Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transl Vis Sci Technol ; 11(1): 6, 2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34985506

RESUMEN

Purpose: Comprehensive genetic testing for inherited retinal dystrophy (IRD) is challenged by difficult-to-sequence genomic regions, which are often mutational hotspots, such as RPGR ORF15. The purpose of this study was to evaluate the diagnostic contribution of RPGR variants in an unselected IRD patient cohort referred for testing in a clinical diagnostic laboratory. Methods: A total of 5201 consecutive patients were analyzed with a clinically validated next-generation sequencing (NGS)-based assay, including the difficult-to-sequence RPGR ORF15 region. Copy number variant (CNV) detection from NGS data was included. Variant interpretation was performed per the American College of Medical Genetics and Genomics guidelines. Results: A confirmed molecular diagnosis in RPGR was found in 4.5% of patients, 24.0% of whom were females. Variants in ORF15 accounted for 74% of the diagnoses; 29% of the diagnostic variants were in the most difficult-to-sequence central region of ORF15 (c.2470-3230). Truncating variants made up the majority (91%) of the diagnostic variants. CNVs explained 2% of the diagnostic cases, of which 80% were one- or two-exon deletions outside of ORF15. Conclusions: Our findings indicate that high-throughput, clinically validated NGS-based testing covering the difficult-to-sequence region of ORF15, in combination with high-resolution CNV detection, can help to maximize the diagnostic yield for patients with IRD. Translational Relevance: These results demonstrate an accurate and scalable method for the detection of RPGR-related variants, including the difficult-to-sequence ORF15 hotspot, which is relevant given current and emerging therapeutic opportunities.


Asunto(s)
Proteínas del Ojo , Distrofias Retinianas , Exones , Proteínas del Ojo/genética , Femenino , Humanos , Linaje , Prevalencia , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/epidemiología , Distrofias Retinianas/genética
2.
Gene Expr Patterns ; 13(8): 473-81, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24045267

RESUMEN

Clarin-1 (CLRN1) is the causative gene in Usher syndrome type 3A, an autosomal recessive disorder characterized by progressive vision and hearing loss. CLRN1 encodes Clarin-1, a glycoprotein with homology to the tetraspanin family of proteins. Previous cell culture studies suggest that Clarin-1 localizes to the plasma membrane and interacts with the cytoskeleton. Mouse models demonstrate a role for the protein in mechanosensory hair bundle integrity, but the function of Clarin-1 in hearing remains unclear. Even less is known of its role in vision, because the Clrn1 knockout mouse does not exhibit a retinal phenotype and expression studies in murine retinas have provided conflicting results. Here, we describe cloning and expression analysis of the zebrafish clrn1 gene, and report protein localization of Clarin-1 in auditory and visual cells from embryonic through adult stages. We detect clrn1 transcripts as early as 24h post-fertilization, and expression is maintained through adulthood. In situ hybridization experiments show clrn1 transcripts enriched in mechanosensory hair cells and supporting cells of the inner ear and lateral line organ, photoreceptors, and cells of the inner retina. In mechanosensory hair cells, Clarin-1 is polarized to the apical cell body and the synapses. In the retina, Clarin-1 localizes to lateral cell contacts between photoreceptors and is associated with the outer limiting membrane and subapical processes emanating from Müller glial cells. We also find Clarin-1 protein in the outer plexiform, inner nuclear and ganglion cell layers of the retina. Given the importance of Clarin-1 function in the human retina, it is imperative to find an animal model with a comparable requirement. Our data provide a foundation for exploring the role of Clarin-1 in retinal cell function and survival in a diurnal, cone-dominant species.


Asunto(s)
Oído Interno/metabolismo , Proteínas de la Membrana/genética , Retina/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Animales , Línea Celular , Clonación Molecular , Cricetinae , Expresión Génica , Humanos , Mecanorreceptores/metabolismo , Proteínas de la Membrana/metabolismo , Especificidad de Órganos , Transporte de Proteínas , Retina/citología , Células Ganglionares de la Retina/metabolismo , Segmento Interno de las Células Fotorreceptoras Retinianas/metabolismo , Homología de Secuencia de Aminoácido , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
3.
JAMA Ophthalmol ; 131(1): 67-74, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22964989

RESUMEN

OBJECTIVE: To study macular structure and function in patients with Usher syndrome type III (USH3) caused by mutations in the Clarin 1 gene (CLRN1). METHODS: High-resolution macular images were obtained by adaptive optics scanning laser ophthalmoscopy and spectral domain optical coherence tomography in 3 patients with USH3 and were compared with those of age-similar control subjects. Vision function measures included best-corrected visual acuity, kinetic and static perimetry, and full-field electroretinography. Coding regions of the CLRN1 gene were sequenced. RESULTS: CLRN1 mutations were present in all the patients; a 20-year-old man showed compound heterozygous mutations (p.N48K and p.S188X), and 2 unrelated women aged 25 and 32 years had homozygous mutations (p.N48K). Best-corrected visual acuity ranged from 20/16 to 20/40, with scotomas beginning at 3° eccentricity. The inner segment-outer segment junction or the inner segment ellipsoid band was disrupted within 1° to 4° of the fovea, and the foveal inner and outer segment layers were significantly thinner than normal. Cones near the fovea in patients 1 and 2 showed normal spacing, and the preserved region ended abruptly. Retinal pigment epithelial cells were visible in patient 3 where cones were lost. CONCLUSIONS: Cones were observed centrally but not in regions with scotomas, and retinal pigment epithelial cells were visible in regions without cones in patients with CLRN1 mutations. High-resolution measures of retinal structure demonstrate patterns of cone loss associated with CLRN1 mutations. CLINICAL RELEVANCE: These findings provide insight into the effect of CLRN1 mutations on macular cone structure, which has implications for the development of treatments for USH3. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT00254605.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Células Fotorreceptoras Retinianas Conos/patología , Enfermedades de la Retina/diagnóstico , Síndromes de Usher/diagnóstico , Adulto , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Oftalmoscopía , Enfermedades de la Retina/genética , Epitelio Pigmentado de la Retina/patología , Escotoma/patología , Tomografía de Coherencia Óptica , Síndromes de Usher/genética , Agudeza Visual/fisiología , Pruebas del Campo Visual , Campos Visuales/fisiología , Adulto Joven
4.
Acta Ophthalmol ; 91(4): 325-34, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22681893

RESUMEN

PURPOSE: The Finnish distribution of clinical Usher syndrome (USH) types is 40% USH3, 34% USH1 and 12% USH2. All patients with USH3 carry the founder mutation in clarin 1 (CLRN1), whereas we recently reported three novel myosin VIIA (MYO7A) mutations in two unrelated patients with USH1. This study was carried out to further investigate the USH mutation spectrum in Finnish patients. METHODS: We analysed samples from nine unrelated USH patients/families without known mutations and two USH3 families with atypically severe phenotype. The Asper Ophthalmics USH mutation chip was used to screen for known mutations and to evaluate the chip in molecular diagnostics of Finnish patients. RESULTS: The chip revealed a heterozygous usherin (USH2A) mutation, p.N346H, in one patient. Sequencing of MYO7A and/or USH2A in three index patients revealed two novel heterozygous mutations, p.R873W in MYO7A and c.14343+2T>C in USH2A. We did not identify definite pathogenic second mutations in the patients, but identified several probably nonpathogenic variations that may modify the disease phenotype. Possible digenism could not be excluded in two families segregating genomic variations in both MYO7A and USH2A, and two families with CLRN1 and USH2A. CONCLUSION: We conclude that there is considerable genetic heterogeneity of USH1 and USH2 in Finland, making molecular diagnostics and genetic counselling of patients and families challenging.


Asunto(s)
ADN/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Mutación , Miosinas/genética , Síndromes de Usher/genética , Adulto , Análisis Mutacional de ADN , Femenino , Finlandia/epidemiología , Genotipo , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Persona de Mediana Edad , Miosina VIIa , Miosinas/metabolismo , Linaje , Prevalencia , Síndromes de Usher/epidemiología , Síndromes de Usher/metabolismo
5.
Otol Neurotol ; 33(1): 38-41, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22143301

RESUMEN

BACKGROUND: Usher syndrome Type 3 (USH3) is an autosomal recessive disorder characterized by variable type and degree of progressive sensorineural hearing loss and retinitis pigmentosa. Cochlear implants are widely used among these patients. OBJECTIVES: To evaluate the results and benefits of cochlear implantation in patients with USH3. STUDY DESIGN: A nationwide multicenter retrospective review. MATERIALS AND METHODS: During the years 1995-2005, in 5 Finnish university hospitals, 19 patients with USH3 received a cochlear implant. Saliva samples were collected to verify the USH3 genotype. Patients answered to 3 questionnaires: Glasgow Benefit Inventory, Glasgow Health Status Inventory, and a self-made questionnaire. Audiological data were collected from patient records. RESULTS: All the patients with USH3 in the study were homozygous for the Finnish major mutation (p.Y176X). Either they had severe sensorineural hearing loss or they were profoundly deaf. The mean preoperative hearing level (pure-tone average, 0.5-4 kHz) was 110 ± 8 dB hearing loss (HL) and the mean aided hearing level was 58 ± 11 dB HL. The postoperative hearing level (34 ± 9 dB HL) and word recognition scores were significantly better than before surgery. According to the Glasgow Benefit Inventory scores and Glasgow Health Status Inventory data related to hearing, the cochlear implantation was beneficial to patients with USH3. CONCLUSION: Cochlear implantation is beneficial to patients with USH3, and patients learn to use the implant without assistance.


Asunto(s)
Implantación Coclear , Trastornos de la Comunicación/terapia , Percepción del Habla/fisiología , Síndromes de Usher/terapia , Adolescente , Adulto , Anciano , Audiometría de Tonos Puros , Niño , Implantación Coclear/efectos adversos , Implantes Cocleares/efectos adversos , Trastornos de la Comunicación/rehabilitación , Análisis Mutacional de ADN , Femenino , Finlandia , Audición/fisiología , Audífonos , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Calidad de Vida , Estudios Retrospectivos , Saliva/química , Encuestas y Cuestionarios , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Síndromes de Usher/genética , Síndromes de Usher/rehabilitación , Visión Ocular/fisiología , Agudeza Visual/fisiología , Adulto Joven
6.
Eur J Hum Genet ; 19(1): 30-5, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20717163

RESUMEN

Clarin 1 (CLRN1) is a four-transmembrane protein expressed in cochlear hair cells and neural retina, and when mutated it causes Usher syndrome type 3 (USH3). The main human splice variant of CLRN1 is composed of three exons that code for a 232-aa protein. In this study, we aimed to refine the structure of CLRN1 by an examination of transcript splice variants and promoter regions. Analysis of human retinal cDNA revealed 11 CLRN1 splice variants, of which 5 have not been previously reported. We studied the regulation of gene expression by several promoter domains using a luciferase assay, and identified 1000 nt upstream of the translation start site of the primary CLRN1 splice variant as the principal promoter region. Our results suggest that the CLRN1 gene is significantly more complex than previously described. The complexity of the CLRN1 gene and the identification of multiple splice variants may partially explain why mutations in CLRN1 result in substantial variation in clinical phenotype.


Asunto(s)
Empalme Alternativo/genética , Proteínas de la Membrana/genética , Síndromes de Usher/fisiopatología , Cóclea/metabolismo , ADN Complementario/genética , ADN Complementario/metabolismo , Regulación de la Expresión Génica , Variación Genética , Humanos , Fenotipo , Regiones Promotoras Genéticas , Retina/citología , Retina/metabolismo , Síndromes de Usher/genética
7.
Mol Vis ; 15: 1806-18, 2009 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-19753315

RESUMEN

PURPOSE: Mutations of clarin 1 (CLRN1) cause Usher syndrome type 3 (USH3). To determine the effects of USH3 mutations on CLRN1 function, we examined the cellular distribution and stability of both normal and mutant CLRN1 in vitro. We also searched for novel disease-causing mutations in a cohort of 59 unrelated Canadian and Finnish USH patients. METHODS: Mutation screening was performed by DNA sequencing. For the functional studies, wild-type (WT) and mutant CLRN1 genes were expressed as hemagglutinin (HA) tagged fusion proteins by transient transfection of BHK-21 cells. Subcellular localization of CLRN1-HA was examined by confocal microscopy. The N-glycosylation status of CLRN1 was studied by using the N-glycosidase F (PNGase F) enzyme and western blotting. Cycloheximide treatment was used to assess the stability of CLRN1 protein. RESULTS: We found three previously reported pathogenic mutations, p.A123D, p.N48K, and p.Y176X, and a novel sequence variant, p.L54P, from the studied USH patients. The WT HA-tagged CLRN1 was correctly trafficked to the plasma membrane, whereas mutant CLRN1-HA proteins were mislocalized and retained in the endoplasmic reticulum. PNGase F treatment of CLRN1-HA resulted in an electrophoretic mobility shift consistent with sugar residue cleavage in WT and in all CLRN1 mutants except in p.N48K mutated CLRN1, in which the mutation abolishes the glycosylation site. Inhibition of protein expression with cycloheximide indicated that WT CLRN1-HA remained stable. In contrast, the CLRN1 mutants showed reduced stability. CONCLUSIONS: WT CLRN1 is a glycoprotein localized to the plasma membrane in transfected BHK-21 cells. Mutant CLRN1 proteins are mislocalized. We suggest that part of the pathogenesis of USH3 may be associated with defective intracellular trafficking as well as decreased stability of mutant CLRN1 proteins.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mutación/genética , Síndromes de Usher/genética , Secuencia de Aminoácidos , Western Blotting , Estudios de Casos y Controles , Secuencia Conservada , Análisis Mutacional de ADN , Humanos , Proteínas de la Membrana/química , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Péptidos/química , Péptidos/metabolismo , Multimerización de Proteína , Estabilidad Proteica , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Tomografía de Coherencia Óptica , Transfección , Síndromes de Usher/patología
8.
PLoS Genet ; 5(8): e1000607, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19680541

RESUMEN

Mutations in the CLRN1 gene cause Usher syndrome type 3 (USH3), a human disease characterized by progressive blindness and deafness. Clarin 1, the protein product of CLRN1, is a four-transmembrane protein predicted to be associated with ribbon synapses of photoreceptors and cochlear hair cells, and recently demonstrated to be associated with the cytoskeleton. To study Clrn1, we created a Clrn1 knockout (KO) mouse and characterized the histological and functional consequences of Clrn1 deletion in the retina and cochlea. Clrn1 KO mice do not develop a retinal degeneration phenotype, but exhibit progressive loss of sensory hair cells in the cochlea and deterioration of the organ of Corti by 4 months. Hair cell stereocilia in KO animals were longer and disorganized by 4 months, and some Clrn1 KO mice exhibited circling behavior by 5-6 months of age. Clrn1 mRNA expression was localized in the retina using in situ hybridization (ISH), laser capture microdissection (LCM), and RT-PCR. Retinal Clrn1 transcripts were found throughout development and adulthood by RT-PCR, although expression peaked at P7 and declined to undetectable levels in adult retina by ISH. LCM localized Clrn1 transcripts to the retinas inner nuclear layer, and WT levels of retinal Clrn1 expression were observed in photoreceptor-less retinas. Examination of Clrn1 KO mice suggests that CLRN1 is unnecessary in the murine retina but essential for normal cochlear development and function. This may reflect a redundancy in the mouse retina not present in human retina. In contrast to mouse KO models of USH1 and USH2, our data indicate that Clrn1 expression in the retina is restricted to the Müller glia. This is a novel finding, as most retinal degeneration associated proteins are expressed in photoreceptors, not in glia. If CLRN1 expression in humans is comparable to the expression pattern observed in mice, this is the first report of an inner retinal protein that, when mutated, causes retinal degeneration.


Asunto(s)
Cóclea/crecimiento & desarrollo , Células Ciliadas Auditivas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Retina/metabolismo , Animales , Cóclea/citología , Cóclea/metabolismo , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas , Retina/crecimiento & desarrollo
9.
Mol Vis ; 12: 1093-7, 2006 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-17093394

RESUMEN

PURPOSE: Usher syndrome (USH) is an autosomal recessive disorder resulting in retinal degeneration and sensorineural deafness caused by mutations in at least 10 gene loci. USH is divided into three main clinical types: USH1 (33-44%), USH2 (56-67%), and USH3. Worldwide, USH1 and USH2 account for most of the Usher syndrome cases with rare occurrence of USH3. In Finland, however, USH3 is the most common type (40%), explained by genetic and geographical isolation accompanied with a founder mutation, while USH1 is estimated to comprise 34% and USH2 12% of all USH cases. METHODS: We examined two unrelated Finnish USH1 patients by sequencing. RESULTS: We found three new myosin VIIA (MYO7A) mutations: p.K923AfsX8, p.Q1896X, and p.E1349K. The p.K923AfsX8 mutation was present in both patients as well as in one of 200 Finnish control chromosomes. CONCLUSIONS: This is the first molecular genetic study of USH1 in Finland. We have found three new pathological mutations causing either premature termination of translation or replacement of an evolutionary conserved MYO7A amino acid.


Asunto(s)
Dineínas/genética , Mutación , Miosinas/genética , Síndromes de Usher/genética , Sustitución de Aminoácidos , Codón sin Sentido , Femenino , Finlandia , Mutación del Sistema de Lectura , Eliminación de Gen , Ácido Glutámico , Glutamina , Heterocigoto , Humanos , Lisina , Masculino , Miosina VIIa , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...