Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
JAMA Ophthalmol ; 134(9): 1049-53, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27389523

ABSTRACT

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.


Subject(s)
Cataract/complications , Immunologic Deficiency Syndromes/complications , Mutation , Nucleotidyltransferases/genetics , Retinal Dystrophies/complications , Adolescent , Cataract/genetics , Cataract/metabolism , Child , Child, Preschool , Exome , Female , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism , Infant , Male , Nucleotidyltransferases/metabolism , Pedigree , Phenotype , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Sequence Analysis, DNA
2.
PLoS One ; 10(7): e0132529, 2015.
Article in English | MEDLINE | ID: mdl-26162006

ABSTRACT

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.


Subject(s)
Chromosome Segregation/genetics , Exome/genetics , Extracellular Matrix Proteins/genetics , Genes, Dominant , Glaucoma, Open-Angle/genetics , Mutation, Missense/genetics , Sequence Analysis, DNA , Black or African American/genetics , Amino Acid Sequence , Animals , Base Sequence , Extracellular Matrix Proteins/chemistry , Eye/metabolism , Family , Female , Gene Expression Regulation , Genetic Association Studies , Genetic Linkage , HEK293 Cells , Haplotypes/genetics , Humans , In Situ Hybridization , Male , Mice , Molecular Sequence Data , RNA Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
Hum Genomics ; 8: 19, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25403472

ABSTRACT

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.


Subject(s)
Cataract/genetics , Connexins/genetics , Exome , gamma-Crystallins/genetics , Cataract/pathology , Connexins/metabolism , Female , Gene Expression Profiling , Genetic Association Studies , Heterozygote , Humans , Male , Mutation, Missense , Pedigree , Polymorphism, Single Nucleotide , gamma-Crystallins/metabolism
5.
PLoS One ; 9(8): e104000, 2014.
Article in English | MEDLINE | ID: mdl-25090642

ABSTRACT

Inherited forms of cataract are a clinically important and genetically heterogeneous cause of visual impairment that usually present at an early age with or without systemic and/or other ocular abnormalities. Here we have identified a new locus for inherited cataract and high-tension glaucoma with variable anterior segment defects, and characterized an underlying mutation in the gene coding for transient receptor potential cation channel, subfamily M, member-3 (TRPM3, melastatin-2). Genome-wide linkage analysis mapped the ocular disease locus to the pericentric region of human chromosome 9. Whole exome and custom-target next-generation sequencing detected a heterozygous A-to-G transition in exon-3 of TRPM3 that co-segregated with disease. As a consequence of alternative splicing this missense mutation was predicted to result in the substitution of isoleucine-to-methionine at codon 65 (c.195A>G; p.I65 M) of TRPM3 transcript variant 9, and at codon 8 (c.24A>G; p.I8 M) of a novel TRPM3 transcript variant expressed in human lens. In both transcript variants the I-to-M substitution was predicted in silico to exert damaging effects on protein function. Furthermore, transient expression studies of a recombinant TRPM3-GFP reporter product predicted that the I-to-M substitution introduced an alternative translation start-site located 89 codons upstream from the native initiator methionine found in eight other TRPM3 transcript variants (1-8). Collectively, these studies have provided the first evidence that TRPM3 is associated with inherited ocular disease in humans, and further provide support for the important role of this cation channel in normal eye development.


Subject(s)
Cataract/genetics , Glaucoma/genetics , Lens, Crystalline/metabolism , Mutation, Missense , TRPM Cation Channels/genetics , Adult , Alternative Splicing , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Cataract/complications , Cataract/congenital , Cataract/pathology , Chromosomes, Human, Pair 9/chemistry , Codon , Exome , Exons , Female , Gene Expression , Genes, Reporter , Genetic Linkage , Genome-Wide Association Study , Glaucoma/complications , Glaucoma/congenital , Glaucoma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Lens, Crystalline/pathology , Male , Molecular Sequence Data , Pedigree
6.
Ophthalmology ; 121(6): 1174-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24480711

ABSTRACT

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.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/pathology , Cytochrome P-450 Enzyme System/genetics , Mutation, Missense , Polymorphism, Single Nucleotide , Retinal Diseases/genetics , Retinal Diseases/pathology , Adult , Aged , Comparative Genomic Hybridization , Cytochrome P450 Family 4 , DNA Mutational Analysis , Electroretinography , Exons/genetics , Female , Fluorescein Angiography , Genetic Association Studies , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Polymerase Chain Reaction , Retinal Pigment Epithelium/pathology , Tomography, Optical Coherence , Visual Acuity/physiology , Young Adult
7.
Hum Mutat ; 35(3): 289-93, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24375934

ABSTRACT

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.


Subject(s)
Frameshift Mutation , Homozygote , Obesity/genetics , Proteins/genetics , Retinitis Pigmentosa/genetics , Adaptor Proteins, Signal Transducing , Child , Chromosome Mapping , Consanguinity , Eye Proteins/genetics , Female , Genes, Recessive , Homeostasis , Humans , Male , Pedigree , United Kingdom , White People/genetics
8.
Hum Mutat ; 34(11): 1537-1546, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23946133

ABSTRACT

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.


Subject(s)
Eye Proteins/genetics , Genetic Association Studies , Leber Congenital Amaurosis/genetics , Microtubule-Associated Proteins/genetics , Mutation , Retinitis Pigmentosa/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , Consanguinity , Female , Fluorescein Angiography , Genotype , Humans , Infant , Infant, Newborn , Leber Congenital Amaurosis/diagnosis , Male , Middle Aged , Pedigree , Phenotype , Retina/pathology , Retinitis Pigmentosa/diagnosis , Young Adult
9.
Ophthalmology ; 120(11): 2324-31, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23769331

ABSTRACT

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.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Mutation , Polymorphism, Single Nucleotide , Adolescent , Adult , Alleles , Child , DNA Mutational Analysis , Electroretinography , Female , Fluorescein Angiography , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Male , Middle Aged , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Retrospective Studies , Stargardt Disease , Tomography, Optical Coherence , Visual Acuity/physiology
10.
Am J Ophthalmol ; 155(6): 1075-1088.e13, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23499370

ABSTRACT

PURPOSE: To investigate the clinical and electrophysiologic natural history of Stargardt disease and correlate with the genotype. DESIGN: Cohort study of 59 patients. METHODS: Clinical history, examination, and electrophysiologic assessment were undertaken in a longitudinal survey. Patients were classified into 3 groups based on electrophysiologic findings, as previously published: Group 1 had dysfunction confined to the macula; Group 2 had macular and generalized cone system dysfunction; and Group 3 had macular and both generalized cone and rod system dysfunction. At baseline, there were 27 patients in Group 1, 17 in Group 2, and 15 in Group 3. Amplitude reduction of >50% in the relevant electroretinogram (ERG) component or a peak time shift of >3 ms for the 30 Hz flicker ERG or bright flash a-wave was considered clinically significant ERG deterioration. Molecular screening of ABCA4 was undertaken. RESULTS: The mean age at baseline was 31.7 years, with the mean follow-up interval being 10.5 years. A total of 22% of patients from Group 1 showed ERG group transition during follow-up, with 11% progressing to Group 2 and 11% to Group 3. Forty-seven percent of patients in Group 2 progressed to Group 3. There was clinically significant ERG deterioration in 54% of all subjects: 22% of Group 1, 65% of Group 2, and 100% of Group 3. At least 1 disease-causing ABCA4 variant was identified in 47 patients. CONCLUSIONS: All patients with initial rod ERG involvement demonstrated clinically significant electrophysiologic deterioration; only 20% of patients with normal full-field ERGs at baseline showed clinically significant progression. Such data assist counseling by providing more accurate prognostic information and are also highly relevant in the design, patient selection, and monitoring of potential therapeutic interventions.


Subject(s)
Macular Degeneration/congenital , Photoreceptor Cells, Vertebrate/pathology , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Cohort Studies , DNA Mutational Analysis , Dark Adaptation , Disease Progression , Electroretinography , Female , Follow-Up Studies , Genetic Association Studies , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Macular Degeneration/physiopathology , Male , Middle Aged , Photic Stimulation , Stargardt Disease , Visual Acuity/physiology , Young Adult
11.
Neurology ; 80(11): 1062-4, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23427326

ABSTRACT

Alpha-synuclein (SNCA) is central to the pathogenesis of Parkinson disease (PD), with 3 missense mutations reported to date. We report a novel mutation (p.H50Q) in a pathologically proven case.


Subject(s)
Mutation, Missense/genetics , Parkinson Disease/diagnosis , Parkinson Disease/genetics , alpha-Synuclein/genetics , Aged, 80 and over , Amino Acid Sequence , Amino Acid Substitution/genetics , Fatal Outcome , Female , Humans , Molecular Sequence Data
12.
Hum Mutat ; 34(3): 506-14, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23281133

ABSTRACT

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.


Subject(s)
Eye Proteins/genetics , Macular Degeneration/genetics , Retina/pathology , Retinitis Pigmentosa/genetics , Adult , Aged , Chromosome Mapping , Cloning, Molecular , Cohort Studies , Consanguinity , Exome , Female , Genes, Recessive , Genetic Loci , Haplotypes , Heterozygote , Homozygote , Humans , Male , Microarray Analysis , Middle Aged , Mutation, Missense , Pedigree , Phenotype , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
13.
Nat Genet ; 44(9): 1040-5, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22842227

ABSTRACT

Leber congenital amaurosis (LCA) is an infantile-onset form of inherited retinal degeneration characterized by severe vision loss(1,2). Two-thirds of LCA cases are caused by mutations in 17 known disease-associated genes(3) (Retinal Information Network (RetNet)). Using exome sequencing we identified a homozygous missense mutation (c.25G>A, p.Val9Met) in NMNAT1 that is likely to be disease causing in two siblings of a consanguineous Pakistani kindred affected by LCA. This mutation segregated with disease in the kindred, including in three other children with LCA. NMNAT1 resides in the previously identified LCA9 locus and encodes the nuclear isoform of nicotinamide mononucleotide adenylyltransferase, a rate-limiting enzyme in nicotinamide adenine dinucleotide (NAD(+)) biosynthesis(4,5). Functional studies showed that the p.Val9Met alteration decreased NMNAT1 enzyme activity. Sequencing NMNAT1 in 284 unrelated families with LCA identified 14 rare mutations in 13 additional affected individuals. These results are the first to link an NMNAT isoform to disease in humans and indicate that NMNAT1 mutations cause LCA.


Subject(s)
Leber Congenital Amaurosis/genetics , Mutation , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Base Sequence , Case-Control Studies , Child , Child, Preschool , DNA Mutational Analysis , Family , Female , Genetic Predisposition to Disease , Humans , Leber Congenital Amaurosis/complications , Male , Mutation/physiology , Nicotinamide-Nucleotide Adenylyltransferase/physiology , Pedigree , Retinal Degeneration/complications , Retinal Degeneration/diagnosis , Retinal Degeneration/genetics
14.
Invest Ophthalmol Vis Sci ; 53(7): 3927-38, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22570351

ABSTRACT

PURPOSE: To report novel variants and characterize the phenotype associated with the autosomal recessive retinal dystrophy caused by mutations in the lecithin retinol acyltransferase (LRAT) gene. METHODS: A total of 149 patients with Leber's congenital amaurosis (LCA) or early onset retinal dystrophy were screened for mutations in LCA-associated genes using an arrayed-primer extension (APEX) genotyping microarray (Asper Ophthalmics). LRAT sequencing was subsequently performed in this 148-patient panel. Patients identified with mutations underwent further detailed phenotyping. RESULTS: APEX analysis identified one patient with a previously reported homozygous LRAT mutation. Sequencing of the panel identified three additional patients with novel homozygous LRAT mutations in exon 2. All four patients had severe progressive nyctalopia, visual field constriction, and photophilia in childhood. Visual acuity ranged from 0.22 logMAR to hand motion. Funduscopy revealed severe retinal pigment epithelial atrophy and minimal retinal pigmentation. Asteroid hyalosis and macular epiretinal fibrosis were frequent. All demonstrated reduced fundus autofluorescence. Optical coherence tomography identified disrupted retinal lamination, outer-retinal debris, and an unidentifiable photoreceptor layer in two cases. Full-field electroretinograms were undetectable or showed severe rod-cone dysfunction. Photopic perimetry revealed severe visual field constriction. Dark-adapted perimetry demonstrated markedly reduced photoreceptor sensitivity. Dark-adapted spectral sensitivity measurements identified functioning rods in two of three patients. All three had severely reduced L- and M-cone sensitivity and poor color discrimination. CONCLUSIONS: LRAT mutations cause a severe, early childhood onset, progressive retinal dystrophy. Phenotypic similarities to the retinal dysfunction associated with RPE-specific protein 65 kDa mutations, another visual cycle gene, suggest that LRAT deficiency may show a good response to novel therapies.


Subject(s)
Acyltransferases/genetics , Mutation , Retinal Dystrophies/genetics , Acyltransferases/deficiency , Child, Preschool , Consanguinity , DNA Mutational Analysis , DNA Primers , Dark Adaptation , Electroretinography , Female , Humans , Infant , Leber Congenital Amaurosis/genetics , Male , Molecular Biology , Night Blindness/genetics , Oligonucleotide Array Sequence Analysis , Phenotype , Photoreceptor Cells, Vertebrate/pathology , Retinal Dystrophies/diagnosis , Retinal Dystrophies/enzymology , Tomography, Optical Coherence , Visual Acuity/physiology , Visual Field Tests , Visual Fields
15.
PLoS One ; 7(3): e32330, 2012.
Article in English | MEDLINE | ID: mdl-22412862

ABSTRACT

Leber Congenital Amaurosis (LCA) and Early Childhood Onset Severe Retinal Dystrophy are clinically and genetically heterogeneous retinal disorders characterised by visual impairment and nystagmus from birth or early infancy. We investigated the prevalence of sequence variants in AIPL1 in a large cohort of such patients (n = 392) and probed the likelihood of disease-causation of the identified variants, subsequently undertaking a detailed assessment of the phenotype of patients with disease-causing mutations. Genomic DNA samples were screened for known variants in the AIPL1 gene using a microarray LCA chip, with 153 of these cases then being directly sequenced. The assessment of disease-causation of identified AIPL1 variants included segregation testing, assessing evolutionary conservation and in silico predictions of pathogenicity. The chip identified AIPL1 variants in 12 patients. Sequencing of AIPL1 in 153 patients and 96 controls found a total of 46 variants, with 29 being novel. In silico analysis suggested that only 6 of these variants are likely to be disease-causing, indicating a previously unrecognized high degree of polymorphism. Seven patients were identified with biallelic changes in AIPL1 likely to be disease-causing. In the youngest subject, electroretinography revealed reduced cone photoreceptor function, but rod responses were within normal limits, with no measurable ERG in other patients. An increasing degree and extent of peripheral retinal pigmentation and degree of maculopathy was noted with increasing age in our series. AIPL1 is significantly polymorphic in both controls and patients, thereby complicating the establishment of disease-causation of identified variants. Despite the associated phenotype being characterised by early-onset severe visual loss in our patient series, there was some evidence of a degree of retinal structural and functional preservation, which was most marked in the youngest patient in our cohort. This data suggests that there are patients who have a reasonable window of opportunity for gene therapy in childhood.


Subject(s)
Carrier Proteins/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Fluorescein Angiography , Gene Expression Profiling , Genetic Therapy , Humans , Leber Congenital Amaurosis/diagnosis , Leber Congenital Amaurosis/therapy , Mutation , RNA Splice Sites , Young Adult
17.
Am J Hum Genet ; 89(6): 782-91, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22137173

ABSTRACT

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.


Subject(s)
Eye Abnormalities/genetics , Group V Phospholipases A2/genetics , Homozygote , Mutation, Missense , Retina/abnormalities , Adult , Aged, 80 and over , Alternative Splicing , Amino Acid Sequence , Base Sequence , Child , Consanguinity , Female , Genetic Association Studies , Group V Phospholipases A2/metabolism , Humans , Male , Molecular Sequence Data , Pedigree , Polymorphism, Single Nucleotide , Protein Transport , Retina/metabolism
18.
Mol Vis ; 17: 2706-16, 2011.
Article in English | MEDLINE | ID: mdl-22065924

ABSTRACT

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.


Subject(s)
Alcohol Oxidoreductases/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Retina/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Age of Onset , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Exons , Female , Genetic Linkage , Genetic Testing , Genotype , Humans , Infant , Leber Congenital Amaurosis/diagnosis , Male , Mutation , Oligonucleotide Array Sequence Analysis , Pedigree , Phenotype , Polymerase Chain Reaction , Retina/pathology , Retinal Degeneration/diagnosis , Retinitis Pigmentosa/diagnosis , United Kingdom
19.
Am J Hum Genet ; 89(1): 183-90, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21763485

ABSTRACT

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.


Subject(s)
Genes, Recessive , Leber Congenital Amaurosis/genetics , Potassium Channels, Inwardly Rectifying/genetics , Adult , Amino Acid Sequence , Codon, Nonsense , Exons , Female , Genetic Variation , Homozygote , Humans , Leber Congenital Amaurosis/pathology , Male , Molecular Sequence Data , Night Blindness/genetics , Pedigree , Potassium Channels/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Structure, Quaternary , Protein Structure, Secondary , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/pathology
20.
Invest Ophthalmol Vis Sci ; 52(6): 3032-8, 2011 May 09.
Article in English | MEDLINE | ID: mdl-21310915

ABSTRACT

PURPOSE: To investigate the prevalence of sequence variants in the gene SPATA7 in patients with Leber congenital amaurosis (LCA) and autosomal recessive, severe, early-onset retinal dystrophy (EORD) and to delineate the ocular phenotype associated with SPATA7 mutations. METHODS: Patients underwent standard ophthalmic evaluation after providing informed consent. One hundred forty-one DNA samples from patients with LCA and EORD had been analyzed for mutations by using a microarray, with negative results. One additional patient underwent SPATA7 screening due to a region of autozygosity surrounding this gene. A further patient was screened who had a compatible ocular phenotype. The entire SPATA7 coding sequence was assayed, including the intron-exon junctions, by using a combination of direct DNA sequencing and high-resolution melting screening. RESULTS: Screening of SPATA7 identified several known and novel single-nucleotide polymorphisms (SNPs). Affected individuals from five unrelated families were identified to have coding changes. Clinical features demonstrated a severe infantile onset retinal dystrophy, similar to Leber congenital amaurosis. The retina had widespread retinal pigment epithelial atrophy, with minimal pigment migration into the neurosensory retina. Fundus autofluorescence imaging showed a parafoveal annulus of increased autofluorescence. High-definition optical coherence tomography showed preservation of the inner segment/outer segment junction at the fovea. CONCLUSIONS: Mutations in SPATA7 are a rare cause of childhood retinal dystrophy accounting for 1.7% of disease in this cohort. Affected patients present in infancy with severe visual loss, but may have some preservation of the photoreceptor structure in the central retina.


Subject(s)
DNA-Binding Proteins/genetics , Leber Congenital Amaurosis/genetics , Mutation , Polymorphism, Single Nucleotide , Retinal Dystrophies/genetics , Vision Disorders/genetics , Adolescent , Adult , Child, Preschool , Consanguinity , DNA Mutational Analysis , DNA Primers/chemistry , Female , Fluorescein Angiography , Genetic Testing , Humans , Leber Congenital Amaurosis/diagnosis , Male , Pedigree , Phenotype , Prevalence , Retinal Dystrophies/diagnosis , Sequence Analysis, DNA , Tomography, Optical Coherence , Vision Disorders/diagnosis , Visual Field Tests , Visual Fields
SELECTION OF CITATIONS
SEARCH DETAIL
...