Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 28(8)2023 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-37110551

RESUMEN

Bestrophin 1 (Best1) is a chloride channel that localises to the plasma membrane of retinal pigment epithelium (RPE) cells. Mutations in the BEST1 gene are associated with a group of untreatable inherited retinal dystrophies (IRDs) called bestrophinopathies, caused by protein instability and loss-of-function of the Best1 protein. 4PBA and 2-NOAA have been shown to rescue the function, expression, and localisation of Best1 mutants; however, it is of interest to find more potent analogues as the concentration of the drugs required is too high (2.5 mM) to be given therapeutically. A virtual docking model of the COPII Sec24a site, where 4PBA has been shown to bind, was generated and a library of 1416 FDA-approved compounds was screened at the site. The top binding compounds were tested in vitro in whole-cell patch-clamp experiments of HEK293T cells expressing mutant Best1. The application of 25 µM tadalafil resulted in full rescue of Cl- conductance, comparable to wild type Best1 levels, for p.M325T mutant Best1 but not for p.R141H or p.L234V mutants.


Asunto(s)
Canales de Cloruro , Epitelio Pigmentado de la Retina , Humanos , Bestrofinas/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Tadalafilo , Células HEK293 , Mutación , Epitelio Pigmentado de la Retina/metabolismo , Proteínas de Transporte Vesicular/genética
2.
Molecules ; 28(15)2023 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-37570721

RESUMEN

Many transmembrane proteins are modulated by intracellular or extracellular pH. Investigation of pH dependence generally proceeds by mutagenesis of a wide set of amino acids, guided by properties such as amino-acid conservation and structure. Prediction of pKas can streamline this process, allowing rapid and effective identification of amino acids of interest with respect to pH dependence. Commencing with the calcium-activated chloride channel bestrophin 1, the carboxylate ligand structure around calcium sites relaxes in the absence of calcium, consistent with a measured lack of pH dependence. By contrast, less relaxation in the absence of calcium in TMEM16A, and maintenance of elevated carboxylate sidechain pKas, is suggested to give rise to pH-dependent chloride channel activity. This hypothesis, modulation of calcium/proton coupling and pH-dependent activity through the extent of structural relaxation, is shown to apply to the well-characterised cytosolic proteins calmodulin (pH-independent) and calbindin D9k (pH-dependent). Further application of destabilised, ionisable charge sites, or electrostatic frustration, is made to other human chloride channels (that are not calcium-activated), ClC-2, GABAA, and GlyR. Experimentally determined sites of pH modulation are readily identified. Structure-based tools for pKa prediction are freely available, allowing users to focus on mutagenesis studies, construct hypothetical proton pathways, and derive hypotheses such as the model for control of pH-dependent calcium activation through structural flexibility. Predicting altered pH dependence for mutations in ion channel disorders can support experimentation and, ultimately, clinical intervention.

3.
J Cell Mol Med ; 25(2): 1319-1322, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33200471

RESUMEN

Dent disease type 1 is caused by mutations in the CLCN5 gene that encodes CLC5, a 2Cl- /H+ exchanger. The CLC5 mutants that have been functionally analysed constitute three major classes based on protein expression, cellular localization and channel function. We tested two small molecules, 4-phenylbutyrate (4PBA) and its analogue 2-naphthoxyacetic acid (2-NOAA), for their effect on mutant CLC5 function and expression by whole-cell patch-clamp and Western blot, respectively. The expression and function of non-Class I CLC5 mutants that have reduced function could be restored by either treatment. Cell viability was reduced in cells treated with 2-NOAA. 4PBA is a FDA-approved drug for the treatment of urea cycle disorders and offers a potential therapy for Dent disease.


Asunto(s)
Quimiocina CCL5/genética , Enfermedad de Dent/genética , Mutación/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Supervivencia Celular/efectos de los fármacos , Quimiocina CCL5/metabolismo , Glicolatos/farmacología , Células HEK293 , Humanos , Fenilbutiratos/farmacología
4.
Proc Natl Acad Sci U S A ; 112(25): E3236-45, 2015 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-26056285

RESUMEN

Ocular developmental disorders, including the group classified as microphthalmia, anophthalmia, and coloboma (MAC) and inherited retinal dystrophies, collectively represent leading causes of hereditary blindness. Characterized by extreme genetic and clinical heterogeneity, the separate groups share many common genetic causes, in particular relating to pathways controlling retinal and retinal pigment epithelial maintenance. To understand these shared pathways and delineate the overlap between these groups, we investigated the genetic cause of an autosomal dominantly inherited condition of retinal dystrophy and bilateral coloboma, present in varying degrees in a large, five-generation family. By linkage analysis and exome sequencing, we identified a previously undescribed heterozygous mutation, n.37 C > T, in the seed region of microRNA-204 (miR-204), which segregates with the disease in all affected individuals. We demonstrated that this mutation determines significant alterations of miR-204 targeting capabilities via in vitro assays, including transcriptome analysis. In vivo injection, in medaka fish (Oryzias latipes), of the mutated miR-204 caused a phenotype consistent with that observed in the family, including photoreceptor alterations with reduced numbers of both cones and rods as a result of increased apoptosis, thereby confirming the pathogenic effect of the n.37 C > T mutation. Finally, knockdown assays in medaka fish demonstrated that miR-204 is necessary for normal photoreceptor function. Overall, these data highlight the importance of miR-204 in the regulation of ocular development and maintenance and provide the first evidence, to our knowledge, of its contribution to eye disease, likely through a gain-of-function mechanism.


Asunto(s)
Coloboma/genética , MicroARNs/genética , Distrofias Retinianas/genética , Secuencia de Bases , Coloboma/complicaciones , Exoma , Femenino , Ligamiento Genético , Humanos , Masculino , Linaje , Distrofias Retinianas/complicaciones , Homología de Secuencia de Ácido Nucleico
5.
Hum Mol Genet ; 24(23): 6565-79, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26395458

RESUMEN

Type 2 brittle cornea syndrome (BCS2) is an inherited connective tissue disease with a devastating ocular phenotype caused by mutations in the transcription factor PR domain containing 5 (PRDM5) hypothesized to exert epigenetic effects through histone and DNA methylation. Here we investigate clinical samples, including skin fibroblasts and retinal tissue from BCS2 patients, to elucidate the epigenetic role of PRDM5 and mechanisms of its dysregulation in disease. First we report abnormal retinal vascular morphology in the eyes of two cousins with BCS2 (PRDM5 Δ exons 9-14) using immunohistochemistry, and mine data from skin fibroblast expression microarrays from patients with PRDM5 mutations p.Arg590* and Δ exons 9-14, as well as from a PRDM5 ChIP-sequencing experiment. Gene ontology analysis of dysregulated PRDM5-target genes reveals enrichment for extracellular matrix (ECM) genes supporting vascular integrity and development. Q-PCR and ChIP-qPCR confirm upregulation of critical mediators of ECM stability in vascular structures (COL13A1, COL15A1, NTN1, CDH5) in patient fibroblasts. We identify H3K9 di-methylation (H3K9me2) at these PRDM5-target genes in fibroblasts, and demonstrate that the BCS2 mutation p.Arg83Cys diminishes interaction of PRDM5 with repressive complexes, including NuRD complex protein CHD4, and the repressive chromatin interactor HP1BP3, by co-immunoprecipitation combined with mass spectrometry. We observe reduced heterochromatin protein 1 binding protein 3 (HP1BP3) staining in the retinas of two cousins lacking exons 9-14 by immunohistochemistry, and dysregulated H3K9me2 in skin fibroblasts of three patients (p.Arg590*, p.Glu134* and Δ exons 9-14) by western blotting. These findings suggest that defective interaction of PRDM5 with repressive complexes, and dysregulation of H3K9me2, play a role in PRDM5-associated disease.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndrome de Ehlers-Danlos/genética , Histonas/metabolismo , Mutación , Vasos Retinianos/patología , Factores de Transcripción/genética , Adulto , Antígenos CD/genética , Cadherinas/genética , Niño , Colágeno/genética , Síndrome de Ehlers-Danlos/metabolismo , Síndrome de Ehlers-Danlos/patología , Femenino , Fibroblastos/metabolismo , Ontología de Genes , Humanos , Masculino , Metilación , Persona de Mediana Edad , Factores de Crecimiento Nervioso/genética , Netrina-1 , Piel/citología , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba , Adulto Joven
6.
Hum Mol Genet ; 23(20): 5527-35, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24895405

RESUMEN

Keratoconus, a common inherited ocular disorder resulting in progressive corneal thinning, is the leading indication for corneal transplantation in the developed world. Genome-wide association studies have identified common SNPs 100 kb upstream of ZNF469 strongly associated with corneal thickness. Homozygous mutations in ZNF469 and PR domain-containing protein 5 (PRDM5) genes result in brittle cornea syndrome (BCS) Types 1 and 2, respectively. BCS is an autosomal recessive generalized connective tissue disorder associated with extreme corneal thinning and a high risk of corneal rupture. Some individuals with heterozygous PRDM5 mutations demonstrate a carrier ocular phenotype, which includes a mildly reduced corneal thickness, keratoconus and blue sclera. We hypothesized that heterozygous variants in PRDM5 and ZNF469 predispose to the development of isolated keratoconus. We found a significant enrichment of potentially pathologic heterozygous alleles in ZNF469 associated with the development of keratoconus (P = 0.00102) resulting in a relative risk of 12.0. This enrichment of rare potentially pathogenic alleles in ZNF469 in 12.5% of keratoconus patients represents a significant mutational load and highlights ZNF469 as the most significant genetic factor responsible for keratoconus identified to date.


Asunto(s)
Queratocono/genética , Queratocono/patología , Factores de Transcripción/genética , Proteínas de Unión al ADN/genética , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patología , Anomalías del Ojo , Estudios de Asociación Genética , Heterocigoto , Homocigoto , Humanos , Inestabilidad de la Articulación/congénito , Mutación , Polimorfismo de Nucleótido Simple , Anomalías Cutáneas
7.
Am J Hum Genet ; 88(6): 767-777, 2011 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-21664999

RESUMEN

Extreme corneal fragility and thinning, which have a high risk of catastrophic spontaneous rupture, are the cardinal features of brittle cornea syndrome (BCS), an autosomal-recessive generalized connective tissue disorder. Enucleation is frequently the only management option for this condition, resulting in blindness and psychosocial distress. Even when the cornea remains grossly intact, visual function could also be impaired by a high degree of myopia and keratoconus. Deafness is another common feature and results in combined sensory deprivation. Using autozygosity mapping, we identified mutations in PRDM5 in families with BCS. We demonstrate that regulation of expression of extracellular matrix components, particularly fibrillar collagens, by PRDM5 is a key molecular mechanism that underlies corneal fragility in BCS and controls normal corneal development and maintenance. ZNF469, encoding a zinc finger protein of hitherto undefined function, has been identified as a quantitative trait locus for central corneal thickness, and mutations in this gene have been demonstrated in Tunisian Jewish and Palestinian kindreds with BCS. We show that ZNF469 and PRDM5, two genes that when mutated cause BCS, participate in the same regulatory pathway.


Asunto(s)
Proteínas de Unión al ADN/genética , Matriz Extracelular/genética , Factores de Transcripción/genética , Niño , Análisis Mutacional de ADN , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patología , Matriz Extracelular/fisiología , Anomalías del Ojo , Femenino , Humanos , Inestabilidad de la Articulación/congénito , Masculino , Mutación , Linaje , Anomalías Cutáneas
8.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24560797

RESUMEN

BEST1 encodes Bestrophin-1 (Best1), a homo-oligomeric, integral membrane protein localized to the basolateral plasma membrane of the retinal pigment epithelium. Mutations in BEST1 cause five distinct retinal degenerative diseases, including adult vitelliform macular dystrophy (AVMD), autosomal recessive bestrophinopathy (ARB), autosomal dominant vitreoretinochoroidopathy (ADVIRC), and retinitis pigmentosa (RP). The mechanisms underlying these diseases and why mutations cause one disease over another are, for the most part, unknown. To gain insights into these four diseases, we expressed 28 Best1 mutants fused to YFP in polarized MDCK monolayers and, via confocal microscopy and immunofluorescence, live-cell FRET, and reciprocal co-immunoprecipitation experiments, screened these mutants for defects in localization and oligomerization. All 28 mutants exhibited comparable FRET efficiencies to and co-immunoprecipitated with WT Best1, indicating unimpaired oligomerization. RP- and ADVIRC-associated mutants were properly localized to the basolateral plasma membrane of cells, while two AVMD and most ARB mutants were mislocalized. When co-expressed, all mislocalized mutants caused mislocalization of WT Best1 to intracellular compartments. Our current and past results indicate that mislocalization of Best1 is not an absolute feature of any individual bestrophinopathy, occurring in AVMD, BVMD, and ARB. Furthermore, some ARB mutants that do not also cause dominant disease cause mislocalization of Best1, indicating that mislocalization is not a cause of disease, and that absence of Best1 activity from the plasma membrane is tolerated. Lastly, we find that the ARB truncation mutants L174Qfs*57 and R200X can form oligomers with WT Best1, indicating that the first ∼174 amino acids of Best1 are sufficient for oligomerization to occur.


Asunto(s)
Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Enfermedades Hereditarias del Ojo/genética , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Mutación Missense , Multimerización de Proteína/fisiología , Enfermedades de la Retina/genética , Adenoviridae/genética , Animales , Proteínas Bacterianas/metabolismo , Bestrofinas , Western Blotting , Enfermedades de la Coroides/genética , Enfermedades de la Coroides/metabolismo , Perros , Electrofisiología , Enfermedades Hereditarias del Ojo/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/metabolismo , Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Proteínas Luminiscentes/metabolismo , Células de Riñón Canino Madin Darby/metabolismo , Microscopía Confocal , Técnicas de Placa-Clamp , Degeneración Retiniana/genética , Degeneración Retiniana/metabolismo , Enfermedades de la Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Transfección , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
9.
Am J Hum Genet ; 86(5): 686-95, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20398886

RESUMEN

Retinitis pigmentosa is a genetically heterogeneous group of inherited ocular disorders characterized by progressive photoreceptor cell loss, night blindness, constriction of the visual field, and progressive visual disability. Homozygosity mapping and gene expression studies identified a 2 exon gene, C2ORF71. The encoded protein has no homologs and is highly expressed in the eye, where it is specifically expressed in photoreceptor cells. Two mutations were found in C2ORF71 in human RP patients: A nonsense mutation (p.W253X) in the first exon is likely to be a null allele; the second, a missense mutation (p.I201F) within a highly conserved region of the protein, leads to proteosomal degradation. Bioinformatic and functional studies identified and validated sites of lipid modification within the first three amino acids of the C2ORF71 protein. Using morpholino oligonucleotides to knockdown c2orf71 expression in zebrafish results in visual defects, confirming that C2ORF71 plays an important role in the development of normal vision. Finally, localization of C2ORF71 to primary cilia in cultured cells suggests that the protein is likely to localize to the connecting cilium or outer segment of photoreceptor cells.


Asunto(s)
Ojo/metabolismo , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas/genética , Retinitis Pigmentosa/genética , Ceguera/genética , Cilios/genética , Cilios/metabolismo , Exones , Proteínas del Ojo/genética , Homocigoto , Humanos , Mutación Missense , Retinitis Pigmentosa/metabolismo
10.
Mol Genet Metab ; 109(3): 289-95, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23680354

RESUMEN

Brittle cornea syndrome (BCS; MIM 229200) is an autosomal recessive generalized connective tissue disorder caused by mutations in ZNF469 and PRDM5. It is characterized by extreme thinning and fragility of the cornea that may rupture in the absence of significant trauma leading to blindness. Keratoconus or keratoglobus, high myopia, blue sclerae, hyperelasticity of the skin without excessive fragility, and hypermobility of the small joints are additional features of BCS. Transcriptional regulation of extracellular matrix components, particularly of fibrillar collagens, by PRDM5 and ZNF469 suggests that they might be part of the same pathway, the disruption of which is likely to cause the features of BCS. In the present study, we have performed molecular analysis of a cohort of 23 BCS affected patients on both ZNF469 and PRDM5, including those who were clinically reported previously [1]; the clinical description of three additional patients is reported in detail. We identified either homozygous or compound heterozygous mutations in ZNF469 in 18 patients while, 4 were found to be homozygous for PRDM5 mutations. In one single patient a mutation in neither ZNF469 nor PRDM5 was identified. Furthermore, we report the 12 novel ZNF469 variants identified in our patient cohort, and show evidence that ZNF469 is a single exon rather than a two exon gene.


Asunto(s)
Síndrome de Ehlers-Danlos/genética , Exones , Matriz Extracelular/genética , Regulación de la Expresión Génica , Mutación , Factores de Transcripción/genética , Adolescente , Niño , Preescolar , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Síndrome de Ehlers-Danlos/diagnóstico , Síndrome de Ehlers-Danlos/terapia , Anomalías del Ojo , Femenino , Genotipo , Humanos , Inestabilidad de la Articulación/congénito , Anomalías Cutáneas
11.
Front Cell Dev Biol ; 11: 1161548, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37206923

RESUMEN

Pathogenic, generally loss-of-function, variants in CACNA1F, encoding the Cav1.4α1 calcium channel, underlie congenital stationary night blindness type 2 (CSNB2), a rare inherited retinal disorder associated with visual disability. To establish the underlying pathomechanism, we investigated 10 clinically derived CACNA1F missense variants located across pore-forming domains, connecting loops, and the carboxy-tail domain of the Cav1.4α subunit. Homology modeling showed that all variants cause steric clashes; informatics analysis correctly predicted pathogenicity for 7/10 variants. In vitro analyses demonstrated that all variants cause a decrease in current, global expression, and protein stability and act through a loss-of-function mechanism and suggested that the mutant Cav1.4α proteins were degraded by the proteasome. We showed that the reduced current for these variants could be significantly increased through treatment with clinical proteasome inhibitors. In addition to facilitating clinical interpretation, these studies suggest that proteasomal inhibition represents an avenue of potential therapeutic intervention for CSNB2.

12.
Am J Hum Genet ; 84(6): 801-6, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19481195

RESUMEN

3-M syndrome is an autosomal-recessive primordial growth disorder characterized by significant intrauterine and postnatal growth restriction. Mutations in the CUL7 gene are known to cause 3-M syndrome. In 3-M syndrome patients that do not carry CUL7 mutations, we performed high-density genome-wide SNP mapping to identify a second locus at 2q35-q36.1. Further haplotype analysis revealed a 1.29 Mb interval in which the underlying gene is located and we subsequently discovered seven distinct null mutations from 10 families within the gene OBSL1. OBSL1 is a putative cytoskeletal adaptor protein that localizes to the nuclear envelope. We were also able to demonstrate that loss of OBSL1 leads to downregulation of CUL7, implying a role for OBSL1 in the maintenance of CUL7 protein levels and suggesting that both proteins are involved within the same molecular pathway.


Asunto(s)
Proteínas del Citoesqueleto/genética , Trastornos del Crecimiento/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Ubiquitinación , Adolescente , Células Cultivadas , Niño , Preescolar , Proteínas Cullin/genética , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto , Femenino , Humanos , Lactante , Riñón/citología , Riñón/metabolismo , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , ARN Interferente Pequeño/farmacología , Síndrome
13.
Am J Hum Genet ; 85(5): 581-92, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19853238

RESUMEN

Bestrophin-1 is preferentially expressed at the basolateral membrane of the retinal pigmented epithelium (RPE) of the retina. Mutations in the BEST1 gene cause the retinal dystrophies vitelliform macular dystrophy, autosomal-dominant vitreochoroidopathy, and autosomal-recessive bestrophinopathy. Here, we describe four missense mutations in bestrophin-1, three that we believe are previously unreported, in patients diagnosed with autosomal-dominant and -recessive forms of retinitis pigmentosa (RP). The physiological function of bestrophin-1 remains poorly understood although its heterologous expression induces a Cl--specific current. We tested the effect of RP-causing variants on Cl- channel activity and cellular localization of bestrophin-1. Two (p.L140V and p.I205T) produced significantly decreased chloride-selective whole-cell currents in comparison to those of wild-type protein. In a model system of a polarized epithelium, two of three mutations (p.L140V and p.D228N) caused mislocalization of bestrophin-1 from the basolateral membrane to the cytoplasm. Mutations in bestrophin-1 are increasingly recognized as an important cause of inherited retinal dystrophy.


Asunto(s)
Canales de Cloruro/genética , Proteínas del Ojo/genética , Mutación Missense , Epitelio Pigmentado de la Retina/patología , Retinitis Pigmentosa/etiología , Retinitis Pigmentosa/genética , Secuencia de Aminoácidos , Bestrofinas , Línea Celular , Cromosomas Humanos Par 11 , Secuencia Conservada , Exones , Femenino , Genes Dominantes , Genes Recesivos , Ligamiento Genético , Homocigoto , Humanos , Riñón/citología , Escala de Lod , Masculino , Datos de Secuencia Molecular , Núcleo Familiar , Linaje , Polimorfismo de Nucleótido Simple , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homología de Secuencia de Aminoácido
14.
Am J Hum Genet ; 82(1): 19-31, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18179881

RESUMEN

We describe a distinct retinal disorder, autosomal-recessive bestrophinopathy (ARB), that is consequent upon biallelic mutation in BEST1 and is associated with central visual loss, a characteristic retinopathy, an absent electro-oculogram light rise, and a reduced electroretinogram. Heterozygous mutations in BEST1 have previously been found to cause the two dominantly inherited disorders, Best macular dystrophy and autosomal-dominant vitreoretinochoroidopathy. The transmembrane protein bestrophin-1, encoded by BEST1, is located at the basolateral membrane of the retinal pigment epithelium in which it probably functions as a Cl(-) channel. We sequenced BEST1 in five families, identifying DNA variants in each of ten alleles. These encoded six different missense variants and one nonsense variant. The alleles segregated appropriately for a recessive disorder in each family. No clinical or electrophysiological abnormalities were identified in any heterozygotes. We conducted whole-cell patch-clamping of HEK293 cells transfected with bestrophin-1 to measure the Cl(-) current. Two ARB missense isoforms severely reduced channel activity. However, unlike two other alleles previously associated with Best disease, cotransfection with wild-type bestrophin-1 did not impair the formation of active wild-type bestrophin-1 channels, consistent with the recessive nature of the condition. We propose that ARB is the null phenotype of bestrophin-1 in humans.


Asunto(s)
Canales de Cloruro/genética , Proteínas del Ojo/genética , Enfermedades de la Retina/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Bestrofinas , Línea Celular , Niño , Preescolar , Canales de Cloruro/química , Codón sin Sentido , Proteínas del Ojo/química , Femenino , Genes Recesivos , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense , Transfección
15.
Front Genet ; 12: 637780, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33584831

RESUMEN

Calcium channels are crucial to a number of cellular functions. The high voltage-gated calcium channel family comprise four heteromeric channels (Cav1.1-1.4) that function in a similar manner, but that have distinct expression profiles. Three of the pore-forming α1 subunits are located on autosomes and the forth on the X chromosome, which has consequences for the type of pathogenic mutation and the disease mechanism associated with each gene. Mutations in this family of channels are associated with malignant hyperthermia (Cav1.1), various QT syndromes (Cav1.2), deafness (Cav1.3), and incomplete congenital stationary night blindness (iCSNB; Cav1.4). In this study we performed a bioinformatic analysis on reported mutations in all four Cav α1 subunits and correlated these with variant frequency in the general population, phenotype and the effect on channel conductance to produce a comprehensive composite Cav1 mutation analysis. We describe regions of mutation clustering, identify conserved residues that are mutated in multiple family members and regions likely to cause a loss- or gain-of-function in Cav1.4. Our research highlights that therapeutic treatments for each of the Cav1 channels will have to consider channel-specific mechanisms, especially for the treatment of X-linked iCSNB.

16.
Mol Vis ; 16: 2916-22, 2010 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-21203346

RESUMEN

PURPOSE: Autosomal recessive bestrophinopathy (ARB) is a newly defined retinal dystrophy caused by biallelic mutations in bestrophin-1 (BEST1) and is hypothesized to represent the null bestrophin-1 phenotype in humans. The aim was to determine whether a synonymous BEST1 variant, c.102C>T, identified in two unrelated ARB patients, alters pre-mRNA splicing of the gene. Additionally a detailed phenotypic characterization of this distinctive condition is presented for both patients. METHODS: BEST1 was analyzed by direct sequencing. Patients underwent standard ophthalmic assessment. In silico and in vitro analysis using a minigene system was performed to assess whether a synonymous variant identified, c.102C>T p.Gly34Gly, alters pre-mRNA splicing of BEST1. RESULTS: Both ARB patients harbored either proven (patient 1; c.102C>T p.Gly34Gly and c.572T>C p.Leu191Pro) or presumed (patient 2; c.102C>T p.Gly34Gly and c.1470_1471delCA, p.His490GlnfsX24) biallelic mutations in BEST1 and were found to have phenotypes consistent with ARB. In vitro analysis of the synonymous variant, c.102C>T p.Gly34Gly, demonstrated it to introduce a cryptic splice donor site 52 nucleotides upstream of the actual splice donor site. CONCLUSIONS: The novel BEST1 variant identified, c.102C>T p.Gly34Gly, alters pre-mRNA splicing in vitro and is potentially pathogenic. In vivo this splicing variant is predicted to lead to the production of an mRNA transcript with a premature termination codon (p.Glu35TrpfsX11) that is predicted to be degraded by NMD.


Asunto(s)
Canales de Cloruro/genética , Codón/genética , Proteínas del Ojo/genética , Genes Recesivos/genética , Mutación/genética , Empalme del ARN/genética , Distrofias Retinianas/genética , Adulto , Bestrofinas , Niño , Biología Computacional , Femenino , Fibronectinas/química , Fibronectinas/genética , Fluorescencia , Fondo de Ojo , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Estructura Terciaria de Proteína , Adulto Joven , Globinas alfa/genética
17.
Invest Ophthalmol Vis Sci ; 61(5): 28, 2020 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-32421148

RESUMEN

Purpose: Bestrophinopathies are a group of untreatable inherited retinal dystrophies caused by mutations in the retinal pigment epithelium (RPE) Cl- channel bestrophin 1. We tested whether sodium phenylbutyrate (4PBA) could rescue the function of mutant bestrophin 1 associated with autosomal dominant and recessive disease. We then sought analogues of 4PBA with increased potency and determined the mode of action for 4PBA and a lead compound 2-naphthoxyacetic acid (2-NOAA). Lastly, we tested if 4PBA and 2-NOAA could functionally rescue bestrophin 1 function in RPE generated from induced pluripotent stem cells (iPSC-RPEs) derived from patients with a dominant or recessive bestrophinopathy. Methods: Global and plasma membrane expression was determined by Western blot and immunofluorescent microscopy, respectively. The effect of 4PBA and 2-NOAA on transcription was measured by quantitative RT-PCR and the rate of protein turnover by cycloheximide chase and Western blot. Channel function was measured by whole-cell patch clamp. Results: 4PBA and 2-NOAA can rescue the global and membrane expression of mutant bestrophin 1 associated with autosomal dominant disease (Best vitelliform macular dystrophy [BVMD]) and autosome recessive bestrophinopathy (ARB), and these small molecules have different modes of action. Both 4PBA and 2-NOAA significantly increased the channel function of mutant BVMD and ARB bestrophin 1 in HEK293T and iPSC-RPE cells derived from patients with BVMD and ARB. For 4PBA, the increased mutant channel function in BVMD and ARB iPSC-RPE was equal to that of wild-type iPSC-RPE bestrophin 1. Conclusions: The restoration of bestrophin 1 function in patient-derived RPE confirms the US Food and Drug Administration-approved drug 4PBA as a promising therapeutic treatment for bestrophinopathies.


Asunto(s)
Antineoplásicos/farmacología , Bestrofinas/genética , Enfermedades Hereditarias del Ojo/tratamiento farmacológico , Regulación de la Expresión Génica/fisiología , Glicolatos/farmacología , Fenilbutiratos/farmacología , Enfermedades de la Retina/tratamiento farmacológico , Epitelio Pigmentado de la Retina/efectos de los fármacos , Western Blotting , Membrana Celular/metabolismo , Canales de Cloruro/metabolismo , Cicloheximida/farmacología , Electroforesis en Gel de Poliacrilamida , Enfermedades Hereditarias del Ojo/genética , Enfermedades Hereditarias del Ojo/metabolismo , Genes Recesivos , Células HEK293/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Microscopía Fluorescente , Técnicas de Placa-Clamp , Reacción en Cadena en Tiempo Real de la Polimerasa , Enfermedades de la Retina/genética , Enfermedades de la Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Transfección
18.
Eur J Hum Genet ; 28(5): 576-586, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31836858

RESUMEN

Thirty percent of all inherited retinal disease (IRD) is accounted for by conditions with extra-ocular features. This study aimed to establish the genetic diagnostic pick-up rate for IRD patients with one or more extra-ocular features undergoing panel-based screening in a clinical setting. One hundred and six participants, tested on a gene panel which contained both isolated and syndromic IRD genes, were retrospectively ascertained from the Manchester Genomic Diagnostics Laboratory database spanning 6 years (2012-2017). Phenotypic features were extracted from the clinical notes and classified according to Human Phenotype Ontology; all identified genetic variants were interpreted in accordance to the American College of Medical Genetics and Genomics guidelines. Overall, 49% (n = 52) of patients received a probable genetic diagnosis. A further 6% (n = 6) had a single disease-associated variant in an autosomal recessive disease-relevant gene. Fifty-two percent (n = 55) of patients had a clinical diagnosis at the time of testing. Of these, 71% (n = 39) received a probable genetic diagnosis. By contrast, for those without a provisional clinical diagnosis (n = 51), only 25% (n = 13) received a probable genetic diagnosis. The clinical diagnosis of Usher (n = 33) and Bardet-Biedl syndrome (n = 10) was confirmed in 67% (n = 22) and 80% (n = 8), respectively. The testing diagnostic rate in patients with clinically diagnosed multisystemic IRD conditions was significantly higher than those without one (71% versus 25%; p value < 0.001). The lower pick-up rate in patients without a clinical diagnosis suggests that panel-based approaches are unlikely to be the most effective means of achieving a molecular diagnosis for this group. Here, we suggest that genome-wide approaches (whole exome or genome) are more appropriate.


Asunto(s)
Enfermedades Hereditarias del Ojo/genética , Pruebas Genéticas/normas , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Enfermedades de la Retina/genética , Análisis de Secuencia de ADN/normas , Adolescente , Adulto , Anciano , Niño , Preescolar , Enfermedades Hereditarias del Ojo/diagnóstico , Femenino , Pruebas Genéticas/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Fenotipo , Enfermedades de la Retina/diagnóstico , Sensibilidad y Especificidad , Análisis de Secuencia de ADN/métodos , Síndrome
19.
Hum Gene Ther Methods ; 30(2): 44-52, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30963787

RESUMEN

Mutations in the human BEST1 gene are responsible for a number of distinct retinal disorders known as bestrophinopathies, for which there are no current treatments. The protein product, bestrophin-1, is expressed in the retinal pigment epithelium (RPE) where it localizes to the basolateral membrane and acts as a Ca2+-activated chloride channel. Recent studies have shown successful BEST1-mediated gene transfer to the RPE, indicating human clinical trials of BEST1 gene therapy may be on the horizon. A critical aspect of such trials is the ability to assess the efficacy of vector prior to patient administration. Here, an assay is presented that enables the quantitative assessment of AAV-mediated BEST1 chloride conductance as a measure of vector efficacy. Expression of BEST1 following transduction of HEK293 cells with AAV.BEST1 vectors was confirmed by liquid chromatography, Western blot, and immunocytochemistry. Whole-cell patch-clamp showed increased chloride conductance in BEST1-transduced cells compared to sham-transduced and untransduced controls. Exogenous chloride current correlated to BEST1 expression level, with an enhanced AAV.BEST1.WPRE vector providing higher expression levels of BEST1 and increases in chloride conductance. This study presents in vitro electrophysical quantification of bestrophin-1 following AAV-mediated gene transfer, providing vital functional data on an AAV gene therapy product that will support a future application for regulatory approval.


Asunto(s)
Bestrofinas/fisiología , Parvovirinae/genética , Bestrofinas/genética , Dependovirus , Vectores Genéticos , Células HEK293 , Humanos , Transducción Genética
20.
Hum Mutat ; 29(8): 1017-27, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18425797

RESUMEN

Klippel-Feil syndrome (KFS) is a congenital disorder of spinal segmentation distinguished by the bony fusion of anterior/cervical vertebrae. Scoliosis, mirror movements, otolaryngological, kidney, ocular, cranial, limb, and/or digit anomalies are often associated. Here we report mutations at the GDF6 gene locus in familial and sporadic cases of KFS including the recurrent missense mutation of an extremely conserved residue c.866T>C (p.Leu289Pro) in association with mirror movements and an inversion breakpoint downstream of the gene in association with carpal, tarsal, and vertebral fusions. GDF6 is expressed at the boundaries of the developing carpals, tarsals, and vertebrae and within the adult vertebral disc. GDF6 knockout mice are best distinguished by fusion of carpals and tarsals and GDF6 knockdown in Xenopus results in a high incidence of anterior axial defects consistent with a role for GDF6 in the etiology, diversity, and variability of KFS.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Síndrome de Klippel-Feil/genética , Secuencia de Aminoácidos , Animales , Proteínas Morfogenéticas Óseas/química , Inversión Cromosómica , Análisis Mutacional de ADN , Femenino , Factor 6 de Diferenciación de Crecimiento , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Missense , Linaje , Alineación de Secuencia , Columna Vertebral/anomalías , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA