Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Med Genet ; 58(8): 570-578, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32817297

RESUMEN

BACKGROUND: Inherited retinal disorders are a clinically and genetically heterogeneous group of conditions and a major cause of visual impairment. Common disease subtypes include vitelliform macular dystrophy (VMD) and retinitis pigmentosa (RP). Despite the identification of over 90 genes associated with RP, conventional genetic testing fails to detect a molecular diagnosis in about one third of patients with RP. METHODS: Exome sequencing was carried out for identifying the disease-causing gene in a family with autosomal dominant RP. Gene panel testing and exome sequencing were performed in 596 RP and VMD families to identified additional IMPG1 variants. In vivo analysis in the medaka fish system by knockdown assays was performed to screen IMPG1 possible pathogenic role. RESULTS: Exome sequencing of a family with RP revealed a splice variant in IMPG1. Subsequently, the same variant was identified in individuals from two families with either RP or VMD. A retrospective study of patients with RP or VMD revealed eight additional families with different missense or nonsense variants in IMPG1. In addition, the clinical diagnosis of the IMPG1 retinopathy-associated variant, originally described as benign concentric annular macular dystrophy, was also revised to RP with early macular involvement. Using morpholino-mediated ablation of Impg1 and its paralog Impg2 in medaka fish, we confirmed a phenotype consistent with that observed in the families, including a decreased length of rod and cone photoreceptor outer segments. CONCLUSION: This study discusses a previously unreported association between monoallelic or biallelic IMPG1 variants and RP. Notably, similar observations have been reported for IMPG2.


Asunto(s)
Proteínas de la Matriz Extracelular , Proteínas del Ojo , Genes Recesivos , Predisposición Genética a la Enfermedad , Mutación , Proteoglicanos , Retinitis Pigmentosa , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Exoma/genética , Secuenciación del Exoma/métodos , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Genes Recesivos/genética , Predisposición Genética a la Enfermedad/genética , Patrón de Herencia/genética , Degeneración Macular/genética , Mutación/genética , Linaje , Fenotipo , Proteoglicanos/genética , Retina/patología , Retinitis Pigmentosa/genética , Estudios Retrospectivos
2.
Retina ; 41(8): 1771-1779, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33315831

RESUMEN

PURPOSE: RTN4IP1 biallelic mutations cause a recessive optic atrophy, sometimes associated to more severe neurological syndromes, but so far, no retinal phenotype has been reported in RTN4IP1 patients, justifying their reappraisal. METHODS: Seven patients from four families carrying biallelic RTN4IP1 variants were retrospectively reviewed, with emphasis on their age of onset, visual acuity, multimodal imaging including color and autofluorescence frames, spectral-domain optical coherence tomography with RNFL and macular analyses. RESULTS: Seven patients from four RTN4IP1 families developed in their first decade of life a bilateral recessive optic atrophy with severe central visual loss, and primary nystagmus developed in 5 of 7 patients. Six patients were legally blind. In a second stage, the seven individuals developed a rod-cone dystrophy, sparing the macular zone and the far periphery. This retinal damage was identified by 55° field fundus autofluorescence frames and also by spectral-domain optical coherence tomography scans of the temporal part of the macular zone in five of the seven patients. Full-field electroretinography measurements disclosed reduced b-wave amplitude of the rod responses in all patients but two. Family 4 with the p.R103H and c.601A > T (p.K201*) truncating mutation had further combined neurological signs with cerebellar ataxia, seizures, and intellectual disability. CONCLUSION: RTN4IP1 recessive optic atrophy is systematically associated to a rod-cone dystrophy, which suggests that both the retinal ganglion cells and the rods are affected as a result of a deficit in the mitochondrial respiratory chain. Thus, systematic widefield autofluorescence frames and temporal macular scans are recommended for the evaluation of patients with optic neuropathies.


Asunto(s)
Proteínas Portadoras/genética , Distrofias de Conos y Bastones/genética , ADN/genética , Proteínas Mitocondriales/genética , Mutación , Adolescente , Adulto , Proteínas Portadoras/metabolismo , Niño , Distrofias de Conos y Bastones/diagnóstico , Distrofias de Conos y Bastones/metabolismo , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Angiografía con Fluoresceína/métodos , Fondo de Ojo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/metabolismo , Linaje , Fenotipo , Estudios Retrospectivos , Tomografía de Coherencia Óptica/métodos , Agudeza Visual , Campos Visuales , Adulto Joven
3.
Hum Mol Genet ; 26(22): 4367-4374, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28973654

RESUMEN

In this study, we report a novel duplication causing North Carolina macular dystrophy (NCMD) identified applying whole genome sequencing performed on eight affected members of two presumed unrelated families mapping to the MCDR1 locus. In our families, the NCMD phenotype was associated with a 98.4 kb tandem duplication encompassing the entire CCNC and PRDM13 genes and a common DNase 1 hypersensitivity site. To study the impact of PRDM13 or CCNC dysregulation, we used the Drosophila eye development as a model. Knock-down and overexpression of CycC and CG13296, Drosophila orthologues of CCNC and PRDM13, respectively, were induced separately during eye development. In flies, eye development was not affected, while knocking down either CycC or CG13296 mutant models. Overexpression of CycC also had no effect. Strikingly, overexpression of CG13296 in Drosophila leads to a severe loss of the imaginal eye-antennal disc. This study demonstrated for the first time in an animal model that overexpression of PRDM13 alone causes a severe abnormal retinal development. It is noteworthy that mutations associated with this autosomal dominant foveal developmental disorder are frequently duplications always including an entire copy of PRDM13, or variants in one DNase 1 hypersensitivity site at this locus.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Ciclina C/genética , N-Metiltransferasa de Histona-Lisina/genética , Adulto , Animales , Mapeo Cromosómico , Cromosomas Humanos Par 6 , Distrofias Hereditarias de la Córnea/metabolismo , Ciclina C/metabolismo , Drosophila melanogaster , Proteínas del Ojo/genética , Femenino , Ligamiento Genético , Haplotipos , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Masculino , Dominios PR-SET , Linaje , Secuenciación Completa del Genoma
4.
Hum Genet ; 138(5): 441-453, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30904946

RESUMEN

Retinitis pigmentosa (RP) is a heterogeneous group of inherited retinal disorders eventually leading to blindness with different ages of onset, progression and severity. Human RP, first characterized by the progressive degeneration of rod photoreceptor cells, shows high genetic heterogeneity with more than 90 genes identified. However, about one-third of patients have no known genetic causes. Interestingly, dogs are also severely affected by similar diseases, called progressive retinal atrophy (PRA). Indeed, RP and PRA have comparable clinical signs, physiopathology and outcomes, similar diagnosis methods and most often, orthologous genes are involved. The many different dog PRAs often segregate in specific breeds. Indeed, undesired alleles have been selected and amplified through drastic selection and excessive use of inbreeding. Out of the 400 breeds, nearly 100 have an inherited form of PRA, which are natural animal models that can be used to investigate the genetics, disease progression and therapies in dogs for the benefit of both dogs and humans. Recent knowledge on the canine genome and access to new genotyping and sequencing technologies now efficiently allows the identification of mutations involved in canine genetic diseases. To date, PRA genes identified in dog breeds correspond to the same genes in humans and represent relevant RP models, and new genes found in dogs represent good candidate for still unknown human RP. We present here a review of the main advantages of the dog models for human RP with the genes already identified and an X-linked PRA in the Border collie as a model for orphan X-linked RPs in human.


Asunto(s)
Enfermedades de los Perros/genética , Degeneración Retiniana/genética , Degeneración Retiniana/veterinaria , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Animales , Modelos Animales de Enfermedad , Enfermedades de los Perros/patología , Perros , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Degeneración Retiniana/patología , Retinitis Pigmentosa/patología
5.
Int J Mol Sci ; 20(10)2019 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-31126147

RESUMEN

: Inherited retinal dystrophies (IRDs) are a clinically and genetically heterogeneous group of diseases with more than 250 causative genes. The most common form is retinitis pigmentosa. IRDs lead to vision impairment for which there is no universal cure. Encouragingly, a first gene supplementation therapy has been approved for an autosomal recessive IRD. However, for autosomal dominant IRDs, gene supplementation therapy is not always pertinent because haploinsufficiency is not the only cause. Disease-causing mechanisms are often gain-of-function or dominant-negative, which usually require alternative therapeutic approaches. In such cases, genome-editing technology has raised hopes for treatment. Genome editing could be used to i) invalidate both alleles, followed by supplementation of the wild type gene, ii) specifically invalidate the mutant allele, with or without gene supplementation, or iii) to correct the mutant allele. We review here the most prevalent genes causing autosomal dominant retinitis pigmentosa and the most appropriate genome-editing strategy that could be used to target their different causative mutations.


Asunto(s)
Edición Génica/métodos , Terapia Genética/métodos , Retinitis Pigmentosa/terapia , Animales , Sistemas CRISPR-Cas , Humanos , Mutación , Retinitis Pigmentosa/genética
6.
Hum Mol Genet ; 25(5): 916-26, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26744326

RESUMEN

Inherited retinal dystrophies are clinically and genetically heterogeneous with significant number of cases remaining genetically unresolved. We studied a large family from the West Indies islands with a peculiar retinal disease, the Martinique crinkled retinal pigment epitheliopathy that begins around the age of 30 with retinal pigment epithelium (RPE) and Bruch's membrane changes resembling a dry desert land and ends with a retinitis pigmentosa. Whole-exome sequencing identified a heterozygous c.518T>C (p.Leu173Pro) mutation in MAPKAPK3 that segregates with the disease in 14 affected and 28 unaffected siblings from three generations. This unknown variant is predicted to be damaging by bioinformatic predictive tools and the mutated protein to be non-functional by crystal structure analysis. MAPKAPK3 is a serine/threonine protein kinase of the p38 signaling pathway that is activated by a variety of stress stimuli and is implicated in cellular responses and gene regulation. In contrast to other tissues, MAPKAPK3 is highly expressed in the RPE, suggesting a crucial role for retinal physiology. Expression of the mutated allele in HEK cells revealed a mislocalization of the protein in the cytoplasm, leading to cytoskeleton alteration and cytodieresis inhibition. In Mapkapk3-/- mice, Bruch's membrane is irregular with both abnormal thickened and thinned portions. In conclusion, we identified the first pathogenic mutation in MAPKAPK3 associated with a retinal disease. These findings shed new lights on Bruch's membrane/RPE pathophysiology and will open studies of this signaling pathway in diseases with RPE and Bruch's membrane alterations, such as age-related macular degeneration.


Asunto(s)
Lámina Basal de la Coroides/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Proteínas Serina-Treonina Quinasas/genética , Distrofias Retinianas/genética , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/genética , Adulto , Edad de Inicio , Anciano de 80 o más Años , Secuencia de Aminoácidos , Animales , Lámina Basal de la Coroides/patología , Exoma , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Distrofias Retinianas/metabolismo , Distrofias Retinianas/patología , Epitelio Pigmentado de la Retina/patología , Alineación de Secuencia , Hermanos
7.
Am J Hum Genet ; 97(5): 754-60, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593267

RESUMEN

Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.


Asunto(s)
Proteínas Portadoras/genética , Fibroblastos/patología , Mitocondrias/patología , Proteínas Mitocondriales/genética , Mutación/genética , Enfermedades del Nervio Óptico/genética , Enfermedades del Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/antagonistas & inhibidores , Proteínas Portadoras/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Complejo I de Transporte de Electrón , Femenino , Fibroblastos/metabolismo , Estudios de Seguimiento , Genes Recesivos , Humanos , Masculino , Ratones , Mitocondrias/genética , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Datos de Secuencia Molecular , Degeneración Nerviosa , Linaje , Pronóstico , Células Ganglionares de la Retina/metabolismo , Homología de Secuencia de Aminoácido , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
8.
Hum Mol Genet ; 24(14): 3948-55, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25901006

RESUMEN

Mitochondrial complex I (CI) deficiencies are causing debilitating neurological diseases, among which, the Leber Hereditary Optic Neuropathy and Leigh Syndrome are the most frequent. Here, we describe the first germinal pathogenic mutation in the NDUFA13/GRIM19 gene encoding a CI subunit, in two sisters with early onset hypotonia, dyskinesia and sensorial deficiencies, including a severe optic neuropathy. Biochemical analysis revealed a drastic decrease in CI enzymatic activity in patient muscle biopsies, and reduction of CI-driven respiration in fibroblasts, while the activities of complex II, III and IV were hardly affected. Western blots disclosed that the abundances of NDUFA13 protein, CI holoenzyme and super complexes were drastically reduced in mitochondrial fractions, a situation that was reproduced by silencing NDUFA13 in control cells. Thus, we established here a correlation between the first mutation yet identified in the NDUFA13 gene, which induces CI instability and a severe but slowly evolving clinical presentation affecting the central nervous system.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Discinesias/genética , Complejo I de Transporte de Electrón/deficiencia , Enfermedades Mitocondriales/genética , Hipotonía Muscular/genética , NADH NADPH Oxidorreductasas/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Niño , Preescolar , Complejo I de Transporte de Electrón/genética , Femenino , Estudios de Seguimiento , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Mutación , NADH NADPH Oxidorreductasas/metabolismo , Sistemas de Lectura Abierta , Linaje
9.
Mol Vis ; 23: 198-209, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28442884

RESUMEN

PURPOSE: Sixteen different mutations in the guanylate cyclase activator 1A gene (GUCA1A), have been previously identified to cause autosomal dominant cone dystrophy (adCOD), cone-rod dystrophy (adCORD), macular dystrophy (adMD), and in an isolated patient, retinitis pigmentosa (RP). The purpose of this study is to report on two novel mutations and the patients' clinical features. METHODS: Clinical investigations included visual acuity and visual field testing, fundus examination, high-resolution spectral-domain optical coherence tomography (OCT), fundus autofluorescence imaging, and full-field and multifocal electroretinogram (ERG) recordings. GUCA1A was screened by Sanger sequencing in a cohort of 12 French families with adCOD, adCORD, and adMD. RESULTS: We found two novel GUCA1A mutations-one amino acid deletion, c.302_304delTAG (p.Val101del), and one missense mutation, c.444T>A (p.Asp148Glu)-each of which was found in one family. The p.Asp148Glu mutation affected one of the Ca2+-binding amino acids of the EF4 hand, while the p.Val101del mutation resulted in the in-frame deletion of Valine-101, localized between two Ca2+-binding aspartic acid residues at positions 100 and 102 of the EF3 hand. Both families complained of visual acuity loss worsening with age. However, the p.Asp148Glu mutation was present in one family with adCOD involving abnormal cone function and an absence of macular atrophy, whereas p.Val101del mutation was encountered in another family with adMD without a generalized cone defect. CONCLUSIONS: The two novel mutations described in this study are associated with distinct phenotypes, MD for p.Val101del and COD for p.Asp148Glu, with no intrafamilial phenotypic heterogeneity.


Asunto(s)
Distrofias de Conos y Bastones/genética , Proteínas Activadoras de la Guanilato-Ciclasa/genética , Degeneración Macular/genética , Mutación Missense , Células Fotorreceptoras Retinianas Conos/patología , Degeneración Retiniana/genética , Eliminación de Secuencia , Adulto , Niño , Distrofias de Conos y Bastones/diagnóstico , Distrofias de Conos y Bastones/fisiopatología , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Genes Dominantes , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/fisiopatología , Masculino , Persona de Mediana Edad , Imagen Óptica , Linaje , Degeneración Retiniana/diagnóstico , Degeneración Retiniana/fisiopatología , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Campos Visuales/fisiología , Adulto Joven
10.
Am J Hum Genet ; 93(3): 571-8, 2013 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-23993198

RESUMEN

Vitelliform macular dystrophies (VMD) are inherited retinal dystrophies characterized by yellow, round deposits visible upon fundus examination and encountered in individuals with juvenile Best macular dystrophy (BMD) or adult-onset vitelliform macular dystrophy (AVMD). Although many BMD and some AVMD cases harbor mutations in BEST1 or PRPH2, the underlying genetic cause remains unknown for many affected individuals. In a large family with autosomal-dominant VMD, gene mapping and whole-exome sequencing led to the identification of a c.713T>G (p.Leu238Arg) IMPG1 mutation, which was subsequently found in two other families with autosomal-dominant VMD and the same phenotype. IMPG1 encodes the SPACR protein, a component of the rod and cone photoreceptor extracellular matrix domains. Structural modeling indicates that the p.Leu238Arg substitution destabilizes the conserved SEA1 domain of SPACR. Screening of 144 probands who had various forms of macular dystrophy revealed three other IMPG1 mutations. Two individuals from one family affected by autosomal-recessive VMD were homozygous for the splice-site mutation c.807+1G>T, and two from another family were compound heterozygous for the mutations c.461T>C (p.Leu154Pro) and c.1519C>T (p.Arg507(∗)). Most cases had a normal or moderately decreased electrooculogram Arden ratio. We conclude that IMPG1 mutations cause both autosomal-dominant and -recessive forms of VMD, thus indicating that impairment of the interphotoreceptor matrix might be a general cause of VMD.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Predisposición Genética a la Enfermedad , Mutación/genética , Proteoglicanos/genética , Distrofia Macular Viteliforme/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos/genética , Proteínas de la Matriz Extracelular/química , Proteínas del Ojo/química , Femenino , Fondo de Ojo , Humanos , Patrón de Herencia/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Fenotipo , Proteoglicanos/química , Adulto Joven
11.
Mol Vis ; 20: 843-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24959063

RESUMEN

PURPOSE: Mutations in genes encoding proteins from the tri-snRNP complex of the spliceosome account for more than 12% of cases of autosomal dominant retinitis pigmentosa (adRP). Although the exact mechanism by which splicing factor defects trigger photoreceptor death is not completely clear, their role in retinitis pigmentosa has been demonstrated by several genetic and functional studies. To test for possible novel associations between splicing factors and adRP, we screened four tri-snRNP splicing factor genes (EFTUD2, PRPF4, NHP2L1, and AAR2) as candidate disease genes. METHODS: We screened up to 303 patients with adRP from Europe and North America who did not carry known RP mutations. Exon-PCR and Sanger methods were used to sequence the NHP2L1 and AAR2 genes, while the sequences of EFTUD2 and PRPF4 were obtained by using long-range PCRs spanning coding and non-coding regions followed by next-generation sequencing. RESULTS: We detected novel missense changes in individual patients in the sequence of the genes PRPF4 and EFTUD2, but the role of these changes in relationship to disease could not be verified. In one other patient we identified a novel nucleotide substitution in the 5' untranslated region (UTR) of NHP2L1, which did not segregate with the disease in the family. CONCLUSIONS: The absence of clearly pathogenic mutations in the candidate genes screened in our cohort suggests that EFTUD2, PRPF4, NHP2L1, and AAR2 are either not involved in adRP or are associated with the disease in rare instances, at least as observed in this study in patients of European and North American origin.


Asunto(s)
Análisis Mutacional de ADN/métodos , Genes Dominantes , Pruebas Genéticas , Empalme del ARN/genética , Retinitis Pigmentosa/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Sistemas de Lectura Abierta/genética , Factores de Elongación de Péptidos/genética , Ribonucleoproteína Nuclear Pequeña U4-U6/genética , Ribonucleoproteína Nuclear Pequeña U5 , Ribonucleoproteínas Nucleares Pequeñas/genética
12.
Ophthalmology ; 121(12): 2406-14, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25085631

RESUMEN

PURPOSE: To assess the frequency of and to characterize the clinical spectrum and optical coherence tomography findings of vitelliform macular dystrophy linked to IMPG1 and IMPG2, 2 new causal genes expressed in the interphotoreceptor matrix. DESIGN: Retrospective epidemiologic, clinical, electrophysiologic, and molecular genetic study. PARTICIPANTS: The database of a national referral center specialized in genetic sensory diseases was screened for patients with a macular vitelliform dystrophy without identified mutation or small deletion or large rearrangement in BEST1 and PRPH2 genes. Forty-nine families were included. METHODS: Clinical, imaging, and electro-oculogram findings were reviewed. Mutation screening of IMPG1 and IMPG2 genes were performed systematically. MAIN OUTCOME MEASURES: Frequency, inheritance, and clinical pattern of vitelliform dystrophy associated with IMPG1 and IMPG2 mutations were characterized. RESULTS: IMPG1 was the causal gene in 3 families (IMPG1 1-3, 11 patients) and IMPG2 in a fourth family (2 patients). With an autosomal dominant transmission, families 1 and 2 had the c.713T→G (p.Leu238Arg) mutation in IMPG1 and family 4 had the c.3230G→T (p.Cys1077Phe) mutation in IMPG2. Patients with IMPG1 or IMPG2 mutations had a late onset and moderate visual impairment (mean visual acuity, 20/40; mean age of onset, 42 years), even in the sporadic case of family 3 with a presumed recessive transmission (age at onset, 38 years; mean visual acuity, 20/50). Drusen-like lesions adjacent to the vitelliform deposits were observed in 9 of 13 patients. The vitelliform material was above the retinal pigment epithelium (RPE) at any stage of the macular dystrophy, and this epithelium was well preserved and maintained its classical reflectivity on spectral-domain optical coherence tomography (SD-OCT). Electro-oculogram results were normal or borderline in 9 cases. CONCLUSIONS: IMPG1 and IMPG2 are new causal genes in 8% of families negative for BEST1 and PRPH2 mutations. These genes should be screened in adult-onset vitelliform dystrophy with (1) moderate visual impairment, (2) drusen-like lesions, (3) normal reflectivity of the RPE line on SD-OCT, and (4) vitelliform deposits located between ellipsoid and interdigitation lines on SD-OCT. These clinical characteristics are not observed in the classical forms of BEST1 or PRPH2 vitelliform dystrophies.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Predisposición Genética a la Enfermedad , Mutación , Proteoglicanos/genética , Distrofia Macular Viteliforme , Adulto , Anciano , Estudios de Casos y Controles , Electrooculografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Epitelio Pigmentado de la Retina/patología , Estudios Retrospectivos , Tomografía de Coherencia Óptica , Agudeza Visual , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/patología , Distrofia Macular Viteliforme/fisiopatología
13.
Mol Vis ; 19: 2487-500, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24339724

RESUMEN

PURPOSE: Autosomal recessive retinitis pigmentosa (arRP) is a genetically heterogeneous disease resulting in progressive loss of photoreceptors that leads to blindness. To date, 36 genes are known to cause arRP, rendering the molecular diagnosis a challenge. The aim of this study was to use homozygosity mapping to identify the causative mutation in a series of inbred families with arRP. METHODS: arRP patients underwent standard ophthalmic examination, Goldman perimetry, fundus examination, retinal OCT, autofluorescence measurement, and full-field electroretinogram. Fifteen consanguineous families with arRP excluded for USH2A and EYS were genotyped on 250 K SNP arrays. Homozygous regions were listed, and known genes within these regions were PCR sequenced. Familial segregation and mutation analyzes were performed. RESULTS: We found ten mutations, seven of which were novel mutations in eight known genes, including RP1, IMPG2, NR2E3, PDE6A, PDE6B, RLBP1, CNGB1, and C2ORF71, in ten out of 15 families. The patients carrying RP1, C2ORF71, and IMPG2 mutations presented with severe RP, while those with PDE6A, PDE6B, and CNGB1 mutations were less severely affected. The five families without mutations in known genes could be a source of identification of novel genes. CONCLUSIONS: Homozygosity mapping combined with systematic screening of known genes results in a positive molecular diagnosis in 66.7% of families.


Asunto(s)
Proteínas del Ojo/genética , Genes Recesivos , Homocigoto , Mutación , Polimorfismo Genético , Retinitis Pigmentosa/genética , Adolescente , Adulto , Consanguinidad , Exones , Femenino , Técnicas de Genotipaje , Humanos , Intrones , Masculino , Persona de Mediana Edad , Linaje , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/patología , Análisis de Secuencia de ADN , Índice de Severidad de la Enfermedad
14.
Genes (Basel) ; 13(9)2022 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-36140676

RESUMEN

Several pathogenic variants have been reported in the IMPG1 gene associated with the inherited retinal disorders vitelliform macular dystrophy (VMD) and retinitis pigmentosa (RP). IMPG1 and its paralog IMPG2 encode for two proteoglycans, SPACR and SPACRCAN, respectively, which are the main components of the interphotoreceptor matrix (IPM), the extracellular matrix surrounding the photoreceptor cells. To determine the role of SPACR in the pathological mechanisms leading to RP and VMD, we generated a knockout mouse model lacking Impg1, the mouse ortholog. Impg1-deficient mice show abnormal accumulation of autofluorescent deposits visible by fundus imaging and spectral-domain optical coherence tomography (SD-OCT) and attenuated electroretinogram responses from 9 months of age. Furthermore, SD-OCT of Impg1-/- mice shows a degeneration of the photoreceptor layer, and transmission electron microscopy shows a disruption of the IPM and the retinal pigment epithelial cells. The decrease in the concentration of the chromophore 11-cis-retinal supports this loss of photoreceptors. In conclusion, our results demonstrate the essential role of SPACR in maintaining photoreceptors. Impg1-/- mice provide a novel model for mechanistic investigations and the development of therapies for VMD and RP caused by IMPG1 pathogenic variants.


Asunto(s)
Proteínas de la Matriz Extracelular , Proteínas del Ojo , Proteoglicanos , Retinitis Pigmentosa , Distrofia Macular Viteliforme , Animales , Matriz Extracelular/genética , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/genética , Proteínas del Ojo/genética , Ratones , Células Fotorreceptoras/patología , Proteoglicanos/genética , Epitelio Pigmentado de la Retina/patología , Pigmentos Retinianos , Retinaldehído , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Distrofia Macular Viteliforme/genética
15.
BMC Med Genet ; 12: 54, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21496248

RESUMEN

BACKGROUND: Rod-cone dystrophy, also known as retinitis pigmentosa (RP), and cone-rod dystrophy (CRD) are degenerative retinal dystrophies leading to blindness. To identify new genes responsible for these diseases, we have studied one large non consanguineous French family with autosomal dominant (ad) CRD. METHODS: Family members underwent detailed ophthalmological examination. Linkage analysis using microsatellite markers and a whole-genome SNP analysis with the use of Affymetrix 250 K SNP chips were performed. Five candidate genes within the candidate region were screened for mutations by direct sequencing. RESULTS: We first excluded the involvement of known adRP and adCRD genes in the family by genotyping and linkage analysis. Then, we undertook a whole-genome scan on 22 individuals in the family. The analysis revealed a 41.3-Mb locus on position 2q24.2-2q33.1. This locus was confirmed by linkage analysis with specific markers of this region. The maximum LOD score was 2.86 at θ = 0 for this locus. Five candidate genes, CERKL, BBS5, KLHL23, NEUROD1, and SF3B1 within this locus, were not mutated. CONCLUSION: A novel locus for adCRD, named CORD12, has been mapped to chromosome 2q24.2-2q33.1 in a non consanguineous French family.


Asunto(s)
Cromosomas Humanos Par 2 , Genes Dominantes , Mapeo Cromosómico , Femenino , Ligamiento Genético , Humanos , Masculino , Repeticiones de Microsatélite/genética , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Retinitis Pigmentosa/genética
16.
Ophthalmology ; 118(6): 1130-6, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21269699

RESUMEN

PURPOSE: To evaluate a genetic approach of BEST1 and PRPH2 screening according to age of onset, family history, and Arden ratio in patients with juvenile vitelliform macular dystrophy (VMD2) or adult-onset vitelliform macular dystrophy (AVMD), which are characterized by autofluorescent deposits. DESIGN: Clinical, electrophysiologic, and molecular retrospective study. PARTICIPANTS: The database of a clinic specialized in genetic sensory diseases was screened for patients with macular vitelliform dystrophy. Patients with an age of onset less than 40 years were included in the VMD2 group (25 unrelated patients), and patients with an age of onset more than 40 years were included in the AVMD group (19 unrelated patients). METHODS: Clinical, fundus photography, and electro-oculogram (EOG) findings were reviewed. Mutation screening of BEST1 and PRPH2 genes was systematically performed. MAIN OUTCOME MEASURES: Relevance of age of onset, family history, and Arden ratio were reviewed. RESULTS: Patients with VMD2 carried a BEST1 mutation in 60% of the cases. Seven novel mutations in BEST1 (p.V9L, p.F80V, p.I73V, p.R130S, pF298C, pD302A, and p.179delN) were found. Patients with VMD2 with a positive family history or a reduced Arden ratio carried a BEST1 mutation in 70.5% of cases and in 83% if both criteria were fulfilled. Patients with AVMD carried a PRPH2 mutation in 10.5% of cases and did not carry a BEST1 mutation. The probability of finding a PRPH2 mutation increased in the case of a family history (2/5 patients). Electro-oculogram was normal in 3 of 15 patients with BEST1 mutations and reduced in the 3 patients with PRPH2 mutations. CONCLUSIONS: Age of onset is a major criterion to distinguish VMD2 from AVMD. Electro-oculogram is not as relevant because decreased or normal Arden ratios have been associated with mutations in both genes and diseases. A positive family history increased the probability of finding a mutation. BEST1 screening should be recommended to patients with an age of onset less than 40 years, and PRPH2 screening should be recommended to patients with an age of onset more than 40 years. For an onset between 30 and 40 years, PRPH2 can be screened if no mutation has been detected in BEST1. FINANCIAL DISCLOSURE(S): The author(s) have no proprietary or commercial interest in any materials discussed in this article.


Asunto(s)
Canales de Cloruro/genética , ADN/genética , Proteínas del Ojo/genética , Proteínas de Filamentos Intermediarios/genética , Glicoproteínas de Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Distrofia Macular Viteliforme/genética , Adulto , Anciano , Bestrofinas , Canales de Cloruro/metabolismo , Análisis Mutacional de ADN , Electrooculografía , Proteínas del Ojo/metabolismo , Femenino , Angiografía con Fluoresceína , Fondo de Ojo , Pruebas Genéticas , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Persona de Mediana Edad , Proteínas del Tejido Nervioso/metabolismo , Linaje , Periferinas , Estudios Retrospectivos , Distrofia Macular Viteliforme/diagnóstico , Distrofia Macular Viteliforme/fisiopatología
17.
Mitochondrion ; 59: 169-174, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34023438

RESUMEN

Mitochondrial complex I (CI) deficiencies (OMIM 252010) are the commonest inherited mitochondrial disorders in children. Acyl-CoA dehydrogenase 9 (ACAD9) is a flavoenzyme involved chiefly in CI assembly and possibly in fatty acid oxidation. Biallelic pathogenic variants result in CI dysfunction, with a phenotype ranging from early onset and sometimes fatal mitochondrial encephalopathy with lactic acidosis to late-onset exercise intolerance. Cardiomyopathy is often associated. We report a patient with childhood-onset optic and peripheral neuropathy without cardiac involvement, related to CI deficiency. Genetic analysis revealed compound heterozygous pathogenic variants in ACAD9, expanding the clinical spectrum associated to ACAD9 mutations. Importantly, riboflavin treatment (15 mg/kg/day) improved long-distance visual acuity and demonstrated significant rescue of CI activity in vitro.


Asunto(s)
Acil-CoA Deshidrogenasas/genética , Mutación del Sistema de Lectura , Enfermedades del Nervio Óptico/tratamiento farmacológico , Riboflavina/administración & dosificación , Edad de Inicio , Niño , Heterocigoto , Humanos , Masculino , Enfermedades del Nervio Óptico/genética , Riboflavina/uso terapéutico , Resultado del Tratamiento
18.
J Clin Invest ; 130(1): 143-156, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31550237

RESUMEN

Mutations in genes encoding components of the mitochondrial DNA (mtDNA) replication machinery cause mtDNA depletion syndromes (MDSs), which associate ocular features with severe neurological syndromes. Here, we identified heterozygous missense mutations in single-strand binding protein 1 (SSBP1) in 5 unrelated families, leading to the R38Q and R107Q amino acid changes in the mitochondrial single-stranded DNA-binding protein, a crucial protein involved in mtDNA replication. All affected individuals presented optic atrophy, associated with foveopathy in half of the cases. To uncover the structural features underlying SSBP1 mutations, we determined a revised SSBP1 crystal structure. Structural analysis suggested that both mutations affect dimer interactions and presumably distort the DNA-binding region. Using patient fibroblasts, we validated that the R38Q variant destabilizes SSBP1 dimer/tetramer formation, affects mtDNA replication, and induces mtDNA depletion. Our study showing that mutations in SSBP1 cause a form of dominant optic atrophy frequently accompanied with foveopathy brings insights into mtDNA maintenance disorders.


Asunto(s)
ADN Mitocondrial/genética , Proteínas de Unión al ADN/genética , Proteínas Mitocondriales/genética , Mutación Missense , Atrofia Óptica Autosómica Dominante/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Replicación del ADN , Proteínas de Unión al ADN/química , Femenino , GTP Fosfohidrolasas/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/química , Atrofia Óptica Autosómica Dominante/etiología , Secuenciación del Exoma
19.
Stem Cell Res ; 38: 101478, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31203166

RESUMEN

We generated an induced pluripotent stem cell (iPSC) line using dermal fibroblasts from a 53 year-old patient with autosomal dominant cone-rod dystrophy (CRD) caused by a missense mutation, c.121C > T, in the CRX gene. Patient fibroblasts were reprogrammed using the non-integrative Sendai virus reprogramming system and the human OSKM transcription factor cocktail. The generated iPSCs contained the congenital mutation in exon 3 of CRX and were pluripotent and genetically stable. This iPSC line will be an important tool for retinal differentiation studies to better understand the CRD phenotype caused by the mutant p.Arg41Trp CRX protein.


Asunto(s)
Técnicas de Reprogramación Celular , Distrofias de Conos y Bastones , Fibroblastos/metabolismo , Proteínas de Homeodominio , Células Madre Pluripotentes Inducidas/metabolismo , Mutación Missense , Transactivadores , Sustitución de Aminoácidos , Línea Celular , Distrofias de Conos y Bastones/genética , Distrofias de Conos y Bastones/metabolismo , Distrofias de Conos y Bastones/patología , Fibroblastos/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/patología , Masculino , Persona de Mediana Edad , Transactivadores/genética , Transactivadores/metabolismo
20.
Stem Cell Res ; 38: 101476, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31247521

RESUMEN

The human induced pluripotent stem cell (iPSC) line, INMi004-A, was generated using dermal fibroblasts from a 6 year-old patient with autosomal dominant Leber Congenital Amaurosis (LCA) caused by the point mutation c.695delC (p.Pro232Argfs*139) in the CRX gene. We used non-integrative Sendai virus vectors containing the human OSKM transcription factor cocktail to reprogram patient fibroblasts. The generated iPSC line contained the congenital deletion c.695delC in exon 4 of CRX, had a normal karyotype, and was capable of differentiation into all three germ layers. This cell line represents an important tool to study the pathophysiology of CRX-associated LCA.


Asunto(s)
Secuencia de Bases , Fibroblastos , Proteínas de Homeodominio , Células Madre Pluripotentes Inducidas , Amaurosis Congénita de Leber , Mutación Puntual , Eliminación de Secuencia , Transactivadores , Línea Celular , Fibroblastos/metabolismo , Fibroblastos/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Amaurosis Congénita de Leber/patología , Transactivadores/genética , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA