Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Eur J Hum Genet ; 28(5): 576-586, 2020 05.
Article in English | MEDLINE | ID: mdl-31836858

ABSTRACT

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.


Subject(s)
Eye Diseases, Hereditary/genetics , Genetic Testing/standards , High-Throughput Nucleotide Sequencing/standards , Retinal Diseases/genetics , Sequence Analysis, DNA/standards , Adolescent , Adult , Aged , Child , Child, Preschool , Eye Diseases, Hereditary/diagnosis , Female , Genetic Testing/methods , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Male , Middle Aged , Phenotype , Retinal Diseases/diagnosis , Sensitivity and Specificity , Sequence Analysis, DNA/methods , Syndrome
2.
Hum Gene Ther Methods ; 30(2): 44-52, 2019 04.
Article in English | MEDLINE | ID: mdl-30963787

ABSTRACT

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.


Subject(s)
Bestrophins/physiology , Parvovirinae/genetics , Bestrophins/genetics , Dependovirus , Genetic Vectors , HEK293 Cells , Humans , Transduction, Genetic
3.
Dis Model Mech ; 9(11): 1317-1328, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27519691

ABSTRACT

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.


Subject(s)
Bestrophins/genetics , Bestrophins/metabolism , Cell Polarity , Epithelial Cells/metabolism , Epithelial Cells/pathology , Mutant Proteins/metabolism , Animals , Biotinylation , Cell Polarity/drug effects , Dogs , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Epithelial Cells/drug effects , Eye Diseases, Hereditary/genetics , Eye Diseases, Hereditary/pathology , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Models, Biological , Mutation/genetics , Patch-Clamp Techniques , Phenylbutyrates/pharmacology , Protein Transport/drug effects , Retinal Diseases/genetics , Retinal Diseases/pathology , Small Molecule Libraries/pharmacology , Transfection
4.
Orphanet J Rare Dis ; 10: 145, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26560304

ABSTRACT

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.


Subject(s)
Bruch Membrane/metabolism , Bruch Membrane/pathology , DNA-Binding Proteins/biosynthesis , Eye Abnormalities/diagnosis , Eye Abnormalities/metabolism , Joint Instability/congenital , Skin Abnormalities/diagnosis , Skin Abnormalities/metabolism , Transcription Factors/biosynthesis , Adolescent , Adult , Aged , Amino Acid Sequence , Cells, Cultured , Child , DNA-Binding Proteins/genetics , Eye Abnormalities/genetics , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Joint Instability/diagnosis , Joint Instability/genetics , Joint Instability/metabolism , Male , Middle Aged , Molecular Sequence Data , Skin Abnormalities/genetics , Transcription Factors/genetics , Young Adult
5.
Hum Mol Genet ; 24(23): 6565-79, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26395458

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , Ehlers-Danlos Syndrome/genetics , Histones/metabolism , Mutation , Retinal Vessels/pathology , Transcription Factors/genetics , Adult , Antigens, CD/genetics , Cadherins/genetics , Child , Collagen/genetics , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/pathology , Female , Fibroblasts/metabolism , Gene Ontology , Humans , Male , Methylation , Middle Aged , Nerve Growth Factors/genetics , Netrin-1 , Skin/cytology , Tumor Suppressor Proteins/genetics , Up-Regulation , Young Adult
6.
Proc Natl Acad Sci U S A ; 112(25): E3236-45, 2015 Jun 23.
Article in English | MEDLINE | ID: mdl-26056285

ABSTRACT

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.


Subject(s)
Coloboma/genetics , MicroRNAs/genetics , Retinal Dystrophies/genetics , Base Sequence , Coloboma/complications , Exome , Female , Genetic Linkage , Humans , Male , Pedigree , Retinal Dystrophies/complications , Sequence Homology, Nucleic Acid
7.
Exp Eye Res ; 121: 74-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24560797

ABSTRACT

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.


Subject(s)
Chloride Channels/genetics , Chloride Channels/metabolism , Eye Diseases, Hereditary/genetics , Eye Proteins/genetics , Eye Proteins/metabolism , Mutation, Missense , Protein Multimerization/physiology , Retinal Diseases/genetics , Adenoviridae/genetics , Animals , Bacterial Proteins/metabolism , Bestrophins , Blotting, Western , Choroid Diseases/genetics , Choroid Diseases/metabolism , Dogs , Electrophysiology , Eye Diseases, Hereditary/metabolism , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/metabolism , Gene Expression , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Luminescent Proteins/metabolism , Madin Darby Canine Kidney Cells/metabolism , Microscopy, Confocal , Patch-Clamp Techniques , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retinal Diseases/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Transfection , Vitelliform Macular Dystrophy/genetics , Vitelliform Macular Dystrophy/metabolism
8.
Invest Ophthalmol Vis Sci ; 55(2): 977-85, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24425855

ABSTRACT

PURPOSE: This study aimed to investigate the potency and specificity of short-interfering RNA (siRNA) treatment for TGFBI-Arg124Cys lattice corneal dystrophy type I (LCDI) using exogenous expression constructs in model systems and endogenous gene targeting in an ex vivo model using corneal epithelial cell cultures. METHODS: A panel of 19 TGFBI-Arg124Cys-specific siRNAs were assessed by a dual-luciferase reporter assay. Further assessment using pyrosequencing and qPCR was used to identify the lead siRNA; suppression of mutant TGFBIp expression was confirmed by Western blot and Congo red aggregation assays. An ex vivo model of LCDI was established using limbal biopsies from corneal dystrophy patients harboring the Arg124Cys mutation. Treatment efficiency of the siRNA was assessed for the inhibition of the mutant allele in the primary patient's corneal epithelial cells using pyrosequencing, quantitative PCR (qPCR), and an ELISA. RESULTS: A lead siRNA was identified, and demonstrated to be potent and specific in inhibiting the TGFBI-Arg124Cys mutant allele at the mRNA and protein levels. Besides high allele specificity, siRNA treatment achieved a 44% reduction of the endogenous Arg124Cys allele in an ex vivo model of LCDI. CONCLUSIONS: We have identified a lead siRNA specific to the TGFBI-Arg124Cys mutant allele associated with LCDI. Silencing of exogenous TGFBI was observed at mRNA and protein levels, and in an ex vivo model of LCDI with an efficient suppression of the endogenous mutant allele. This result indicates the potential of siRNA treatment as a personalized medicine approach for the management of heritable TGFBI-associated corneal dystrophies.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Gene Silencing , Point Mutation , RNA, Small Interfering/genetics , Transforming Growth Factor beta/genetics , Alleles , Blotting, Western , Cell Culture Techniques , Enzyme-Linked Immunosorbent Assay , Humans , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Transfection
9.
Orphanet J Rare Dis ; 8: 68, 2013 May 04.
Article in English | MEDLINE | ID: mdl-23642083

ABSTRACT

Brittle cornea syndrome (BCS) is an autosomal recessive disorder characterised by extreme corneal thinning and fragility. Corneal rupture can therefore occur either spontaneously or following minimal trauma in affected patients. Two genes, ZNF469 and PRDM5, have now been identified, in which causative pathogenic mutations collectively account for the condition in nearly all patients with BCS ascertained to date. Therefore, effective molecular diagnosis is now available for affected patients, and those at risk of being heterozygous carriers for BCS. We have previously identified mutations in ZNF469 in 14 families (in addition to 6 reported by others in the literature), and in PRDM5 in 8 families (with 1 further family now published by others). Clinical features include extreme corneal thinning with rupture, high myopia, blue sclerae, deafness of mixed aetiology with hypercompliant tympanic membranes, and variable skeletal manifestations. Corneal rupture may be the presenting feature of BCS, and it is possible that this may be incorrectly attributed to non-accidental injury. Mainstays of management include the prevention of ocular rupture by provision of protective polycarbonate spectacles, careful monitoring of visual and auditory function, and assessment for skeletal complications such as developmental dysplasia of the hip. Effective management depends upon appropriate identification of affected individuals, which may be challenging given the phenotypic overlap of BCS with other connective tissue disorders.


Subject(s)
Ehlers-Danlos Syndrome , Adolescent , DNA-Binding Proteins/genetics , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Ehlers-Danlos Syndrome/therapy , Eye Abnormalities , Female , Humans , Joint Instability/congenital , Mutation , Skin Abnormalities , Transcription Factors/genetics
10.
Mol Genet Metab ; 109(3): 289-95, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23680354

ABSTRACT

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.


Subject(s)
Ehlers-Danlos Syndrome/genetics , Exons , Extracellular Matrix/genetics , Gene Expression Regulation , Mutation , Transcription Factors/genetics , Adolescent , Child , Child, Preschool , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/therapy , Eye Abnormalities , Female , Genotype , Humans , Joint Instability/congenital , Skin Abnormalities
11.
Invest Ophthalmol Vis Sci ; 53(9): 5594-608, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22807293

ABSTRACT

PURPOSE: We investigated the retinal disease due to mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene in human patients and in an Rpgr conditional knockout (cko) mouse model. METHODS: XLRP patients with RPGR-ORF15 mutations (n = 35, ages at first visit 5-72 years) had clinical examinations, and rod and cone perimetry. Rpgr-cko mice, in which the proximal promoter and first exon were deleted ubiquitously, were back-crossed onto a BALB/c background, and studied with optical coherence tomography and electroretinography (ERG). Retinal histopathology was performed on a subset. RESULTS: Different patterns of rod and cone dysfunction were present in patients. Frequently, there were midperipheral losses with residual rod and cone function in central and peripheral retina. Longitudinal data indicated that central rod loss preceded peripheral rod losses. Central cone-only vision with no peripheral function was a late stage. Less commonly, patients had central rod and cone dysfunction, but preserved, albeit abnormal, midperipheral rod and cone vision. Rpgr-cko mice had progressive retinal degeneration detectable in the first months of life. ERGs indicated relatively equal rod and cone disease. At late stages, there was greater inferior versus superior retinal degeneration. CONCLUSIONS: RPGR mutations lead to progressive loss of rod and cone vision, but show different patterns of residual photoreceptor disease expression. Knowledge of the patterns should guide treatment strategies. Rpgr-cko mice had onset of degeneration at relatively young ages and progressive photoreceptor disease. The natural history in this model will permit preclinical proof-of-concept studies to be designed and such studies should advance progress toward human therapy.


Subject(s)
Eye Proteins/genetics , Genetic Diseases, X-Linked/genetics , Mutation/genetics , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Adolescent , Adult , Aged , Animals , Child , Child, Preschool , Disease Models, Animal , Female , Genetic Diseases, X-Linked/physiopathology , Humans , Male , Mice , Mice, Knockout , Middle Aged , Retinal Cone Photoreceptor Cells/physiology , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/physiopathology , Visual Acuity , Young Adult
14.
Invest Ophthalmol Vis Sci ; 52(11): 7859-65, 2011 Oct 04.
Article in English | MEDLINE | ID: mdl-21896847

ABSTRACT

PURPOSE: Primary open angle glaucoma is the most prevalent type of glaucoma and the leading cause of irreversible blindness worldwide. The genetic basis is poorly understood. Of 14 loci associated with this disease, only two genes have been identified, accounting for approximately 4% of cases. The authors investigated the genetic cause of primary open angle glaucoma in a large four-generation family with an apparent autosomal dominant mode of inheritance. METHODS: Twenty-three family members underwent comprehensive phenotyping by a single ophthalmologist, and the MYOC gene was sequenced in all affected family members for whom DNA was available. Parametric genomewide linkage analysis was performed on 10 affected family members and one unaffected family member. Within the critical region, mutation analysis of candidate genes LRP2BP, CYP4V2, and UFSP2 was carried out by direct sequencing. RESULTS: No mutations were identified in MYOC. Genomewide linkage analysis generated one significant LOD score of 3.1 (maximum affected-only LOD score of 2.8) centered on chromosome 4 at 4q35.1-q35.2, a critical region that does not contain any of the previously reported primary open angle glaucoma loci. A 1.866-Mb (7.2 cM) region was identified containing 17 known or hypothetical genes. No mutations were identified in the candidate genes LRPB2BP, CYP4V2, and UFSP2. CONCLUSIONS: This study identifies a new primary open angle glaucoma locus, GLC1Q, in a region on chromosome 4 not previously associated with glaucoma.


Subject(s)
Chromosomes, Human, Pair 4/genetics , Genes, Dominant , Genetic Loci , Glaucoma, Open-Angle/genetics , Adaptor Proteins, Signal Transducing , Adult , Aged , Carrier Proteins/genetics , Cysteine Endopeptidases/genetics , Cytochrome P-450 Enzyme System/genetics , Cytochrome P450 Family 4 , Cytoskeletal Proteins/genetics , DNA Mutational Analysis , Eye Proteins/genetics , Female , Genetic Linkage , Genome-Wide Association Study , Glaucoma, Open-Angle/diagnosis , Glycoproteins/genetics , Haplotypes , Humans , Lod Score , Low Density Lipoprotein Receptor-Related Protein-2 , Male , Middle Aged , Pedigree , Phenotype , Polymerase Chain Reaction
16.
Am J Hum Genet ; 88(6): 767-777, 2011 Jun 10.
Article in English | MEDLINE | ID: mdl-21664999

ABSTRACT

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.


Subject(s)
DNA-Binding Proteins/genetics , Extracellular Matrix/genetics , Transcription Factors/genetics , Child , DNA Mutational Analysis , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Extracellular Matrix/physiology , Eye Abnormalities , Female , Humans , Joint Instability/congenital , Male , Mutation , Pedigree , Skin Abnormalities
17.
FEBS Lett ; 585(14): 2187-92, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21689651

ABSTRACT

A novel gene, TMEM114, was annotated as a member of the claudin gene family and was subsequently associated as a cause of autosomal dominant cataract because of a translocation in its putative promoter. Our bioinformatic and molecular analyses of TMEM114, and the closely related TMEM235, demonstrate that these proteins are more closely related to members of the voltage dependent calcium channel gamma subunit family. TMEM114 and TMEM235 differed from claudins in terms of localisation in polarised epithelial cells and by the presence of N-linked glycans. By gene expression knockdown in Xenopus tropicalis we also demonstrate a role for Tmem114 in eye development.


Subject(s)
Claudins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Amino Acid Sequence , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cataract/genetics , Cell Line , Claudins/genetics , Embryo, Nonmammalian/anatomy & histology , Embryo, Nonmammalian/physiology , Eye/embryology , Eye/growth & development , Eye/metabolism , Eye/pathology , Humans , Membrane Glycoproteins/classification , Membrane Proteins/classification , Molecular Sequence Data , Phylogeny , Sequence Alignment , Xenopus
18.
Invest Ophthalmol Vis Sci ; 52(6): 3730-6, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21330666

ABSTRACT

PURPOSE: Autosomal recessive bestrophinopathy (ARB) is a retinal dystrophy affecting macular and retinal pigmented epithelium function resulting from homozygous or compound heterozygous mutations in BEST1. In this study we characterize the functional implications of missense bestrophin-1 mutations that cause ARB by investigating their effect on bestrophin-1's chloride conductance, cellular localization, and stability. METHODS: The chloride conductance of wild-type bestropin-1 and a series of ARB mutants were determined by whole-cell patch-clamping of transiently transfected HEK cells. The effect of ARB mutations on the cellular localization of bestrophin-1 was determined by confocal immunofluorescence on transiently transfected MDCK II cells that had been polarized on Transwell filters. Protein stability of wild-type and ARB mutant forms of bestrophin-l was determined by the addition of proteasomal or lysosomal inhibitors to transiently transfected MDCK II cells. Lysates were then analyzed by Western blot analysis. RESULTS: All ARB mutants investigated produced significantly smaller chloride currents compared to wild-type bestrophin-1. Additionally, co-transfection of compound heterozygous mutants abolished chloride conductance in contrast to co-transfections of a single mutant with wild-type bestrophin-l, reflecting the recessive nature of the condition. In control experiments, expression of two dominant vitelliform macular dystrophy mutants was shown to inhibit wild-type currents. Cellular localization of ARB mutants demonstrated that the majority did not traffic correctly to the plasma membrane and that five of these seven mutants were rapidly degraded by the proteasome. Two ARB-associated mutants (p.D312N and p.V317M) that were not trafficked correctly nor targeted to the proteasome had a distinctive appearance, possibly indicative of aggresome or aggresome-like inclusion bodies. CONCLUSIONS: Differences in cellular processing mechanisms for different ARB associated mutants lead to the same disease phenotype. The existence of distinct pathogenic disease mechanisms has important ramifications for potential gene replacement therapies since we show that missense mutations associated with an autosomal recessive disease have a pathogenic influence beyond simple loss of function.


Subject(s)
Chloride Channels/genetics , Eye Proteins/genetics , Genes, Recessive , Mutation, Missense/physiology , Retinal Degeneration/genetics , Animals , Bestrophins , Blotting, Western , Cell Culture Techniques , Dogs , Humans , Kidney/cytology , Kidney/embryology , Microscopy, Confocal , Patch-Clamp Techniques , Phenotype , Retinal Pigment Epithelium/metabolism , Transfection
20.
Am J Hum Genet ; 86(5): 686-95, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20398886

ABSTRACT

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.


Subject(s)
Eye/metabolism , Mutation , Photoreceptor Cells, Vertebrate/metabolism , Proteins/genetics , Retinitis Pigmentosa/genetics , Blindness/genetics , Cilia/genetics , Cilia/metabolism , Exons , Eye Proteins/genetics , Homozygote , Humans , Mutation, Missense , Retinitis Pigmentosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...