Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Am J Ophthalmol ; 263: 35-49, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38311152

RESUMEN

PURPOSE: The NIGHT study aimed to assess the natural history of choroideremia (CHM), an X-linked inherited chorioretinal degenerative disease leading to blindness, and determine which outcomes would be the most sensitive for monitoring disease progression. DESIGN: A prospective, observational, multicenter cohort study. METHODS: Males aged ≥18 years with genetically confirmed CHM, visible active disease within the macular region, and best-corrected visual acuity (BCVA) ≥34 Early Treatment Diabetic Retinopathy Study (ETDRS) letters at baseline were assessed for 20 months. The primary outcome was the change in BCVA over time at Months 4, 8, 12, 16, and 20. A range of functional and anatomical secondary outcome measures were assessed up to Month 12, including retinal sensitivity, central ellipsoid zone (EZ) area, and total area of fundus autofluorescence (FAF). Additional ocular assessments for safety were performed. RESULTS: A total of 220 participants completed the study. The mean BCVA was stable over 20 months. Most participants (81.4% in the worse eye and 77.8% in the better eye) had change from baseline > -5 ETDRS letters at Month 20. Interocular symmetry was low overall. Reductions from baseline to Month 12 were observed (worse eye, better eye) for retinal sensitivity (functional outcome; -0.68 dB, -0.48 dB), central EZ area (anatomical outcome; -0.276 mm2, -0.290 mm2), and total area of FAF (anatomical outcome; -0.605 mm2, -0.533 mm2). No assessment-related serious adverse events occurred. CONCLUSIONS: Retinal sensitivity, central EZ area, and total area of FAF are more sensitive than BCVA in measuring the natural progression of CHM.


Asunto(s)
Coroideremia , Progresión de la Enfermedad , Angiografía con Fluoresceína , Tomografía de Coherencia Óptica , Agudeza Visual , Humanos , Coroideremia/fisiopatología , Coroideremia/diagnóstico , Masculino , Estudios Prospectivos , Agudeza Visual/fisiología , Adulto , Persona de Mediana Edad , Angiografía con Fluoresceína/métodos , Anciano , Retina/fisiopatología , Adulto Joven , Estudios de Seguimiento , Adolescente
2.
Nat Med ; 29(10): 2464-2472, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37814062

RESUMEN

Choroideremia is a rare, X-linked retinal degeneration resulting in progressive vision loss. A randomized, masked, phase 3 clinical trial evaluated the safety and efficacy over 12 months of follow-up in adult males with choroideremia randomized to receive a high-dose (1.0 × 1011 vector genomes (vg); n = 69) or low-dose (1.0 × 1010 vg; n = 34) subretinal injection of the AAV2-vector-based gene therapy timrepigene emparvovec versus non-treated control (n = 66). Most treatment-emergent adverse events were mild or moderate. The trial did not meet its primary endpoint of best-corrected visual acuity (BCVA) improvement. In the primary endpoint analysis, three of 65 participants (5%) in the high-dose group, one of 34 (3%) participants in the low-dose group and zero of 62 (0%) participants in the control group had ≥15-letter Early Treatment Diabetic Retinopathy Study (ETDRS) improvement from baseline BCVA at 12 months (high dose, P = 0.245 versus control; low dose, P = 0.354 versus control). As the primary endpoint was not met, key secondary endpoints were not tested for significance. In a key secondary endpoint, nine of 65 (14%), six of 35 (18%) and one of 62 (2%) participants in the high-dose, low-dose and control groups, respectively, experienced ≥10-letter ETDRS improvement from baseline BCVA at 12 months. Potential opportunities to enhance future gene therapy studies for choroideremia include optimization of entry criteria (more preserved retinal area), surgical techniques and clinical endpoints. EudraCT registration: 2015-003958-41 .


Asunto(s)
Coroideremia , Retinopatía Diabética , Masculino , Humanos , Adulto , Coroideremia/genética , Coroideremia/terapia , Agudeza Visual , Terapia Genética/efectos adversos , Terapia Genética/métodos , Retina
3.
Acta Ophthalmol ; 101(2): 215-221, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36128853

RESUMEN

PURPOSE: To report clinical features and potential disease markers of inherited retinal dystrophy (IRD) caused by the biallelic c.148delG variant in the tubby-like protein 1 (TULP1) gene. METHODS: A retrospective observational study of 16 IRD patients carrying a homozygous pathogenic TULP1 c.148delG variant. Clinical data including fundus spectral-domain optical coherence tomography (SD-OCT) were assessed. A meta-analysis of visual acuity of previously reported other pathogenic TULP1 variants was performed for reference. RESULTS: The biallelic TULP1 variant c.148delG was associated with infantile and early childhood onset IRD. Retinal ophthalmoscopy was primarily normal converting to peripheral pigmentary retinopathy and maculopathy characterized by progressive extra-foveal loss of the ellipsoid zone (EZ), the outer plexiform layer (OPL), and the outer nuclear layer (ONL) bands in the SD-OCT images. The horizontal width of the foveal EZ showed significant regression with the best-corrected visual acuity (BCVA) of the eye (p < 0.0001, R2  = 0.541, F = 26.0), the age of the patient (p < 0.0001, R2  = 0.433, F = 16.8), and mild correlation with the foveal OPL-ONL thickness (p = 0.014, R2  = 0.245, F = 7.2). Modelling of the BCVA data suggested a mean annual loss of logMAR 0.027. The level of visual loss was similar to that previously reported in patients carrying other truncating TULP1 variants. CONCLUSIONS: This study describes the progression of TULP1 IRD suggesting a potential time window for therapeutic interventions. The width of the foveal EZ and the thickness of the foveal OPL-ONL layers could serve as biomarkers of the disease stage.


Asunto(s)
Distrofias Retinianas , Preescolar , Humanos , Biomarcadores , Proteínas del Ojo/genética , Fondo de Ojo , Estudios Observacionales como Asunto , Retina , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/genética , Retinitis Pigmentosa/genética , Tomografía de Coherencia Óptica/métodos
4.
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
5.
Ophthalmic Genet ; 42(6): 664-673, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34223797

RESUMEN

Atypical Usher syndrome (USH) is poorly defined with a broad clinical spectrum. Here, we characterize the clinical phenotype of disease caused by variants in CEP78, CEP250, ARSG, and ABHD12.Chart review evaluating demographic, clinical, imaging, and genetic findings of 19 patients from 18 families with a clinical diagnosis of retinal disease and confirmed disease-causing variants in CEP78, CEP250, ARSG, or ABHD12.CEP78-related disease included sensorineural hearing loss (SNHL) in 6/7 patients and demonstrated a broad phenotypic spectrum including: vascular attenuation, pallor of the optic disc, intraretinal pigment, retinal pigment epithelium mottling, areas of mid-peripheral hypo-autofluorescence, outer retinal atrophy, mild pigmentary changes in the macula, foveal hypo-autofluorescence, and granularity of the ellipsoid zone. Nonsense and frameshift variants in CEP250 showed mild retinal disease with progressive, non-congenital SNHL. ARSG variants resulted in a characteristic pericentral pattern of hypo-autofluorescence with one patient reporting non-congenital SNHL. ABHD12-related disease showed rod-cone dystrophy with macular involvement, early and severe decreased best corrected visual acuity, and non-congenital SNHL ranging from unreported to severe.This study serves to expand the clinical phenotypes of atypical USH. Given the variable findings, atypical USH should be considered in patients with peripheral and macular retinal disease even without the typical RP phenotype especially when SNHL is noted. Additionally, genetic screening may be useful in patients who have clinical symptoms and retinal findings even in the absence of known SNHL given the variability of atypical USH.


Asunto(s)
Arilsulfatasas/genética , Autoantígenos/genética , Proteínas de Ciclo Celular/genética , Codón sin Sentido/genética , Mutación del Sistema de Lectura/genética , Monoacilglicerol Lipasas/genética , Síndromes de Usher/genética , Adolescente , Adulto , Anciano , Distrofias de Conos y Bastones/diagnóstico , Distrofias de Conos y Bastones/genética , Femenino , Pruebas Genéticas , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Humanos , Masculino , Persona de Mediana Edad , Imagen Multimodal , Fenotipo , Epitelio Pigmentado de la Retina/patología , Estudios Retrospectivos , Tomografía de Coherencia Óptica , Síndromes de Usher/diagnóstico por imagen , Agudeza Visual/fisiología , Adulto Joven
6.
Clin Genet ; 99(1): 193-198, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32901921

RESUMEN

Conventional next-generation sequencing methods, used in most gene panels, cannot separate maternally and paternally derived sequence information of distant variants. In recessive diseases, two or more equally plausible causative variants with unsolved phase information prevent accurate molecular diagnosis. In reality, close relatives might be unavailable for segregation analysis. Here, we utilized whole genome linked-read sequencing to assign variants to haplotypes in two patients with inherited retinal dystrophies. Patient 1 with macular dystrophy had variants c.3442T>C, p.(Cys1148Arg), c.4209G>T, p.(Glu1403Asp), and c.1182C>T, p.(Cys394=) in CRB1, and Patient 2 with nonsyndromic retinitis pigmentosa had c.1328T>A, p.(Val443Asp) and c.3032C>G, p.(Ser1011*) in AHI1. The relatives were not available for genotyping. Using whole genome linked-read sequencing we phased the variants to haplotypes providing genetic background for the retinal dystrophies. In future, when the price of sequencing methods that provides long-read data decreases and their read-depth and accuracy increases, they are probably considered the primary or adjunctive sequencing methods in genetic testing, allowing the immediate collection of phase information and thus obviating the need for the carrier testing and segregation analysis.


Asunto(s)
Proteínas del Ojo/genética , Pruebas Genéticas , Haplotipos/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Distrofias Retinianas/genética , Adulto , Femenino , Genoma Humano/genética , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación/genética , Linaje , Distrofias Retinianas/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Secuenciación Completa del Genoma
7.
Br J Ophthalmol ; 103(7): 933-937, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30297337

RESUMEN

BACKGROUND/AIMS: To better understand the pattern of degeneration progression in cases with choroideremia. METHODS: A cohort of genotypically confirmed choroideremia cases who underwent optical coherence tomography (OCT) and fundus autofluorescence (FAF) imaging was studied. Using HEYEX review software, the foveal centre was marked on FAF images under guidance of corresponding OCT images, followed by application of an ETDRS grid. The boundaries of preserved autofluorescence (AF) were manually segmented in each individual ETDRS subfield. The regional distribution of preserved AF was assessed by comparing its area among the various subfields. RESULTS: A total of 168 eyes from 84 choroideremia cases were enrolled. There was a statistically significant difference in the amount of preserved AF area between inner subfields as determined by one-way analysis of variance (F (3,668)=9.997, p<0.001) and also between outer subfields (F (3,668)=8.348, p<0.001). A Tukey posthoc test revealed that the preserved AF area in the nasal subfields in both the inner and outer subfields was significantly smaller compared with analogue subfields. CONCLUSION: The asymmetric spatial distribution of preserved AF in choroideremia (corresponding to the stellate shaped nature of these regions) suggests that the progression of degeneration has directional preference.


Asunto(s)
Coroideremia/diagnóstico , Angiografía con Fluoresceína/métodos , Fóvea Central/patología , Imagen Multimodal , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica/métodos , Agudeza Visual , Fondo de Ojo , Humanos
8.
Acta Ophthalmol ; 96(2): 183-191, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29068140

RESUMEN

PURPOSE: To study the genetic aetiology of retinal dystrophies (RD) in Finnish patients. METHODS: A targeted next-generation sequencing (NGS) panel of 105 retinal dystrophy genes was used in a cohort of 55 RD patients. RESULTS: The overall diagnostic yield was 60% demonstrating the power of this approach. Interestingly, a missense mutation c.375C>G p.(Cys125Trp) in the CERKL gene was found in 18% of the patients in either a homozygous or compound heterozygous state. Data from Exome Aggregation Consortium (ExAC) Browser show that the CERKL c.375C>G p.(Cys125Trp) allele is enriched in the Finnish population and thus is a founder mutation. Furthermore, we report the clinical picture of 18 patients with mutations in the CERKL gene. CERKL mutations cause a macular-onset disease, in which symptoms first become apparent at the second decade. We also detected other novel founder mutations in the CERKL, EYS, RP1, ABCA4 and GUCY2D genes. CONCLUSION: Our report indicates that the first diagnostic test for Finnish patients with sporadic or autosomal recessive RD should be a targeted test for founder mutations in the CERKL, EYS, RP1, ABCA4 and GUCY2D genes. These results confirm the utility of NGS-based gene panels as a powerful method for mutation identification in RD, thus enabling improved genetic counselling for these families.


Asunto(s)
Efecto Fundador , Mutación Missense , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Distrofias Retinianas/genética , Transportadoras de Casetes de Unión a ATP/genética , Adolescente , Adulto , Anciano , Niño , Estudios de Cohortes , Análisis Mutacional de ADN , Electrorretinografía , Proteínas del Ojo/genética , Femenino , Finlandia , Guanilato Ciclasa/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Proteínas Asociadas a Microtúbulos , Persona de Mediana Edad , Linaje , Receptores de Superficie Celular/genética , Retina/fisiopatología , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/fisiopatología , Agudeza Visual/fisiología , Campos Visuales/fisiología
9.
Nat Commun ; 8(1): 1289, 2017 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-29097701

RESUMEN

Familial growth hormone deficiency provides an opportunity to identify new genetic causes of short stature. Here we combine linkage analysis with whole-genome resequencing in patients with growth hormone deficiency and maternally inherited gingival fibromatosis. We report that patients from three unrelated families harbor either of two missense mutations, c.347G>T p.(Arg116Leu) or c.1106C>T p.(Pro369Leu), in KCNQ1, a gene previously implicated in the long QT interval syndrome. Kcnq1 is expressed in hypothalamic GHRH neurons and pituitary somatotropes. Co-expressing KCNQ1 with the KCNE2 ß-subunit shows that both KCNQ1 mutants increase current levels in patch clamp analyses and are associated with reduced pituitary hormone secretion from AtT-20 cells. In conclusion, our results reveal a role for the KCNQ1 potassium channel in the regulation of human growth, and show that growth hormone deficiency associated with maternally inherited gingival fibromatosis is an allelic disorder with cardiac arrhythmia syndromes caused by KCNQ1 mutations.


Asunto(s)
Fibromatosis Gingival/genética , Hormona de Crecimiento Humana/deficiencia , Canal de Potasio KCNQ1/genética , Mutación Missense , Adolescente , Hormona Adrenocorticotrópica/metabolismo , Adulto , Alelos , Sustitución de Aminoácidos , Animales , Arritmias Cardíacas/genética , Niño , Preescolar , Femenino , Fibromatosis Gingival/metabolismo , Humanos , Canal de Potasio KCNQ1/química , Canal de Potasio KCNQ1/metabolismo , Masculino , Herencia Materna/genética , Ratones , Persona de Mediana Edad , Modelos Moleculares , Linaje , Mapas de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Adulto Joven
10.
Am J Ophthalmol ; 179: 110-117, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28499705

RESUMEN

PURPOSE: To identify valid and reproducible methods for quantifying anatomic outcome measures for eyes with choroideremia (CHM) in clinical trials. DESIGN: Reliability analysis study. METHODS: In this multicenter study, patients with confirmed genetic diagnosis of CHM were enrolled. All cases underwent spectral-domain optical coherence tomography (SDOCT) and fundus autofluorescence (FAF) imaging. Two graders independently delineated boundaries of preserved autofluorescence (PAF) and preserved ellipsoid zone (EZ) on FAF and OCT images, respectively. The results of the 2 independent gradings of both FAF and OCT images were compared to assess the reproducibility of the grading methods. RESULTS: A total of 148 eyes from 75 cases were included. In 21% of eyes PAF and in 43% of eyes preserved EZ had extended beyond the image capture area. After exclusion of these eyes and low-quality images, 114 FAF and 77 OCT images were graded. The mean PAF areas from 2 independent gradings were 3.720 ± 3.340 mm2 and 3.692 ± 3.253 mm2, respectively. Intraclass correlation coefficient (ICC) for these gradings was 0.996. The mean preserved EZ areas from 2 independent gradings were 2.746 ± 2.319 mm2 and 2.858 ± 2.446 mm2, respectively. ICC for these gradings was 0.991. CONCLUSIONS: Quantifying preserved retinal pigment epithelium and EZ areas on FAF and OCT images, respectively, in CHM patients is highly reproducible. These variables would be potential anatomic outcome measures for CHM clinical trials and could be studied and tracked longitudinally in choroideremia.


Asunto(s)
Coroideremia/diagnóstico , Angiografía con Fluoresceína/métodos , Oftalmoscopía/métodos , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica/métodos , Agudeza Visual , Anciano , Coroideremia/fisiopatología , Femenino , Fondo de Ojo , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
J Biol Chem ; 284(28): 18980-93, 2009 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-19423712

RESUMEN

Clarin-1 is the protein product encoded by the gene mutated in Usher syndrome III. Although the molecular function of clarin-1 is unknown, its primary structure predicts four transmembrane domains similar to a large family of membrane proteins that include tetraspanins. Here we investigated the role of clarin-1 by using heterologous expression and in vivo model systems. When expressed in HEK293 cells, clarin-1 localized to the plasma membrane and concentrated in low density compartments distinct from lipid rafts. Clarin-1 reorganized actin filament structures and induced lamellipodia. This actin-reorganizing function was absent in the modified protein encoded by the most prevalent North American Usher syndrome III mutation, the N48K form of clarin-1 deficient in N-linked glycosylation. Proteomics analyses revealed a number of clarin-1-interacting proteins involved in cell-cell adhesion, focal adhesions, cell migration, tight junctions, and regulation of the actin cytoskeleton. Consistent with the hypothesized role of clarin-1 in actin organization, F-actin-enriched stereocilia of auditory hair cells evidenced structural disorganization in Clrn1(-/-) mice. These observations suggest a possible role for clarin-1 in the regulation and homeostasis of actin filaments, and link clarin-1 to the interactive network of Usher syndrome gene products.


Asunto(s)
Actinas/química , Proteínas de la Membrana/fisiología , Síndromes de Usher/metabolismo , Animales , Adhesión Celular , Membrana Celular/metabolismo , Adhesiones Focales/metabolismo , Células Ciliadas Auditivas/metabolismo , Humanos , Microdominios de Membrana/química , Proteínas de la Membrana/química , Ratones , Ratones Transgénicos , Estructura Terciaria de Proteína , Tunicamicina/farmacología
19.
Invest Ophthalmol Vis Sci ; 48(6): 2498-502, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17525176

RESUMEN

PURPOSE: Aland Island eye disease (AIED), also known as Forsius-Eriksson syndrome, is an X-linked recessive retinal disease characterized by a combination of fundus hypopigmentation, decreased visual acuity, nystagmus, astigmatism, protan color vision defect, progressive myopia, and defective dark adaptation. Electroretinography reveals abnormalities in both photopic and scotopic functions. The gene locus for AIED has been mapped to the pericentromeric region of the X-chromosome, but the causative gene is unknown. The purpose of this study was to identify the mutated gene underlying the disease phenotype in the original AIED-affected family. METHODS: All exons of the CACNA1F gene were studied by DNA sequencing. CACNA1F mRNA from cultured lymphoblasts was analyzed by RT-PCR and cDNA sequencing. RESULTS: A novel deletion covering exon 30 and portions of flanking introns of the CACNA1F gene was identified in patients with AIED. Results from expression studies were consistent with the DNA studies and showed that mRNA lacked exon 30. The identified in-frame deletion mutation is predicted to cause a deletion of a transmembrane segment and an extracellular loop within repeat domain IV, and consequently an altered membrane topology of the encoded alpha1-subunit of the Ca(v)1.4 calcium channel. CONCLUSIONS: Mutations in CACNA1F are known to cause the incomplete form of X-linked congenital stationary night blindness (CSNB2). Since the clinical picture of AIED is quite similar to CSNB2, it has long been discussed whether these disorders are allelic or form a single entity. The present study clearly indicates that AIED is also caused by a novel CACNA1F gene mutation.


Asunto(s)
Canales de Calcio Tipo L/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Mutación , Ceguera Nocturna/genética , Eliminación de Secuencia , Análisis Mutacional de ADN , Exones/genética , Femenino , Humanos , Intrones/genética , Masculino , Linaje , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
J Med Genet ; 44(2): 153-60, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16963483

RESUMEN

BACKGROUND: Usher syndrome, a combination of retinitis pigmentosa (RP) and sensorineural hearing loss with or without vestibular dysfunction, displays a high degree of clinical and genetic heterogeneity. Three clinical subtypes can be distinguished, based on the age of onset and severity of the hearing impairment, and the presence or absence of vestibular abnormalities. Thus far, eight genes have been implicated in the syndrome, together comprising 347 protein-coding exons. METHODS: To improve DNA diagnostics for patients with Usher syndrome, we developed a genotyping microarray based on the arrayed primer extension (APEX) method. Allele-specific oligonucleotides corresponding to all 298 Usher syndrome-associated sequence variants known to date, 76 of which are novel, were arrayed. RESULTS: Approximately half of these variants were validated using original patient DNAs, which yielded an accuracy of >98%. The efficiency of the Usher genotyping microarray was tested using DNAs from 370 unrelated European and American patients with Usher syndrome. Sequence variants were identified in 64/140 (46%) patients with Usher syndrome type I, 45/189 (24%) patients with Usher syndrome type II, 6/21 (29%) patients with Usher syndrome type III and 6/20 (30%) patients with atypical Usher syndrome. The chip also identified two novel sequence variants, c.400C>T (p.R134X) in PCDH15 and c.1606T>C (p.C536S) in USH2A. CONCLUSION: The Usher genotyping microarray is a versatile and affordable screening tool for Usher syndrome. Its efficiency will improve with the addition of novel sequence variants with minimal extra costs, making it a very useful first-pass screening tool.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos , Síndromes de Usher/genética , ADN/genética , Cartilla de ADN , Europa (Continente) , Variación Genética , Genotipo , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...