Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 111(2): 393-402, 2024 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-38272031

RESUMEN

Sterile alpha motif domain containing 7 (SAMD7) is a component of the Polycomb repressive complex 1, which inhibits transcription of many genes, including those activated by the transcription factor Cone-Rod Homeobox (CRX). Here we report bi-allelic mutations in SAMD7 as a cause of autosomal-recessive macular dystrophy with or without cone dysfunction. Four of these mutations affect splicing, while another mutation is a missense variant that alters the repressive effect of SAMD7 on CRX-dependent promoter activity, as shown by in vitro assays. Immunostaining of human retinal sections revealed that SAMD7 is localized in the nuclei of both rods and cones, as well as in those of cells belonging to the inner nuclear layer. These results place SAMD7 as a gene crucial for human retinal function and demonstrate a significant difference in the role of SAMD7 between the human and the mouse retina.


Asunto(s)
Anomalías del Ojo , Degeneración Macular , Ratones , Animales , Humanos , Transactivadores/genética , Proteínas de Homeodominio/genética , Retina , Mutación/genética , Degeneración Macular/genética
2.
Am J Hum Genet ; 109(3): 498-507, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35120629

RESUMEN

Recurrence risk calculations in autosomal recessive diseases are complicated when the effect of genetic variants and their population frequencies and penetrances are unknown. An example of this is Stargardt disease (STGD1), a frequent recessive retinal disease caused by bi-allelic pathogenic variants in ABCA4. In this cross-sectional study, 1,619 ABCA4 variants from 5,579 individuals with STGD1 were collected and categorized by (1) severity based on statistical comparisons of their frequencies in STGD1-affected individuals versus the general population, (2) their observed versus expected homozygous occurrence in STGD1-affected individuals, (3) their occurrence in combination with established mild alleles in STGD1-affected individuals, and (4) previous functional and clinical studies. We used the sum allele frequencies of these severity categories to estimate recurrence risks for offspring of STGD1-affected individuals and carriers of pathogenic ABCA4 variants. The risk for offspring of an STGD1-affected individual with the "severe|severe" genotype or a "severe|mild with complete penetrance" genotype to develop STGD1 at some moment in life was estimated at 2.8%-3.1% (1 in 36-32 individuals) and 1.6%-1.8% (1 in 62-57 individuals), respectively. The risk to develop STGD1 in childhood was estimated to be 2- to 4-fold lower: 0.68%-0.79% (1 in 148-126) and 0.34%-0.39% (1 in 296-252), respectively. In conclusion, we established personalized recurrence risk calculations for STGD1-affected individuals with different combinations of variants. We thus propose an expanded genotype-based personalized counseling to appreciate the variable recurrence risks for STGD1-affected individuals. This represents a conceptual breakthrough because risk calculations for STGD1 may be exemplary for many other inherited diseases.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Asesoramiento Genético , Transportadoras de Casetes de Unión a ATP/genética , Estudios Transversales , Humanos , Mutación , Enfermedad de Stargardt/genética
3.
Mol Ther ; 32(3): 837-851, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38243599

RESUMEN

The high allelic heterogeneity in Stargardt disease (STGD1) complicates the design of intervention strategies. A significant proportion of pathogenic intronic ABCA4 variants alters the pre-mRNA splicing process. Antisense oligonucleotides (AONs) are an attractive yet mutation-specific therapeutic strategy to restore these splicing defects. In this study, we experimentally assessed the potential of a splicing modulation therapy to target multiple intronic ABCA4 variants. AONs were inserted into U7snRNA gene cassettes and tested in midigene-based splice assays. Five potent antisense sequences were selected to generate a multiple U7snRNA cassette construct, and this combination vector showed substantial rescue of all of the splicing defects. Therefore, the combination cassette was used for viral synthesis and assessment in patient-derived photoreceptor precursor cells (PPCs). Simultaneous delivery of several modified U7snRNAs through a single AAV, however, did not show substantial splicing correction, probably due to suboptimal transduction efficiency in PPCs and/or a heterogeneous viral population containing incomplete AAV genomes. Overall, these data demonstrate the potential of the U7snRNA system to rescue multiple splicing defects, but also suggest that AAV-associated challenges are still a limiting step, underscoring the need for further optimization before implementing this strategy as a potential treatment for STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Empalme del ARN , Humanos , Transportadoras de Casetes de Unión a ATP/genética , Enfermedad de Stargardt/genética , Mutación , Células Fotorreceptoras
4.
Clin Genet ; 106(2): 127-139, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38468396

RESUMEN

Leber congenital amaurosis (LCA) and early-onset retinal degeneration (EORD) are inherited retinal diseases (IRD) characterized by early-onset vision impairment. Herein, we studied 15 Saudi families by whole exome sequencing (WES) and run-of-homozygosity (ROH) detection via AutoMap in 12/15 consanguineous families. This revealed (likely) pathogenic variants in 11/15 families (73%). A potential founder variant was found in RPGRIP1. Homozygous pathogenic variants were identified in known IRD genes (ATF6, CRB1, CABP4, RDH12, RIMS2, RPGRIP1, SPATA7). We established genotype-driven clinical reclassifications for ATF6, CABP4, and RIMS2. Specifically, we observed isolated IRD in the individual with the novel RIMS2 variant, and we found a retina-enriched RIMS2 isoform conserved but not annotated in mouse. The latter illustrates potential different phenotypic consequences of pathogenic variants depending on the particular tissue/cell-type specific isoforms they affect. Lastly, a compound heterozygous genotype in GUCY2D in one non-consanguineous family was demonstrated, and homozygous variants in novel candidate genes ATG2B and RUFY3 were found in the two remaining consanguineous families. Reporting these genes will allow to validate them in other IRD cohorts. Finally, the missing heritability of the two unsolved IRD cases may be attributed to variants in non-coding regions or structural variants that remained undetected, warranting future WGS studies.


Asunto(s)
Consanguinidad , Secuenciación del Exoma , Linaje , Fenotipo , Humanos , Femenino , Masculino , Retina/patología , Homocigoto , Enfermedades de la Retina/genética , Isoformas de Proteínas/genética , Exoma/genética , Mutación , Niño , Predisposición Genética a la Enfermedad , Amaurosis Congénita de Leber/genética , Estudios de Cohortes , Genotipo , Estudios de Asociación Genética/métodos
5.
Clin Genet ; 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39199020

RESUMEN

Usher syndrome (USH) is the most common cause of deafblindness. USH is autosomal recessively inherited and characterized by rod-cone dystrophy or retinitis pigmentosa (RP), often accompanied by sensorineural hearing loss. Variants in >15 genes have been identified as causative for clinically and genetically distinct subtypes. Among the ultra-rare and recently discovered genes is ARSG, coding for the lysosomal sulfatase Arylsulfatase G. This subtype was assigned as "USH IV" with a late onset of RP and usually late-onset progressive SNHL without vestibular involvement. Here, we describe nine new subjects and the clinical description of four cases with the USH IV phenotype bearing seven novel and two known pathogenic variants. Functional experiments indicated the complete loss of sulfatase enzymatic activity upon ectopic expression of mutated ARSG cDNA. Interestingly, we identified a homozygous missense variant, p.(Arg99His), previously described in dogs with neuronal ceroid lipofuscinosis. Our study expands the genetic landscape of ARSG-USH IV and the number of known subjects by more than 30%. These findings highlight that USH IV likely has been underdiagnosed and emphasize the need to test molecularly unresolved subjects with deafblindness syndrome. Finally, testing of ARSG should be considered for the genetic work-up of apparent isolated inherited retinal diseases.

6.
Proc Natl Acad Sci U S A ; 117(16): 9001-9012, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32265282

RESUMEN

The interplay of transcription factors and cis-regulatory elements (CREs) orchestrates the dynamic and diverse genetic programs that assemble the human central nervous system (CNS) during development and maintain its function throughout life. Genetic variation within CREs plays a central role in phenotypic variation in complex traits including the risk of developing disease. We took advantage of the retina, a well-characterized region of the CNS known to be affected by pathogenic variants in CREs, to establish a roadmap for characterizing regulatory variation in the human CNS. This comprehensive analysis of tissue-specific regulatory elements, transcription factor binding, and gene expression programs in three regions of the human visual system (retina, macula, and retinal pigment epithelium/choroid) reveals features of regulatory element evolution that shape tissue-specific gene expression programs and defines regulatory elements with the potential to contribute to Mendelian and complex disorders of human vision.


Asunto(s)
Evolución Molecular , Regulación del Desarrollo de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retina/patología , Enfermedades de la Retina/genética , Adulto , Animales , Análisis Mutacional de ADN , Epigenómica , Femenino , Variación Genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Mutación , RNA-Seq , Retina/crecimiento & desarrollo , Enfermedades de la Retina/patología , Especificidad de la Especie
7.
Genet Med ; 22(7): 1235-1246, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32307445

RESUMEN

PURPOSE: Missing heritability in human diseases represents a major challenge, and this is particularly true for ABCA4-associated Stargardt disease (STGD1). We aimed to elucidate the genomic and transcriptomic variation in 1054 unsolved STGD and STGD-like probands. METHODS: Sequencing of the complete 128-kb ABCA4 gene was performed using single-molecule molecular inversion probes (smMIPs), based on a semiautomated and cost-effective method. Structural variants (SVs) were identified using relative read coverage analyses and putative splice defects were studied using in vitro assays. RESULTS: In 448 biallelic probands 14 known and 13 novel deep-intronic variants were found, resulting in pseudoexon (PE) insertions or exon elongations in 105 alleles. Intriguingly, intron 13 variants c.1938-621G>A and c.1938-514G>A resulted in dual PE insertions consisting of the same upstream, but different downstream PEs. The intron 44 variant c.6148-84A>T resulted in two PE insertions and flanking exon deletions. Eleven distinct large deletions were found, two of which contained small inverted segments. Uniparental isodisomy of chromosome 1 was identified in one proband. CONCLUSION: Deep sequencing of ABCA4 and midigene-based splice assays allowed the identification of SVs and causal deep-intronic variants in 25% of biallelic STGD1 cases, which represents a model study that can be applied to other inherited diseases.


Asunto(s)
Degeneración Macular , Transcriptoma , Transportadoras de Casetes de Unión a ATP/genética , Genómica , Humanos , Intrones , Degeneración Macular/genética , Mutación , Linaje , Enfermedad de Stargardt
8.
Clin Genet ; 97(3): 426-436, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31721179

RESUMEN

Biallelic MFSD8 variants are an established cause of severe late-infantile subtype of neuronal ceroid lipofuscinosis (v-LINCL), a severe lysosomal storage disorder, but have also been associated with nonsyndromic adult-onset maculopathy. Here, we functionally characterized two novel MFSD8 variants found in a child with juvenile isolated maculopathy, in order to establish a refined prognosis. ABCA4 locus resequencing was followed by the analysis of other inherited retinal disease genes by whole exome sequencing (WES). Minigene assays and cDNA sequencing were used to assess the effect of a novel MFSD8 splice variant. MFSD8 expression was quantified with qPCR and overexpression studies were analyzed by immunoblotting. Transmission electron microscopy (TEM) was performed on a skin biopsy and ophthalmological and neurological re-examinations were conducted. WES revealed two novel MFSD8 variants: c.[590del];[439+3A>C] p.[Gly197Valfs*2];[Ile67Glufs*3]. Characterization of the c.439+3A>C variant via splice assays showed exon-skipping (p.Ile67Glufs*3), while overexpression studies of the corresponding protein indicated expression of a truncated polypeptide. In addition, a significantly reduced MFSD8 RNA expression was noted in patient's lymphocytes. TEM of a skin biopsy revealed typical v-LINCL lipopigment inclusions while neurological imaging of the proband displayed subtle cerebellar atrophy. Functional characterization demonstrated the pathogenicity of two novel MFSD8 variants, found in a child with an initial diagnosis of juvenile isolated maculopathy but likely evolving to v-LINCL with a protracted disease course. Our study allowed a refined neurological prognosis in the proband and expands the natural history of MFSD8-associated disease.


Asunto(s)
Degeneración Macular/genética , Proteínas de Transporte de Membrana/genética , Lipofuscinosis Ceroideas Neuronales/genética , Niño , Femenino , Variación Genética , Homocigoto , Humanos , Degeneración Macular/diagnóstico por imagen , Degeneración Macular/fisiopatología , Microscopía Electrónica de Transmisión , Mutación , Lipofuscinosis Ceroideas Neuronales/fisiopatología , Retina/diagnóstico por imagen , Retina/fisiopatología , Secuenciación del Exoma
9.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-27486781

RESUMEN

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Asunto(s)
Alelos , Factores de Intercambio de Guanina Nucleótido/genética , Mutación Missense/genética , Distrofias Retinianas/genética , Ubiquitinación/genética , Adolescente , Adulto , Edad de Inicio , Niño , Consanguinidad , Proteínas Cullin/metabolismo , Exoma/genética , Femenino , Efecto Fundador , Genes Recesivos , Haplotipos/genética , Homocigoto , Humanos , Linfocitos/metabolismo , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Linaje , Fenotipo , ARN Mensajero/genética , Retina/metabolismo , Síndrome , Turquía
10.
Genet Med ; 21(8): 1998, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30297699

RESUMEN

The original version of this Article contained an error in the spelling of the author Anja K. Mayer, which was incorrectly given as Anja Kathrin Mayer. This has now been corrected in both the PDF and HTML versions of the Article.

11.
Genet Med ; 21(8): 1761-1771, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30670881

RESUMEN

PURPOSE: ABCA4-associated disease, a recessive retinal dystrophy, is hallmarked by a large proportion of patients with only one pathogenic ABCA4 variant, suggestive for missing heritability. METHODS: By locus-specific analysis of ABCA4, combined with extensive functional studies, we aimed to unravel the missing alleles in a cohort of 67 patients (p), with one (p = 64) or no (p = 3) identified coding pathogenic variants of ABCA4. RESULTS: We identified eight pathogenic (deep-)intronic ABCA4 splice variants, of which five are novel and six structural variants, four of which are novel, including two duplications. Together, these variants account for the missing alleles in 40.3% of patients. Furthermore, two novel variants with a putative cis-regulatory effect were identified. The common hypomorphic variant c.5603A>T p.(Asn1868Ile) was found as a candidate second allele in 43.3% of patients. Overall, we have elucidated the missing heritability in 83.6% of our cohort. In addition, we successfully rescued three deep-intronic variants using antisense oligonucleotide (AON)-mediated treatment in HEK 293-T cells and in patient-derived fibroblast cells. CONCLUSION: Noncoding pathogenic variants, novel structural variants, and a common hypomorphic allele of the ABCA4 gene explain the majority of unsolved cases with ABCA4-associated disease, rendering this retinopathy a model for missing heritability in autosomal recessive disorders.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Genes Recesivos/genética , Oligonucleótidos Antisentido/genética , Distrofias Retinianas/genética , Adulto , Alelos , Estudios de Cohortes , Exones/genética , Femenino , Frecuencia de los Genes , Células HEK293 , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Mutación/genética , Oligonucleótidos Antisentido/farmacología , Linaje , Fenotipo , Distrofias Retinianas/patología
12.
Genet Med ; 21(8): 1751-1760, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30643219

RESUMEN

PURPOSE: Using exome sequencing, the underlying variants in many persons with autosomal recessive diseases remain undetected. We explored autosomal recessive Stargardt disease (STGD1) as a model to identify the missing heritability. METHODS: Sequencing of ABCA4 was performed in 8 STGD1 cases with one variant and p.Asn1868Ile in trans, 25 cases with one variant, and 3 cases with no ABCA4 variant. The effect of intronic variants was analyzed using in vitro splice assays in HEK293T cells and patient-derived fibroblasts. Antisense oligonucleotides were used to correct splice defects. RESULTS: In 24 of the probands (67%), one known and five novel deep-intronic variants were found. The five novel variants resulted in messenger RNA pseudoexon inclusions, due to strengthening of cryptic splice sites or by disrupting a splicing silencer motif. Variant c.769-784C>T showed partial insertion of a pseudoexon and was found in cis with c.5603A>T (p.Asn1868Ile), so its causal role could not be fully established. Variant c.4253+43G>A resulted in partial skipping of exon 28. Remarkably, antisense oligonucleotides targeting the aberrant splice processes resulted in (partial) correction of all splicing defects. CONCLUSION: Our data demonstrate the importance of assessing noncoding variants in genetic diseases, and show the great potential of splice modulation therapy for deep-intronic variants.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Oligonucleótidos Antisentido/genética , Isoformas de Proteínas/genética , Enfermedad de Stargardt/genética , Adolescente , Adulto , Anciano , Niño , Exones/genética , Células HEK293 , Humanos , Intrones/genética , Persona de Mediana Edad , Mutación/genética , Oligonucleótidos Antisentido/farmacología , Linaje , Polimorfismo de Nucleótido Simple/genética , Empalme del ARN/genética , Enfermedad de Stargardt/patología , Adulto Joven
13.
Genet Med ; 20(2): 202-213, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28749477

RESUMEN

PurposePart of the hidden genetic variation in heterogeneous genetic conditions such as inherited retinal diseases (IRDs) can be explained by copy-number variations (CNVs). Here, we explored the genomic landscape of IRD genes listed in RetNet to identify and prioritize those genes susceptible to CNV formation.MethodsRetNet genes underwent an assessment of genomic features and of CNV occurrence in the Database of Genomic Variants and literature. CNVs identified in an IRD cohort were characterized using targeted locus amplification (TLA) on extracted genomic DNA.ResultsExhaustive literature mining revealed 1,345 reported CNVs in 81 different IRD genes. Correlation analysis between rankings of genomic features and CNV occurrence demonstrated the strongest correlation between gene size and CNV occurrence of IRD genes. Moreover, we identified and delineated 30 new CNVs in IRD cases, 13 of which are novel and three of which affect noncoding, putative cis-regulatory regions. Finally, the breakpoints of six complex CNVs were determined using TLA in a hypothesis-neutral manner.ConclusionWe propose a ranking of CNV-prone IRD genes and demonstrate the efficacy of TLA for the characterization of CNVs on extracted DNA. Finally, this IRD-oriented CNV study can serve as a paradigm for other genetically heterogeneous Mendelian diseases with hidden genetic variation.


Asunto(s)
Mapeo Cromosómico , Variaciones en el Número de Copia de ADN , Genoma Humano , Genómica , Sistemas de Lectura Abierta , ARN no Traducido , Enfermedades de la Retina/genética , Alelos , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Bases de Datos Genéticas , Proteínas del Ojo/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genómica/métodos , Humanos , Secuencias Reguladoras de Ácidos Nucleicos , Enfermedades de la Retina/diagnóstico , Análisis de Secuencia de ADN , Eliminación de Secuencia
14.
Genet Med ; 19(4): 457-466, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27608171

RESUMEN

PURPOSE: Our goal was to design a customized microarray, arrEYE, for high-resolution copy number variant (CNV) analysis of known and candidate genes for inherited retinal dystrophy (iRD) and retina-expressed noncoding RNAs (ncRNAs). METHODS: arrEYE contains probes for the full genomic region of 106 known iRD genes, including those implicated in retinitis pigmentosa (RP) (the most frequent iRD), cone-rod dystrophies, macular dystrophies, and an additional 60 candidate iRD genes and 196 ncRNAs. Eight CNVs in iRD genes identified by other techniques were used as positive controls. The test cohort consisted of 57 patients with autosomal dominant, X-linked, or simplex RP. RESULTS: In an RP patient, a novel heterozygous deletion of exons 7 and 8 of the HGSNAT gene was identified: c.634-408_820+338delinsAGAATATG, p.(Glu212Glyfs*2). A known variant was found on the second allele: c.1843G>A, p.(Ala615Thr). Furthermore, we expanded the allelic spectrum of USH2A and RCBTB1 with novel CNVs. CONCLUSION: The arrEYE platform revealed subtle single-exon to larger CNVs in iRD genes that could be characterized at the nucleotide level, facilitated by the high resolution of the platform. We report the first CNV in HGSNAT that, combined with another mutation, leads to RP, further supporting its recently identified role in nonsyndromic iRD.Genet Med 19 4, 457-466.


Asunto(s)
Hibridación Genómica Comparativa/métodos , Variaciones en el Número de Copia de ADN , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Distrofias Retinianas/genética , Acetiltransferasas/genética , Proteínas de la Matriz Extracelular/genética , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Masculino , ARN no Traducido/genética , Eliminación de Secuencia
15.
Ophthalmology ; 123(6): 1375-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26976702

RESUMEN

PURPOSE: To elucidate the functional effect of the ABCA4 variant c.5461-10T→C, one of the most frequent variants associated with Stargardt disease (STGD1). DESIGN: Case series. PARTICIPANTS: Seventeen persons with STGD1 carrying ABCA4 variants and 1 control participant. METHODS: Haplotype analysis of 4 homozygotes and 11 heterozygotes for c.5461-10T→C and sequence analysis of the ABCA4 gene for a homozygous proband. Fibroblasts were reprogrammed from 3 persons with STGD1 into induced pluripotent stem cells, which were differentiated into photoreceptor progenitor cells (PPCs). The effect of the c.5461-10T→C variant on RNA splicing by reverse-transcription polymerase chain reaction was analyzed using PPC mRNA. In vitro assays were performed with minigene constructs containing ABCA4 exon 39. We analyzed the natural history and ophthalmologic characteristics of 4 persons homozygous for c.5461-10T→C. MAIN OUTCOME MEASURES: Haplotype and rare variant data for ABCA4, RNA splice defects, age at diagnosis, visual acuity, fundus appearance, visual field, electroretinography (ERG) results, fluorescein angiography results, and fundus autofluorescence findings. RESULTS: The frequent ABCA4 variant c.5461-10T→C has a subtle effect on splicing based on prediction programs. A founder haplotype containing c.5461-10T→C was found to span approximately 96 kb of ABCA4 and did not contain other rare sequence variants. Patient-derived PPCs showed skipping of exon 39 or exons 39 and 40 in the mRNA. HEK293T cell transduction with minigenes carrying exon 39 showed that the splice defects were the result of the c.5461-10T→C variant. All 4 subjects carrying the c.5461-10T→C variant in a homozygous state showed a young age of STGD1 onset, with low visual acuity at presentation and abnormal cone ERG results. All 4 demonstrated severe cone-rod dystrophy before 20 years of age and were legally blind by 25 years of age. CONCLUSIONS: The ABCA4 variant c.5461-10T→C is located on a founder haplotype lacking other disease-causing rare sequence variants. In vitro studies revealed that it leads to mRNA exon skipping and ABCA4 protein truncation. Given the severe phenotype in persons homozygous for this variant, we conclude that this variant results in the absence of ABCA4 activity.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Empalme Alternativo , Exones/genética , Degeneración Macular/congénito , Células Fotorreceptoras de Vertebrados/fisiología , ARN Mensajero/genética , Células Madre/fisiología , Adulto , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Fibroblastos/metabolismo , Angiografía con Fluoresceína , Células HEK293 , Haplotipos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Degeneración Macular/fisiopatología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Sitios de Empalme de ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Enfermedad de Stargardt , Transfección , Agudeza Visual/fisiología , Campos Visuales/fisiología , Adulto Joven
16.
Hum Mutat ; 36(12): 1188-96, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26316326

RESUMEN

Leber congenital amaurosis (LCA) is a severe autosomal-recessive retinal dystrophy leading to congenital blindness. A recently identified LCA gene is NMNAT1, located in the LCA9 locus. Although most mutations in blindness genes are coding variations, there is accumulating evidence for hidden noncoding defects or structural variations (SVs). The starting point of this study was an LCA9-associated consanguineous family in which no coding mutations were found in the LCA9 region. Exploring the untranslated regions of NMNAT1 revealed a novel homozygous 5'UTR variant, c.-70A>T. Moreover, an adjacent 5'UTR variant, c.-69C>T, was identified in a second consanguineous family displaying a similar phenotype. Both 5'UTR variants resulted in decreased NMNAT1 mRNA abundance in patients' lymphocytes, and caused decreased luciferase activity in human retinal pigment epithelial RPE-1 cells. Second, we unraveled pseudohomozygosity of a coding NMNAT1 mutation in two unrelated LCA patients by the identification of two distinct heterozygous partial NMNAT1 deletions. Molecular characterization of the breakpoint junctions revealed a complex Alu-rich genomic architecture. Our study uncovered hidden genetic variation in NMNAT1-associated LCA and emphasized a shift from coding to noncoding regulatory mutations and repeat-mediated SVs in the molecular pathogenesis of heterogeneous recessive disorders such as hereditary blindness.


Asunto(s)
Regiones no Traducidas 5' , Variaciones en el Número de Copia de ADN , Amaurosis Congénita de Leber/genética , Mutación , Nicotinamida-Nucleótido Adenililtransferasa/genética , Alelos , Elementos Alu , Niño , Puntos de Rotura del Cromosoma , Mapeo Cromosómico , Biología Computacional/métodos , Consanguinidad , Exones , Femenino , Expresión Génica , Estudios de Asociación Genética , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Amaurosis Congénita de Leber/diagnóstico , Masculino , Linaje , Fenotipo , ARN Mensajero/genética , Análisis de Secuencia de ADN , Adulto Joven
17.
Hum Mutat ; 36(1): 39-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25346251

RESUMEN

Autosomal-recessive Stargardt disease (STGD1) is hallmarked by a large proportion of patients with a single heterozygous causative variant in the disease gene ABCA4. Braun et al. () reported deep intronic variants of ABCA4 in STGD1 patients with one coding variant, prompting us to perform an augmented screen in 131 Belgian STGD1 patients with one or no ABCA4 variant to uncover deep intronic causal ABCA4 variants. This revealed a second variant in 28.6% of cases. Twenty-six percent of these carry the same causal variant c.4539+2001G>A (V4). Haplotyping in V4 carriers showed a common region of 63 kb, suggestive of a founder mutation. Genotype-phenotype correlations suggest a moderate-to-severe impact of V4 on the STGD1 phenotype. In conclusion, V4 occurs in a high fraction of Belgian STGD1 patients and represents the first deep intronic founder mutation in ABCA4. This emphasizes the importance of augmented molecular genetic testing of ABCA4 in Belgian STGD1.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Efecto Fundador , Degeneración Macular/congénito , Bélgica , Estudios de Asociación Genética , Haplotipos , Humanos , Intrones , Degeneración Macular/genética , Mutación , Enfermedad de Stargardt
18.
Genet Med ; 17(4): 291-9, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25122145

RESUMEN

PURPOSE: The aim of this study was to identify the genetic cause of early-onset autosomal recessive cerebellar ataxia associated with retinal dystrophy in a consanguineous family. METHODS: An affected 6-month-old child underwent neurological and ophthalmological examinations. Genetic analyses included homozygosity mapping, copy number analysis, conventional polymerase chain reaction, Sanger sequencing, quantitative polymerase chain reaction, and whole-exome sequencing. Expression analysis of GRID2 was performed by quantitative polymerase chain reaction and immunohistochemistry. RESULTS: A homozygous deletion of exon 2 of GRID2 (p.Gly30_Glu81del) was identified in the proband. GRID2 encodes an ionotropic glutamate receptor known to be selectively expressed in cerebellar Purkinje cells. Here, we demonstrated GRID2 expression in human adult retina and retinal pigment epithelium. In addition, Grid2 expression was demonstrated in different stages of murine retinal development. GRID2 immunostaining was shown in murine and human retina. Whole-exome sequencing in the proband did not provide arguments for other disease-causing mutations, supporting the idea that the phenotype observed represents a single clinical entity. CONCLUSION: We identified GRID2 as an underlying disease gene of early-onset autosomal recessive cerebellar ataxia with retinal dystrophy, expanding the clinical spectrum of GRID2 deletion mutants. We demonstrated for the first time GRID2 expression and localization in human and murine retina, providing evidence for a novel functional role of GRID2 in the retina.


Asunto(s)
Receptores de Glutamato/genética , Distrofias Retinianas/genética , Degeneraciones Espinocerebelosas/genética , Animales , Preescolar , Variaciones en el Número de Copia de ADN/genética , Exones/genética , Femenino , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Ratones , Linaje , Receptores de Glutamato/biosíntesis , Retina/metabolismo , Retina/patología , Distrofias Retinianas/complicaciones , Distrofias Retinianas/patología , Eliminación de Secuencia , Degeneraciones Espinocerebelosas/complicaciones , Degeneraciones Espinocerebelosas/patología
19.
Ophthalmic Res ; 54(4): 181-94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26492201

RESUMEN

PURPOSE: To investigate the type and severity of acquired colour vision deficiencies (CVDs) in molecularly proven Stargardt disease (STD) and to establish whether a relationship exists between best-corrected visual acuity (BCVA) and full-field electroretinography (ffERG), and the degree of CVD. METHODS: A retrospective, cross-sectional study of 73 patients with a molecularly proven diagnosis of STD, who underwent extensive colour vision evaluation, using pseudo-isochromatic and arrangement tests. Thirteen patients underwent Nagel anomaloscopy. RESULTS: Normal colour vision was found in almost 20% of patients. Red/green (R/G) CVDs increased as BCVA declined. About 45% of all R/G CVDs were of the deutan type, although protan type CVDs became progressively apparent when moving from the high to the low BCVA group. An additional blue/yellow CVD was noted in about 25% of patients. In 10/13 patients, a pseudoprotanomaly was noted on anomaloscopy. Severe CVDs leading to scotopization were noted in patients with low BCVA and/or long-standing disease. No statistically significant differences in ERG results were found between groups with or without a CVD. CONCLUSIONS: The degree and type of colour vision deficiency in STD patients correlate better with BCVA than with ffERG results. The presence of specific CVDs may help to establish a diagnosis of STD. A battery of colour vision tests is required to properly evaluate CVDs in STD.


Asunto(s)
Defectos de la Visión Cromática/fisiopatología , Degeneración Macular/congénito , Transportadoras de Casetes de Unión a ATP/genética , Adulto , Pruebas de Percepción de Colores , Defectos de la Visión Cromática/diagnóstico , Estudios Transversales , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Degeneración Macular/fisiopatología , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Mutación , Estudios Retrospectivos , Enfermedad de Stargardt , Agudeza Visual/fisiología
20.
Genet Med ; 16(9): 671-80, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24625443

RESUMEN

PURPOSE: Autosomal recessive retinal dystrophies are clinically and genetically heterogeneous, which hampers molecular diagnosis. We evaluated identity-by-descent-guided Sanger sequencing or whole-exome sequencing in 26 families with nonsyndromic (19) or syndromic (7) autosomal recessive retinal dystrophies to identify disease-causing mutations. METHODS: Patients underwent genome-wide identity-by-descent mapping followed by Sanger sequencing (16) or whole-exome sequencing (10). Whole-exome sequencing data were filtered against identity-by-descent regions and known retinal dystrophy genes. The medical history was reviewed in mutation-positive families. RESULTS: We identified mutations in 14 known retinal dystrophy genes in 20/26 (77%) families: ABCA4, CERKL, CLN3, CNNM4, C2orf71, IQCB1, LRAT, MERTK, NMNAT1, PCDH15, PDE6B, RDH12, RPGRIP1, and USH2A. Whole-exome sequencing in single individuals revealed mutations in either the largest or smaller identity-by-descent regions, and a compound heterozygous genotype in NMNAT1. Moreover, a novel deletion was found in PCDH15. In addition, we identified mutations in CLN3, CNNM4, and IQCB1 in patients initially diagnosed with nonsyndromic retinal dystrophies. CONCLUSION: Our study emphasized that identity-by-descent-guided mutation analysis and/or whole-exome sequencing are powerful tools for the molecular diagnosis of retinal dystrophy. Our approach uncovered unusual molecular findings and unmasked syndromic retinal dystrophies, guiding future medical management. Finally, elucidating ABCA4, LRAT, and MERTK mutations offers potential gene-specific therapeutic perspectives.


Asunto(s)
Consanguinidad , Análisis Mutacional de ADN , Exoma , Mutación , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/genética , Adolescente , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Niño , Preescolar , Femenino , Genes Recesivos , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , Masculino , Mutación Missense , Oftalmoscopios , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Diente/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA