Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Genet Med ; 26(6): 101106, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38420906

RESUMEN

PURPOSE: Inherited retinal diseases (IRDs) are a group of monogenic conditions that can lead to progressive blindness. Their missing heritability is still considerable, due in part to the presence of disease genes that await molecular identification. The purpose of this work was to identify novel genetic associations with IRDs. METHODS: Patients underwent a comprehensive ophthalmological evaluation using standard-of-care tests, such as detailed retinal imaging (macular optical coherence tomography and short-wavelength fundus autofluorescence) and electrophysiological testing. Exome and genome sequencing, as well as computer-assisted data analysis were used for genotyping and detection of DNA variants. A minigene-driven splicing assay was performed to validate the deleterious effects of 1 of such variants. RESULTS: We identified 8 unrelated families from Hungary, the United States, Israel, and The Netherlands with members presenting with a form of autosomal recessive and nonsyndromic retinal degeneration, predominantly described as rod-cone dystrophy but also including cases of cone/cone-rod dystrophy. Age of disease onset was very variable, with some patients experiencing first symptoms during their fourth decade of life or later. Myopia greater than 5 diopters was present in 5 of 7 cases with available refractive data, and retinal detachment was reported in 2 cases. All ascertained patients carried biallelic loss-of-function variants in UBAP1L (HGNC: 40028), a gene with unknown function and with homologies to UBAP1, encoding a protein involved in ubiquitin metabolism. One of these pathogenic variants, the intronic NM_001163692.2:c.910-7G>A substitution, was identified in 5 unrelated families. Minigene-driven splicing assays in HEK293T cells confirmed that this DNA change is responsible for the creation of a new acceptor splice site, resulting in aberrant splicing. CONCLUSION: We identified UBAP1L as a novel IRD gene. Although its function is currently unknown, UBAP1L is almost exclusively expressed in photoreceptors and the retinal pigment epithelium, hence possibly explaining the link between pathogenic variants in this gene and an ocular phenotype.


Asunto(s)
Linaje , Degeneración Retiniana , Humanos , Masculino , Femenino , Adulto , Degeneración Retiniana/genética , Persona de Mediana Edad , Mutación con Pérdida de Función , Genes Recesivos , Niño , Adolescente , Distrofias de Conos y Bastones/genética , Hungría , Adulto Joven , Predisposición Genética a la Enfermedad
2.
Artículo en Inglés | MEDLINE | ID: mdl-38565268

RESUMEN

Retinal disorders caused by genetic or environmental factors cause severe visual impairment and often result in blindness. The past ten years have seen rapid progress in our understanding of the biological basis of these conditions, as well as significant advances towards gene and cell-based therapies. Regulatory challenges remain, but there is reason to hope that creative approaches will lead to safe and effective breakthrough treatments for these conditions in the near future.

3.
Genes (Basel) ; 15(5)2024 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-38790275

RESUMEN

Inherited retinal diseases (IRDs) are a clinically and genetically heterogeneous group of diseases which cause visual loss due to Mendelian mutations in over 250 genes, making genetic diagnosis challenging and time-consuming. Here, we developed a new tool, CDIP (Cost-effective Deep-sequencing IRD Panel) in which a simultaneous sequencing of common mutations is performed. CDIP is based on simultaneous amplification of 47 amplicons harboring common mutations followed by next-generation sequencing (NGS). Following five rounds of calibration of NGS-based steps, CDIP was used in 740 IRD samples. The analysis revealed 151 mutations in 131 index cases. In 54 (7%) of these cases, CDIP identified the genetic cause of disease (the remaining were single-heterozygous recessive mutations). These include a patient that was clinically diagnosed with retinoschisis and found to be homozygous for NR2E3-c.932G>A (p.R311Q), and a patient with RP who is hemizygous for an RPGR variant, c.292C>A (p.H98N), which was not included in the analysis but is located in proximity to one of these mutations. CDIP is a cost-effective deep sequencing panel for simultaneous detection of common founder mutations. This protocol can be implemented for additional populations as well as additional inherited diseases, and mainly in populations with strong founder effects.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Mutación , Humanos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Enfermedades de la Retina/genética , Enfermedades de la Retina/diagnóstico , Efecto Fundador , Masculino , Femenino , Pruebas Genéticas/métodos , Pruebas Genéticas/economía , Análisis Costo-Beneficio , Linaje
4.
Genes (Basel) ; 15(7)2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39062705

RESUMEN

Inherited retinal diseases (IRDs) are extremely heterogeneous with at least 350 causative genes, complicating the process of genetic diagnosis. We analyzed samples of 252 index cases with IRDs using the Blueprint Genetics panel for "Retinal Dystrophy" that includes 351 genes. The cause of disease could be identified in 55% of cases. A clear difference was obtained between newly recruited cases (74% solved) and cases that were previously analyzed by panels or whole exome sequencing (26% solved). As for the mode of inheritance, 75% of solved cases were autosomal recessive (AR), 10% were X-linked, 8% were autosomal dominant, and 7% were mitochondrial. Interestingly, in 12% of solved cases, structural variants (SVs) were identified as the cause of disease. The most commonly identified genes were ABCA4, EYS and USH2A, and the most common mutations were MAK-c.1297_1298ins353 and FAM161A-c.1355_1356del. In line with our previous IRD carrier analysis, we identified heterozygous AR mutations that were not the cause of disease in 36% of cases. The studied IRD panel was found to be efficient in gene identification. Some variants were misinterpreted by the pipeline, and therefore, multiple analysis tools are recommended to obtain a more accurate annotation of potential disease-causing variants.


Asunto(s)
Mutación , Enfermedades de la Retina , Humanos , Masculino , Femenino , Enfermedades de la Retina/genética , Transportadoras de Casetes de Unión a ATP/genética , Proteínas del Ojo/genética , Pruebas Genéticas/métodos , Secuenciación del Exoma/métodos , Proteínas de la Matriz Extracelular/genética , Linaje , Distrofias Retinianas/genética , Predisposición Genética a la Enfermedad
5.
J Clin Med ; 13(7)2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38610827

RESUMEN

Background: Visual acuity (VA) assessments are crucial in ophthalmology but traditionally rely on in-clinic evaluations. The emergence of telemedicine has spurred interest in creating dependable self-administered VA tests for use beyond standard clinical environments. This study evaluated the practicality and validity of a self-administered near VA card test against traditional Snellen and Rosenbaum Pocket Vision Screener (RPVS) methods for home monitoring and enhancing clinical workflow. Methods: In a cross-sectional study, a near VA card (Hadassah Self-Visual Acuity Screener (HSVA)) was developed with written and videotaped instructions for self-use. Patients with a minimal best-corrected VA (BCVA) of 1.0 LogMAR in at least one eye were recruited from ophthalmology and optometry clinics. Outcomes included the mean BCVA difference between the self-administered values and those obtained by the examiner, and correlations between BCVA values obtained by the Snellen, RPVS, HSVA, and previous distance BCVA methods according to the patients' electronic medical records. Results: A total of 275 participants (mean age: 42.5 ± 19.4 years; range: 18-89 years; 47% female) were included. Test-retest reliability analysis of the HSVA demonstrated a very good correlation and repeatability (n = 38 patients; Rs = 1.0; p < 0.001). Accuracy analysis revealed the mean LogMAR BCVA values of an additional 237 patients obtained by the Snellen, RPVS, and HSVA methods were similar (p = 0.10). The self-test BCVA results obtained by the HSVA agreed with the masked examiner-tested VA results (n = 67 patients; p = 0.17; Rs = 0.87; ICC = 0.96). Similar results were obtained when stratification by median age (42 years) was performed. Bland-Altman analysis of the HSVA and RPVS methods demonstrated a good agreement. To assess whether the HSVA could predict the VA results in the clinically used charts, multivariate analysis was used and revealed that the HSVA predicted the RPVS results (ß = 0.91; p = 0.001; R2 = 0.88), and the self-test HSVA predicted the Snellen VA results within two lines (ß = 0.93; p = 0.01; R2 = 0.36). Conclusions: The home-based HSVA assessment exhibited high test-retest reliability, accuracy, and alignment with clinical-standard VA tests. Its efficacy in self-testing mirrored examiner-conducted VA assessments and accurately predicted Snellen VA outcomes, indicating the HSVA's suitability for self-monitoring in chronic ocular conditions or when access to conventional examinations is limited. The utility of self-administered VA tests may extend beyond ophthalmology and optometry, potentially benefiting primary care, emergency medicine, and neurology. Further research is needed to explore and validate the practical applications of remote VA testing.

6.
Mol Ther Nucleic Acids ; 35(1): 102130, 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38375504

RESUMEN

Adenosine deaminases acting on RNA (ADARs) are endogenous enzymes catalyzing the deamination of adenosines to inosines, which are then read as guanosines during translation. This ability to recode makes ADAR an attractive therapeutic tool to edit genetic mutations and reprogram genetic information at the mRNA level. Using the endogenous ADARs and guiding them to a selected target has promising therapeutic potential. Indeed, different studies have reported several site-directed RNA-editing approaches for making targeted base changes in RNA molecules. The basic strategy has been to use guide RNAs (gRNAs) that hybridize and form a double-stranded RNA (dsRNA) structure with the desired RNA target because of ADAR activity in regions of dsRNA formation. Here we report on a novel pipeline for identifying disease-causing variants as candidates for RNA editing, using a yeast-based screening system to select efficient gRNAs for editing of nonsense mutations, and test them in a human cell line reporter system. We have used this pipeline to modify the sequence of transcripts carrying nonsense mutations that cause inherited retinal diseases in the FAM161A, KIZ, TRPM1, and USH2A genes. Our approach can serve as a basis for gene therapy intervention in knockin mouse models and ultimately in human patients.

7.
EMBO Mol Med ; 16(4): 805-822, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38504136

RESUMEN

For 15 years, gene therapy has been viewed as a beacon of hope for inherited retinal diseases. Many preclinical investigations have centered around vectors with maximal gene expression capabilities, yet despite efficient gene transfer, minimal physiological improvements have been observed in various ciliopathies. Retinitis pigmentosa-type 28 (RP28) is the consequence of bi-allelic null mutations in the FAM161A, an essential protein for the structure of the photoreceptor connecting cilium (CC). In its absence, cilia become disorganized, leading to outer segment collapses and vision impairment. Within the human retina, FAM161A has two isoforms: the long one with exon 4, and the short one without it. To restore CC in Fam161a-deficient mice shortly after the onset of cilium disorganization, we compared AAV vectors with varying promoter activities, doses, and human isoforms. While all vectors improved cell survival, only the combination of both isoforms using the weak FCBR1-F0.4 promoter enabled precise FAM161A expression in the CC and enhanced retinal function. Our investigation into FAM161A gene replacement for RP28 emphasizes the importance of precise therapeutic gene regulation, appropriate vector dosing, and delivery of both isoforms. This precision is pivotal for secure gene therapy involving structural proteins like FAM161A.


Asunto(s)
Retinitis Pigmentosa , Animales , Ratones , Humanos , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/terapia , Retinitis Pigmentosa/metabolismo , Retina/metabolismo , Exones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Terapia Genética , Proteínas del Ojo/genética , Proteínas del Ojo/química , Proteínas del Ojo/metabolismo
8.
Genes (Basel) ; 15(6)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38927740

RESUMEN

Retinitis pigmentosa (RP) is a heterogeneous inherited retinal disorder. Mutations in KIZ cause autosomal recessive (AR) RP. We aimed to characterize the genotype, expression pattern, and phenotype in a large cohort of KIZ cases. Sanger and whole exome sequencing were used to identify the KIZ variants. Medical records were reviewed and analyzed. Thirty-one patients with biallelic KIZ mutations were identified: 28 homozygous for c.226C>T (p.R76*), 2 compound heterozygous for p.R76* and c.3G>A (p.M1?), and one homozygous for c.247C>T (p.R83*). c.226C>T is a founder mutation among patients of Jewish descent. The clinical parameters were less severe in KIZ compared to DHDDS and FAM161A cases. RT-PCR analysis in fibroblast cells revealed the presence of four different transcripts in both WT and mutant samples with a lower percentage of the WT transcript in patients. Sequence analysis identified an exonic sequence enhancer (ESE) that includes the c.226 position which is affected by the mutation. KIZ mutations are an uncommon cause of IRD worldwide but are not rare among Ashkenazi Jews. Our data indicate that p.R76* affect an ESE which in turn results in the pronounced skipping of exon 3. Therefore, RNA-based therapies might show low efficacy since the mutant transcripts are spliced.


Asunto(s)
Mutación , Retinitis Pigmentosa , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Secuenciación del Exoma/métodos , Proteínas del Ojo/genética , Judíos/genética , Linaje , Fenotipo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
9.
Invest Ophthalmol Vis Sci ; 65(2): 39, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38411968

RESUMEN

Purpose: To review all reported disease-causing mutations in BEST1, perform genotype-phenotype correlation, and estimate disease prevalence in the Israeli population. Methods: Medical records of patients diagnosed with Best disease and allied diseases from nine Israeli medical centers over the past 20 years were collected, as were clinical data including ocular findings, electrophysiology results, and retina imaging. Mutation detection involved mainly whole exome sequencing and candidate gene analysis. Demographic data were obtained from the Israeli Bureau of Statistics (January 2023). A bibliometric study was also conducted to gather mutation data from online sources. Results: A total of 134 patients were clinically diagnosed with Best disease and related conditions. The estimated prevalence of Best disease was calculated to be 1 in 127,000, with higher rates among Arab Muslims (1 in 76,000) than Jews (1 in 145,000). Genetic causes were identified in 76 individuals (57%), primarily showing autosomal-dominant inheritance due to BEST1 mutations (58 patients). Critical conserved domains were identified consisting of a high percentage of dominant missense mutations, primarily in transmembrane domains and the intracellular region (Ca2+ binding domain) of the BEST1 protein. Conclusions: This study represents the largest cohort of patients with Best disease reported in Israel and globally. The prevalence in Israel is akin to that in Denmark but is lower than that in the United States. Critical conserved domains within the BEST1 protein are pivotal for normal functioning, and even minor missense alterations in these areas lead to a dominant disease manifestation. Genetic testing is indispensable as the gold standard for Best disease diagnosis due to the variable clinical presentation of the disease.


Asunto(s)
Distrofia Macular Viteliforme , Humanos , Israel/epidemiología , Prevalencia , Mutación , Estudios de Asociación Genética , Bestrofinas
10.
JAMA Ophthalmol ; 142(7): 609-616, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38753338

RESUMEN

Importance: Data regarding the prevalence of various inherited retinal diseases (IRDs) are limited and vary across populations; moreover, nationwide prevalence studies may be limited to a specific IRD phenotype, potentially leading to inaccurate prevalence estimations. Therefore, nationwide prevalence data are needed. Objective: To determine the prevalence of 67 IRD phenotypes in the Israeli population. Design, Setting, and Participants: This cohort study collected nationwide data regarding the number of individuals affected with IRD phenotypes assessed in 10 clinical and academic centers in Israel as part of the research activity of the Israeli inherited retinal disease consortium. Data were collected in May 2023 on 9396 individuals residing in Israel who were diagnosed by an ophthalmologist with an IRD using either electroretinography or retinal imaging where included. Individuals with retinal diseases known to have a nonmendelian basis or without a clear genetic basis and those who were reported as deceased at the time of data collection were excluded from this study. Main Outcomes and Measures: Prevalence of 67 IRD phenotypes. Results: Among the 9396 participants in our cohort, the most common IRD in Israel was retinitis pigmentosa with a disease prevalence of approximately 1:2400 individuals, followed by cone-rod dystrophy (approximately 1:14 000), Stargardt disease (approximately 1:16 000), Usher syndrome (approximately 1:16,000), and congenital stationary night blindness (approximately 1:18 000). The prevalence of all IRDs combined was 1:1043 individuals. Conclusions and Relevance: The current study provides large prevalence dataset of 67 IRD phenotypes, some of which are extremely rare, with only a single identified case. This analysis highlights the potential importance of performing additional nationwide prevalence studies to potentially assist with determining the prevalence of IRDs worldwide.


Asunto(s)
Electrorretinografía , Enfermedades de la Retina , Humanos , Israel/epidemiología , Prevalencia , Masculino , Femenino , Adulto , Enfermedades de la Retina/epidemiología , Enfermedades de la Retina/genética , Enfermedades de la Retina/diagnóstico , Persona de Mediana Edad , Fenotipo , Adolescente , Adulto Joven , Anciano , Niño , Enfermedades Hereditarias del Ojo/epidemiología , Enfermedades Hereditarias del Ojo/genética , Preescolar
11.
Biomolecules ; 14(3)2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38540785

RESUMEN

Inherited macular dystrophies (iMDs) are a group of genetic disorders, which affect the central region of the retina. To investigate the genetic basis of iMDs, we used single-molecule Molecular Inversion Probes to sequence 105 maculopathy-associated genes in 1352 patients diagnosed with iMDs. Within this cohort, 39.8% of patients were considered genetically explained by 460 different variants in 49 distinct genes of which 73 were novel variants, with some affecting splicing. The top five most frequent causative genes were ABCA4 (37.2%), PRPH2 (6.7%), CDHR1 (6.1%), PROM1 (4.3%) and RP1L1 (3.1%). Interestingly, variants with incomplete penetrance were revealed in almost one-third of patients considered solved (28.1%), and therefore, a proportion of patients may not be explained solely by the variants reported. This includes eight previously reported variants with incomplete penetrance in addition to CDHR1:c.783G>A and CNGB3:c.1208G>A. Notably, segregation analysis was not routinely performed for variant phasing-a limitation, which may also impact the overall diagnostic yield. The relatively high proportion of probands without any putative causal variant (60.2%) highlights the need to explore variants with incomplete penetrance, the potential modifiers of disease and the genetic overlap between iMDs and age-related macular degeneration. Our results provide valuable insights into the genetic landscape of iMDs and warrant future exploration to determine the involvement of other maculopathy genes.


Asunto(s)
Degeneración Macular , Humanos , Mutación , Penetrancia , Linaje , Degeneración Macular/genética , Retina , Fenotipo , Transportadoras de Casetes de Unión a ATP/genética , Proteínas del Ojo , Proteínas Relacionadas con las Cadherinas , Proteínas del Tejido Nervioso/genética
12.
Br J Ophthalmol ; 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39079892

RESUMEN

BACKGROUND/AAIMS: Congenital stationary night blindness (CSNB) is an inherited retinal disease that is often associated with high myopia and can be caused by pathological variants in multiple genes, most commonly CACNA1F, NYX and TRPM1. High myopia is associated with retinal degeneration and increased risk for retinal detachment. Slowing the progression of myopia in patients with CSNB would likely be beneficial in reducing risk, but before interventions can be considered, it is important to understand the natural history of myopic progression. METHODS: This multicentre, retrospective study explored CSNB caused by variants in CACNA1F, NYX or TRPM1 in patients who had at least 6 measurements of their spherical equivalent of refraction (SER) before the age of 18. A mixed-effect model was used to predict progression of SER overtime and differences between genotypes were evaluated. RESULTS: 78 individuals were included in this study. All genotypes showed a significant myopic predicted SER at birth (-3.076D, -5.511D and -5.386D) for CACNA1F, NYX and TRPM1 respectively. Additionally, significant progression of myopia per year (-0.254D, -0.257D and -0.326D) was observed for all three genotypes CACNA1F, NYX and TRPM1, respectively. CONCLUSIONS: Patients with CSNB tend to be myopic from an early age and progress to become more myopic with age. Patients may benefit from long-term myopia slowing treatment in the future and further studies are indicated. Additionally, CSNB should be considered in the differential diagnosis for early-onset myopia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA