Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 112(25): E3236-45, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26056285

RESUMO

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.


Assuntos
Coloboma/genética , MicroRNAs/genética , Distrofias Retinianas/genética , Sequência de Bases , Coloboma/complicações , Exoma , Feminino , Ligação Genética , Humanos , Masculino , Linhagem , Distrofias Retinianas/complicações , Homologia de Sequência do Ácido Nucleico
2.
Hum Mol Genet ; 24(23): 6565-79, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26395458

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Síndrome de Ehlers-Danlos/genética , Histonas/metabolismo , Mutação , Vasos Retinianos/patologia , Fatores de Transcrição/genética , Adulto , Antígenos CD/genética , Caderinas/genética , Criança , Colágeno/genética , Síndrome de Ehlers-Danlos/metabolismo , Síndrome de Ehlers-Danlos/patologia , Feminino , Fibroblastos/metabolismo , Ontologia Genética , Humanos , Masculino , Metilação , Pessoa de Meia-Idade , Fatores de Crescimento Neural/genética , Netrina-1 , Pele/citologia , Proteínas Supressoras de Tumor/genética , Regulação para Cima , Adulto Jovem
3.
Am J Hum Genet ; 88(6): 767-777, 2011 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-21664999

RESUMO

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.


Assuntos
Proteínas de Ligação a DNA/genética , Matriz Extracelular/genética , Fatores de Transcrição/genética , Criança , Análise Mutacional de DNA , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/patologia , Matriz Extracelular/fisiologia , Anormalidades do Olho , Feminino , Humanos , Instabilidade Articular/congênito , Masculino , Mutação , Linhagem , Anormalidades da Pele
4.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24560797

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Oftalmopatias Hereditárias/genética , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Mutação de Sentido Incorreto , Multimerização Proteica/fisiologia , Doenças Retinianas/genética , Adenoviridae/genética , Animais , Proteínas de Bactérias/metabolismo , Bestrofinas , Western Blotting , Doenças da Coroide/genética , Doenças da Coroide/metabolismo , Cães , Eletrofisiologia , Oftalmopatias Hereditárias/metabolismo , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/metabolismo , Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Proteínas Luminescentes/metabolismo , Células Madin Darby de Rim Canino/metabolismo , Microscopia Confocal , Técnicas de Patch-Clamp , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Doenças Retinianas/metabolismo , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Transfecção , Distrofia Macular Viteliforme/genética , Distrofia Macular Viteliforme/metabolismo
5.
Am J Hum Genet ; 86(5): 686-95, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20398886

RESUMO

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.


Assuntos
Olho/metabolismo , Mutação , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas/genética , Retinose Pigmentar/genética , Cegueira/genética , Cílios/genética , Cílios/metabolismo , Éxons , Proteínas do Olho/genética , Homozigoto , Humanos , Mutação de Sentido Incorreto , Retinose Pigmentar/metabolismo
6.
Mol Genet Metab ; 109(3): 289-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23680354

RESUMO

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.


Assuntos
Síndrome de Ehlers-Danlos/genética , Éxons , Matriz Extracelular/genética , Regulação da Expressão Gênica , Mutação , Fatores de Transcrição/genética , Adolescente , Criança , Pré-Escolar , Análise Mutacional de DNA , Proteínas de Ligação a DNA/genética , Síndrome de Ehlers-Danlos/diagnóstico , Síndrome de Ehlers-Danlos/terapia , Anormalidades do Olho , Feminino , Genótipo , Humanos , Instabilidade Articular/congênito , Anormalidades da Pele
7.
Am J Hum Genet ; 84(6): 801-6, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19481195

RESUMO

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.


Assuntos
Proteínas do Citoesqueleto/genética , Transtornos do Crescimento/genética , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Ubiquitinação , Adolescente , Células Cultivadas , Criança , Pré-Escolar , Proteínas Culina/genética , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto , Feminino , Humanos , Lactente , Rim/citologia , Rim/metabolismo , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , RNA Interferente Pequeno/farmacologia , Síndrome
8.
Am J Hum Genet ; 85(5): 581-92, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19853238

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Proteínas do Olho/genética , Mutação de Sentido Incorreto , Epitélio Pigmentado da Retina/patologia , Retinose Pigmentar/etiologia , Retinose Pigmentar/genética , Sequência de Aminoácidos , Bestrofinas , Linhagem Celular , Cromossomos Humanos Par 11 , Sequência Conservada , Éxons , Feminino , Genes Dominantes , Genes Recessivos , Ligação Genética , Homozigoto , Humanos , Rim/citologia , Escore Lod , Masculino , Dados de Sequência Molecular , Núcleo Familiar , Linhagem , Polimorfismo de Nucleotídeo Único , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Homologia de Sequência de Aminoácidos
9.
Am J Hum Genet ; 82(1): 19-31, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18179881

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Proteínas do Olho/genética , Doenças Retinianas/genética , Adolescente , Adulto , Sequência de Aminoácidos , Bestrofinas , Linhagem Celular , Criança , Pré-Escolar , Canais de Cloreto/química , Códon sem Sentido , Proteínas do Olho/química , Feminino , Genes Recessivos , Humanos , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Transfecção
10.
Mol Vis ; 16: 2916-22, 2010 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21203346

RESUMO

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.


Assuntos
Canais de Cloreto/genética , Códon/genética , Proteínas do Olho/genética , Genes Recessivos/genética , Mutação/genética , Splicing de RNA/genética , Distrofias Retinianas/genética , Adulto , Bestrofinas , Criança , Biologia Computacional , Feminino , Fibronectinas/química , Fibronectinas/genética , Fluorescência , Fundo de Olho , Células HEK293 , Humanos , Masculino , Pessoa de Meia-Idade , Estrutura Terciária de Proteína , Adulto Jovem , alfa-Globinas/genética
11.
Eur J Hum Genet ; 28(5): 576-586, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31836858

RESUMO

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.


Assuntos
Oftalmopatias Hereditárias/genética , Testes Genéticos/normas , Sequenciamento de Nucleotídeos em Larga Escala/normas , Doenças Retinianas/genética , Análise de Sequência de DNA/normas , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Oftalmopatias Hereditárias/diagnóstico , Feminino , Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Fenótipo , Doenças Retinianas/diagnóstico , Sensibilidade e Especificidade , Análise de Sequência de DNA/métodos , Síndrome
12.
Hum Gene Ther Methods ; 30(2): 44-52, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30963787

RESUMO

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.


Assuntos
Bestrofinas/fisiologia , Parvovirinae/genética , Bestrofinas/genética , Dependovirus , Vetores Genéticos , Células HEK293 , Humanos , Transdução Genética
13.
Hum Mutat ; 29(8): 1017-27, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18425797

RESUMO

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.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Síndrome de Klippel-Feil/genética , Sequência de Aminoácidos , Animais , Proteínas Morfogenéticas Ósseas/química , Inversão Cromossômica , Análise Mutacional de DNA , Feminino , Fator 6 de Diferenciação de Crescimento , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Linhagem , Alinhamento de Sequência , Coluna Vertebral/anormalidades , Xenopus laevis
15.
Invest Ophthalmol Vis Sci ; 47(1): 348-56, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16384984

RESUMO

PURPOSE: The present study examined the developmental and tissue expression of the retinitis pigmentosa GTPase regulator (RPGR) gene in Xenopus laevis. METHODS: The cDNA for X. laevis RPGR (XRPGR) was isolated from adult eye mRNA by reverse transcription-polymerase chain reaction (RT-PCR) and rapid amplification of cDNA ends. The deduced peptide sequence was aligned with RPGR orthologues. Gene expression was examined by whole-mount in situ hybridization and RT-PCR. The localization of XRPGR in X. laevis photoreceptor cells and XTC-2 cells was determined by immunostaining. RESULTS: The XRPGR(ex1-19) isoform encodes a protein of 727 amino acids containing an RCC1 domain and a C-terminal isoprenylation anchorage motif. It shares 33% to 41% amino acid identity with human, mouse, and dog RPGR. The C-terminal exon of the alternatively spliced RPGR(ORF15) isoform is also conserved across species. XRPGR is expressed at the earliest stages of X. laevis development and persists into adulthood, where expression is highest in the eye. XRPGR is expressed in presumptive eye fields (stages 18 to 22), becoming largely restricted to the central retina (stages 28 to 40). XRPGR protein colocalizes with beta-tubulin at the X. laevis ciliary axoneme and with gamma-tubulin at centrosomes in XTC-2 cells. CONCLUSIONS: XRPGR is widely expressed throughout development but shows highest expression after the appearance of the eye primordium and persists in the eye into adulthood. The data are consistent with XRPGR expression in a single microtubular organelle-the centriole or basal body and associated ciliary transitional zone found in modified sensory cilia of photoreceptors and motile cilia.


Assuntos
Embrião não Mamífero/metabolismo , Proteínas do Olho/genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Troca do Nucleotídeo Guanina/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Proteínas de Xenopus/genética , Sequência de Aminoácidos , Animais , Linhagem Celular , Centrossomo/metabolismo , Clonagem Molecular , DNA Complementar/análise , Proteínas do Olho/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hibridização In Situ , Dados de Sequência Molecular , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Tubulina (Proteína)/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia
16.
Trends Mol Med ; 11(10): 449-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16153893

RESUMO

Molecular genetics has provided relatively few insights into late-onset eye disorders, but epidemiological data indicate that genetic factors are important in some late-onset eye disorders that cause major health burdens. Much clinical genetic research is based on the belief that developmental and late-onset disorders are not necessarily the result of defects in different genes, but are often caused by different mutations in the same collection of genes. Thus, mutations that either abolish or radically change gene function might cause early-onset disorders, whereas more-subtle changes in gene expression might underlie late-onset diseases. We present arguments and examples that indicate that this principle might be a fruitful guide to investigating the causes of late-onset eye disorders.


Assuntos
Idade de Início , Oftalmopatias Hereditárias/etiologia , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Fenótipo , Humanos , Mutação/genética , Fatores de Transcrição/genética
17.
Am J Ophthalmol ; 141(2): 418-20, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16458719

RESUMO

PURPOSE: To present the detailed phenotype of a subject with MRCS (microcornea, retinal dystrophy, cataract, and posterior staphyloma) syndrome and to investigate the underlying molecular genetic basis. DESIGN: Interventional case report. METHODS: Clinical examination, electrophysiologic assessment, B-scan ultrasonography, and mutation screening of the gene VMD2. The protocol of the study was approved by the local ethics committee and informed consent was obtained. RESULTS: A 12-year-old boy was identified with bilateral microcornea, rod-cone dystrophy, congenital cataracts, and posterior staphylomata associated with high myopia (MRCS). Mutation screening failed to identify disease-causing sequence variants in VMD2, the gene associated with MRCS syndrome. All previous subjects have had pathogenic VMD2 sequence alterations. CONCLUSIONS: We present a further report of the MRCS syndrome and provide evidence in support of genetic heterogeneity in this phenotype.


Assuntos
Catarata/congênito , Córnea/anormalidades , Heterogeneidade Genética , Retinose Pigmentar/genética , Doenças da Esclera/genética , Bestrofinas , Criança , Canais de Cloreto , Análise Mutacional de DNA , Dilatação Patológica , Eletrorretinografia , Proteínas do Olho/genética , Humanos , Masculino , Fenótipo , Síndrome
18.
Dis Model Mech ; 9(11): 1317-1328, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27519691

RESUMO

Autosomal recessive bestrophinopathy (ARB) is a retinopathy caused by mutations in the bestrophin-1 protein, which is thought to function as a Ca2+-gated Cl- channel in the basolateral surface of the retinal pigment epithelium (RPE). Using a stably transfected polarised epithelial cell model, we show that four ARB mutant bestrophin-1 proteins were mislocalised and subjected to proteasomal degradation. In contrast to the wild-type bestrophin-1, each of the four mutant proteins also failed to conduct Cl- ions in transiently transfected cells as determined by whole-cell patch clamp. We demonstrate that a combination of two clinically approved drugs, bortezomib and 4-phenylbutyrate (4PBA), successfully restored the expression and localisation of all four ARB mutant bestrophin-1 proteins. Importantly, the Cl- conductance function of each of the mutant bestrophin-1 proteins was fully restored to that of wild-type bestrophin-1 by treatment of cells with 4PBA alone. The functional rescue achieved with 4PBA is significant because it suggests that this drug, which is already approved for long-term use in infants and adults, might represent a promising therapy for the treatment of ARB and other bestrophinopathies resulting from missense mutations in BEST1.


Assuntos
Bestrofinas/genética , Bestrofinas/metabolismo , Polaridade Celular , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas Mutantes/metabolismo , Animais , Biotinilação , Polaridade Celular/efeitos dos fármacos , Cães , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Epiteliais/efeitos dos fármacos , Oftalmopatias Hereditárias/genética , Oftalmopatias Hereditárias/patologia , Células HEK293 , Humanos , Células Madin Darby de Rim Canino , Modelos Biológicos , Mutação/genética , Técnicas de Patch-Clamp , Fenilbutiratos/farmacologia , Transporte Proteico/efeitos dos fármacos , Doenças Retinianas/genética , Doenças Retinianas/patologia , Bibliotecas de Moléculas Pequenas/farmacologia , Transfecção
20.
Orphanet J Rare Dis ; 10: 145, 2015 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-26560304

RESUMO

BACKGROUND: Brittle cornea syndrome (BCS) is a rare, generalized connective tissue disorder associated with extreme corneal thinning and a high risk of corneal rupture. Recessive mutations in transcription factors ZNF469 and PRDM5 cause BCS. Both transcription factors are suggested to act on a common pathway regulating extracellular matrix genes, particularly fibrillar collagens. We identified bilateral myopic choroidal neovascularization as the presenting feature of BCS in a 26-year-old-woman carrying a novel PRDM5 mutation (p.Glu134*). We performed immunohistochemistry of anterior and posterior segment ocular tissues, as expression of PRDM5 in the eye has not been described, or the effects of PRDM5-associated disease on the retina, particularly the extracellular matrix composition of Bruch's membrane. METHODS: Immunohistochemistry using antibodies against PRDM5, collagens type I, III, and IV was performed on the eyes of two unaffected controls and two patients (both with Δ9-14 PRDM5). Expression of collagens, integrins, tenascin and fibronectin in skin fibroblasts of a BCS patient with a novel p.Glu134* PRDM5 mutation was assessed using immunofluorescence. RESULTS: PRDM5 is expressed in the corneal epithelium and retina. We observe reduced expression of major components of Bruch's membrane in the eyes of two BCS patients with a PRDM5 Δ9-14 mutation. Immunofluorescence performed on skin fibroblasts from a patient with p.Glu134* confirms the generalized nature of extracellular matrix abnormalities in BCS. CONCLUSIONS: PDRM5-related disease is known to affect the cornea, skin and joints. Here we demonstrate, to the best of our knowledge for the first time, that PRDM5 localizes not only in the human cornea, but is also widely expressed in the retina. Our findings suggest that ECM abnormalities in PRDM5-associated disease are more widespread than previously reported.


Assuntos
Lâmina Basilar da Corioide/metabolismo , Lâmina Basilar da Corioide/patologia , Proteínas de Ligação a DNA/biossíntese , Anormalidades do Olho/diagnóstico , Anormalidades do Olho/metabolismo , Instabilidade Articular/congênito , Anormalidades da Pele/diagnóstico , Anormalidades da Pele/metabolismo , Fatores de Transcrição/biossíntese , Adolescente , Adulto , Idoso , Sequência de Aminoácidos , Células Cultivadas , Criança , Proteínas de Ligação a DNA/genética , Anormalidades do Olho/genética , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Instabilidade Articular/diagnóstico , Instabilidade Articular/genética , Instabilidade Articular/metabolismo , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Anormalidades da Pele/genética , Fatores de Transcrição/genética , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA