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.
Hum Genomics ; 8: 19, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25403472

RESUMEN

BACKGROUND: Inherited cataract is a clinically important and genetically heterogeneous cause of visual impairment. Typically, it presents at an early age with or without other ocular/systemic signs and lacks clear phenotype-genotype correlation rendering both clinical classification and molecular diagnosis challenging. Here we have utilized trio-based whole exome sequencing to discover mutations in candidate genes underlying autosomal dominant cataract segregating in three nuclear families. RESULTS: In family A, we identified a recurrent heterozygous mutation in exon-2 of the gene encoding γD-crystallin (CRYGD; c.70C > A, p.Pro24Thr) that co-segregated with 'coralliform' lens opacities. Families B and C were found to harbor different novel variants in exon-2 of the gene coding for gap-junction protein α8 (GJA8; c.20T > C, p.Leu7Pro and c.293A > C, p.His98Pro). Each novel variant co-segregated with disease and was predicted in silico to have damaging effects on protein function. CONCLUSIONS: Exome sequencing facilitates concurrent mutation-profiling of the burgeoning list of candidate genes for inherited cataract, and the results can provide enhanced clinical diagnosis and genetic counseling for affected families.


Asunto(s)
Catarata/genética , Conexinas/genética , Exoma , gamma-Cristalinas/genética , Catarata/patología , Conexinas/metabolismo , Femenino , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Heterocigoto , Humanos , Masculino , Mutación Missense , Linaje , Polimorfismo de Nucleótido Simple , gamma-Cristalinas/metabolismo
2.
Hum Mutat ; 35(3): 289-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24375934

RESUMEN

Inherited retinal dystrophies are a major cause of childhood blindness. Here, we describe the identification of a homozygous frameshift mutation (c.1194_1195delAG, p.Arg398Serfs*9) in TUB in a child from a consanguineous UK Caucasian family investigated using autozygosity mapping and whole-exome sequencing. The proband presented with obesity, night blindness, decreased visual acuity, and electrophysiological features of a rod cone dystrophy. The mutation was also found in two of the proband's siblings with retinal dystrophy and resulted in mislocalization of the truncated protein. In contrast to known forms of retinal dystrophy, including those caused by mutations in the tubby-like protein TULP-1, loss of function of TUB in the proband and two affected family members was associated with early-onset obesity, consistent with an additional role for TUB in energy homeostasis.


Asunto(s)
Mutación del Sistema de Lectura , Homocigoto , Obesidad/genética , Proteínas/genética , Retinitis Pigmentosa/genética , Proteínas Adaptadoras Transductoras de Señales , Niño , Mapeo Cromosómico , Consanguinidad , Proteínas del Ojo/genética , Femenino , Genes Recesivos , Homeostasis , Humanos , Masculino , Linaje , Reino Unido , Población Blanca/genética
3.
Am J Hum Genet ; 89(1): 183-90, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21763485

RESUMEN

Inherited retinal degenerations, including retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA), comprise a group of disorders showing high genetic and allelic heterogeneity. The determination of a full catalog of genes that can, when mutated, cause human retinal disease is a powerful means to understand the molecular physiology and pathology of the human retina. As more genes are found, remaining ones are likely to be rarer and/or unexpected candidates. Here, we identify a family in which all known RP/LCA-related genes are unlikely to be associated with their disorder. A combination of homozygosity mapping and exome sequencing identifies a homozygous nonsense mutation, c.496C>T (p.Arg166X), in a gene, KCNJ13, encoding a potassium channel subunit Kir7.1. A screen of a further 333 unrelated individuals with recessive retinal degeneration identified an additional proband, homozygous for a missense mutation, c.722T>C (p.Leu241Pro), in the same gene. The three affected members of the two families have been diagnosed with LCA. All have a distinct and unusual retinal appearance and a similar early onset of visual loss, suggesting both impaired retinal development and progressive retinal degeneration, involving both rod and cone pathways. Examination of heterozygotes revealed no ocular disease. This finding implicates Kir7.1 as having an important role in human retinal development and maintenance. This disorder adds to a small diverse group of diseases consequent upon loss or reduced function of inwardly rectifying potassium channels affecting various organs. The distinct retinal phenotype that results from biallelic mutations in KCNJ13 should facilitate the molecular diagnosis in further families.


Asunto(s)
Genes Recesivos , Amaurosis Congénita de Leber/genética , Canales de Potasio de Rectificación Interna/genética , Adulto , Secuencia de Aminoácidos , Codón sin Sentido , Exones , Femenino , Variación Genética , Homocigoto , Humanos , Amaurosis Congénita de Leber/patología , Masculino , Datos de Secuencia Molecular , Ceguera Nocturna/genética , Linaje , Canales de Potasio/genética , Canales de Potasio de Rectificación Interna/metabolismo , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/patología
4.
Am J Hum Genet ; 89(6): 782-91, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22137173

RESUMEN

Flecked-retina syndromes, including fundus flavimaculatus, fundus albipunctatus, and benign fleck retina, comprise a group of disorders with widespread or limited distribution of yellow-white retinal lesions of various sizes and configurations. Three siblings who have benign fleck retina and were born to consanguineous parents are the basis of this report. A combination of homozygosity mapping and exome sequencing helped to identify a homozygous missense mutation, c.133G>T (p.Gly45Cys), in PLA2G5, a gene encoding a secreted phospholipase (group V phospholipase A(2)). A screen of a further four unrelated individuals with benign fleck retina detected biallelic variants in the same gene in three patients. In contrast, no loss of function or common (minor-allele frequency>0.05%) nonsynonymous PLA2G5 variants have been previously reported (EVS, dbSNP, 1000 Genomes Project) or were detected in an internal database of 224 exomes (from subjects with adult onset neurodegenerative disease and without a diagnosis of ophthalmic disease). All seven affected individuals had fundoscopic features compatible with those previously described in benign fleck retina and no visual or electrophysiological deficits. No medical history of major illness was reported. Levels of low-density lipoprotein were mildly elevated in two patients. Optical coherence tomography and fundus autofluorescence findings suggest that group V phospholipase A(2) plays a role in the phagocytosis of photoreceptor outer-segment discs by the retinal pigment epithelium. Surprisingly, immunohistochemical staining of human retinal tissue revealed localization of the protein predominantly in the inner and outer plexiform layers.


Asunto(s)
Anomalías del Ojo/genética , Fosfolipasas A2 Grupo V/genética , Homocigoto , Mutación Missense , Retina/anomalías , Adulto , Anciano de 80 o más Años , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Consanguinidad , Femenino , Estudios de Asociación Genética , Fosfolipasas A2 Grupo V/metabolismo , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Retina/metabolismo
5.
Ophthalmology ; 121(6): 1174-84, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24480711

RESUMEN

OBJECTIVE: To provide a detailed phenotype/genotype characterization of Bietti crystalline dystrophy (BCD). DESIGN: Observational case series. PARTICIPANTS: Twenty patients from 17 families recruited from a multiethnic British population. METHODS: Patients underwent color fundus photography, near-infrared (NIR) imaging, fundus autofluorescence (FAF) imaging, spectral domain optical coherence tomography (SD-OCT), and electroretinogram (ERG) assessment. The gene CYP4V2 was sequenced. MAIN OUTCOME MEASURES: Clinical, imaging, electrophysiologic, and molecular genetics findings. RESULTS: Patients ranged in age from 19 to 72 years (median, 40 years), with a visual acuity of 6/5 to perception of light (median, 6/12). There was wide intrafamilial and interfamilial variability in clinical severity. The FAF imaging showed well-defined areas of retinal pigment epithelium (RPE) loss that corresponded on SD-OCT to well-demarcated areas of outer retinal atrophy. Retinal crystals were not evident on FAF imaging and were best visualized with NIR imaging. Spectral domain OCT showed them to be principally located on or in the RPE/Bruch's membrane complex. Disappearance of the crystals, revealed by serial recording, was associated with severe disruption and thinning of the RPE/Bruch's membrane complex. Cases with extensive RPE degeneration (N = 5) had ERGs consistent with generalized rod and cone dysfunction, but those with more focal RPE atrophy showed amplitude reduction without delay (N = 3), consistent with restricted loss of function, or that was normal (N = 2). Likely disease-causing variants were identified in 34 chromosomes from 17 families. Seven were novel, including p.Met66Arg, found in all 11 patients from 8 families of South Asian descent. This mutation appears to be associated with earlier onset (median age, 30 years) compared with other substitutions (median age, 41 years). Deletions of exon 7 were associated with more severe disease. CONCLUSIONS: The phenotype is highly variable. Several novel variants are reported, including a highly prevalent substitution in patients of South Asian descent that is associated with earlier-onset disease. Autofluorescence showed sharply demarcated areas of RPE loss that coincided with abrupt edges of outer retinal atrophy on SD-OCT; crystals were generally situated on or in the RPE/Bruch's complex but could disappear over time with associated RPE disruption. These results support a role for the RPE in disease pathogenesis.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/patología , Sistema Enzimático del Citocromo P-450/genética , Mutación Missense , Polimorfismo de Nucleótido Simple , Enfermedades de la Retina/genética , Enfermedades de la Retina/patología , Adulto , Anciano , Hibridación Genómica Comparativa , Familia 4 del Citocromo P450 , Análisis Mutacional de ADN , Electrorretinografía , Exones/genética , Femenino , Angiografía con Fluoresceína , Estudios de Asociación Genética , Humanos , Hibridación Fluorescente in Situ , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Epitelio Pigmentado de la Retina/patología , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología , Adulto Joven
6.
Hum Mutat ; 34(3): 506-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23281133

RESUMEN

In one consanguineous family with retinitis pigmentosa (RP), a condition characterized by progressive visual loss due to retinal degeneration, homozygosity mapping, and candidate gene sequencing suggested a novel locus. Exome sequencing identified a homozygous frameshifting mutation, c.601delG, p.Lys203Argfs*28, in RP1L1 encoding RP 1-like1, a photoreceptor-specific protein. A screen of a further 285 unrelated individuals with autosomal recessive RP identified an additional proband, homozygous for a missense variant, c.1637G>C, p.Ser546Thr, in RP1L1. A distinct retinal disorder, occult macular dystrophy (OCMD) solely affects the central retinal cone photoreceptors and has previously been reported to be associated with variants in the same gene. The association between mutations in RP1L1 and the disorder OCMD was explored by screening a cohort of 28 unrelated individuals with the condition; 10 were found to harbor rare (minor allele frequency ≤0.5% in the 1,000 genomes dataset) heterozygous RP1L1 missense variants. Analysis of family members revealed many unaffected relatives harboring the same variant. Linkage analysis excluded the possibility of a recessive mode of inheritance, and sequencing of RP1, a photoreceptor protein that interacts with RP1L1, excluded a digenic mechanism involving this gene. These findings imply an important and diverse role for RP1L1 in human retinal physiology and disease.


Asunto(s)
Proteínas del Ojo/genética , Degeneración Macular/genética , Retina/patología , Retinitis Pigmentosa/genética , Adulto , Anciano , Mapeo Cromosómico , Clonación Molecular , Estudios de Cohortes , Consanguinidad , Exoma , Femenino , Genes Recesivos , Sitios Genéticos , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Mutación Missense , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ADN
7.
Hum Mutat ; 34(11): 1537-1546, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23946133

RESUMEN

This study was undertaken to investigate the prevalence of sequence variants in LCA5 in patients with Leber congenital amaurosis (LCA), early-onset retinal dystrophy (EORD), and autosomal recessive retinitis pigmentosa (arRP); to delineate the ocular phenotypes; and to provide an overview of all published LCA5 variants in an online database. Patients underwent standard ophthalmic evaluations after providing informed consent. In selected patients, optical coherence tomography (OCT) and fundus autofluorescence imaging were possible. DNA samples from 797 unrelated patients with LCA and 211 with the various types of retinitis pigmentosa (RP) were screened by Sanger sequence analysis of all LCA5 exons and intron/exon junctions. Some LCA patients were prescreened by APEX technology or selected based on homozygosity mapping. In silico analyses were performed to assess the pathogenicity of the variants. Segregation analysis was performed where possible. Published and novel LCA5 variants were collected, amended for their correct nomenclature, and listed in a Leiden Open Variation Database (LOVD). Sequence analysis identified 18 new probands with 19 different LCA5 variants. Seventeen of the 19 LCA5 variants were novel. Except for two missense variants and one splice site variant, all variants were protein-truncating mutations. Most patients expressed a severe phenotype, typical of LCA. However, some LCA subjects had better vision and intact inner segment/outer segment (IS/OS) junctions on OCT imaging. In two families with LCA5 variants, the phenotype was more compatible with EORD with affected individuals displaying preserved islands of retinal pigment epithelium. One of the families with a milder phenotype harbored a homozygous splice site mutation; a second family was found to have a combination of a stop mutation and a missense mutation. This is the largest LCA5 study to date. We sequenced 1,008 patients (797 with LCA, 211 with arRP) and identified 18 probands with LCA5 mutations. Mutations in LCA5 are a rare cause of childhood retinal dystrophy accounting for ∼2% of disease in this cohort, and the majority of LCA5 mutations are likely null. The LCA5 protein truncating mutations are predominantly associated with LCA. However, in two families with the milder EORD, the LCA5 gene analysis revealed a homozygous splice site mutation in one and a stop mutation in combination with a missense mutation in a second family, suggesting that this milder phenotype is due to residual function of lebercilin and expanding the currently known phenotypic spectrum to include the milder early onset RP. Some patients have remaining foveal cone structures (intact IS/OS junctions on OCT imaging) and remaining visual acuities, which may bode well for upcoming treatment trials.


Asunto(s)
Proteínas del Ojo/genética , Estudios de Asociación Genética , Amaurosis Congénita de Leber/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación , Retinitis Pigmentosa/genética , Adolescente , Adulto , Alelos , Niño , Preescolar , Consanguinidad , Femenino , Angiografía con Fluoresceína , Genotipo , Humanos , Lactante , Recién Nacido , Amaurosis Congénita de Leber/diagnóstico , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Retina/patología , Retinitis Pigmentosa/diagnóstico , Adulto Joven
8.
Ophthalmology ; 120(11): 2324-31, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23769331

RESUMEN

PURPOSE: To describe the phenotypic presentation of a cohort of individuals with homozygous disease-associated ABCA4 variants. DESIGN: Retrospective case series. PARTICIPANTS: Eighteen affected individuals from 13 families ascertained from a total cohort of 214 families with ABCA4-related retinal disease presenting to a single center. METHODS: A detailed history was obtained, and color fundus photography, autofluorescence (AF) imaging, optical coherence tomography (OCT), and electrophysiologic assessment were performed. Phenotypes based on ophthalmoscopy, AF, and electrophysiology were assigned using previously reported characteristics. ABCA4 mutation detection was performed using the ABCR400 microarray (Asper Biotech, Tartu, Estonia) and high-throughput DNA sequencing, with direct sequencing used to assess segregation. MAIN OUTCOME MEASURES: Detailed clinical, electrophysiologic, and molecular genetic findings. RESULTS: Eleven disease-associated homozygous ABCA4 alleles were identified, including 1 frame shift, 2 stops, 1 intronic variant causing splice-site alteration, 2 complex missense variants, and 5 missense variants: p.Glu905fsX916, p.Arg1300X, p.Gln2220X, c.4253+4 C>T, p.Leu541Pro and p.Ala1038Val (homozygosity for complex allele), p.Val931Met and p.Arg1705Gln (complex allele), p.Arg212Cys, p.Cys1488Arg, p.Arg1640Trp, p.Gly1961Glu, and p.Leu2027Phe. Eight of these 11 homozygous alleles have not been reported previously. Six of 7 patients with homozygous null alleles had early-onset (<10 years) disease, with all 7 having a severe phenotype. Two patients with homozygous missense variants (p.Leu541Pro and p.Ala1038Val [complex], and p.Arg1640Trp) presented with a severe phenotype. Three patients with homozygous p.Gly1961Glu had adult-onset disease and a mild phenotype. One patient with homozygous p.Leu2027Phe showed a spared fovea and preserved visual acuity. CONCLUSIONS: The phenotypes represented in patients identified as homozygous for presumed disease-associated ABCA4 variants gives insight into the effect of individual alleles. Null alleles have severe functional effects, and certain missense variants are similar to nulls, suggesting complete abrogation of protein function. The common alleles identified, p.Gly1961Glu and p. Leu2027Phe, both have a mild structural and functional effect on the adult retina; the latter is associated with relatively retained photoreceptor architecture and function at the fovea.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Mutación , Polimorfismo de Nucleótido Simple , Adolescente , Adulto , Alelos , Niño , Análisis Mutacional de ADN , Electrorretinografía , Femenino , Angiografía con Fluoresceína , Humanos , Degeneración Macular/diagnóstico , Degeneración Macular/genética , Masculino , Persona de Mediana Edad , Fenotipo , Células Fotorreceptoras de Vertebrados/patología , Estudios Retrospectivos , Enfermedad de Stargardt , Tomografía de Coherencia Óptica , Agudeza Visual/fisiología
9.
Am J Hum Genet ; 85(5): 711-9, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19878917

RESUMEN

Complete congenital stationary night blindness (cCSNB) is associated with loss of function of rod and cone ON bipolar cells in the mammalian retina. In humans, mutations in NYX and GRM6 have been shown to cause the condition. Through the analysis of a consanguineous family and screening of nine additional pedigrees, we have identified three families with recessive mutations in the gene TRPM1 encoding transient receptor potential cation channel, subfamily M, member 1, also known as melastatin. A number of other variants of unknown significance were found. All patients had myopia, reduced central vision, nystagmus, and electroretinographic evidence of ON bipolar cell dysfunction. None had abnormalities of skin pigmentation, although other skin conditions were reported. RNA derived from human retina and skin was analyzed and alternate 5' exons were determined. The most 5' exon is likely to harbor an initiation codon, and the protein sequence is highly conserved across vertebrate species. These findings suggest an important role of this specific cation channel for the normal function of ON bipolar cells in the human retina.


Asunto(s)
Mutación , Ceguera Nocturna/congénito , Ceguera Nocturna/genética , Células Fotorreceptoras Retinianas Bastones/fisiología , Canales Catiónicos TRPM/genética , Secuencia de Aminoácidos , Secuencia de Bases , Estudios de Casos y Controles , Mapeo Cromosómico , Cromosomas Humanos Par 15 , Estudios de Cohortes , Consanguinidad , Electrorretinografía , Femenino , Heterocigoto , Homocigoto , Humanos , Masculino , Datos de Secuencia Molecular , Linaje
10.
Mol Vis ; 17: 2706-16, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22065924

RESUMEN

PURPOSE: To identify patients with autosomal recessive retinal dystrophy caused by mutations in the gene, retinal dehydrogenase 12 (RDH12), and to report the associated phenotype. METHODS: After giving informed consent, all patients underwent full clinical evaluation. Patients were selected for mutation analysis based upon positive results from the Asper Ophthalmics Leber congenital amaurosis arrayed primer extansion (APEX) microarray screening, linkage analysis, or their clinical phenotype. All coding exons of RDH12 were screened by direct Sanger sequencing. Potential variants were checked for segregation in the respective families and screened in controls, and their pathogenicity analyzed using in silico prediction programs. RESULTS: Screening of 389 probands by the APEX microarray and/or direct sequencing identified bi-allelic mutations in 29 families. Seventeen novel mutations were identified. The phenotype in these patients presented with a severe early-onset rod-cone dystrophy. Funduscopy showed severe generalized retinal pigment epithelial and retinal atrophy, which progressed to dense, widespread intraretinal pigment migration by adulthood. The macula showed severe atrophy, with pigmentation and yellowing, and corresponding loss of fundus autofluorescence. Optical coherence tomography revealed marked retinal thinning and excavation at the macula. CONCLUSIONS: RDH12 mutations account for approximately 7% of disease in our cohort of patients diagnosed with Leber congenital amaurosis and early-onset retinal dystrophy. The clinical features of this disorder are highly characteristic and facilitate candidate gene screening. The term RDH12 retinopathy is proposed as a more accurate description.


Asunto(s)
Oxidorreductasas de Alcohol/genética , Proteínas del Ojo/genética , Amaurosis Congénita de Leber/genética , Retina/metabolismo , Degeneración Retiniana/genética , Retinitis Pigmentosa/genética , Edad de Inicio , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Exones , Femenino , Ligamiento Genético , Pruebas Genéticas , Genotipo , Humanos , Lactante , Amaurosis Congénita de Leber/diagnóstico , Masculino , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa , Retina/patología , Degeneración Retiniana/diagnóstico , Retinitis Pigmentosa/diagnóstico , Reino Unido
11.
Mol Vis ; 16: 540-8, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20361016

RESUMEN

PURPOSE: To determine the spectrum of mutations and phenotypic variability within patients with mutations in membrane-type frizzled related protein gene (MFRP). METHODS: Individuals were initially ascertained based on a phenotype similar to that previously published in association with MFRP mutations. Affected patients underwent a full ophthalmic examination (best-corrected visual acuity, slit-lamp examination, applanation tonometry, and fundoscopy), color fundus photography, optical coherence tomography, autofluorescence imaging, and electrophysiology. MFRP was identified by a genome-wide scan in the fourth-largest autozygous region in one consanguineous family. Sanger sequencing of all the exons and intron-exon boundaries of MFRP was undertaken in the affected individuals. RESULTS: Seven affected individuals from four families were identified as having mutations in MFRP. Patients from two families were homozygous for mutations already previously described (c.1143_1144 insC and c.492 delC), while those from the other two were compound heterozygous for mutations (c.201G>A and c.491_492 insT, and c.492 delC, and c.1622_1625 delTCTG), three of which were novel. There was considerable phenotypic variability within and among families. Autofluorescence imaging revealed the central macula to be relatively well preserved. Foveal cysts and optic nerve head drusen were present in two of the four families. Electrophysiology results showed rod-cone dystrophy with mild to moderate reduction in macular function in all affected members. CONCLUSIONS: We report three novel MFRP mutations and expand the phenotypic data available on patients with MFRP mutations.


Asunto(s)
Oftalmopatías/genética , Proteínas de la Membrana/genética , Adulto , Secuencia de Bases , Fenómenos Electrofisiológicos , Electrorretinografía , Oftalmopatías/fisiopatología , Femenino , Fluorescencia , Fondo de Ojo , Humanos , Masculino , Proteínas de la Membrana/química , Persona de Mediana Edad , Datos de Secuencia Molecular , Mutación/genética , Linaje , Fenotipo , Tomografía de Coherencia Óptica
12.
Mol Vis ; 16: 46-52, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20087419

RESUMEN

PURPOSE: To describe the clinical findings and mutations in affected members of two families with an autosomal recessive retinal dystrophy associated with mutations in the protocadherin-21 (PCDH21) gene. METHODS: A full genome scan of members of two consanguineous families segregating an autosomal recessive retinal dystrophy was performed and regions identical by descent identified. Positional candidate genes were identified and sequenced. All patients had a detailed ophthalmic examination, including electroretinography and retinal imaging. RESULTS: Affected members of both families showed identical homozygosity for an overlapping region of chromosome 10q. Sequencing of a candidate gene, PCDH21, showed two separate homozygous single-base deletions, c.337delG (p.G113AfsX1) and c.1459delG (p.G487GfsX20), which were not detected in 282 control chromosomes. Affected members of the two families first reported nyctalopia in late teenage years and retained good central vision until their late 30s. No color vision was detected in any proband. The fundus appearance included the later development of characteristic circular patches of pigment epithelial atrophy at the macula and in the peripheral retina. CONCLUSIONS: Biallelic mutations in the photoreceptor-specific gene PCDH21 cause recessive retinal degeneration in humans.


Asunto(s)
Alelos , Cadherinas/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Degeneración Retiniana/genética , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas Relacionadas con las Cadherinas , Cadherinas/química , Segregación Cromosómica/genética , Análisis Mutacional de ADN , Electrorretinografía , Exones/genética , Familia , Femenino , Fondo de Ojo , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Linaje , Estructura Terciaria de Proteína
13.
Mol Vis ; 16: 369-77, 2010 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-20300561

RESUMEN

PURPOSE: To report the clinical phenotype in patients with a retinal dystrophy associated with novel mutations in the MER tyrosine kinase (MERTK) gene. METHODS: A consanguineous family of Middle Eastern origin was identified, and affected members underwent a full clinical evaluation. Linkage analysis was performed using the Affymetrix 50K chip. Regions of homozygosity were identified. The positional candidate genes protocadherin 21 (PCDH21), retinal G protein-coupled receptor (RGR), and MERTK were polymerase chain reaction (PCR) amplified and sequenced. Long-range PCR was performed to characterize the deletion. Two hundred and ninety-two probands with autosomal recessive, childhood onset, retinal dystrophies were analyzed using the Asper Ophthalmics Leber congenital amaurosis chip to screen for known MERTK mutations. RESULTS: Analysis of a 50K-Affymetrix whole genome scan identified three regions of homozygosity on chromosomes 2 and 10. Screening of the candidate gene MERTK showed a possible deletion of exon 8. Long-range PCR identified a ~9 kb deletion within MERTK that removes exon 8. Screening of DNA from a panel of Saudi Arabian patients with autosomal recessive retinitis pigmentosa identified a second consanguineous family with the same mutation. One patient with a known MERTK mutation (p.R651X) was identified using the Asper Ophthalmics Leber congenital amaurosis chip. Further screening of the gene identified a second novel splice site mutation in intron 1. The phenotype associated with these identified MERTK mutations is of a childhood onset rod-cone dystrophy with early macular atrophy. The optical coherence tomography (OCT) appearance is distinctive with evidence of debris beneath the sensory retina. CONCLUSIONS: Mutations in MERTK are a rare cause of retinal dystrophy. Non homologous recombination between Alu Y repeats near or within disease genes may be an important cause of retinal dystrophies.


Asunto(s)
Predisposición Genética a la Enfermedad , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Retinitis Pigmentosa/epidemiología , Retinitis Pigmentosa/genética , Adolescente , Adulto , Edad de Inicio , Niño , Análisis Mutacional de ADN , Electroforesis en Gel de Agar , Exones/genética , Familia , Femenino , Fondo de Ojo , Genoma Humano/genética , Haplotipos/genética , Humanos , Amaurosis Congénita de Leber/enzimología , Amaurosis Congénita de Leber/genética , Masculino , Linaje , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética , Retinitis Pigmentosa/enzimología , Adulto Joven , Tirosina Quinasa c-Mer
15.
Invest Ophthalmol Vis Sci ; 48(2): 500-8, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17251442

RESUMEN

PURPOSE: To characterize the optical properties of lenses from mice deficient in the gene for lens intrinsic membrane protein-2 (Lim2), which encodes the second most abundant integral protein (Lim2) of lens fiber cell plasma membranes. METHODS: Lim2-deficient mice were derived from a library of gene-trap embryo stem cells. Genotyping was performed by polymerase chain reaction (PCR) amplification of tail genomic DNA and resequencing. Lim2 expression was analyzed by reverse transcription (RT)-PCR and Northern blotting of lens total RNA, immunoblotting of lens membrane extracts, and immunofluorescence confocal microscopy of lens sections. Lens morphology was assessed by light microscopy, and lens refractive properties were quantified with a laser imaging system. RESULTS: Genomic PCR amplification and resequencing indicated that the gene-trap vector had disrupted intron 3 of Lim2, effectively resulting in a null allele (Lim2(Gt)), as verified by RT-PCR amplification and sequencing, RNA blotting, immunoblotting, and immunofluorescence confocal microscopy. Heterozygous Lim2 gene-trap lenses (Lim2(Gt/+)) were morphologically indistinguishable from wild type, whereas homozygous Lim2 gene-trap lenses (Lim2(Gt/Gt)) consistently developed faint, central pulverulent cataracts. Laser imaging analysis indicated that rays passing through the peripheral cortex of the Lim2(Gt/Gt) lens were more strongly refracted than normal, suggesting that the internal gradient refractive index of the lens was disturbed. CONCLUSIONS: These data show that heterozygous loss of Lim2 is insufficient to trigger cataracts in mice, and they provide the first direct evidence that Lim2 plays a critical role in establishing the correct internal refractive properties of the crystalline lens.


Asunto(s)
Catarata/genética , Proteínas del Ojo/genética , Silenciador del Gen/fisiología , Glicoproteínas de Membrana/genética , Errores de Refracción/genética , Secuencia de Aminoácidos , Animales , Northern Blotting , Catarata/patología , Femenino , Genotipo , Cristalino/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Datos de Secuencia Molecular , Fenotipo , Errores de Refracción/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
JAMA Ophthalmol ; 134(9): 1049-53, 2016 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-27389523

RESUMEN

IMPORTANCE: A multiorgan syndromic disorder characterized by sideroblastic anemia, immunodeficiency, periodic fever, and developmental delay with an uncharacterized retinal dystrophy is caused by TRNT1. This report of a family with a homozygous mutation in TRNT1 expands the ocular phenotype to include cataract and inner retinal dysfunction and details a mild systemic phenotype. OBSERVATIONS: A consanguineous family with 3 affected children was investigated. Key clinical features comprised hypogammaglobulinemia, short stature with microcephaly, cataract, and inner retinal dysfunction without sideroblastic anemia or developmental delay. Two siblings had poor balance and 1 sibling had sensorineural hearing loss. The oldest sibling had primary ovarian failure diagnosed at age 14.5 years. Exome sequencing identified a homozygous missense variant in TRNT1, c.295C>T (p.Arg99Trp) in all 3 patients. The sibling with hearing loss also harbored a homozygous mutation in GJB2, c.71G>A (p.Trp24*), which is an established cause of sensorineural hearing loss. CONCLUSIONS AND RELEVANCE: This family expands the ocular and systemic phenotypes associated with mutations in TRNT1, demonstrating phenotypic variability and highlighting the need for ophthalmic review of these patients.


Asunto(s)
Catarata/complicaciones , Síndromes de Inmunodeficiencia/complicaciones , Mutación , Nucleotidiltransferasas/genética , Distrofias Retinianas/complicaciones , Adolescente , Catarata/genética , Catarata/metabolismo , Niño , Preescolar , Exoma , Femenino , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Lactante , Masculino , Nucleotidiltransferasas/metabolismo , Linaje , Fenotipo , Distrofias Retinianas/genética , Distrofias Retinianas/metabolismo , Análisis de Secuencia de ADN
19.
PLoS One ; 10(7): e0132529, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26162006

RESUMEN

Primary open-angle glaucoma (POAG) is a clinically important and genetically heterogeneous cause of progressive vision loss as a result of retinal ganglion cell death. Here we have utilized trio-based, whole-exome sequencing to identify the genetic defect underlying an autosomal dominant form of adult-onset POAG segregating in an African-American family. Exome sequencing identified a novel missense variant (c.418C>T, p.Arg140Trp) in exon-5 of the gene coding for epidermal growth factor (EGF) containing fibulin-like extracellular matrix protein 1 (EFEMP1) that co-segregated with disease in the family. Linkage and haplotype analyses with microsatellite markers indicated that the disease interval overlapped a known POAG locus (GLC1H) on chromosome 2p. The p.Arg140Trp substitution was predicted in silico to have damaging effects on protein function and transient expression studies in cultured cells revealed that the Trp140-mutant protein exhibited increased intracellular accumulation compared with wild-type EFEMP1. In situ hybridization of the mouse eye with oligonucleotide probes detected the highest levels of EFEMP1 transcripts in the ciliary body, cornea, inner nuclear layer of the retina, and the optic nerve head. The recent finding that a common variant near EFEMP1 was associated with optic nerve-head morphology supports the possibility that the EFEMP1 variant identified in this POAG family may be pathogenic.


Asunto(s)
Segregación Cromosómica/genética , Exoma/genética , Proteínas de la Matriz Extracelular/genética , Genes Dominantes , Glaucoma de Ángulo Abierto/genética , Mutación Missense/genética , Análisis de Secuencia de ADN , Negro o Afroamericano/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de la Matriz Extracelular/química , Ojo/metabolismo , Familia , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética , Ligamiento Genético , Células HEK293 , Haplotipos/genética , Humanos , Hibridación in Situ , Masculino , Ratones , Datos de Secuencia Molecular , Transporte de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo
20.
Eur J Hum Genet ; 11(10): 784-93, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14512969

RESUMEN

Hereditary cataract is a clinically and genetically heterogeneous lens disease that accounts for a significant proportion of visual impairment and blindness in childhood. The alphaA-crystallin (CRYAA) gene (CRYAA) encodes a member of the small-heat-shock protein (sHSP) family of molecular chaperones and is primarily and abundantly expressed in the ocular lens. Here, we have used linkage analysis to identify a novel missense mutation in CRYAA that underlies an autosomal dominant form of 'nuclear' cataract segregating in a four-generation Caucasian family. A maximum two-point LOD score (Z(max)) of 2.19 (maximum recombination fraction, theta(max)=0) and multipoint Z(max) of 3.3 (theta(max)=0) was obtained at marker D21S1885. Haplotype analysis indicated that the disease gene lay in the approximately 2.7 Mb physical interval between D21S1912 and D21S1260 flanking CRYAA on 21q22.3. Sequence analysis identified a C --> T transition in exon 1 of CRYAA from affected individuals that was predicted to result in the nonconservative substitution of cysteine for arginine at codon 49 (R49C). Transfection studies of lens epithelial cells revealed that, unlike wild-type CRYAA, the R49C mutant protein was abnormally localized to the nucleus and failed to protect from staurosporine-induced apoptotic cell death. This study has identified the first dominant cataract mutation in CRYAA located outside the phylogenetically conserved 'alpha-crystallin core domain' of the sHSP family.


Asunto(s)
Cromosomas Humanos Par 21 , Cadena A de alfa-Cristalina/genética , Alelos , Apoptosis , Muerte Celular , Núcleo Celular/metabolismo , Separación Celular , Células Cultivadas , Microscopía por Crioelectrón , Análisis Mutacional de ADN , Cartilla de ADN/genética , ADN Complementario/metabolismo , Exones , Femenino , Citometría de Flujo , Genes Dominantes , Ligamiento Genético , Genotipo , Haplotipos , Humanos , Immunoblotting , Intrones , Escala de Lod , Masculino , Microscopía Fluorescente , Microscopía Inmunoelectrónica , Modelos Genéticos , Mutagénesis Sitio-Dirigida , Mutación , Mutación Missense , Linaje , Estructura Terciaria de Proteína , Estaurosporina/farmacología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA