Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 24(1): 230-42, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25168386

RESUMEN

Primary cilia are sensory organelles present on most mammalian cells. The assembly and maintenance of primary cilia are facilitated by intraflagellar transport (IFT), a bidirectional protein trafficking along the cilium. Mutations in genes coding for IFT components have been associated with a group of diseases called ciliopathies. These genetic disorders can affect a variety of organs including the retina. Using whole exome sequencing in three families, we identified mutations in Intraflagellar Transport 172 Homolog [IFT172 (Chlamydomonas)] that underlie an isolated retinal degeneration and Bardet-Biedl syndrome. Extensive functional analyses of the identified mutations in cell culture, rat retina and in zebrafish demonstrated their hypomorphic or null nature. It has recently been reported that mutations in IFT172 cause a severe ciliopathy syndrome involving skeletal, renal, hepatic and retinal abnormalities (Jeune and Mainzer-Saldino syndromes). Here, we report for the first time that mutations in this gene can also lead to an isolated form of retinal degeneration. The functional data for the mutations can partially explain milder phenotypes; however, the involvement of modifying alleles in the IFT172-associated phenotypes cannot be excluded. These findings expand the spectrum of disease associated with mutations in IFT172 and suggest that mutations in genes originally reported to be associated with syndromic ciliopathies should also be considered in subjects with non-syndromic retinal dystrophy.


Asunto(s)
Síndrome de Bardet-Biedl/genética , Síndrome de Bardet-Biedl/patología , Proteínas Portadoras/genética , Retina/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Adulto , Animales , Células Cultivadas , Proteínas del Citoesqueleto , Exoma , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación , Linaje , Ratas , Retina/metabolismo , Análisis de Secuencia de ADN , Adulto Joven , Pez Cebra
2.
J Med Genet ; 51(3): 143-51, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24401286

RESUMEN

Many proteins depend on post-translational prenylation for a correct subcellular localisation and membrane anchoring. This involves the covalent attachment of farnesyl or geranylgeranyl residues to cysteines residing in consensus motifs at the C-terminal parts of proteins. Retinal photoreceptor cells are highly compartmentalised and membranous structures, and therefore it can be expected that the proper function of many retinal proteins depends on prenylation, which has been proven for several proteins that are absent or defective in different inherited retinal diseases (IRDs). These include proteins involved in the phototransduction cascade, such as GRK1, the phosphodiesterase 6 subunits and the transducin γ subunit, or proteins involved in transport processes, such as RAB28 and retinitis pigmentosa GTPase regulator (RPGR). In addition, there is another class of general prenylation defects due to mutations in proteins such as AIPL1, PDE6D and rab escort protein-1 (REP-1), which can act as chaperones for subsets of prenylated retinal proteins that are associated with IRDs. REP-1 also is a key accessory protein of geranylgeranyltransferase II, an enzyme involved in the geranylgeranylation of almost all members of a large family of Rab GTPases. Finally, mutations in the mevalonate kinase (MVK) gene, which were known to be principally associated with mevalonic aciduria, were recently associated with non-syndromic retinitis pigmentosa. We hypothesise that MVK deficiency results in a depletion of prenyl moieties that affects the prenylation of many proteins synthesised specifically in the retina, including Rabs. In this review, we discuss the entire spectrum of prenylation defects underlying progressive degeneration of photoreceptors, the retinal pigment epithelium and the choroid.


Asunto(s)
Prenilación de Proteína , Enfermedades de la Retina , Animales , Humanos , Ratones
3.
Am J Hum Genet ; 89(2): 253-64, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21835304

RESUMEN

A fundamental challenge in analyzing exome-sequence data is distinguishing pathogenic mutations from background polymorphisms. To address this problem in the context of a genetically heterogeneous disease, retinitis pigmentosa (RP), we devised a candidate-gene prioritization strategy called cis-regulatory mapping that utilizes ChIP-seq data for the photoreceptor transcription factor CRX to rank candidate genes. Exome sequencing combined with this approach identified a homozygous nonsense mutation in male germ cell-associated kinase (MAK) in the single affected member of a consanguineous Turkish family with RP. MAK encodes a cilium-associated mitogen-activated protein kinase whose function is conserved from the ciliated alga, Chlamydomonas reinhardtii, to humans. Mutations in MAK orthologs in mice and other model organisms result in abnormally long cilia and, in mice, rapid photoreceptor degeneration. Subsequent sequence analyses of additional individuals with RP identified five probands with missense mutations in MAK. Two of these mutations alter amino acids that are conserved in all known kinases, and an in vitro kinase assay indicates that these mutations result in a loss of kinase activity. Thus, kinase activity appears to be critical for MAK function in humans. This study highlights a previously underappreciated role for CRX as a direct transcriptional regulator of ciliary genes in photoreceptors. In addition, it demonstrates the effectiveness of CRX-based cis-regulatory mapping in prioritizing candidate genes from exome data and suggests that this strategy should be generally applicable to a range of retinal diseases.


Asunto(s)
Cilios/genética , Exones/genética , Mutación/genética , Proteínas Serina-Treonina Quinasas/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retinitis Pigmentosa/genética , Análisis de Secuencia de ADN , Adulto , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cilios/enzimología , Femenino , Genes Recesivos/genética , Sitios Genéticos/genética , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Retinitis Pigmentosa/enzimología , Rodopsina/genética , Transactivadores/metabolismo , Transcripción Genética , Adulto Joven
4.
Mol Vis ; 20: 753-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24940029

RESUMEN

PURPOSE: The gene encoding nicotinamide nucleotide adenylyltransferase 1 (NMNAT1) was recently found to be mutated in a subset of patients with Leber congenital amaurosis (LCA) with macular atrophy. The aim of this study was to determine the occurrence and frequency of NMNAT1 mutations and associated phenotypes in different types of inherited retinal dystrophies. METHODS: DNA samples of 161 patients with LCA without genetic diagnosis were analyzed for variants in NMNAT1 using Sanger sequencing. Variants in exon 5 of NMNAT1, which harbors the majority of the previously identified mutations, were screened in 532 additional patients with retinal dystrophies. This cohort encompassed 108 persons with isolated or autosomal recessive cone-rod dystrophy (CRD), 271 with isolated or autosomal recessive retinitis pigmentosa (RP), and 49 with autosomal dominant RP, as well as 104 persons with LCA in whom the causative mutation was previously identified. RESULTS: Compound heterozygous alterations were found in six patients with LCA and in one person with early-onset RP. All except one carried the common p.E257K variant on one allele. Macular atrophy was absent in one patient, who carried this variant in combination with a truncating mutation on the other allele. The p.E257K alteration was also found in a heterozygous state in five individuals with LCA and one with RP while no mutation was detected on the other allele. Two individuals with LCA carried other NMNAT1 variants in a heterozygous state, whereas no NMNAT1 variants in exon 5 were identified in individuals with CRD. The p.E257K variant was found to be enriched in a heterozygous state in individuals with LCA (0.94%) compared to Caucasian controls (0.18%), although the difference was statistically insignificant (p=0.12). CONCLUSIONS: Although macular atrophy can occur in LCA and CRD, no NMNAT1 mutations were found in the latter cohort. NMNAT1 variants were also not found in a large group of patients with sporadic or autosomal recessive RP. The enrichment of p.E257K in a heterozygous state in patients with LCA versus controls suggests that this allele could act as a modifier in other genetic subtypes of LCA.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Heterocigoto , Amaurosis Congénita de Leber/enzimología , Amaurosis Congénita de Leber/genética , Nicotinamida-Nucleótido Adenililtransferasa/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Adulto Joven
5.
Am J Hum Genet ; 87(2): 199-208, 2010 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-20673862

RESUMEN

Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal diseases caused by progressive degeneration of the photoreceptor cells. Using autozygosity mapping, we identified two families, each with three affected siblings sharing large overlapping homozygous regions that harbored the IMPG2 gene on chromosome 3. Sequence analysis of IMPG2 in the two index cases revealed homozygous mutations cosegregating with the disease in the respective families: three affected siblings of Iraqi Jewish ancestry displayed a nonsense mutation, and a Dutch family displayed a 1.8 kb genomic deletion that removes exon 9 and results in the absence of seven amino acids in a conserved SEA domain of the IMPG2 protein. Transient transfection of COS-1 cells showed that a construct expressing the wild-type SEA domain is properly targeted to the plasma membrane, whereas the mutant lacking the seven amino acids appears to be retained in the endoplasmic reticulum. Mutation analysis in ten additional index cases that were of Dutch, Israeli, Italian, and Pakistani origin and had homozygous regions encompassing IMPG2 revealed five additional mutations; four nonsense mutations and one missense mutation affecting a highly conserved phenylalanine residue. Most patients with IMPG2 mutations showed an early-onset form of RP with progressive visual-field loss and deterioration of visual acuity. The patient with the missense mutation, however, was diagnosed with maculopathy. The IMPG2 gene encodes the interphotoreceptor matrix proteoglycan IMPG2, which is a constituent of the interphotoreceptor matrix. Our data therefore show that mutations in a structural component of the interphotoreceptor matrix can cause arRP.


Asunto(s)
Genes Recesivos/genética , Mutación/genética , Proteoglicanos/genética , Retinitis Pigmentosa/genética , Adulto , Anciano , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Mapeo Cromosómico , Segregación Cromosómica/genética , Análisis Mutacional de ADN , Femenino , Fondo de Ojo , Ligamiento Genético , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Linaje , Proteoglicanos/química , Fracciones Subcelulares/metabolismo
6.
Ophthalmology ; 120(12): 2697-2705, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24084495

RESUMEN

OBJECTIVE: Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous disorder characterized by night blindness and peripheral vision loss, and in many cases leads to blindness. Despite extensive knowledge about genes involved in the pathogenesis of RP, the genetic cause remains elusive in many patients. In this study, we aimed to identify novel genes that are involved in the cause of RP. DESIGN: We present a case series with mutations in the mevalonate kinase (MVK) gene. PARTICIPANTS: A total of 769 patients with nonsyndromic RP and 174 Dutch control individuals participated in this study. METHODS: Exome sequencing analysis was performed in a proband of Dutch origin who was initially diagnosed with nonsyndromic autosomal recessive RP. Mutations in MVK were identified and subsequently tested for segregation within the patient's family and screened in a large cohort of patients with genetically unsolved RP. Patients with mutations underwent extensive clinical reexamination. MAIN OUTCOME MEASURES: Digital fundus photography, spectral-domain optical coherence tomography (OCT), and fundus autofluorescence analysis were performed in patients with MVK mutations. Mevalonate kinase (MK) enzyme activity was analyzed in cultured lymphoblastoid cells, and mevalonic acid levels were measured in urine samples. RESULTS: Exome variant filtering and prioritization led to the identification of compound heterozygous mutations in MVK (p.I268T and p.A334T) in the proband and her affected brother. Screening of our nonsyndromic RP patient cohort revealed an additional individual who was homozygous for the p.A334T alteration. Clinical reevaluation of all 3 patients showed a classic form of RP with variable extraocular symptoms, such as history of recurrent childhood febrile crises in 2 patients, mild ataxia in 1, and renal failure in 1. All 3 affected individuals showed a significantly decreased MK activity and highly elevated levels of urinary mevalonic acid. CONCLUSIONS: Although the MK activity in cells and mevalonic acid concentrations in urine are strongly aberrant and comparable to that in patients with systemic mevalonate kinase deficiency (MKD), only mild clinical symptoms related to this syndrome were observed in our patients. In the current article, we add another phenotype to the spectrum of diverging disorders associated with mutations in MVK.


Asunto(s)
Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Retinitis Pigmentosa/genética , Adulto , Análisis Mutacional de ADN , Electrorretinografía , Exoma/genética , Femenino , Angiografía con Fluoresceína , Humanos , Masculino , Ácido Mevalónico/orina , Persona de Mediana Edad , Linaje , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/orina , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología
7.
Hum Mutat ; 33(6): 963-72, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22334370

RESUMEN

Molecular diagnostics for patients with retinitis pigmentosa (RP) has been hampered by extreme genetic and clinical heterogeneity, with 52 causative genes known to date. Here, we developed a comprehensive next-generation sequencing (NGS) approach for the clinical molecular diagnostics of RP. All known inherited retinal disease genes (n = 111) were captured and simultaneously analyzed using NGS in 100 RP patients without a molecular diagnosis. A systematic data analysis pipeline was developed and validated to prioritize and predict the pathogenicity of all genetic variants identified in each patient, which enabled us to reduce the number of potential pathogenic variants from approximately 1,200 to zero to nine per patient. Subsequent segregation analysis and in silico predictions of pathogenicity resulted in a molecular diagnosis in 36 RP patients, comprising 27 recessive, six dominant, and three X-linked cases. Intriguingly, De novo mutations were present in at least three out of 28 isolated cases with causative mutations. This study demonstrates the enormous potential and clinical utility of NGS in molecular diagnosis of genetically heterogeneous diseases such as RP. De novo dominant mutations appear to play a significant role in patients with isolated RP, having major implications for genetic counselling.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Retinitis Pigmentosa/diagnóstico , Alelos , Biología Computacional/métodos , Femenino , Variación Genética , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Mutación , Linaje , Fenotipo , Control de Calidad , Reproducibilidad de los Resultados , Retinitis Pigmentosa/genética
8.
Mol Vis ; 18: 2411-9, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077400

RESUMEN

PURPOSE: The purpose of this study was to identify the underlying molecular genetic defect in an Indonesian family with three affected individuals who had received a diagnosis of retinitis pigmentosa (RP). METHODS: Clinical evaluation of the family members included measuring visual acuity and fundoscopy, and assessing visual field and color vision. Genomic DNA of the three affected individuals was analyzed with Illumina 700k single nucleotide polymorphism (SNP) arrays, and homozygous regions were identified using PLINK software. Mutation analysis was performed with sequence analysis of the retinitis pigmentosa 1 (RP1) gene that resided in one of the homozygous regions. The frequency of the identified mutation in the Indonesian population was determined with TaqI restriction fragment length polymorphism analysis. RESULTS: A novel homozygous nonsense mutation in exon 4 of the RP1 gene, c.1012C>T (p.R338*), was identified in the proband and her two affected sisters. Unaffected family members either carried two wild-type alleles or were heterozygous carriers of the mutation. The mutation was not present in 184 Indonesian control samples. CONCLUSIONS: Most of the previously reported RP1 mutations are inherited in an autosomal dominant mode, and appear to cluster in exon 4. Here, we identified a novel homozygous p.R338* mutation in exon 4 of RP1, and speculate on the mutational mechanisms of different RP1 mutations underlying dominant and recessive RP.


Asunto(s)
Pueblo Asiatico/genética , Proteínas del Ojo/genética , Retina/metabolismo , Retinitis Pigmentosa/genética , Adulto , Estudios de Casos y Controles , Codón sin Sentido , Análisis Mutacional de ADN , Electrorretinografía , Exones , Femenino , Mutación del Sistema de Lectura , Genes Recesivos , Genotipo , Heterocigoto , Homocigoto , Humanos , Indonesia , Masculino , Proteínas Asociadas a Microtúbulos , Persona de Mediana Edad , Linaje , Retina/patología , Agudeza Visual
9.
Mol Vis ; 18: 2447-53, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077403

RESUMEN

PURPOSE: The purpose of this study is to identify the genetic defect in a Turkish family with autosomal recessive retinitis pigmentosa, nanophthalmos, and optic disc drusen. METHODS: Ophthalmological examinations consisted of measuring the best-corrected visual acuity and the refractive error, electroretinography, optical coherence tomography, B-mode ultrasonography, and fundus photography. The involvement of the membrane frizzled-related protein (MFRP) gene in this family was studied with direct DNA sequencing of the coding exons of MFRP and with linkage analysis with microsatellite markers. After MFRP was excluded, genome-wide homozygosity mapping was performed with 250 K single nucleotide polymorphism (SNP) microarrays. Mutation analysis of the crumbs homolog 1 (CRB1) gene was performed with direct sequencing. RESULTS: Ophthalmological evaluation of both affected individuals in the family revealed a decreased axial length (18-19 mm), retinal dystrophy, macular edema, and hyperopia of >+8.0 diopters. Sequencing of MFRP did not reveal any pathogenic changes, and microsatellite marker analysis showed that the chromosomal region did not segregate within the disease in this family. Genome-wide homozygosity mapping using single nucleotide polymorphism microarrays revealed a 28-Mb homozygous region encompassing the CRB1 gene, and direct sequencing disclosed a novel homozygous missense mutation (p.Gly833Asp) in CRB1. CONCLUSIONS: Previous studies associated mutations in the MFRP gene with the syndrome nanophthalmos-retinitis pigmentosa-foveoschisis-optic disc drusen. In this study, we demonstrated that a similar disease complex can be caused by mutations in the CRB1 gene.


Asunto(s)
Proteínas del Ojo/genética , Proteínas de la Membrana/genética , Microftalmía/genética , Mutación Missense , Proteínas del Tejido Nervioso/genética , Drusas del Disco Óptico/genética , Retinitis Pigmentosa/genética , Adolescente , Secuencia de Bases , Niño , Consanguinidad , Electrorretinografía , Exones , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Microftalmía/complicaciones , Microftalmía/patología , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Drusas del Disco Óptico/complicaciones , Drusas del Disco Óptico/patología , Linaje , Retinitis Pigmentosa/complicaciones , Retinitis Pigmentosa/patología , Análisis de Secuencia de ADN
10.
Mol Vis ; 17: 3013-24, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22128245

RESUMEN

PURPOSE: Retinitis pigmentosa (RP) is a clinically and genetically heterogeneous retinal disorder. Despite tremendous knowledge about the genes involved in RP, little is known about the genetic causes of RP in Indonesia. Here, we aim to identify the molecular genetic causes underlying RP in a small cohort of Indonesian patients, using genome-wide homozygosity mapping. METHODS: DNA samples from affected and healthy individuals from 14 Indonesian families segregating autosomal recessive, X-linked, or isolated RP were collected. Homozygosity mapping was conducted using Illumina 6k or Affymetrix 5.0 single nucleotide polymorphism (SNP) arrays. Known autosomal recessive RP (arRP) genes residing in homozygous regions and X-linked RP genes were sequenced for mutations. RESULTS: In ten out of the 14 families, homozygous regions were identified that contained genes known to be involved in the pathogenesis of RP. Sequence analysis of these genes revealed seven novel homozygous mutations in ATP-binding cassette, sub-family A, member 4 (ABCA4), crumbs homolog 1 (CRB1), eyes shut homolog (Drosophila) (EYS), c-mer proto-oncogene tyrosine kinase (MERTK), nuclear receptor subfamily 2, group E, member 3 (NR2E3) and phosphodiesterase 6A, cGMP-specific, rod, alpha (PDE6A), all segregating in the respective families. No mutations were identified in the X-linked genes retinitis pigmentosa GTPase regulator (RPGR) and retinitis pigmentosa 2 (X-linked recessive; RP2). CONCLUSIONS: Homozygosity mapping is a powerful tool to identify the genetic defects underlying RP in the Indonesian population. Compared to studies involving patients from other populations, the same genes appear to be implicated in the etiology of recessive RP in Indonesia, although all mutations that were discovered are novel and as such may be unique for this population.


Asunto(s)
Pueblo Asiatico , Mapeo Cromosómico/métodos , Proteínas del Ojo/genética , Genes Recesivos , Genoma Humano , Homocigoto , Retinitis Pigmentosa/genética , Adolescente , Adulto , Anciano , Niño , Estudios de Cohortes , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Genes Ligados a X , Pruebas Genéticas , Estudio de Asociación del Genoma Completo , Humanos , Indonesia/epidemiología , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación , Linaje , Polimorfismo de Nucleótido Simple , Proto-Oncogenes Mas , Retinitis Pigmentosa/etnología , Retinitis Pigmentosa/patología
11.
Ophthalmic Genet ; 38(2): 127-132, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27029556

RESUMEN

BACKGROUND: Retinitis pigmentosa (RP) is the most common cause of inherited retinal degeneration and can occur in non-syndromic and syndromic forms. Syndromic RP is accompanied by other symptoms such as intellectual disability, hearing loss, or congenital abnormalities. Both forms are known to exhibit complex genetic interactions that can modulate the penetrance and expressivity of the phenotype. MATERIALS AND METHODS: In an individual with atypical RP, hearing loss, ataxia and cerebellar atrophy, whole exome sequencing was performed. The candidate pathogenic variants were tested by developing an in vivo zebrafish model and assaying for retinal and cerebellar integrity. RESULTS: Exome sequencing revealed a complex heterozygous protein-truncating mutation in RP1L1, p.[(Lys111Glnfs*27; Gln2373*)], and a heterozygous nonsense mutation in C2orf71, p.(Ser512*). Mutations in both genes have previously been implicated in autosomal recessive non-syndromic RP, raising the possibility of a digenic model in this family. Functional testing in a zebrafish model for two key phenotypes of the affected person showed that the combinatorial suppression of rp1l1 and c2orf71l induced discrete pathology in terms of reduction of eye size with concomitant loss of rhodopsin in the photoreceptors, and disorganization of the cerebellum. CONCLUSIONS: We propose that the combination of heterozygous loss-of-function mutations in these genes drives syndromic retinal dystrophy, likely through the genetic interaction of at least two loci. Haploinsufficiency at each of these loci is insufficient to induce overt pathology.


Asunto(s)
Proteínas del Ojo/genética , Predisposición Genética a la Enfermedad , Patrón de Herencia , Mutación/genética , Retinitis Pigmentosa/genética , Adulto , Animales , Hibridación Genómica Comparativa , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Embrión no Mamífero , Exoma/genética , Femenino , Silenciador del Gen , Heterocigoto , Humanos , Linaje , Retinitis Pigmentosa/patología , Rodopsina/genética , Análisis de Secuencia de ADN , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
12.
Acta Ophthalmol ; 93(1): 83-94, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25385675

RESUMEN

PURPOSE: Defects in MAK, encoding a protein localized to the photoreceptor connecting cilium, have recently been associated with autosomal recessive retinitis pigmentosa (RP). The aim of this study is to describe our detailed clinical observations in patients with MAK-associated RP, including an assessment of syndromic symptoms frequently observed in ciliopathies. METHODS: In this international collaborative study, 11 patients carrying nonsense or missense mutations in MAK were clinically evaluated, including extensive assessment of the medical history, slit-lamp biomicroscopy, ophthalmoscopy, kinetic perimetry, electroretinography (ERG), spectral-domain optical coherence tomography (SD-OCT), autofluorescence imaging and fundus photography. Additionally, we used a questionnaire to evaluate the presence of syndromic features and tested the olfactory function. RESULTS: MAK-associated RP is not associated with syndromic features, not even with subclinical dysfunction of the olfactory apparatus. All patients experienced typical RP symptoms of night blindness followed by visual field constriction. Symptoms initiated between childhood and the age of 43 (mean: 23 years). Although some patients experienced vision loss, the visual acuity remained normal in most patients. ERG and ophthalmoscopy revealed classic RP characteristics, and SD-OCT demonstrated thinning of the overall retina, outer nuclear layer and photoreceptor-pigment epithelium complex. CONCLUSION: Nonsense and missense mutations in MAK give rise to a non-syndromic recessive RP phenotype without apparent extra-ocular features. When compared to other retinal ciliopathies, MAK-associated RP appears to be relatively mild and shows remarkable resemblance to RP1-associated RP, which could be explained by the close functional relation of these proteins.


Asunto(s)
Codón sin Sentido , Mutación Missense , Cilio Conector de los Fotorreceptores/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Retinitis Pigmentosa/genética , Adulto , Anciano , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Retinitis Pigmentosa/patología , Encuestas y Cuestionarios , Tomografía de Coherencia Óptica , Pruebas del Campo Visual , Adulto Joven
13.
Artículo en Inglés | MEDLINE | ID: mdl-24939053

RESUMEN

In view of their high degree of genetic heterogeneity, inherited retinal diseases (IRDs) pose a significant challenge for identifying novel genetic causes. Thus far, more than 200 genes have been found to be mutated in IRDs, which together contain causal variants in >80% of the cases. Accurate genetic diagnostics is particularly important for isolated cases, in which X-linked and de novo autosomal dominant variants are not uncommon. In addition, new gene- or mutation-specific therapies are emerging, underlining the importance of identifying causative mutations in each individual. Sanger sequencing of selected genes followed by cost-effective targeted next-generation sequencing (NGS) can identify defects in known IRD-associated genes in the majority of the cases. Exome NGS in combination with genetic linkage or homozygosity mapping studies can aid the identification of the remaining causal genes. As these are thought to be mutated in <1% of the cases, validation through functional modeling in, for example, zebrafish and/or replication through the genotyping of large patient cohorts is required. In the near future, whole genome NGS in combination with transcriptome NGS may reveal mutations that are currently hidden in the noncoding regions of the human genome.


Asunto(s)
Genómica/métodos , Mutación/genética , Degeneración Retiniana/genética , Variaciones en el Número de Copia de ADN/genética , Exoma/genética , Perfilación de la Expresión Génica/métodos , Frecuencia de los Genes/genética , Ligamiento Genético/genética , Técnicas Genéticas , Pruebas Genéticas/métodos , Estudio de Asociación del Genoma Completo/métodos , Humanos
14.
JAMA Ophthalmol ; 132(8): 1002-4, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24830548

RESUMEN

IMPORTANCE: The NMNAT1 gene was recently found to be mutated in a subset of patients with Leber congenital amaurosis and macular atrophy. The most prevalent NMNAT1 variant was p.Glu257Lys, which was observed in 38 of 106 alleles (35.8%). On the basis of functional assays, it was deemed a severe variant. OBSERVATIONS: The p.Glu257Lys variant was 80-fold less frequent in a homozygous state in patients with Leber congenital amaurosis than predicted based on its heterozygosity frequency in the European American population. Moreover, we identified this variant in a homozygous state in a patient with no ocular abnormalities. CONCLUSIONS AND RELEVANCE: On the basis of these results, the p.Glu257Lys variant is considered not fully penetrant. Homozygotes of the p.Glu257Lys variant in most persons are therefore not associated with ocular disease. Consequently, genetic counselors should exercise great caution in the interpretation of this variant.


Asunto(s)
Amaurosis Congénita de Leber/genética , Mutación , Nicotinamida-Nucleótido Adenililtransferasa/genética , Humanos
15.
Invest Ophthalmol Vis Sci ; 55(6): 3939-53, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24876279

RESUMEN

PURPOSE: To provide the first detailed clinical description in patients with RP caused by recessive mutations in IMPG2. METHODS: This international collaborative study includes 17 RP patients with inherited retinal disease caused by mutations in IMPG2. The patients were clinically (re-)examined, including extensive medical history taking, slit-lamp biomicroscopy, ophthalmoscopy, perimetry, ERG, optical coherence tomography (OCT), fundus autofluorescence (FAF) imaging, fundus photography, and color vision tests. The main outcome measures included mean age at onset, initial symptom, best-corrected visual acuity, fundus appearance, perimetry results, ERG responses, OCT images, FAF imaging, color vision test reports and DNA sequence variants. RESULTS: The mean age at onset was 10.5 years (range, 4-20 years). Initial symptoms included night blindness in 59% of patients, a decreased visual acuity in 35%, and visual field loss in 6%. Fundus abnormalities were typical of RP: optic disc pallor, attenuated vessels, bone spicules, and generalized atrophy of the retina and choriocapillaris. Additionally, we observed macular abnormalities in all patients, ranging from subtle mottling of the macular pigment epithelium (two patients) and a bull's eye maculopathy (seven patients) to macular chorioretinal atrophy (seven patients). CONCLUSIONS: Mutations in IMPG2 cause a severe form of RP with symptoms manifesting in the first 2 decades of life. IMPG2-associated RP is frequently accompanied by macular involvement, ranging from mild pigment alterations to profound chorioretinal atrophy. The resulting decrease in central vision in combination with the severe tunnel vision leads to severe visual impairment in patients with IMPG2-associated RP.


Asunto(s)
Genes Recesivos , Mutación , Proteoglicanos/genética , Retinitis Pigmentosa/genética , Adulto , Edad de Inicio , Anciano , Pruebas de Percepción de Colores , Distrofias Hereditarias de la Córnea/diagnóstico , Distrofias Hereditarias de la Córnea/genética , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Ceguera Nocturna/diagnóstico , Ceguera Nocturna/genética , Oftalmoscopía , Linaje , Retinitis Pigmentosa/diagnóstico , Tomografía de Coherencia Óptica , Trastornos de la Visión/diagnóstico , Trastornos de la Visión/genética , Agudeza Visual/fisiología , Pruebas del Campo Visual , Campos Visuales/fisiología , Adulto Joven
16.
Invest Ophthalmol Vis Sci ; 52(5): 2227-39, 2011 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-21217109

RESUMEN

PURPOSE: To determine the genetic defects underlying autosomal recessive retinitis pigmentosa (arRP) in the Dutch population and in a subset of patients originating from other countries. The hypothesis was that, because there has been little migration over the past centuries in certain areas of The Netherlands, a significant fraction of Dutch arRP patients carry their genetic defect in the homozygous state. METHODS: High-resolution genome-wide SNP genotyping on SNP arrays and subsequent homozygosity mapping were performed in a large cohort of 186 mainly nonconsanguineous arRP families living in The Netherlands. Candidate genes residing in homozygous regions were sequenced. RESULTS: In ~94% of the affected individuals, large homozygous sequences were identified in their genome. In 42 probands, at least one of these homozygous regions contained one of the 26 known arRP genes. Sequence analysis of the corresponding genes in each of these patients revealed 21 mutations and two possible pathogenic changes, 14 of which were novel. All mutations were identified in only a single family, illustrating the genetic diversity within the Dutch population. CONCLUSIONS: This report demonstrates that homozygosity mapping is a powerful tool for identifying the genetic defect underlying genetically heterogeneous recessive disorders like RP, even in populations with little consanguinity.


Asunto(s)
Mapeo Cromosómico , Estudio de Asociación del Genoma Completo/métodos , Homocigoto , Mutación/genética , Polimorfismo de Nucleótido Simple , Retinitis Pigmentosa/genética , Consanguinidad , Análisis Mutacional de ADN , Genes Recesivos , Genotipo , Humanos , Análisis por Micromatrices , Países Bajos , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA