: Pathogenic variants in GJB2 are the most common cause of autosomal recessive sensorineural hearing loss. The classification of c.101T>C/p.Met34Thr and c.109G>A/p.Val37Ile in GJB2 are controversial. Therefore, an expert consensus is required for the interpretation of these two variants. METHODS: The ClinGen Hearing Loss Expert Panel collected published data and shared unpublished information from contributing laboratories and clinics regarding the two variants. Functional, computational, allelic, and segregation data were also obtained. Case-control statistical analyses were performed. RESULTS: The panel reviewed the synthesized information, and classified the p.Met34Thr and p.Val37Ile variants utilizing professional variant interpretation guidelines and professional judgment. We found that p.Met34Thr and p.Val37Ile are significantly overrepresented in hearing loss patients, compared with population controls. Individuals homozygous or compound heterozygous for p.Met34Thr or p.Val37Ile typically manifest mild to moderate hearing loss. Several other types of evidence also support pathogenic roles for these two variants. CONCLUSION: Resolving controversies in variant classification requires coordinated effort among a panel of international multi-institutional experts to share data, standardize classification guidelines, review evidence, and reach a consensus. We concluded that p.Met34Thr and p.Val37Ile variants in GJB2 are pathogenic for autosomal recessive nonsyndromic hearing loss with variable expressivity and incomplete penetrance.


Asunto(s)
Conexinas/genética , Pérdida Auditiva/genética , Alelos , Estudios de Casos y Controles , Conexina 26/genética , Conexinas/metabolismo , Sordera/genética , Femenino , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Mutación , Polimorfismo de Nucleótido Simple/genética
5.
J Clin Immunol ; 36(3): 220-34, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26951490

RESUMEN

PURPOSE: Spondyloenchondrodysplasia is a rare immuno-osseous dysplasia caused by biallelic mutations in ACP5. We aimed to provide a survey of the skeletal, neurological and immune manifestations of this disease in a cohort of molecularly confirmed cases. METHODS: We compiled clinical, genetic and serological data from a total of 26 patients from 18 pedigrees, all with biallelic ACP5 mutations. RESULTS: We observed a variability in skeletal, neurological and immune phenotypes, which was sometimes marked even between affected siblings. In total, 22 of 26 patients manifested autoimmune disease, most frequently autoimmune thrombocytopenia and systemic lupus erythematosus. Four patients were considered to demonstrate no clinical autoimmune disease, although two were positive for autoantibodies. In the majority of patients tested we detected upregulated expression of interferon-stimulated genes (ISGs), in keeping with the autoimmune phenotype and the likely immune-regulatory function of the deficient protein tartrate resistant acid phosphatase (TRAP). Two mutation positive patients did not demonstrate an upregulation of ISGs, including one patient with significant autoimmune disease controlled by immunosuppressive therapy. CONCLUSIONS: Our data expand the known phenotype of SPENCD. We propose that the OMIM differentiation between spondyloenchondrodysplasia and spondyloenchondrodysplasia with immune dysregulation is no longer appropriate, since the molecular evidence that we provide suggests that these phenotypes represent a continuum of the same disorder. In addition, the absence of an interferon signature following immunomodulatory treatments in a patient with significant autoimmune disease may indicate a therapeutic response important for the immune manifestations of spondyloenchondrodysplasia.


Asunto(s)
Enfermedades Autoinmunes/genética , Discapacidad Intelectual/genética , Lupus Eritematoso Sistémico/genética , Mutación , Osteocondrodisplasias/genética , Púrpura Trombocitopénica Idiopática/genética , Fosfatasa Ácida Tartratorresistente/genética , Adolescente , Adulto , Alelos , Autoanticuerpos/biosíntesis , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Huesos/inmunología , Huesos/patología , Encéfalo/inmunología , Encéfalo/patología , Niño , Preescolar , Femenino , Expresión Génica , Genotipo , Humanos , Discapacidad Intelectual/inmunología , Discapacidad Intelectual/patología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Masculino , Osteocondrodisplasias/inmunología , Osteocondrodisplasias/patología , Linaje , Fenotipo , Púrpura Trombocitopénica Idiopática/inmunología , Púrpura Trombocitopénica Idiopática/patología , Fosfatasa Ácida Tartratorresistente/deficiencia , Fosfatasa Ácida Tartratorresistente/inmunología
6.
Hum Mutat ; 36(11): 1052-63, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26178382

RESUMEN

Neurofibromatosis type 1 (NF1) is one of the most frequent genetic disorders, affecting 1:3,000 worldwide. Identification of genotype-phenotype correlations is challenging because of the wide range clinical variability, the progressive nature of the disorder, and extreme diversity of the mutational spectrum. We report 136 individuals with a distinct phenotype carrying one of five different NF1 missense mutations affecting p.Arg1809. Patients presented with multiple café-au-lait macules (CALM) with or without freckling and Lisch nodules, but no externally visible plexiform neurofibromas or clear cutaneous neurofibromas were found. About 25% of the individuals had Noonan-like features. Pulmonic stenosis and short stature were significantly more prevalent compared with classic cohorts (P < 0.0001). Developmental delays and/or learning disabilities were reported in over 50% of patients. Melanocytes cultured from a CALM in a segmental NF1-patient showed two different somatic NF1 mutations, p.Arg1809Cys and a multi-exon deletion, providing genetic evidence that p.Arg1809Cys is a loss-of-function mutation in the melanocytes and causes a pigmentary phenotype. Constitutional missense mutations at p.Arg1809 affect 1.23% of unrelated NF1 probands in the UAB cohort, therefore this specific NF1 genotype-phenotype correlation will affect counseling and management of a significant number of patients.


Asunto(s)
Sustitución de Aminoácidos , Codón , Mutación Missense , Neurofibromina 1/genética , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Fenotipo , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Enanismo/genética , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Neurofibromina 1/química , Adulto Joven
8.
World J Gastroenterol ; 20(46): 17656-60, 2014 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-25516681

RESUMEN

Wilson's disease (WD) is an autosomal recessive inherited disorder of hepatic copper metabolism. WD can be present in different clinical conditions, with the most common ones being liver disease and neuropsychiatric disturbances. Most cases present symptoms at < 40 years of age. However, few reports exist in the literature on patients in whom the disease presented beyond this age. In this report, we present a case of late onset fulminant WD in a 58-year-old patient in whom the diagnosis was established clinically, by genetic analysis of the ATP7B gene disclosing rare mutations (G1099S and c.1707+3insT) as well as by high hepatic copper content. We also reviewed the relevant literature. The diagnosis of WD with late onset presentation is easily overlooked. The diagnostic features and the genetic background in patients with late onset WD are not different from those in patients with early onset WD, except for the age. Effective treatments for this disorder that can be fatal are available and will prevent or reverse many manifestations if the disease is discovered early.


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Degeneración Hepatolenticular/diagnóstico , Mutación , Adenosina Trifosfatasas/metabolismo , Edad de Inicio , Biopsia , Proteínas de Transporte de Catión/metabolismo , ATPasas Transportadoras de Cobre , Análisis Mutacional de ADN , Resultado Fatal , Femenino , Predisposición Genética a la Enfermedad , Degeneración Hepatolenticular/genética , Degeneración Hepatolenticular/metabolismo , Degeneración Hepatolenticular/terapia , Humanos , Hígado/metabolismo , Hígado/patología , Persona de Mediana Edad , Fenotipo , Valor Predictivo de las Pruebas , Factores de Tiempo
9.
J Basic Clin Physiol Pharmacol ; 25(3): 289-92, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25153233

RESUMEN

BACKGROUND: Connexin 26 (GJB2) mutations are associated with various types of hearing loss, either without associated symptoms or with skin disease, constituting a form of syndromic hearing loss. These mutations can lead to deafness in either a recessive or a dominant autosomal form of inheritance. METHODS: Ascertainment of a Jewish Ashkenazi family with nonsyndromic hearing loss led to the construction of a pedigree for a four-generation family, with hearing loss detected in three successive generations. The entire coding region of the GJB2 gene was amplified and sequenced by Sanger sequencing. RESULTS: Audiological analysis revealed that the age of onset and severity of hearing loss were earlier and more severe, respectively, in each successive generation of an Ashkenazi Jewish family. A mutation, c.224G>A, leading to missense p.Arg75Gln was detected only in the affected members of the family. CONCLUSIONS: The entire coding region of GJB2 should be checked in hearing-impaired patients by Sanger sequencing, rather than examination only of the two most prevalent mutations, regardless of mode of inheritance or ethnicity. Furthermore, predictions regarding phenotype based on genotype can be difficult to make due to clinical variability in multigenerational families, as demonstrated in the family presented in this study.


Asunto(s)
Conexinas/genética , Sordera/genética , Conexina 26 , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Mutación , Linaje , Fenotipo
10.
Hum Mutat ; 35(7): 779-90, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24692096

RESUMEN

Mutations affecting skeletal muscle isoforms of the tropomyosin genes may cause nemaline myopathy, cap myopathy, core-rod myopathy, congenital fiber-type disproportion, distal arthrogryposes, and Escobar syndrome. We correlate the clinical picture of these diseases with novel (19) and previously reported (31) mutations of the TPM2 and TPM3 genes. Included are altogether 93 families: 53 with TPM2 mutations and 40 with TPM3 mutations. Thirty distinct pathogenic variants of TPM2 and 20 of TPM3 have been published or listed in the Leiden Open Variant Database (http://www.dmd.nl/). Most are heterozygous changes associated with autosomal-dominant disease. Patients with TPM2 mutations tended to present with milder symptoms than those with TPM3 mutations, DA being present only in the TPM2 group. Previous studies have shown that five of the mutations in TPM2 and one in TPM3 cause increased Ca(2+) sensitivity resulting in a hypercontractile molecular phenotype. Patients with hypercontractile phenotype more often had contractures of the limb joints (18/19) and jaw (6/19) than those with nonhypercontractile ones (2/22 and 1/22), whereas patients with the non-hypercontractile molecular phenotype more often (19/22) had axial contractures than the hypercontractile group (7/19). Our in silico predictions show that most mutations affect tropomyosin-actin association or tropomyosin head-to-tail binding.


Asunto(s)
Estudios de Asociación Genética , Enfermedades Musculares/congénito , Enfermedades Musculares/genética , Mutación , Tropomiosina/genética , Actinas/metabolismo , Adolescente , Adulto , Secuencia de Aminoácidos , Niño , Preescolar , Bases de Datos Genéticas , Femenino , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Enfermedades Musculares/diagnóstico , Fenotipo , Fosforilación , Unión Proteica , Alineación de Secuencia , Tropomiosina/química , Tropomiosina/metabolismo , Adulto Joven
11.
Eur J Hum Genet ; 22(6): 768-75, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24105371

RESUMEN

Hereditary hearing loss is genetically heterogeneous, with a large number of genes and mutations contributing to this sensory, often monogenic, disease. This number, as well as large size, precludes comprehensive genetic diagnosis of all known deafness genes. A combination of targeted genomic capture and massively parallel sequencing (MPS), also referred to as next-generation sequencing, was applied to determine the deafness-causing genes in hearing-impaired individuals from Israeli Jewish and Palestinian Arab families. Among the mutations detected, we identified nine novel mutations in the genes encoding myosin VI, myosin VIIA and myosin XVA, doubling the number of myosin mutations in the Middle East. Myosin VI mutations were identified in this population for the first time. Modeling of the mutations provided predicted mechanisms for the damage they inflict in the molecular motors, leading to impaired function and thus deafness. The myosin mutations span all regions of these molecular motors, leading to a wide range of hearing phenotypes, reinforcing the key role of this family of proteins in auditory function. This study demonstrates that multiple mutations responsible for hearing loss can be identified in a relatively straightforward manner by targeted-gene MPS technology and concludes that this is the optimal genetic diagnostic approach for identification of mutations responsible for hearing loss.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Pérdida Auditiva/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mutación , Cadenas Pesadas de Miosina/genética , Miosinas/genética , Árabes/genética , Secuencia de Bases , Salud de la Familia , Femenino , Genómica/métodos , Humanos , Israel , Judíos/genética , Masculino , Miosina VIIa , Linaje , Fenotipo
12.
Hum Mutat ; 34(8): 1102-10, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23606368

RESUMEN

POU3F4 is a POU domain transcription factor that is required for hearing. In the ear, POU3F4 is essential for mesenchymal remodeling of the bony labyrinth and is the causative gene for DFNX2 human nonsyndromic deafness. Ear abnormalities underlie this form of deafness, characterized previously in multiple spontaneous, radiation-induced and transgenic mouse mutants. Here, we report three novel mutations in the POU3F4 gene that result in profound hearing loss in both humans and mice. A p.Gln79* mutation was identified in a child from an Israeli family, revealed by massively parallel sequencing (MPS). This strategy demonstrates the strength of MPS for diagnosis with only one affected individual. A second mutation, p.Ile285Argfs*43, was identified by Sanger sequencing. A p.Cys300* mutation was found in an ENU-induced mutant mouse, schwindel (sdl), by positional cloning. The mutation leads to a predicted truncated protein, similar to the human mutations, providing a relevant mouse model. The p.Ile285Argfs*43 and p.Cys300* mutations lead to a shift of Pou3f4 nuclear localization to the cytoplasm, demonstrated in cellular localization studies and in the inner ears of the mutant mice. The discovery of these mutations facilitates a deeper comprehension of the molecular basis of inner ear defects due to mutations in the POU3F4 transcription factor.


Asunto(s)
Citoplasma/metabolismo , Sordera/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Animales , Células COS , Núcleo Celular/metabolismo , Niño , Chlorocebus aethiops , Sordera/metabolismo , Oído Interno/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
13.
J Clin Invest ; 123(2): 740-50, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23348741

RESUMEN

Hereditary hearing loss is the most common sensory deficit. We determined that progressive high-frequency hearing loss in 2 families of Iraqi Jewish ancestry was due to homozygosity for the protein truncating mutation SYNE4 c.228delAT. SYNE4, a gene not previously associated with hearing loss, encodes nesprin-4 (NESP4), an outer nuclear membrane (ONM) protein expressed in the hair cells of the inner ear. The truncated NESP4 encoded by the families' mutation did not localize to the ONM. NESP4 and SUN domain-containing protein 1 (SUN1), which localizes to the inner nuclear membrane (INM), are part of the linker of nucleoskeleton and cytoskeleton (LINC) complex in the nuclear envelope. Mice lacking either Nesp4 or Sun1 were evaluated for hair cell defects and hearing loss. In both Nesp4-/- and Sun1-/- mice, OHCs formed normally, but degenerated as hearing matured, leading to progressive hearing loss. The nuclei of OHCs from mutant mice failed to maintain their basal localization, potentially affecting cell motility and hence the response to sound. These results demonstrate that the LINC complex is essential for viability and normal morphology of OHCs and suggest that the position of the nucleus in sensory epithelial cells is critical for maintenance of normal hearing.


Asunto(s)
Audición/fisiología , Complejos Multiproteicos/fisiología , Animales , Análisis Mutacional de ADN , Femenino , Compuestos Ferrosos , Células Ciliadas Auditivas Externas/patología , Células Ciliadas Auditivas Externas/fisiología , Audición/genética , Pérdida Auditiva de Alta Frecuencia/etiología , Pérdida Auditiva de Alta Frecuencia/genética , Pérdida Auditiva de Alta Frecuencia/fisiopatología , Humanos , Irak/etnología , Israel , Judíos/genética , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Complejos Multiproteicos/deficiencia , Complejos Multiproteicos/genética , Mutación , Membrana Nuclear/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Linaje
14.
Eur J Hum Genet ; 21(3): 343-6, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22872102

RESUMEN

Small genomic rearrangements and copy-number variations (CNVs) involving a single gene have been associated recently with many neurocognitive phenotypes, including intellectual disability (ID), behavioral abnormalities, and autistic spectrum disorders (ASDs). Such small CNVs in the Autism susceptibility candidate 2 (AUTS2) gene have been shown to be associated with seizures, ID, and ASDs. We report four patients with small CNVs ranging in size between 133-319 kb that disrupt AUTS2. Two patients have duplications involving single exons, whereas two have deletions that removed multiple exons. All patients had developmental delay, whereas two patients had a diagnosis of ASDs. The CNVs were detected by an exon-targeted array CGH with dense oligonucleotide coverage in exons of genes known or hypothesized to be causative of multiple human phenotypes. Our report further shows that disruption of AUTS2 results in a variety of neurobehavioral phenotypes. More importantly, it demonstrates the utility of targeted exon array as a highly sensitive clinical diagnostic tool for the detection of small genomic rearrangements in the clinically relevant regions of the human genome.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Discapacidades del Desarrollo/genética , Proteínas/genética , Adolescente , Trastornos Generalizados del Desarrollo Infantil/etiología , Preescolar , Hibridación Genómica Comparativa/métodos , Proteínas del Citoesqueleto , Variaciones en el Número de Copia de ADN , Discapacidades del Desarrollo/etiología , Exones , Predisposición Genética a la Enfermedad , Humanos , Lactante , Factores de Transcripción
15.
Prenat Diagn ; 32(1): 70-4, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22367672

RESUMEN

OBJECTIVE: To increase awareness to the possibility of nemaline myopathy (NM) when abnormal prenatal ultrasound findings appear together with a carrier state for the common exon 55 deletion in the nebulin gene (NEB) of an Ashkenazi Jewish parent. METHODS: We describe four unrelated pregnancies with abnormal prenatal ultrasound findings resulting in the birth of newborns with NM, where one or both parents were of Ashkenazi Jewish origin. Data was collected retrospectively from the patients' medical files. Molecular analysis of NEB was performed on the DNA from the patients and parents. RESULTS: Prenatal ultrasound findings included polyhydramnios, decreased fetal movements, club feet, and arthrogryposis. A biopsy from two of the newborns was consistent with NM. In all of the newborns, the common NEB exon 55 deletion was detected in the heterozygote state and in three of them, a second novel mutation was found. CONCLUSIONS: Ultrasonographic findings suggestive of a myopathy and a carrier state for the NEB exon 55 deletion in one of the parents should trigger a thorough investigation for NM. The extreme size of NEB imposes great difficulties when searching for a second mutation, especially under the time constraints of an ongoing pregnancy.


Asunto(s)
Eliminación de Gen , Tamización de Portadores Genéticos/métodos , Heterocigoto , Proteínas Musculares/genética , Miopatías Nemalínicas/diagnóstico por imagen , Miopatías Nemalínicas/genética , Ultrasonografía Prenatal , Adulto , Codón sin Sentido , Exones/genética , Femenino , Predisposición Genética a la Enfermedad , Humanos , Recién Nacido , Judíos/genética , Masculino , Linaje , Embarazo
16.
J Med Screen ; 18(4): 169-72, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22156145

RESUMEN

OBJECTIVE: To assess the frequency of the D1152H mutation in the CFTR gene in normal individuals, in cystic fibrosis (CF) patients and in the setting of prenatal diagnosis. SETTING: A database analysis of sequential screening results seen at the Sheba Medical Center, Israel, between 2001 and 2010. METHODS: We retrospectively analyzed the frequency of D1152H in a large cohort of healthy individuals who were screened as part of a routine prenatal care programme, in individuals referred due to CF-related symptoms and in the setting of prenatal diagnosis. RESULTS: We found one asymptomatic homozygous female and 195 D1152H carriers among 49,940 healthy individuals screened, establishing a carrier rate of 1:255 for this mutation. We detected D1152H in nine of 103 individuals referred due to CF-related symptoms. Four suffered from respiratory symptoms and five from congenital bilateral absence of the vas deferens (CBAVD). During this period D1152H was detected in three pregnancies, two of which were aborted. CONCLUSION: The increased frequency of D1152H in individuals referred due to CF-related symptoms compared with healthy individuals included in the CF carrier screening programme (P < 0.001) clearly indicates that it is a disease-causing mutation.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/diagnóstico , Fibrosis Quística/genética , Tamización de Portadores Genéticos , Pruebas Genéticas , Mutación Missense , Diagnóstico Prenatal , Adolescente , Adulto , Niño , Estudios de Cohortes , Fibrosis Quística/epidemiología , Femenino , Frecuencia de los Genes , Humanos , Israel/epidemiología , Judíos/genética , Masculino , Embarazo , Estudios Retrospectivos , Adulto Joven
17.
Genome Biol ; 12(9): R89, 2011 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-21917145

RESUMEN

BACKGROUND: Identification of genes responsible for medically important traits is a major challenge in human genetics. Due to the genetic heterogeneity of hearing loss, targeted DNA capture and massively parallel sequencing are ideal tools to address this challenge. Our subjects for genome analysis are Israeli Jewish and Palestinian Arab families with hearing loss that varies in mode of inheritance and severity. RESULTS: A custom 1.46 MB design of cRNA oligonucleotides was constructed containing 246 genes responsible for either human or mouse deafness. Paired-end libraries were prepared from 11 probands and bar-coded multiplexed samples were sequenced to high depth of coverage. Rare single base pair and indel variants were identified by filtering sequence reads against polymorphisms in dbSNP132 and the 1000 Genomes Project. We identified deleterious mutations in CDH23, MYO15A, TECTA, TMC1, and WFS1. Critical mutations of the probands co-segregated with hearing loss. Screening of additional families in a relevant population was performed. TMC1 p.S647P proved to be a founder allele, contributing to 34% of genetic hearing loss in the Moroccan Jewish population. CONCLUSIONS: Critical mutations were identified in 6 of the 11 original probands and their families, leading to the identification of causative alleles in 20 additional probands and their families. The integration of genomic analysis into early clinical diagnosis of hearing loss will enable prediction of related phenotypes and enhance rehabilitation. Characterization of the proteins encoded by these genes will enable an understanding of the biological mechanisms involved in hearing loss.


Asunto(s)
Pruebas Genéticas/métodos , Pérdida Auditiva/genética , Judíos/genética , Alelos , Animales , Árabes/genética , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Biología Computacional , Análisis Mutacional de ADN/métodos , Exones , Proteínas de la Matriz Extracelular/genética , Efecto Fundador , Proteínas Ligadas a GPI/genética , Frecuencia de los Genes , Biblioteca de Genes , Predisposición Genética a la Enfermedad , Genética de Población , Genoma Humano , Pérdida Auditiva/epidemiología , Humanos , Mutación INDEL , Patrón de Herencia , Proteínas de la Membrana/genética , Ratones , Medio Oriente/epidemiología , Miosinas/genética , Linaje
18.
Am J Hum Genet ; 88(6): 827-838, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21636066

RESUMEN

Congenital cataracts (CCs), responsible for about one-third of blindness in infants, are a major cause of vision loss in children worldwide. Autosomal-recessive congenital cataracts (arCC) form a clinically diverse and genetically heterogeneous group of disorders of the crystalline lens. To identify the genetic cause of arCC in consanguineous Pakistani families, we performed genome-wide linkage analysis and fine mapping and identified linkage to 3p21-p22 with a summed LOD score of 33.42. Mutations in the gene encoding FYVE and coiled-coil domain containing 1 (FYCO1), a PI(3)P-binding protein family member that is associated with the exterior of autophagosomes and mediates microtubule plus-end-directed vesicle transport, were identified in 12 Pakistani families and one Arab Israeli family in which arCC had previously been mapped to the overlapping CATC2 region. Nine different mutations were identified, including c.3755 delC (p.Ala1252AspfsX71), c.3858_3862dupGGAAT (p.Leu1288TrpfsX37), c.1045 C>T (p.Gln349X), c.2206C>T (p.Gln736X), c.2761C>T (p.Arg921X), c.2830C>T (p.Arg944X), c.3150+1 G>T, c.4127T>C (p.Leu1376Pro), and c.1546C>T (p.Gln516X). Fyco1 is expressed in the mouse embryonic and adult lens and peaks at P12d. Expressed mutant proteins p.Leu1288TrpfsX37 and p.Gln736X are truncated on immunoblots. Wild-type and p.L1376P FYCO1, the only missense mutant identified, migrate at the expected molecular mass. Both wild-type and p. Leu1376Pro FYCO1 proteins expressed in human lens epithelial cells partially colocalize to microtubules and are found adjacent to Golgi, but they primarily colocalize to autophagosomes. Thus, FYCO1 is involved in lens development and transparency in humans, and mutations in this gene are one of the most common causes of arCC in the Pakistani population.


Asunto(s)
Catarata/congénito , Catarata/genética , Proteínas de Unión al ADN/genética , Genes Recesivos , Factores de Transcripción/genética , Secuencia de Aminoácidos , Catarata/patología , Pruebas Genéticas , Estudio de Asociación del Genoma Completo , Humanos , Proteínas Asociadas a Microtúbulos , Datos de Secuencia Molecular , Mutación , Pakistán , Linaje
19.
J Appl Genet ; 52(4): 437-41, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21584729

RESUMEN

The purpose of this study was to identify a gene causing non-syndromic X-linked mental retardation in an extended family, taking advantage of the X chromosome inactivation status of the females in order to determine their carrier state. X inactivation in the females was determined with the androgen receptor methylation assay; thereafter, the X chromosome was screened with evenly spaced polymorphic markers. Once initial linkage was identified, the region of interest was saturated with additional markers and the males were added to the analysis. Candidate genes were sequenced. Ten females showed skewed inactivation, while six revealed a normal inactivation pattern. A maximal lod score of 5.54 at θ = 0.00 was obtained with the marker DXS10151. Recombination events mapped the disease gene to a 17.4-Mb interval between the markers DXS10153 and DXS10157. Three candidate genes in the region were sequenced and a previously described missense mutation (P375L) was identified in the ACSL4/FACL4 gene. On the basis of the female X inactivation status, we have mapped and identified the causative mutation in a gene causing non-syndromic X-linked mental retardation.


Asunto(s)
Cromosomas Humanos X/genética , Discapacidad Intelectual Ligada al Cromosoma X/diagnóstico , Inactivación del Cromosoma X , Adolescente , Adulto , Femenino , Estudios de Asociación Genética , Tamización de Portadores Genéticos , Ligamiento Genético , Marcadores Genéticos , Pruebas Genéticas , Haplotipos , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/genética , Linaje
20.
J Pediatr ; 159(3): 466-471.e1, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21429505

RESUMEN

OBJECTIVE: To define the neurologic characteristics and course of ataxia-telangiectasia (A-T). STUDY DESIGN: Retrospective cross-sectional chart study of 57 children (ages 2 to 19 years) followed at an A-T clinic. Cerebellar and extracerebellar symptoms were graded according to degree of functional impairment. Head circumferences were plotted from the charts and z-scores were calculated and compared with that of family members. RESULTS: Ataxia was present in 87.7%, followed by dysarthria (82.1%), dysmetria (75.4%), bradykinesia (69.2%), hyperkinetic movements (58.9%), and dystonia (15.8%). All features aggravated with age. The most striking clinical observation in our patients was low head circumference (z-score below 1), which was present in 60.9%; 17% had true microcephaly (z-score below 2). Microcephaly appeared postnatally, was proportionate to height and weight, and did not correlate with severity of ataxia or genotype. CONCLUSIONS: In addition to cerebellar ataxia, extrapyramidal symptoms, especially bradykinesia, were frequent and disabling. Microcephaly is an integral part of A-T; understanding its pathogenesis may shed light on the mechanism by which ATM mutation causes dysfunction in the nervous system.


Asunto(s)
Ataxia Telangiectasia/epidemiología , Cefalometría , Microcefalia/epidemiología , Adolescente , Envejecimiento , Ataxia Telangiectasia/genética , Niño , Preescolar , Estudios Transversales , Disartria/epidemiología , Disartria/etiología , Discinesias/epidemiología , Discinesias/etiología , Femenino , Humanos , Masculino , Mutación , Trastornos de la Motilidad Ocular/epidemiología , Trastornos de la Motilidad Ocular/etiología , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...