Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 141(11): 1785-1794, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35536377

RESUMEN

The evolutionary conserved Polo-like kinase 4 (PLK4) is essential for centriole duplication, spindle assembly, and de novo centriole formation. In man, homozygous mutations in PLK4 lead to primary microcephaly, altered PLK4 expression is associated with aneuploidy in human embryos. Here, we report on a consanguineous four-generation family with 8 affected individuals compound heterozygous for a novel missense variant, c.881 T > G, and a deletion of the PLK4 gene. The clinical phenotype of the adult patients is mild compared to individuals with previously described PLK4 mutations. One individual was homozygous for the variant c.881G and phenotypically unaffected. The deletion was inherited by 14 of 16 offspring and thus exhibits transmission ratio distortion (TRD). Moreover, based on the already published families with PLK4 mutations, it could be shown that due to the preferential transmission of the mutant alleles, the number of affected offspring is significantly increased. It is assumed that reduced expression of PLK4 decreases the intrinsically high error rate of the first cell divisions after fertilization, increases the number of viable embryos and thus leads to preferential transmission of the deleted/mutated alleles.


Asunto(s)
Proteínas de Ciclo Celular , Centriolos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , División Celular , Centriolos/genética , Centriolos/metabolismo , Humanos , Mutación , Proteínas Serina-Treonina Quinasas/genética
2.
Am J Hum Genet ; 100(2): 297-315, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28132687

RESUMEN

Homozygous SMN1 loss causes spinal muscular atrophy (SMA), the most common lethal genetic childhood motor neuron disease. SMN1 encodes SMN, a ubiquitous housekeeping protein, which makes the primarily motor neuron-specific phenotype rather unexpected. SMA-affected individuals harbor low SMN expression from one to six SMN2 copies, which is insufficient to functionally compensate for SMN1 loss. However, rarely individuals with homozygous absence of SMN1 and only three to four SMN2 copies are fully asymptomatic, suggesting protection through genetic modifier(s). Previously, we identified plastin 3 (PLS3) overexpression as an SMA protective modifier in humans and showed that SMN deficit impairs endocytosis, which is rescued by elevated PLS3 levels. Here, we identify reduction of the neuronal calcium sensor Neurocalcin delta (NCALD) as a protective SMA modifier in five asymptomatic SMN1-deleted individuals carrying only four SMN2 copies. We demonstrate that NCALD is a Ca2+-dependent negative regulator of endocytosis, as NCALD knockdown improves endocytosis in SMA models and ameliorates pharmacologically induced endocytosis defects in zebrafish. Importantly, NCALD knockdown effectively ameliorates SMA-associated pathological defects across species, including worm, zebrafish, and mouse. In conclusion, our study identifies a previously unknown protective SMA modifier in humans, demonstrates modifier impact in three different SMA animal models, and suggests a potential combinatorial therapeutic strategy to efficiently treat SMA. Since both protective modifiers restore endocytosis, our results confirm that endocytosis is a major cellular mechanism perturbed in SMA and emphasize the power of protective modifiers for understanding disease mechanism and developing therapies.


Asunto(s)
Endocitosis/genética , Atrofia Muscular Espinal/genética , Neurocalcina/metabolismo , Animales , Caenorhabditis elegans/genética , Línea Celular , Clonación Molecular , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas Motoras/patología , Atrofia Muscular Espinal/terapia , Neurocalcina/genética , Células PC12 , Linaje , Ratas , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Proteína 2 para la Supervivencia de la Neurona Motora/metabolismo , Transcriptoma , Pez Cebra/genética
3.
Clin Genet ; 98(1): 32-42, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32279305

RESUMEN

Nonsyndromic hearing loss is an extremely heterogeneous disorder. Thus, clinical diagnostics is challenging, in particular due to differences in the etiology of hearing loss between populations. With this study, we wanted to elucidate the genetic basis of hearing loss in 61 consanguineous Egyptian families. In 25 families, linkage analysis was used as a prescreening to identify regions for targeted sequencing of candidate genes. Initially, the coding regions of 12 and later of 94 genes associated with hearing loss were enriched and subjected to massively parallel sequencing (MPS) with diagnostic yields of 36% and 75%, respectively. Causative variants were identified in 48 families (79%). They were found in 23 different genes with the majority being located in MYO15A (15.3%), SLC26A4 (9.7%), GJB2 (8.3%), and MYO7A (6.4%). As many as 32 variants were novel ones at the time of detection. Five variants were shared by two, three, or even four families. Our study provides a first survey of the mutational spectrum of deaf patients in Egypt revealing less GJB2 variants than in many European populations. It underlines the value of targeted enrichment of well-selected deafness genes in combination with MPS in the diagnostics of this frequent and genetically heterogeneous disorder.


Asunto(s)
Sordera/genética , Pérdida Auditiva Sensorineural/genética , Egipto , Familia , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Linaje
4.
Ann Neurol ; 86(3): 368-383, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31298765

RESUMEN

OBJECTIVE: Autosomal dominant optic atrophy (ADOA) starts in early childhood with loss of visual acuity and color vision deficits. OPA1 mutations are responsible for the majority of cases, but in a portion of patients with a clinical diagnosis of ADOA, the cause remains unknown. This study aimed to identify novel ADOA-associated genes and explore their causality. METHODS: Linkage analysis and sequencing were performed in multigeneration families and unrelated patients to identify disease-causing variants. Functional consequences were investigated in silico and confirmed experimentally using the zebrafish model. RESULTS: We defined a new ADOA locus on 7q33-q35 and identified 3 different missense variants in SSBP1 (NM_001256510.1; c.113G>A [p.(Arg38Gln)], c.320G>A [p.(Arg107Gln)] and c.422G>A [p.(Ser141Asn)]) in affected individuals from 2 families and 2 singletons with ADOA and variable retinal degeneration. The mutated arginine residues are part of a basic patch that is essential for single-strand DNA binding. The loss of a positive charge at these positions is very likely to lower the affinity of SSBP1 for single-strand DNA. Antisense-mediated knockdown of endogenous ssbp1 messenger RNA (mRNA) in zebrafish resulted in compromised differentiation of retinal ganglion cells. A similar effect was achieved when mutated mRNAs were administered. These findings point toward an essential role of ssbp1 in retinal development and the dominant-negative nature of the identified human variants, which is consistent with the segregation pattern observed in 2 multigeneration families studied. INTERPRETATION: SSBP1 is an essential protein for mitochondrial DNA replication and maintenance. Our data have established pathogenic variants in SSBP1 as a cause of ADOA and variable retinal degeneration. ANN NEUROL 2019;86:368-383.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad/genética , Proteínas Mitocondriales/genética , Atrofia Óptica Autosómica Dominante/genética , Animales , Diferenciación Celular/genética , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético/genética , Humanos , Masculino , Ratones , Mutación Missense , Atrofia Óptica Autosómica Dominante/patología , Linaje , ARN Mensajero/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Pez Cebra/genética
5.
Mol Cell ; 48(4): 641-6, 2012 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-23063529

RESUMEN

Together with GTP and initiator methionyl-tRNA, translation initiation factor eIF2 forms a ternary complex that binds the 40S ribosome and then scans an mRNA to select the AUG start codon for protein synthesis. Here, we show that a human X-chromosomal neurological disorder characterized by intellectual disability and microcephaly is caused by a missense mutation in eIF2γ (encoded by EIF2S3), the core subunit of the heterotrimeric eIF2 complex. Biochemical studies of human cells overexpressing the eIF2γ mutant and of yeast eIF2γ with the analogous mutation revealed a defect in binding the eIF2ß subunit to eIF2γ. Consistent with this loss of eIF2 integrity, the yeast eIF2γ mutation impaired translation start codon selection and eIF2 function in vivo in a manner that was suppressed by overexpressing eIF2ß. These findings directly link intellectual disability to impaired translation initiation, and provide a mechanistic basis for the human disease due to partial loss of eIF2 function.


Asunto(s)
Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Discapacidad Intelectual/genética , Iniciación de la Cadena Peptídica Traduccional/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Secuencia de Aminoácidos , Secuencia de Bases , Factor 2 Eucariótico de Iniciación/química , Humanos , Modelos Moleculares , Mutación Missense , Proteínas de Saccharomyces cerevisiae/química
6.
Hum Mol Genet ; 26(20): 4055-4066, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29016863

RESUMEN

Congenital cranial dysinnervation disorders (CCDDs) comprise a heterogeneous spectrum of diseases characterized by congenital, non-progressive impairment of eye, eyelid and/or facial movements including Möbius syndrome, Duane retraction syndrome, congenital ptosis, and congenital fibrosis of the extraocular muscles. Over the last 20 years, several CCDDs have been identified as neurodevelopmental disorders that are caused by mutations of genes involved in brain and cranial nerve development, e.g. KIF21A and TUBB3 that each plays a pivotal role for microtubule function. In a five-generation pedigree, we identified a heterozygous mutation of TUBB6, a gene encoding a class V tubulin which has not been linked to a human hereditary disease so far. The missense mutation (p.Phe394Ser) affects an amino acid residue highly conserved in evolution, and co-segregates with a phenotype characterized by congenital non-progressive bilateral facial palsy and congenital velopharyngeal dysfunction presenting with varying degrees of hypomimia, rhinophonia, impaired gag reflex and bilateral ptosis. Expression of the mutated protein in yeast led to an impaired viability compared to wildtype cells when exposed to the microtubule-poison benomyl. Our findings enlarge the spectrum of tubulinopathies and emphasize that mutations of TUBB6 should be considered in patients with congenital non-progressive facial palsy. Further studies are needed to verify whether this phenotype is indeed part of the CCDD spectrum.


Asunto(s)
Blefaroptosis/complicaciones , Blefaroptosis/genética , Parálisis Facial/congénito , Parálisis Facial/genética , Tubulina (Proteína)/genética , Insuficiencia Velofaríngea/congénito , Insuficiencia Velofaríngea/genética , Blefaroptosis/patología , Preescolar , Parálisis Facial/patología , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Mutación , Músculos Oculomotores/patología , Linaje , Insuficiencia Velofaríngea/patología
7.
Genome Res ; 26(2): 183-91, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26755636

RESUMEN

The CRISPR/Cas technology enables targeted genome editing and the rapid generation of transgenic animal models for the study of human genetic disorders. Here we describe an autosomal recessive human disease in two unrelated families characterized by a split-foot defect, nail abnormalities of the hands, and hearing loss, due to mutations disrupting the SAM domain of the protein kinase ZAK. ZAK is a member of the MAPKKK family with no known role in limb development. We show that Zak is expressed in the developing limbs and that a CRISPR/Cas-mediated knockout of the two Zak isoforms is embryonically lethal in mice. In contrast, a deletion of the SAM domain induces a complex hindlimb defect associated with down-regulation of Trp63, a known split-hand/split-foot malformation disease gene. Our results identify ZAK as a key player in mammalian limb patterning and demonstrate the rapid utility of CRISPR/Cas genome editing to assign causality to human mutations in the mouse in <10 wk.


Asunto(s)
Deformidades Congénitas de las Extremidades/genética , Quinasas Quinasa Quinasa PAM/genética , Proteínas Quinasas/genética , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas , Proteína 9 Asociada a CRISPR , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Cocultivo , Endonucleasas , Exoma , Femenino , Humanos , Escala de Lod , Quinasas Quinasa Quinasa PAM/química , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Mutación Missense , Linaje , Polimorfismo de Nucleótido Simple , Proteínas Quinasas/química , Análisis de Secuencia de ADN
8.
Genet Med ; 20(6): 614-621, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29309402

RESUMEN

PurposeHearing loss is genetically extremely heterogeneous, making it suitable for next-generation sequencing (NGS). We identified a four-generation family with nonsyndromic mild to severe hearing loss of the mid- to high frequencies and onset from early childhood to second decade in seven members.MethodsNGS of 66 deafness genes, Sanger sequencing, genome-wide linkage analysis, whole-exome sequencing (WES), semiquantitative reverse-transcriptase polymerase chain reaction.ResultsWe identified a heterozygous nonsense mutation, c.6881G>A (p.Trp2294*), in the last coding exon of PTPRQ. PTPRQ has been linked with recessive (DFNB84A), but not dominant deafness. NGS and Sanger sequencing of all exons (including alternatively spliced 5' and N-scan-predicted exons of a putative "extended" transcript) did not identify a second mutation. The highest logarithm of the odds score was in the PTPRQ-containing region on chromosome 12, and p.Trp2294* cosegregated with hearing loss. WES did not identify other cosegregating candidate variants from the mapped region. PTPRQ expression in patient fibroblasts indicated that the mutant allele escapes nonsense-mediated decay (NMD).ConclusionKnown PTPRQ mutations are recessive and do not affect the C-terminal exon. In contrast to recessive loss-of-function mutations, c.6881G>A transcripts may escape NMD. PTPRQTrp2294* protein would lack only six terminal residues and could exert a dominant-negative effect, a possible explanation for allelic deafness, DFNA73, clinically and genetically distinct from DFNB84A.


Asunto(s)
Sordera/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Secuencia de Aminoácidos , Codón sin Sentido/genética , Exoma/genética , Exones/genética , Familia , Femenino , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Pérdida Auditiva/genética , Humanos , Masculino , Mutación , Linaje , Secuenciación del Exoma
10.
Hum Mol Genet ; 24(9): 2594-603, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25616960

RESUMEN

Determination of variant pathogenicity represents a major challenge in the era of high-throughput sequencing. Erroneous categorization may result if variants affect genes that are in fact dispensable. We demonstrate that this also applies to rare, apparently unambiguous truncating mutations of an established disease gene. By whole-exome sequencing (WES) in a consanguineous family with congenital non-syndromic deafness, we unexpectedly identified a homozygous nonsense variant, p.Arg1066*, in AHI1, a gene associated with Joubert syndrome (JBTS), a severe recessive ciliopathy. None of four homozygotes expressed any signs of JBTS, and one of them had normal hearing, which also ruled out p.Arg1066* as the cause of deafness. Homozygosity mapping and WES in the only other reported JBTS family with a homozygous C-terminal truncation (p.Trp1088Leufs*16) confirmed AHI1 as disease gene, but based on a more N-terminal missense mutation impairing WD40-repeat formation. Morpholinos against N-terminal zebrafish Ahi1, orthologous to where human mutations cluster, produced a ciliopathy, but targeting near human p.Arg1066 and p.Trp1088 did not. Most AHI1 mutations in JBTS patients result in truncated protein lacking WD40-repeats and the SH3 domain; disease was hitherto attributed to loss of these protein interaction modules. Our findings indicate that normal development does not require the C-terminal SH3 domain. This has far-reaching implications, considering that variants like p.Glu984* identified by preconception screening ('Kingsmore panel') do not necessarily indicate JBTS carriership. Genomes of individuals with consanguineous background are enriched for homozygous variants that may unmask dispensable regions of disease genes and unrecognized false positives in diagnostic large-scale sequencing and preconception carrier screening.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Estudios de Asociación Genética , Mutación , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras del Transporte Vesicular , Animales , Encéfalo/patología , Cerebelo/anomalías , Mapeo Cromosómico , Consanguinidad , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Evolución Molecular , Exoma , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Femenino , Orden Génico , Genes Recesivos , Sitios Genéticos , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/genética , Imagen por Resonancia Magnética , Masculino , Modelos Moleculares , Linaje , Conformación Proteica , Retina/anomalías , Pez Cebra/genética
11.
Am J Hum Genet ; 95(5): 622-32, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439729

RESUMEN

Filippi syndrome is a rare, presumably autosomal-recessive disorder characterized by microcephaly, pre- and postnatal growth failure, syndactyly, and distinctive facial features, including a broad nasal bridge and underdeveloped alae nasi. Some affected individuals have intellectual disability, seizures, undescended testicles in males, and teeth and hair abnormalities. We performed homozygosity mapping and whole-exome sequencing in a Sardinian family with two affected children and identified a homozygous frameshift mutation, c.571dupA (p.Ile191Asnfs(∗)6), in CKAP2L, encoding the protein cytoskeleton-associated protein 2-like (CKAP2L). The function of this protein was unknown until it was rediscovered in mice as Radmis (radial fiber and mitotic spindle) and shown to play a pivotal role in cell division of neural progenitors. Sanger sequencing of CKAP2L in a further eight unrelated individuals with clinical features consistent with Filippi syndrome revealed biallelic mutations in four subjects. In contrast to wild-type lymphoblastoid cell lines (LCLs), dividing LCLs established from the individuals homozygous for the c.571dupA mutation did not show CKAP2L at the spindle poles. Furthermore, in cells from the affected individuals, we observed an increase in the number of disorganized spindle microtubules owing to multipolar configurations and defects in chromosome segregation. The observed cellular phenotypes are in keeping with data from in vitro and in vivo knockdown studies performed in human cells and mice, respectively. Our findings show that loss-of-function mutations in CKAP2L are a major cause of Filippi syndrome.


Asunto(s)
Proteínas del Citoesqueleto/genética , Trastornos del Crecimiento/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Sindactilia/genética , Animales , Secuencia de Bases , Análisis Citogenético , Facies , Mutación del Sistema de Lectura/genética , Componentes del Gen , Genes Recesivos/genética , Trastornos del Crecimiento/patología , Humanos , Discapacidad Intelectual/patología , Italia , Masculino , Ratones , Microcefalia/patología , Microscopía Confocal , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Sindactilia/patología
12.
Audiol Neurootol ; 22(1): 30-40, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28601886

RESUMEN

BACKGROUND: Auditory synaptopathy/neuropathy (AS/AN) is a heterogeneous disorder, which may be caused by environmental factors like postnatal hyperbilirubinemia or by genetic factors. The genetic forms are subdivided into syndromic and non-syndromic types, and show different inheritance patterns with a strong preponderance of autosomal-recessive forms. To date, only a single locus for non-syndromic autosomal-dominant AS/AN (AUNA1) has been reported in a single family, in which a non-coding DIAPH3 mutation was subsequently described as causative. MATERIALS AND METHODS: Here, we report detailed clinical data on a large German AS/AN family with slowly progressive postlingual hearing loss. Affected family members developed their first symptoms in their second decade. Moderate hearing loss in the fourth decade then progressed to profound hearing impairment in older family members. Comprehensive audiological and neurological tests were performed in the affected family members. Genetic testing comprised linkage analyses with polymorphic markers and a genome-wide linkage analysis using the Affymetrix GeneChip® Human Mapping 250K. RESULTS AND CONCLUSION: We identified a large family with autosomal-dominant AS/AN. By means of linkage analyses, the AUNA1 locus was excluded, and putatively linked regions on chromosomal bands 12q24 and 13q34 were identified as likely carrying the second locus for autosomal-dominant AS/AN (AUNA2). AUNA2 is associated with a slowly progressive postlingual hearing loss without any evidence for additional symptoms in other organ systems.


Asunto(s)
Cromosomas Humanos Par 12/genética , Cromosomas Humanos Par 13/genética , Nervio Coclear/fisiopatología , Pérdida Auditiva Central/genética , Linaje , Enfermedades del Nervio Vestibulococlear/genética , Adolescente , Adulto , Anciano , Audiometría de Respuesta Evocada , Audiometría de Tonos Puros , Niño , Progresión de la Enfermedad , Femenino , Ligamiento Genético , Alemania , Pérdida Auditiva Central/fisiopatología , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Enfermedades del Nervio Vestibulococlear/fisiopatología , Población Blanca/genética
13.
Nat Genet ; 40(9): 1113-8, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18711368

RESUMEN

Pontocerebellar hypoplasias (PCH) represent a group of neurodegenerative autosomal recessive disorders with prenatal onset, atrophy or hypoplasia of the cerebellum, hypoplasia of the ventral pons, microcephaly, variable neocortical atrophy and severe mental and motor impairments. In two subtypes, PCH2 and PCH4, we identified mutations in three of the four different subunits of the tRNA-splicing endonuclease complex. Our findings point to RNA processing as a new basic cellular impairment in neurological disorders.


Asunto(s)
Cerebelo/anomalías , Endorribonucleasas/genética , Mutación , Puente/anomalías , Encéfalo/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 17 , Humanos , Modelos Moleculares , Polimorfismo de Nucleótido Simple , Síndrome
14.
Hum Mutat ; 37(2): 170-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26593283

RESUMEN

Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.


Asunto(s)
Adenosina Trifosfatasas/genética , Trastornos Sordoceguera/genética , Hipoplasia del Esmalte Dental/genética , Microcefalia/genética , Mutación Missense , Degeneración Retiniana/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Ameloblastos/metabolismo , Ameloblastos/patología , Secuencia de Aminoácidos , Animales , Niño , Cilios/metabolismo , Cilios/patología , Consanguinidad , Trastornos Sordoceguera/metabolismo , Trastornos Sordoceguera/patología , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/patología , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Microcefalia/metabolismo , Microcefalia/patología , Datos de Secuencia Molecular , Odontoblastos/metabolismo , Odontoblastos/patología , Linaje , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Hermanos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Adulto Joven
15.
Hum Mutat ; 37(9): 847-64, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27302555

RESUMEN

Kabuki syndrome (KS) is a rare but recognizable condition that consists of a characteristic face, short stature, various organ malformations, and a variable degree of intellectual disability. Mutations in KMT2D have been identified as the main cause for KS, whereas mutations in KDM6A are a much less frequent cause. Here, we report a mutation screening in a case series of 347 unpublished patients, in which we identified 12 novel KDM6A mutations (KS type 2) and 208 mutations in KMT2D (KS type 1), 132 of them novel. Two of the KDM6A mutations were maternally inherited and nine were shown to be de novo. We give an up-to-date overview of all published mutations for the two KS genes and point out possible mutation hot spots and strategies for molecular genetic testing. We also report the clinical details for 11 patients with KS type 2, summarize the published clinical information, specifically with a focus on the less well-defined X-linked KS type 2, and comment on phenotype-genotype correlations as well as sex-specific phenotypic differences. Finally, we also discuss a possible role of KDM6A in Kabuki-like Turner syndrome and report a mutation screening of KDM6C (UTY) in male KS patients.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Histona Demetilasas/genética , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/patología , Cara/patología , Femenino , Genes Ligados a X , Predisposición Genética a la Enfermedad , Enfermedades Hematológicas/patología , Humanos , Masculino , Herencia Materna , Síndrome de Noonan/genética , Análisis de Secuencia de ADN , Enfermedades Vestibulares/patología
16.
Stroke ; 47(12): 2904-2909, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27827325

RESUMEN

BACKGROUND AND PURPOSE: Carotid intima-media thickness is a marker for subclinical atherosclerosis that predicts subsequent clinical cardiovascular events. The aim of this study was to identify chromosomal loci with linkage or association to common carotid intima-media thickness. METHODS: Nuclear families were recruited using the single parental proband sib-pair design. Genotype data were available for 546 individuals from 132 nuclear families of the Bonn IMT Family Study using the Affymetrix GeneChip Human Mapping 250K Sty chip. Multipoint logarithm of the odds (LOD) scores were determined with the quantitative trait locus statistic implemented in multipoint engine for rapid likelihood. Linkage analysis and family-based association tests were conducted. Data from 2471 German participants from the HNR (Heinz Nixdorf Recall) Study were used for subsequent replication. RESULTS: Two new genomic regions with suggestive linkage (LOD>2) were identified on chromosome 4 (LOD=2.26) and on chromosome 17 (LOD=2.01). Previously reported linkage findings were replicated on chromosomes 13 and 14. Fifteen single nucleotide polymorhisms, located on chromosomes 4, 6, and 9, revealed P<10-4 in the family-based association analyses. One of these signals was replicated in HNR (rs2416804, 1-sided P=1.60×10-3, located in the gene TRAF1). CONCLUSIONS: This study presents the first genome-wide linkage and association study of common carotid intima-media thickness in the German population. Alleles of rs2416804 in TRAF1 were identified as being linked and associated with carotid intima-media thickness. Further studies are needed to evaluate the contribution of this locus to the development of atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Grosor Intima-Media Carotídeo , Factor 1 Asociado a Receptor de TNF/genética , Adulto , Anciano , Femenino , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Alemania , Humanos , Masculino , Persona de Mediana Edad , Núcleo Familiar
17.
Hum Mol Genet ; 23(22): 5940-9, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24951542

RESUMEN

Asymmetric cell division is essential for normal human brain development. Mutations in several genes encoding centrosomal proteins that participate in accurate cell division have been reported to cause autosomal recessive primary microcephaly (MCPH). By homozygosity mapping including three affected individuals from a consanguineous MCPH family from Pakistan, we delineated a critical region of 18.53 Mb on Chromosome 1p21.3-1p13.1. This region contains the gene encoding HsSAS-6, a centrosomal protein primordial for seeding the formation of new centrioles during the cell cycle. Both next-generation and Sanger sequencing revealed a homozygous c.185T>C missense mutation in the HsSAS-6 gene, resulting in a p.Ile62Thr substitution within a highly conserved region of the PISA domain of HsSAS-6. This variant is neither present in any single-nucleotide polymorphism or exome sequencing databases nor in a Pakistani control cohort. Experiments in tissue culture cells revealed that the Ile62Thr mutant of HsSAS-6 is substantially less efficient than the wild-type protein in sustaining centriole formation. Together, our findings demonstrate a dramatic impact of the mutation p.Ile62Thr on HsSAS-6 function and add this component to the list of genes mutated in primary microcephaly.


Asunto(s)
Pueblo Asiatico/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Mutación Missense , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Niño , Preescolar , Exoma , Femenino , Humanos , Masculino , Microcefalia/genética , Microcefalia/metabolismo , Persona de Mediana Edad , Datos de Secuencia Molecular , Pakistán , Linaje , Polimorfismo de Nucleótido Simple , Estructura Terciaria de Proteína , Alineación de Secuencia
18.
Hum Genet ; 135(2): 157-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26621532

RESUMEN

Primary microcephaly is a disorder characterized by a small head and brain associated with impaired cognitive capabilities. Mutations in 13 different genes encoding centrosomal proteins and cell cycle regulators have been reported to cause the disease. CASC5, a gene encoding a protein important for kinetochore formation and proper chromosome segregation during mitosis, has been suggested to be associated with primary microcephaly-4 (MCPH4). This was based on one mutation only and circumstantial functional evidence. By combining homozygosity mapping and whole-exome sequencing in an MCPH family from Pakistan, we identified a second mutation (NM_170589.4;c.6673-19T>A) in CASC5. This mutation induced skipping of exon 25 of CASC5 resulting in a frameshift and the introduction of a premature stop codon (p.Met2225Ilefs*7). The C-terminally truncated protein lacks 118 amino acids that encompass the region responsible for the interaction with the hMIS12 complex, which is essential for proper chromosome alignment and segregation. Furthermore, we showed a down-regulation of CASC5 mRNA and reduction of the amount of CASC5 protein by quantitative RT-PCR and western blot analysis, respectively. As a further sign of functional deficits, we observed dispersed dots of CASC5 immunoreactive material outside the metaphase plate of dividing patient fibroblasts. Normally, CASC5 is a component of the kinetochore of metaphase chromosomes. A higher mitotic index in patient cells indicated a mitotic arrest in the cells carrying the mutation. We also observed lobulated and fragmented nuclei as well as micronuclei in the patient cells. Moreover, we detected an altered DNA damage response with higher levels of γH2AX and 53BP1 in mutant as compared to control fibroblasts. Our findings substantiate the proposed role of CASC5 for primary microcephaly and suggest that it also might be relevant for genome stability.


Asunto(s)
Pueblo Asiatico/genética , Homocigoto , Microcefalia/genética , Proteínas Asociadas a Microtúbulos/genética , Empalme del ARN , Secuencia de Aminoácidos , Células Cultivadas , Segregación Cromosómica , Codón sin Sentido/genética , Codón sin Sentido/metabolismo , Daño del ADN/genética , Regulación hacia Abajo , Exones , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Mutación del Sistema de Lectura , Ligamiento Genético , Sitios Genéticos , Estudio de Asociación del Genoma Completo , Humanos , Cinetocoros/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Mitosis , Datos de Secuencia Molecular , Pakistán , Linaje , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
19.
Am J Hum Genet ; 93(4): 727-34, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24035193

RESUMEN

In guanosine diphosphate (GDP)-mannose pyrophosphorylase A (GMPPA), we identified a homozygous nonsense mutation that segregated with achalasia and alacrima, delayed developmental milestones, and gait abnormalities in a consanguineous Pakistani pedigree. Mutations in GMPPA were subsequently found in ten additional individuals from eight independent families affected by the combination of achalasia, alacrima, and neurological deficits. This autosomal-recessive disorder shows many similarities with triple A syndrome, which is characterized by achalasia, alacrima, and variable neurological deficits in combination with adrenal insufficiency. GMPPA is a largely uncharacterized homolog of GMPPB. GMPPB catalyzes the formation of GDP-mannose, which is an essential precursor of glycan moieties of glycoproteins and glycolipids and is associated with congenital and limb-girdle muscular dystrophies with hypoglycosylation of α-dystroglycan. Surprisingly, GDP-mannose pyrophosphorylase activity was unchanged and GDP-mannose levels were strongly increased in lymphoblasts of individuals with GMPPA mutations. This suggests that GMPPA might serve as a GMPPB regulatory subunit mediating feedback inhibition of GMPPB instead of displaying catalytic enzyme activity itself. Thus, a triple-A-like syndrome can be added to the growing list of congenital disorders of glycosylation, in which dysregulation rather than mere enzyme deficiency is the basal pathophysiological mechanism.


Asunto(s)
Codón sin Sentido , Genes Recesivos/genética , Guanosina Difosfato Manosa/genética , Discapacidad Intelectual/genética , Nucleotidiltransferasas/genética , Adolescente , Insuficiencia Suprarrenal/genética , Adulto , Niño , Consanguinidad , Acalasia del Esófago/genética , Enfermedades Hereditarias del Ojo/genética , Glicosilación , Guanosina Difosfato Manosa/metabolismo , Homocigoto , Humanos , Discapacidad Intelectual/enzimología , Enfermedades del Aparato Lagrimal/genética , Enfermedades del Sistema Nervioso/genética , Nucleotidiltransferasas/metabolismo , Linaje , Adulto Joven
20.
Am J Hum Genet ; 93(1): 181-90, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23830518

RESUMEN

Myopathies are a clinically and etiologically heterogeneous group of disorders that can range from limb girdle muscular dystrophy (LGMD) to syndromic forms with associated features including intellectual disability. Here, we report the identification of mutations in transport protein particle complex 11 (TRAPPC11) in three individuals of a consanguineous Syrian family presenting with LGMD and in five individuals of Hutterite descent presenting with myopathy, infantile hyperkinetic movements, ataxia, and intellectual disability. By using a combination of whole-exome or genome sequencing with homozygosity mapping, we identified the homozygous c.2938G>A (p.Gly980Arg) missense mutation within the gryzun domain of TRAPPC11 in the Syrian LGMD family and the homozygous c.1287+5G>A splice-site mutation resulting in a 58 amino acid in-frame deletion (p.Ala372_Ser429del) in the foie gras domain of TRAPPC11 in the Hutterite families. TRAPPC11 encodes a component of the multiprotein TRAPP complex involved in membrane trafficking. We demonstrate that both mutations impair the binding ability of TRAPPC11 to other TRAPP complex components and disrupt the Golgi apparatus architecture. Marker trafficking experiments for the p.Ala372_Ser429del deletion indicated normal ER-to-Golgi trafficking but dramatically delayed exit from the Golgi to the cell surface. Moreover, we observed alterations of the lysosomal membrane glycoproteins lysosome-associated membrane protein 1 (LAMP1) and LAMP2 as a consequence of TRAPPC11 dysfunction supporting a defect in the transport of secretory proteins as the underlying pathomechanism.


Asunto(s)
Discapacidad Intelectual/genética , Trastornos del Movimiento/genética , Enfermedades Musculares/genética , Distrofia Muscular de Cinturas/genética , Eliminación de Secuencia , Proteínas de Transporte Vesicular/metabolismo , Adolescente , Adulto , Ataxia/genética , Mapeo Cromosómico , Consanguinidad , Creatina Quinasa/sangre , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Exoma , Femenino , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Homocigoto , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Masculino , Trastornos del Movimiento/patología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Enfermedades Musculares/patología , Distrofia Muscular de Cinturas/patología , Linaje , Unión Proteica , Transporte de Proteínas , Sitios de Empalme de ARN , Siria , Proteínas de Transporte Vesicular/genética , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA