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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 11(2): e0148678, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26872363

RESUMEN

The significance of the ubiquitin-proteasome system (UPS) for protein degradation has been highlighted in the context of neurodegenerative diseases, including retinal dystrophies. TOPORS, a dual E3 ubiquitin and SUMO1 ligase, forms a component of the UPS and selected substrates for its enzymatic activities, such as DJ-1/PARK7 and APOBEC2, are important for neuronal as well as retinal homeostasis, respectively. TOPORS is ubiquitously expressed, yet its mutations are only known to result in autosomal dominant retinitis pigmentosa. We performed a yeast two-hybrid (Y2H) screen of a human retinal cDNA library in order to identify interacting protein partners of TOPORS from the retina, and thus begin delineating the putative disease mechanism(s) associated with the retina-specific phenotype resulting from mutations in TOPORS. The screen led to isolation of the 26 S protease regulatory subunit 4 (P26s4/ PSMC1), an ATPase indispensable for correct functioning of UPS-mediated proteostasis. The interaction between endogenous TOPORS and P26s4 proteins was validated by co-immuno-precipitation from mammalian cell extracts and further characterised by immunofluorescent co-localisation studies in cell lines and retinal sections. Findings from hTERT-RPE1 and 661W cells demonstrated that TOPORS and P26s4 co-localise at the centrosome in cultured cells. Immunofluorescent staining of mouse retinae revealed a strong P26s4 reactivity at the interface between retinal pigmented epithelium (RPE) layer and the photoreceptors outer segments (OS). This finding leads us to speculate that P26s4, along with TOPORS, may have a role(s) in RPE phagocytosis, in addition to contributing to the overall photoreceptor and retinal homeostasis via the UPS.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Mapas de Interacción de Proteínas , Retina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/análisis , Adenosina Trifosfatasas/genética , Animales , Línea Celular , ADN Complementario/genética , Humanos , Ratones , Mutación , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Retina/citología , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/análisis , Ubiquitina-Proteína Ligasas/genética
2.
Mol Vis ; 20: 1307-17, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25352739

RESUMEN

PURPOSE: To describe the genotype-phenotype correlation and serial observations in a five-generation Czech family with X-linked retinitis pigmentosa (XLRP) associated with severe visual impairment in women. METHODS: Comprehensive ophthalmological examination including spectral domain optical coherence tomography (SD-OCT) was performed. Based on the pedigree structure and women being severely affected, autosomal dominant inheritance was suspected, and screening for known mutations by genotyping microarray was performed. Subsequently, direct sequencing of ORF15 RPGR was undertaken. RESULTS: Eighteen family members (nine women and nine men) were examined. A pathogenic variant, c.2543del in ORF15 of RPGR, was found to segregate with disease. The oldest woman and her two sisters had no perception of light in their sixth decade. Four women and five men had signs and symptoms of typical XLRP, including moderate to high myopia. Three other women also had moderate to high myopia and myopic astigmatism but without the presence of bone spicule-like formation. Severe disruption of macular architecture on SD-OCT was equally common in both sexes. Only one 32-year-old female carrier had clinically normal findings. Subfoveal choroidal thickness was decreased in all affected men and in all female carriers, except the only carrier with a normal fundus examination. CONCLUSIONS: The c.2543del mutation in ORF15 of RPGR is associated with a severe phenotype in the women in this family. The presence of a significant myopic refractive error, in the absence of male-to-male transmission, may be indicative of X-linked inheritance. Measurements of choroidal thickness may help in clinically identifying carrier status.


Asunto(s)
Secuencia de Bases , Proteínas del Ojo/genética , Genes Ligados a X , Retina/metabolismo , Retinitis Pigmentosa/genética , Eliminación de Secuencia , Adolescente , Adulto , Anciano , Niño , Coroides/metabolismo , Coroides/patología , Femenino , Genes Dominantes , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Linaje , Retina/patología , Retinitis Pigmentosa/patología , Índice de Severidad de la Enfermedad , Factores Sexuales , Tomografía de Coherencia Óptica
3.
Hum Mol Genet ; 23(21): 5827-37, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899048

RESUMEN

Neurodegenerative diseases affecting the macula constitute a major cause of incurable vision loss and exhibit considerable clinical and genetic heterogeneity, from early-onset monogenic disease to multifactorial late-onset age-related macular degeneration (AMD). As part of our continued efforts to define genetic causes of macular degeneration, we performed whole exome sequencing in four individuals of a two-generation family with autosomal dominant maculopathy and identified a rare variant p.Glu1144Lys in Fibrillin 2 (FBN2), a glycoprotein of the elastin-rich extracellular matrix (ECM). Sanger sequencing validated the segregation of this variant in the complete pedigree, including two additional affected and one unaffected individual. Sequencing of 192 maculopathy patients revealed additional rare variants, predicted to disrupt FBN2 function. We then undertook additional studies to explore the relationship of FBN2 to macular disease. We show that FBN2 localizes to Bruch's membrane and its expression appears to be reduced in aging and AMD eyes, prompting us to examine its relationship with AMD. We detect suggestive association of a common FBN2 non-synonymous variant, rs154001 (p.Val965Ile) with AMD in 10 337 cases and 11 174 controls (OR = 1.10; P-value = 3.79 × 10(-5)). Thus, it appears that rare and common variants in a single gene--FBN2--can contribute to Mendelian and complex forms of macular degeneration. Our studies provide genetic evidence for a key role of elastin microfibers and Bruch's membrane in maintaining blood-retina homeostasis and establish the importance of studying orphan diseases for understanding more common clinical phenotypes.


Asunto(s)
Estudios de Asociación Genética , Variación Genética , Degeneración Macular/genética , Proteínas de Microfilamentos/genética , Adulto , Anciano , Secuencia de Aminoácidos , Lámina Basal de la Coroides/metabolismo , Análisis Mutacional de ADN , Exoma , Matriz Extracelular/metabolismo , Fibrilina-2 , Fibrilinas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Degeneración Macular/diagnóstico , Masculino , Metaanálisis como Asunto , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Linaje , Conformación Proteica , Estabilidad Proteica , Retina/metabolismo , Retina/patología , Alineación de Secuencia
4.
Hum Mol Genet ; 22(8): 1507-15, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23297361

RESUMEN

Ataxia-telangiectasia and Rad3 (ATR), a sensor of DNA damage, is associated with the regulation and control of cell division. ATR deficit is known to cause Seckel syndrome, characterized by severe proportionate short stature and microcephaly. We used a mouse model for Seckel disease to study the effect of ATR deficit on retinal development and function and we have found a new role for ATR, which is critical for the postnatal development of the photoreceptor (PR) layer in mouse retina. The structural and functional characterization of the ATR(+/s) mouse retinas displayed a specific, severe and early degeneration of rod and cone cells resembling some characteristics of human retinal degenerations. A new localization of ATR in the cilia of PRs and the fact that mutant mice have shorter cilia suggests that the PR degeneration here described results from a ciliary defect.


Asunto(s)
Proteínas de Ciclo Celular/genética , Células Fotorreceptoras de Vertebrados , Proteínas Serina-Treonina Quinasas/genética , Retina/metabolismo , Degeneración Retiniana/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Enanismo/genética , Enanismo/patología , Facies , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Microcefalia/genética , Microcefalia/patología , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Cilio Conector de los Fotorreceptores/metabolismo , Cilio Conector de los Fotorreceptores/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Retina/crecimiento & desarrollo , Degeneración Retiniana/patología
5.
J Mol Genet Med ; 7(2)2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25729402

RESUMEN

Mutations in PRPF31 have been implicated in retinitis pigmentosa, a blinding disease caused by degeneration of rod photoreceptors. The disease mechanism in the majority of cases is haploinsufficiency. Crucially, attempts at generation of animal models of disease have proved unsuccessful, yielding animals with a visual phenotype that does not mirror human disease. This suggests that, in these animals, the transcriptional regulation of PRPF31 is different to humans and compared to other species. Study of the evolution of the PRPF31 core promoter has important implications for our understanding of human disease, as disease phenotype is modified by differentially expressed alleles in the population. PRPF31 lies in a head-to-head arrangement with TFPT, a gene involved in cellular apoptosis. The two genes were shown to share common regulatory elements in the human genome. In this study, the core promoters of PRPF31 and TFPT were characterised by dual-luciferase reporter assay using genomic DNA from the green monkey, domestic dog and house mouse. It was found that the core promoters were conserved between human and monkey. In dog, the TFPT core promoter was conserved, but different PRPF31 gene architecture meant the gene was controlled by a long-range promoter lying some 2000bp from the transcription start site. There was very low level of conservation (<20%) of the PRPF31 5' region between mouse and human. It was shown that mouse populations did not show variable Prpf31 expression levels, revealing a potential explanation for the lack of phenotype observed in the Prpf31 knock-out mouse model.

6.
Hum Mol Genet ; 21(18): 4126-37, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22723017

RESUMEN

PRPF31, a gene located at chromosome 19q13.4, encodes the ubiquitous splicing factor PRPF31. The gene lies in a head-to-head arrangement with TFPT, a poorly characterized gene with a role in cellular apoptosis. Mutations in PRPF31 have been implicated in autosomal dominant retinitis pigmentosa (adRP), a frequent and important cause of blindness worldwide. Disease associated with PRPF31 mutations is unusual, in that there is often non-penetrance of the disease phenotype in affected families, caused by differential expression of PRPF31. This study aimed to characterize the basic promoter elements of PRPF31 and TFPT. Luciferase reporter constructs were made, using genomic DNA from an asymptomatic individual with a heterozygous deletion of the entire putative promoter region. Fragments were tested by the dual-luciferase reporter assay in HeLa and RPE-1 cell lines. A comparison was made between the promoter regions of symptomatic and asymptomatic mutation-carrying individuals. A patient (CAN493) with adRP was identified, harbouring a regulatory region mutation; both alleles were assayed by the dual-luciferase reporter assay. Luciferase assays led to the identification of core promoters for both PRPF31 and TFPT; despite their shared gene architecture, the two genes appear to be controlled by slightly different regulatory regions. One functional polymorphism was identified in the PRPF31 promoter that increased transcriptional activation. The change was not, however, consistent with the observed symptomatic-asymptomatic phenotypes in a family affected by PRPF31-adRP. Analysis of the mutant promoter fragment from CAN493 showed a >50% reduction in promoter activity, suggesting a disease mechanism of functional haploinsufficiency-the first report of this disease mechanism in adRP.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas del Ojo/genética , Regulación de la Expresión Génica , Retinitis Pigmentosa/genética , Transcripción Genética , Anciano , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Estudios de Casos y Controles , Clonación Molecular , Secuencia Conservada , Análisis Mutacional de ADN , Proteínas del Ojo/metabolismo , Femenino , Genes Dominantes , Genes Reporteros , Estudios de Asociación Genética , Células HeLa , Humanos , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Masculino , Datos de Secuencia Molecular , Filogenia , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Eliminación de Secuencia , Estadísticas no Paramétricas
7.
Mol Vis ; 18: 581-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22419850

RESUMEN

PURPOSE: To identify and functionally characterize the mutation responsible for autosomal dominant retinitis pigmentosa (adRP) in a large, six-generation French family. METHODS: Twenty individuals from this family participated in the genetic investigation. Six affected and 14 unaffected individuals from three-generations were available for linkage analysis using microsatellite markers flanking the rhodopsin (RHO) gene. A two-point logarithm of odds (LOD) score calculation was undertaken using GENEMARKER and MLINK software. Sanger sequencing of RHO was performed. Cellular localization of the mutant protein was performed by transforming SK-N-SH cells with pEGFP-N1-Rho, pEGFP-N1-Rho(P23H), and pEGFP-N1-Rho(c.614-622del). RESULTS: The proband had nyctalopia, visual field constriction, peripheral bone spicule pigmentation of the fundus, central acuity (6/24 RE; 6/12 LE) at 55 years of age. Linkage analysis of this family suggested RHO as a possible candidate since the flanking marker D3S1292 yielded a LOD score of 2.43 at θ=0. Cloning of an exon 3 PCR product and direct sequencing of single clones identified a novel deletion in the third exon of RHO, c.614-622del (p.Y206-F208del). The deleted mutant protein localized to the endoplasmic reticulum and formed inclusion bodies. CONCLUSIONS: This novel deletion in exon 3 of the RHO gene, c.614-622del results in a classical form of adRP in a multi-generation French family. Protein expression analyses confirmed that the deletion led to protein misfolding and suggest this is a class II mutation, similar to P23H, the most common class II mutation seen in North America.


Asunto(s)
Secuencia de Bases , Deficiencias en la Proteostasis/genética , Retinitis Pigmentosa/genética , Rodopsina/genética , Eliminación de Secuencia/genética , Población Blanca/genética , Adolescente , Estudios de Casos y Controles , Línea Celular , Exones , Femenino , Francia , Genes Dominantes , Ligamiento Genético , Humanos , Repeticiones de Microsatélite , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Plásmidos , Análisis de Secuencia de ADN , Transformación Genética
8.
Hum Mol Genet ; 20(5): 975-87, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21159800

RESUMEN

We recently reported that mutations in the widely expressed nuclear protein TOPORS (topoisomerase I-binding arginine/serine rich) are associated with autosomal dominant retinal degeneration. However, the precise localization and a functional role of TOPORS in the retina remain unknown. Here, we demonstrate that TOPORS is a novel component of the photoreceptor sensory cilium, which is a modified primary cilium involved with polarized trafficking of proteins. In photoreceptors, TOPORS localizes primarily to the basal bodies of connecting cilium and in the centrosomes of cultured cells. Morpholino-mediated silencing of topors in zebrafish embryos demonstrates in another species a comparable retinal problem as seen in humans, resulting in defective retinal development and failure to form outer segments. These defects can be rescued by mRNA encoding human TOPORS. Taken together, our data suggest that TOPORS may play a key role in regulating primary cilia-dependent photoreceptor development and function. Additionally, it is well known that mutations in other ciliary proteins cause retinal degeneration, which may explain why mutations in TOPORS result in the same phenotype.


Asunto(s)
Centrosoma/metabolismo , Cilios/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Degeneración Retiniana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Cilios/genética , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Células Fotorreceptoras/metabolismo , Transporte de Proteínas , Retina/metabolismo , Degeneración Retiniana/genética , Ubiquitina-Proteína Ligasas/genética , Pez Cebra
10.
Nat Rev Genet ; 11(4): 273-84, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20212494

RESUMEN

The retina provides exquisitely sensitive vision that relies on the integrity of a uniquely vulnerable cell, the photoreceptor (PR). The genetic and mechanistic causes of retinal degeneration due to PR cell death--which occurs in conditions such as retinitis pigmentosa and age-related macular degeneration--are being successfully dissected. Over one hundred loci, some containing common variants but most containing rare variants, are implicated in the genetic architecture of this complex trait. This genetic heterogeneity results in equally diverse disease mechanisms that affect almost every aspect of PR function but converge on a common cell death pathway. Although genetic and mechanistic diversity creates challenges for therapy, some approaches--particularly gene-replacement therapy--are showing considerable promise.


Asunto(s)
Degeneración Retiniana/genética , Muerte Celular/genética , Activación de Complemento/genética , Metabolismo Energético , Humanos , Inflamación/genética , Peroxidación de Lípido/genética , Células Fotorreceptoras de Vertebrados/patología , Células Fotorreceptoras de Vertebrados/fisiología , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Procesamiento Postranscripcional del ARN , Degeneración Retiniana/patología , Degeneración Retiniana/terapia , Estrés Fisiológico
11.
Invest Ophthalmol Vis Sci ; 50(12): 5927-33, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19578015

RESUMEN

PURPOSE: Pre-mRNA processing factor 31 (PRPF31) is a ubiquitous protein needed for the assembly of the pre-mRNA splicing machinery. It has been shown that mutations in this gene cause autosomal dominant retinitis pigmentosa 11 (RP11), which is characterized by rod-cell degeneration. Interestingly, mutations in this ubiquitously expressed gene do not lead to phenotypes other than retinal malfunction. Furthermore, the dominant inheritance pattern has shown incomplete penetrance, which poses interesting questions about the disease mechanism of RP11. METHODS: To characterize PRPF31 function in the rod cells, two animal models have been generated. One was a heterozygous knock-in mouse (Prpf31(A216P/+)) carrying a point mutation p.A216P, which has previously been identified in RP11 patients. The second was a heterozygous knockout mouse (Prpf31(+/-)). Retinal degeneration in RP11 mouse models was monitored by electroretinography and histology. RESULTS: Generation of the mouse models is presented, as are results of ERGs and retinal morphology. No degenerative phenotype on fundus examination was found in Prpf31(A216P/+) and Prpf31(+/-) mice. Prpf31(A216P/A216P) and Prpf31(-/-) genotypes were embryonic lethal. CONCLUSIONS: The results imply that Prpf31 is necessary for survival, and there is no compensation mechanism in mouse for the lack of this splicing factor. The authors suggest that p.A216P mutation in Prpf31 does not exert a dominant negative effect and that one Prpf31 wild-type allele is sufficient for maintenance of the healthy retina in mice.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Genes Dominantes , Retinitis Pigmentosa/genética , Animales , Western Blotting , Electroforesis en Gel de Poliacrilamida , Electrorretinografía , Femenino , Técnicas de Sustitución del Gen , Técnicas de Inactivación de Genes , Marcación de Gen , Genotipo , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Oftalmoscopía , Mutación Puntual , Retina/fisiopatología , Retinitis Pigmentosa/fisiopatología
12.
Am J Hum Genet ; 84(6): 792-800, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19520207

RESUMEN

Retinitis pigmentosa (RP) refers to a genetically heterogeneous group of progressive neurodegenerative diseases that result in dysfunction and/or death of rod and cone photoreceptors in the retina. So far, 18 genes have been identified for autosomal-dominant (ad) RP. Here, we describe an adRP locus (RP42) at chromosome 7p15 through linkage analysis in a six-generation Scandinavian family and identify a disease-causing mutation, c.449G-->A (p.S150N), in exon 6 of the KLHL7 gene. Mutation screening of KLHL7 in 502 retinopathy probands has revealed three different missense mutations in six independent families. KLHL7 is widely expressed, including expression in rod photoreceptors, and encodes a 75 kDa protein of the BTB-Kelch subfamily within the BTB superfamily. BTB-Kelch proteins have been implicated in ubiquitination through Cullin E3 ligases. Notably, all three putative disease-causing KLHL7 mutations are within a conserved BACK domain; homology modeling suggests that mutant amino acid side chains can potentially fill the cleft between two helices, thereby affecting the ubiquitination complexes. Mutations in an identical region of another BTB-Kelch protein, gigaxonin, have previously been associated with giant axonal neuropathy. Our studies suggest an additional role of the ubiquitin-proteasome protein-degradation pathway in maintaining neuronal health and in disease.


Asunto(s)
Autoantígenos/genética , Genes Dominantes , Mutación Missense/genética , Polimorfismo de Nucleótido Simple/genética , Retinitis Pigmentosa/genética , Secuencia de Aminoácidos , Autoantígenos/metabolismo , Cromosomas Humanos Par 7/genética , Ensayo de Inmunoadsorción Enzimática , Perfilación de la Expresión Génica , Ligamiento Genético , Humanos , Immunoblotting , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
13.
Nat Genet ; 41(6): 739-45, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19430481

RESUMEN

Despite rapid advances in the identification of genes involved in disease, the predictive power of the genotype remains limited, in part owing to poorly understood effects of second-site modifiers. Here we demonstrate that a polymorphic coding variant of RPGRIP1L (retinitis pigmentosa GTPase regulator-interacting protein-1 like), a ciliary gene mutated in Meckel-Gruber (MKS) and Joubert (JBTS) syndromes, is associated with the development of retinal degeneration in individuals with ciliopathies caused by mutations in other genes. As part of our resequencing efforts of the ciliary proteome, we identified several putative loss-of-function RPGRIP1L mutations, including one common variant, A229T. Multiple genetic lines of evidence showed this allele to be associated with photoreceptor loss in ciliopathies. Moreover, we show that RPGRIP1L interacts biochemically with RPGR, loss of which causes retinal degeneration, and that the Thr229-encoded protein significantly compromises this interaction. Our data represent an example of modification of a discrete phenotype of syndromic disease and highlight the importance of a multifaceted approach for the discovery of modifier alleles of intermediate frequency and effect.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Variación Genética , Degeneración Retiniana/genética , Alelos , Animales , Síndrome de Bardet-Biedl/genética , Cuerpo Ciliar/fisiopatología , Europa (Continente)/epidemiología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Humanos , Mutación , Polimorfismo de Nucleótido Simple , ARN Mensajero/genética , Degeneración Retiniana/epidemiología , Degeneración Retiniana/prevención & control , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/genética , Uveítis/epidemiología , Uveítis/genética , Pez Cebra/genética
14.
Ophthalmic Genet ; 30(2): 96-8, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19373681

RESUMEN

Mutations in TOPORS cause autosomal dominant retinitis pigmentosa (adRP). Examination of 160 adRP patients from continental Europe revealed nine exonic single nucleotide variants, eight of which reside in the coding region; three synonymous single nucleotide polymorphisms (SNPs; c.2319T > C, c.2991T > C and c.1560A > G), three nonsynonymous SNPs (c.58C > T/p.P20S, c.74C >G/p.S25W and c.1730C > A/p.S577Y) and two novel missense mutations (c.1205A > C/p.Q402P and c.1818T > G/p.S606R). Whether the latter two variants represent adRP causing mutations awaits further analysis.


Asunto(s)
Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Retinitis Pigmentosa/genética , Ubiquitina-Proteína Ligasas/genética , Europa (Continente) , Genes Dominantes , Humanos , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Polimorfismo Conformacional Retorcido-Simple , ARN Mensajero/metabolismo
15.
Mol Vis ; 14: 683-90, 2008 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-18431455

RESUMEN

PURPOSE: Missense mutations in the splicing factor gene PRPF31 cause a dominant form of retinitis pigmentosa (RP11) with reduced penetrance. Missense mutations in PRPF31 have previously been shown to cause reduced protein solubility, suggesting insufficiency of functional protein as the disease mechanism. Here we examine in further detail the effect of the A216P mutation on splicing function. METHODS: Splicing activity was assayed using an in vivo assay in transfected mammalian cells with rhodopsin (RHO) and transducin (GNAT1) splicing templates. Pull-down assays were used to study the interaction between PRPF31 and one of its cognate partners in the spliceosome, PRPF6. RESULTS: Splicing of RHO intron 3 and GNAT1 introns 3-5 mini-gene templates was inefficient with both spliced and unspliced products clearly detected. Assays using the RHO minigene template revealed a direct negative effect on splicing efficiency of the mutant. However, no effect of the mutation on splicing efficiency could be detected using the longer GNAT1 minigene template or using a full-length RHO transcript, splicing of which had an efficiency of 100%. No unspliced RHO transcripts could be detected in RNA from human retina. Pull-down assays between PRPF31 and PRPF6 proteins showed a stronger interaction for the mutant than wild type, suggesting a mechanism for the negative effect. CONCLUSIONS: Splicing of full-length RHO is more efficient than splicing of the minigene, and assays using a full-length template more accurately mimic splicing in photoreceptors. The RP11 missense mutations exert their pathology mainly via a mechanism based on protein insufficiency due to protein insolubility, but there is also a minor direct negative effect on function.


Asunto(s)
Proteínas del Ojo/genética , Mutación Missense/genética , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología , Línea Celular , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Intrones/genética , Modelos Genéticos , Proteínas Mutantes/metabolismo , Unión Proteica , Precursores del ARN/genética , Empalme del ARN/genética , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética , Retina/patología , Opsinas de Bastones/genética , Moldes Genéticos , Factores de Transcripción/genética , Transducina , Transfección
16.
Am J Hum Genet ; 81(5): 1098-103, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17924349

RESUMEN

We report mutations in the gene for topoisomerase I-binding RS protein (TOPORS) in patients with autosomal dominant retinitis pigmentosa (adRP) linked to chromosome 9p21.1 (locus RP31). A positional-cloning approach, together with the use of bioinformatics, identified TOPORS (comprising three exons and encoding a protein of 1,045 aa) as the gene responsible for adRP. Mutations that include an insertion and a deletion have been identified in two adRP-affected families--one French Canadian and one German family, respectively. Interestingly, a distinct phenotype is noted at the earlier stages of the disease, with an unusual perivascular cuff of retinal pigment epithelium atrophy, which was found surrounding the superior and inferior arcades in the retina. TOPORS is a RING domain-containing E3 ubiquitin ligase and localizes in the nucleus in speckled loci that are associated with promyelocytic leukemia bodies. The ubiquitous nature of TOPORS expression and a lack of mutant protein in patients are highly suggestive of haploinsufficiency, rather than a dominant negative effect, as the molecular mechanism of the disease and make rescue of the clinical phenotype amenable to somatic gene therapy.


Asunto(s)
Genes Dominantes , Mutación/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Epitelio Pigmentado Ocular/irrigación sanguínea , Epitelio Pigmentado Ocular/patología , Retinitis Pigmentosa/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Cromosomas Humanos , Análisis Mutacional de ADN , Exones/genética , Femenino , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Linaje , Ubiquitina-Proteína Ligasas/metabolismo
17.
Mol Vis ; 12: 909-14, 2006 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-16917484

RESUMEN

PURPOSE: To identify the disease-causing mutations in two large Bulgarian Romani (Gypsy) pedigrees: one with autosomal dominant retinitis pigmentosa (adRP) with partial penetrance and the other with severe X-linked RP (xlRP). METHODS: Detailed clinical investigations were undertaken and genomic DNA was extracted from blood samples. DNA was analyzed by PCR amplification with gene-specific primers and direct genomic sequencing. RESULTS: Analysis of the complete coding sequence of PRPF31 in the adRP family led to the identification of a new heterozygous splice site mutation IVS6+1G>T. RPGR mutation screening in affected male individuals in the X-linked RP family identified a hemizygous c.ORF15+652_653delAG mutation. Interestingly this mutation was found in a homozygous state in one severely affected female from the family. CONCLUSIONS: In this first report of molecular genetic analysis of retinitis pigmentosa in Romani families, we describe a novel PRPF31 mutation and present the first case of a homozygous mutation in the RPGR gene in a female individual.


Asunto(s)
Proteínas del Ojo/genética , Biología Molecular , Mutación , Retinitis Pigmentosa/genética , Romaní/genética , Adolescente , Adulto , Secuencia de Bases , Cromosomas Humanos X , Femenino , Genes Dominantes , Ligamiento Genético , Guanina , Homocigoto , Humanos , Intrones , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Penetrancia , Timina
18.
Hum Genet ; 118(3-4): 501-3, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16189705

RESUMEN

Retinitis pigmentosa (RP) is a debilitating disease of the retina affecting approximately 1.5 million people worldwide. RP shows remarkable heterogeneity both clinically and genetically, with more than 40 genetic loci implicated, 12 of which account for the autosomal dominant form (adRP) of inheritance. We have recently identified a French Canadian family that presents with early onset adRP. After exclusion of all known loci for adRP, a genome-wide search established firm linkage with a marker from the short arm of chromosome 9 (LOD score of 6.3 at recombination fraction theta=0). The linked region is flanked by markers D9S285 and D9S1874, corresponding to a genetic distance of 31 cM, in the region 9p22-p13.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 9 , Retinitis Pigmentosa/genética , Edad de Inicio , Ligamiento Genético , Humanos , Patrón de Herencia , Linaje
19.
Psychiatr Genet ; 14(2): 101-6, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15167697

RESUMEN

Several studies provide suggestive evidence of a susceptibility locus for bipolar disorder at chromosome 21q22-23. In an attempt to replicate these findings, we have analyzed linkage to 11 polymorphic markers from this region in 18 Bulgarian pedigrees with affective disorder. Two-point linkage analysis under assumption of homogeneity and a dominant model with reduced penetrance produced modest positive values for some of the markers tested under a 'narrow' phenotype definition, including bipolar I and II, and schizoaffective disorder. The maximum two-point score (lod=1.76, theta=0.00) was at marker D21S1919. Non-parametric linkage analysis under the same phenotype model, yielded positive NPLall values (P<0.05) over the region between markers D21S211 and D21S416, with a peak at D21S1252 (NPL Zall=2.32, P=0.0003). The multipoint lod score (GENEHUNTER) reached a suggestive value for linkage (lod=2.10) also at marker D21S1252. The results under a recessive model were completely negative. These data add to the evidence for the existence of a susceptibility locus for bipolar affective disorder on chromosome 21q22.


Asunto(s)
Trastorno Bipolar/genética , Cromosomas Humanos Par 21/genética , Predisposición Genética a la Enfermedad/genética , Bulgaria , Canadá , Mapeo Cromosómico , Femenino , Humanos , Judíos , Escala de Lod , Masculino , Modelos Genéticos , Linaje , Estados Unidos
20.
Hum Mol Genet ; 11(1): 87-92, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11773002

RESUMEN

Retinitis pigmentosa (RP), the commonest form of inherited retinal dystrophies is a clinically and genetically heterogeneous disorder. It is characterized by progressive degeneration of the peripheral retina leading to night blindness and loss of peripheral visual field. RP is inherited either in an autosomal dominant, autosomal recessive or X-linked mode. A locus (RP18) for autosomal dominant RP was previously mapped by linkage analysis in two large pedigrees to chromosome 1p13-q21. The human HPRP3 gene, the orthologue of the yeast pre-mRNA splicing factor (PRP3), localizes within the RP18 disease interval. The recent identification of mutations in human splicing factors, PRPF31 and PRPC8, led us to screen HPRP3 as a candidate in three chromosome 1q-linked families. So far, two different missense mutations in two English, a Danish family and in three RP individuals have been identified. Both mutations are clustered within a two-codon stretch in the 11th exon of the HPRP3 gene. Interestingly, one of the mutations (T494M) is seen repeatedly in apparently unlinked families raising the possibility of a mutation hot spot. This has been confirmed by haplotype analysis using SNPs spanning the HPRP3 gene region supporting multiple origins of the mutation. The altered HPRP3 amino acids, which are highly conserved in all known HPRP3 orthologues, indicate a major function of that domain in the splicing process. The identification of mutations in a third pre-mRNA splicing factor gene further highlights a novel mechanism of photoreceptor degeneration due to defects in the splicing process.


Asunto(s)
Mutación , Proteínas Nucleares/genética , Empalme del ARN , Retina/metabolismo , Retinitis Pigmentosa/genética , Ribonucleoproteína Nuclear Pequeña U4-U6/genética , Secuencia de Aminoácidos , Cromosomas Humanos Par 1/genética , Cartilla de ADN/química , Femenino , Genes Dominantes , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Linaje , Reacción en Cadena de la Polimerasa , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Homología de Secuencia de Aminoácido , Cromosoma X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...