Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 119(27): e2115538119, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35759666

ABSTRACT

Blue cone monochromacy (BCM) is an X-linked retinal disorder characterized by low vision, photoaversion, and poor color discrimination. BCM is due to the lack of long-wavelength-sensitive and middle-wavelength-sensitive cone photoreceptor function and caused by mutations in the OPN1LW/OPN1MW gene cluster on Xq28. Here, we investigated the prevalence and the landscape of submicroscopic structural variants (SVs) at single-base resolution in BCM patients. We found that about one-third (n = 73) of the 213 molecularly confirmed BCM families carry an SV, most commonly deletions restricted to the OPN1LW/OPN1MW gene cluster. The structure and precise breakpoints of the SVs were resolved in all but one of the 73 families. Twenty-two families-all from the United States-showed the same SV, and we confirmed a common ancestry of this mutation. In total, 42 distinct SVs were identified, including 40 previously unreported SVs, thereby quadrupling the number of precisely mapped SVs underlying BCM. Notably, there was no "region of overlap" among these SVs. However, 90% of SVs encompass the upstream locus control region, an essential enhancer element. Its minimal functional extent based on deletion mapping in patients was refined to 358 bp. Breakpoint analyses suggest diverse mechanisms underlying SV formation as well as in one case the gene conversion-based exchange of a 142-bp deletion between opsin genes. Using parsimonious assumptions, we reconstructed the composition and copy number of the OPN1LW/OPN1MW gene cluster prior to the mutation event and found evidence that large gene arrays may be predisposed to the occurrence of SVs at this locus.


Subject(s)
Color Vision Defects , Rod Opsins , Color Vision Defects/genetics , Gene Deletion , Humans , Multigene Family/genetics , Retinal Cone Photoreceptor Cells , Rod Opsins/genetics
2.
Hum Mutat ; 43(7): 832-858, 2022 07.
Article in English | MEDLINE | ID: mdl-35332618

ABSTRACT

Achromatopsia (ACHM) is a congenital cone photoreceptor disorder characterized by impaired color discrimination, low visual acuity, photosensitivity, and nystagmus. To date, six genes have been associated with ACHM (CNGA3, CNGB3, GNAT2, PDE6C, PDE6H, and ATF6), the majority of these being implicated in the cone phototransduction cascade. CNGA3 encodes the CNGA3 subunit of the cyclic nucleotide-gated ion channel in cone photoreceptors and is one of the major disease-associated genes for ACHM. Herein, we provide a comprehensive overview of the CNGA3 variant spectrum in a cohort of 1060 genetically confirmed ACHM patients, 385 (36.3%) of these carrying "likely disease-causing" variants in CNGA3. Compiling our own genetic data with those reported in the literature and in public databases, we further extend the CNGA3 variant spectrum to a total of 316 variants, 244 of which we interpreted as "likely disease-causing" according to ACMG/AMP criteria. We report 48 novel "likely disease-causing" variants, 24 of which are missense substitutions underlining the predominant role of this mutation class in the CNGA3 variant spectrum. In addition, we provide extensive in silico analyses and summarize reported functional data of previously analyzed missense, nonsense and splicing variants to further advance the pathogenicity assessment of the identified variants.


Subject(s)
Color Vision Defects , Cyclic Nucleotide-Gated Cation Channels , Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Humans , Mutation , Retinal Cone Photoreceptor Cells
3.
J Med Genet ; 58(8): 570-578, 2021 08.
Article in English | MEDLINE | ID: mdl-32817297

ABSTRACT

BACKGROUND: Inherited retinal disorders are a clinically and genetically heterogeneous group of conditions and a major cause of visual impairment. Common disease subtypes include vitelliform macular dystrophy (VMD) and retinitis pigmentosa (RP). Despite the identification of over 90 genes associated with RP, conventional genetic testing fails to detect a molecular diagnosis in about one third of patients with RP. METHODS: Exome sequencing was carried out for identifying the disease-causing gene in a family with autosomal dominant RP. Gene panel testing and exome sequencing were performed in 596 RP and VMD families to identified additional IMPG1 variants. In vivo analysis in the medaka fish system by knockdown assays was performed to screen IMPG1 possible pathogenic role. RESULTS: Exome sequencing of a family with RP revealed a splice variant in IMPG1. Subsequently, the same variant was identified in individuals from two families with either RP or VMD. A retrospective study of patients with RP or VMD revealed eight additional families with different missense or nonsense variants in IMPG1. In addition, the clinical diagnosis of the IMPG1 retinopathy-associated variant, originally described as benign concentric annular macular dystrophy, was also revised to RP with early macular involvement. Using morpholino-mediated ablation of Impg1 and its paralog Impg2 in medaka fish, we confirmed a phenotype consistent with that observed in the families, including a decreased length of rod and cone photoreceptor outer segments. CONCLUSION: This study discusses a previously unreported association between monoallelic or biallelic IMPG1 variants and RP. Notably, similar observations have been reported for IMPG2.


Subject(s)
Extracellular Matrix Proteins , Eye Proteins , Genes, Recessive , Genetic Predisposition to Disease , Mutation , Proteoglycans , Retinitis Pigmentosa , Aged , Female , Humans , Male , Middle Aged , Exome/genetics , Exome Sequencing/methods , Extracellular Matrix Proteins/genetics , Eye Proteins/genetics , Genes, Recessive/genetics , Genetic Predisposition to Disease/genetics , Inheritance Patterns/genetics , Macular Degeneration/genetics , Mutation/genetics , Pedigree , Phenotype , Proteoglycans/genetics , Retina/pathology , Retinitis Pigmentosa/genetics , Retrospective Studies
4.
Int J Mol Sci ; 23(8)2022 Apr 13.
Article in English | MEDLINE | ID: mdl-35457110

ABSTRACT

Biallelic gene defects in MFSD8 are not only a cause of the late-infantile form of neuronal ceroid lipofuscinosis, but also of rare isolated retinal degeneration. We report clinical and genetic data of seven patients compound heterozygous or homozygous for variants in MFSD8, issued from a French cohort with inherited retinal degeneration, and two additional patients retrieved from a Swiss cohort. Next-generation sequencing of large panels combined with whole-genome sequencing allowed for the identification of twelve variants from which seven were novel. Among them were one deep intronic variant c.998+1669A>G, one large deletion encompassing exon 9 and 10, and a silent change c.750A>G. Transcript analysis performed on patients' lymphoblastoid cell lines revealed the creation of a donor splice site by c.998+1669A>G, resulting in a 140 bp pseudoexon insertion in intron 10. Variant c.750A>G produced exon 8 skipping. In silico and in cellulo studies of these variants allowed us to assign the pathogenic effect, and showed that the combination of at least one severe variant with a moderate one leads to isolated retinal dystrophy, whereas the combination in trans of two severe variants is responsible for early onset severe retinal dystrophy in the context of late-infantile neuronal ceroid lipofuscinosis.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Retinal Dystrophies , Exons/genetics , Homozygote , Humans , Membrane Transport Proteins/genetics , Mutation , Neuronal Ceroid-Lipofuscinoses/genetics , Retinal Dystrophies/genetics
5.
Retina ; 41(8): 1771-1779, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-33315831

ABSTRACT

PURPOSE: RTN4IP1 biallelic mutations cause a recessive optic atrophy, sometimes associated to more severe neurological syndromes, but so far, no retinal phenotype has been reported in RTN4IP1 patients, justifying their reappraisal. METHODS: Seven patients from four families carrying biallelic RTN4IP1 variants were retrospectively reviewed, with emphasis on their age of onset, visual acuity, multimodal imaging including color and autofluorescence frames, spectral-domain optical coherence tomography with RNFL and macular analyses. RESULTS: Seven patients from four RTN4IP1 families developed in their first decade of life a bilateral recessive optic atrophy with severe central visual loss, and primary nystagmus developed in 5 of 7 patients. Six patients were legally blind. In a second stage, the seven individuals developed a rod-cone dystrophy, sparing the macular zone and the far periphery. This retinal damage was identified by 55° field fundus autofluorescence frames and also by spectral-domain optical coherence tomography scans of the temporal part of the macular zone in five of the seven patients. Full-field electroretinography measurements disclosed reduced b-wave amplitude of the rod responses in all patients but two. Family 4 with the p.R103H and c.601A > T (p.K201*) truncating mutation had further combined neurological signs with cerebellar ataxia, seizures, and intellectual disability. CONCLUSION: RTN4IP1 recessive optic atrophy is systematically associated to a rod-cone dystrophy, which suggests that both the retinal ganglion cells and the rods are affected as a result of a deficit in the mitochondrial respiratory chain. Thus, systematic widefield autofluorescence frames and temporal macular scans are recommended for the evaluation of patients with optic neuropathies.


Subject(s)
Carrier Proteins/genetics , Cone-Rod Dystrophies/genetics , DNA/genetics , Mitochondrial Proteins/genetics , Mutation , Adolescent , Adult , Carrier Proteins/metabolism , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Electroretinography , Female , Fluorescein Angiography/methods , Fundus Oculi , Humans , Male , Middle Aged , Mitochondrial Proteins/metabolism , Pedigree , Phenotype , Retrospective Studies , Tomography, Optical Coherence/methods , Visual Acuity , Visual Fields , Young Adult
6.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203883

ABSTRACT

Variants of the TTLL5 gene, which encodes tubulin tyrosine ligase-like family member five, are a rare cause of cone dystrophy (COD) or cone-rod dystrophy (CORD). To date, only a few TTLL5 patients have been clinically and genetically described. In this study, we report five patients harbouring biallelic variants of TTLL5. Four adult patients presented either COD or CORD with onset in the late teenage years. The youngest patient had a phenotype of early onset severe retinal dystrophy (EOSRD). Genetic analysis was performed by targeted next generation sequencing of gene panels and assessment of copy number variants (CNV). We identified eight variants, of which six were novel, including two large multiexon deletions in patients with COD or CORD, while the EOSRD patient harboured the novel homozygous p.(Trp640*) variant and three distinct USH2A variants, which might explain the observed rod involvement. Our study highlights the role of TTLL5 in COD/CORD and the importance of large deletions. These findings suggest that COD or CORD patients lacking variants in known genes may harbour CNVs to be discovered in TTLL5, previously undetected by classical sequencing methods. In addition, variable phenotypes in TTLL5-associated patients might be due to the presence of additional gene defects.


Subject(s)
Carrier Proteins/genetics , Cone-Rod Dystrophies/genetics , Eye Diseases, Hereditary/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation/genetics , Retinal Dystrophies/genetics , Adult , Aged , Child , Chromosome Breakpoints , Computer Simulation , Cone-Rod Dystrophies/physiopathology , DNA Copy Number Variations/genetics , Electroretinography , Eye Diseases, Hereditary/physiopathology , Female , Genotype , Humans , Male , Middle Aged , Phenotype , Retinal Dystrophies/physiopathology
7.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884448

ABSTRACT

Pathogenic variants in CRB1 lead to diverse recessive retinal disorders from severe Leber congenital amaurosis to isolated macular dystrophy. Until recently, no clear phenotype-genotype correlation and no appropriate mouse models existed. Herein, we reappraise the phenotype-genotype correlation of 50 patients with regards to the recently identified CRB1 isoforms: a canonical long isoform A localized in Müller cells (12 exons) and a short isoform B predominant in photoreceptors (7 exons). Twenty-eight patients with early onset retinal dystrophy (EORD) consistently had a severe Müller impairment, with variable impact on the photoreceptors, regardless of isoform B expression. Among them, two patients expressing wild type isoform B carried one variant in exon 12, which specifically damaged intracellular protein interactions in Müller cells. Thirteen retinitis pigmentosa patients had mainly missense variants in laminin G-like domains and expressed at least 50% of isoform A. Eight patients with the c.498_506del variant had macular dystrophy. In one family homozygous for the c.1562C>T variant, the brother had EORD and the sister macular dystrophy. In contrast with the mouse model, these data highlight the key role of Müller cells in the severity of CRB1-related dystrophies in humans, which should be taken into consideration for future clinical trials.


Subject(s)
Ependymoglial Cells/pathology , Eye Proteins/genetics , Eye Proteins/metabolism , Macular Degeneration/pathology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Retinal Dystrophies/pathology , Retinitis Pigmentosa/pathology , Adolescent , Age of Onset , Alternative Splicing , Child , Child, Preschool , Ependymoglial Cells/metabolism , Eye Proteins/chemistry , Female , Genetic Association Studies , Humans , Infant , Macular Degeneration/genetics , Macular Degeneration/metabolism , Male , Membrane Proteins/chemistry , Models, Molecular , Mutation, Missense , Nerve Tissue Proteins/chemistry , Point Mutation , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retrospective Studies , Sequence Deletion , Young Adult
8.
Hum Mutat ; 41(1): 255-264, 2020 01.
Article in English | MEDLINE | ID: mdl-31544997

ABSTRACT

Our comprehensive cohort of 1100 unrelated achromatopsia (ACHM) patients comprises a considerable number of cases (~5%) harboring only a single pathogenic variant in the major ACHM gene CNGB3. We sequenced the entire CNGB3 locus in 33 of these patients to find a second variant which eventually explained the patients' phenotype. Forty-seven intronic CNGB3 variants were identified in 28 subjects after a filtering step based on frequency and the exclusion of variants found in cis with pathogenic alleles. In a second step, in silico prediction tools were used to filter out those variants with little odds of being deleterious. This left three variants that were analyzed using heterologous splicing assays. Variant c.1663-1205G>A, found in 14 subjects, and variant c.1663-2137C>T, found in two subjects, were indeed shown to exert a splicing defect by causing pseudoexon insertion into the transcript. Subsequent screening of further unsolved CNGB3 subjects identified four additional cases harboring the c.1663-1205G>A variant which makes it the eighth most frequent CNGB3 variant in our cohort. Compound heterozygosity could be validated in ten cases. Our study demonstrates that whole gene sequencing can be a powerful approach to identify the second pathogenic allele in patients apparently harboring only one disease-causing variant.


Subject(s)
Color Vision Defects/diagnosis , Color Vision Defects/genetics , Cyclic Nucleotide-Gated Cation Channels/genetics , Exons , Genetic Variation , Introns , Pseudogenes , Alleles , Amino Acid Substitution , Base Sequence , Computational Biology/methods , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Mutation , Phenotype , RNA Splicing
9.
Hum Mutat ; 40(8): 1145-1155, 2019 08.
Article in English | MEDLINE | ID: mdl-31058429

ABSTRACT

Achromatopsia (ACHM) is a hereditary cone photoreceptor disorder characterized by the inability to discriminate colors, nystagmus, photophobia, and low-visual acuity. Six genes have been associated with this rare autosomal recessively inherited disease, including the GNAT2 gene encoding the catalytic α-subunit of the G-protein transducin which is expressed in the cone photoreceptor outer segment. Out of a cohort of 1,116 independent families diagnosed with a primary clinical diagnosis of ACHM, we identified 23 patients with ACHM from 19 independent families with likely causative mutations in GNAT2, representing 1.7% of our large ACHM cohort. In total 22 different potentially disease-causing variants, of which 12 are novel, were identified. The mutation spectrum also includes a novel copy number variation, a heterozygous duplication of exon 4, of which the breakpoint matches exactly that of the previously reported exon 4 deletion. Two patients carry just a single heterozygous variant. In addition to our previous study on GNAT2-ACHM, we also present detailed clinical data of these patients.


Subject(s)
Color Vision Defects/genetics , Heterotrimeric GTP-Binding Proteins/genetics , Mutation , Sequence Analysis, DNA/methods , Adolescent , Adult , Aged , Child , Child, Preschool , DNA Copy Number Variations , Exons , Female , Genetic Predisposition to Disease , Humans , Infant , Male , Middle Aged , Pedigree , Young Adult
10.
Hum Mol Genet ; 26(22): 4367-4374, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28973654

ABSTRACT

In this study, we report a novel duplication causing North Carolina macular dystrophy (NCMD) identified applying whole genome sequencing performed on eight affected members of two presumed unrelated families mapping to the MCDR1 locus. In our families, the NCMD phenotype was associated with a 98.4 kb tandem duplication encompassing the entire CCNC and PRDM13 genes and a common DNase 1 hypersensitivity site. To study the impact of PRDM13 or CCNC dysregulation, we used the Drosophila eye development as a model. Knock-down and overexpression of CycC and CG13296, Drosophila orthologues of CCNC and PRDM13, respectively, were induced separately during eye development. In flies, eye development was not affected, while knocking down either CycC or CG13296 mutant models. Overexpression of CycC also had no effect. Strikingly, overexpression of CG13296 in Drosophila leads to a severe loss of the imaginal eye-antennal disc. This study demonstrated for the first time in an animal model that overexpression of PRDM13 alone causes a severe abnormal retinal development. It is noteworthy that mutations associated with this autosomal dominant foveal developmental disorder are frequently duplications always including an entire copy of PRDM13, or variants in one DNase 1 hypersensitivity site at this locus.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Cyclin C/genetics , Histone-Lysine N-Methyltransferase/genetics , Adult , Animals , Chromosome Mapping , Chromosomes, Human, Pair 6 , Corneal Dystrophies, Hereditary/metabolism , Cyclin C/metabolism , Drosophila melanogaster , Eye Proteins/genetics , Female , Genetic Linkage , Haplotypes , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , PR-SET Domains , Pedigree , Whole Genome Sequencing
11.
Int J Mol Sci ; 20(10)2019 May 23.
Article in English | MEDLINE | ID: mdl-31126147

ABSTRACT

: Inherited retinal dystrophies (IRDs) are a clinically and genetically heterogeneous group of diseases with more than 250 causative genes. The most common form is retinitis pigmentosa. IRDs lead to vision impairment for which there is no universal cure. Encouragingly, a first gene supplementation therapy has been approved for an autosomal recessive IRD. However, for autosomal dominant IRDs, gene supplementation therapy is not always pertinent because haploinsufficiency is not the only cause. Disease-causing mechanisms are often gain-of-function or dominant-negative, which usually require alternative therapeutic approaches. In such cases, genome-editing technology has raised hopes for treatment. Genome editing could be used to i) invalidate both alleles, followed by supplementation of the wild type gene, ii) specifically invalidate the mutant allele, with or without gene supplementation, or iii) to correct the mutant allele. We review here the most prevalent genes causing autosomal dominant retinitis pigmentosa and the most appropriate genome-editing strategy that could be used to target their different causative mutations.


Subject(s)
Gene Editing/methods , Genetic Therapy/methods , Retinitis Pigmentosa/therapy , Animals , CRISPR-Cas Systems , Humans , Mutation , Retinitis Pigmentosa/genetics
12.
Hum Mutat ; 39(10): 1366-1371, 2018 10.
Article in English | MEDLINE | ID: mdl-30080950

ABSTRACT

Biallelic PDE6C mutations are a known cause for rod monochromacy, better known as autosomal recessive achromatopsia (ACHM), and early-onset cone photoreceptor dysfunction. PDE6C encodes the catalytic α'-subunit of the cone photoreceptor phosphodiesterase, thereby constituting an essential part of the phototransduction cascade. Here, we present the results of a study comprising 176 genetically preselected patients who remained unsolved after Sanger sequencing of the most frequent genes accounting for ACHM, and were subsequently screened for exonic and splice site variants in PDE6C applying a targeted next generation sequencing approach. We were able to identify potentially pathogenic biallelic variants in 15 index cases. The mutation spectrum comprises 18 different alleles, 15 of which are novel. Our study significantly contributes to the mutation spectrum of PDE6C and allows for a realistic estimate of the prevalence of PDE6C mutations in ACHM since our entire ACHM cohort comprises 1,074 independent families.


Subject(s)
Catalytic Domain/genetics , Color Vision Defects/diagnosis , Color Vision Defects/genetics , Cyclic Nucleotide Phosphodiesterases, Type 6/genetics , Eye Proteins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Mutation , Alleles , Child, Preschool , Computational Biology/methods , Cyclic Nucleotide Phosphodiesterases, Type 6/chemistry , Databases, Genetic , Eye Proteins/chemistry , Genotype , Humans , Infant , Infant, Newborn , Phenotype , Retinal Cone Photoreceptor Cells/metabolism , Sequence Analysis, DNA
13.
Hum Mol Genet ; 25(5): 916-26, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26744326

ABSTRACT

Inherited retinal dystrophies are clinically and genetically heterogeneous with significant number of cases remaining genetically unresolved. We studied a large family from the West Indies islands with a peculiar retinal disease, the Martinique crinkled retinal pigment epitheliopathy that begins around the age of 30 with retinal pigment epithelium (RPE) and Bruch's membrane changes resembling a dry desert land and ends with a retinitis pigmentosa. Whole-exome sequencing identified a heterozygous c.518T>C (p.Leu173Pro) mutation in MAPKAPK3 that segregates with the disease in 14 affected and 28 unaffected siblings from three generations. This unknown variant is predicted to be damaging by bioinformatic predictive tools and the mutated protein to be non-functional by crystal structure analysis. MAPKAPK3 is a serine/threonine protein kinase of the p38 signaling pathway that is activated by a variety of stress stimuli and is implicated in cellular responses and gene regulation. In contrast to other tissues, MAPKAPK3 is highly expressed in the RPE, suggesting a crucial role for retinal physiology. Expression of the mutated allele in HEK cells revealed a mislocalization of the protein in the cytoplasm, leading to cytoskeleton alteration and cytodieresis inhibition. In Mapkapk3-/- mice, Bruch's membrane is irregular with both abnormal thickened and thinned portions. In conclusion, we identified the first pathogenic mutation in MAPKAPK3 associated with a retinal disease. These findings shed new lights on Bruch's membrane/RPE pathophysiology and will open studies of this signaling pathway in diseases with RPE and Bruch's membrane alterations, such as age-related macular degeneration.


Subject(s)
Bruch Membrane/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Protein Serine-Threonine Kinases/genetics , Retinal Dystrophies/genetics , Retinal Pigment Epithelium/metabolism , Signal Transduction/genetics , Adult , Age of Onset , Aged, 80 and over , Amino Acid Sequence , Animals , Bruch Membrane/pathology , Exome , Female , Gene Expression Regulation , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , Retinal Dystrophies/metabolism , Retinal Dystrophies/pathology , Retinal Pigment Epithelium/pathology , Sequence Alignment , Siblings
15.
Hum Genet ; 137(2): 111-127, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29305691

ABSTRACT

Cerebellar ataxia, areflexia, pes cavus, optic atrophy and sensorineural hearing impairment (CAPOS) is a rare clinically distinct syndrome caused by a single dominant missense mutation, c.2452G>A, p.Glu818Lys, in ATP1A3, encoding the neuron-specific alpha subunit of the Na+/K+-ATPase α3. Allelic mutations cause the neurological diseases rapid dystonia Parkinsonism and alternating hemiplegia of childhood, disorders which do not encompass hearing or visual impairment. We present detailed clinical phenotypic information in 18 genetically confirmed patients from 11 families (10 previously unreported) from Denmark, Sweden, UK and Germany indicating a specific type of hearing impairment-auditory neuropathy (AN). All patients were clinically suspected of CAPOS and had hearing problems. In this retrospective analysis of audiological data, we show for the first time that cochlear outer hair cell activity was preserved as shown by the presence of otoacoustic emissions and cochlear microphonic potentials, but the auditory brainstem responses were grossly abnormal, likely reflecting neural dyssynchrony. Poor speech perception was observed, especially in noise, which was beyond the hearing level obtained in the pure tone audiograms in several of the patients presented here. Molecular modelling and in vitro electrophysiological studies of the specific CAPOS mutation were performed. Heterologous expression studies of α3 with the p.Glu818Lys mutation affects sodium binding to, and release from, the sodium-specific site in the pump, the third ion-binding site. Molecular dynamics simulations confirm that the structure of the C-terminal region is affected. In conclusion, we demonstrate for the first time evidence for auditory neuropathy in CAPOS syndrome, which may reflect impaired propagation of electrical impulses along the spiral ganglion neurons. This has implications for diagnosis and patient management. Auditory neuropathy is difficult to treat with conventional hearing aids, but preliminary improvement in speech perception in some patients suggests that cochlear implantation may be effective in CAPOS patients.


Subject(s)
Cerebellar Ataxia/genetics , Foot Deformities, Congenital/genetics , Hearing Loss, Central/genetics , Hearing Loss, Sensorineural/genetics , Optic Atrophy/genetics , Reflex, Abnormal/genetics , Sodium-Potassium-Exchanging ATPase/genetics , Adolescent , Adult , Cerebellar Ataxia/epidemiology , Cerebellar Ataxia/physiopathology , Child , Child, Preschool , Denmark/epidemiology , Female , Foot Deformities, Congenital/epidemiology , Foot Deformities, Congenital/physiopathology , Germany/epidemiology , Hearing Loss, Central/epidemiology , Hearing Loss, Central/physiopathology , Hearing Loss, Sensorineural/epidemiology , Hearing Loss, Sensorineural/physiopathology , Humans , Male , Molecular Dynamics Simulation , Mutation, Missense/genetics , Optic Atrophy/epidemiology , Optic Atrophy/physiopathology , Phenotype , Retrospective Studies , Sodium-Potassium-Exchanging ATPase/chemistry , Sweden/epidemiology , Young Adult
16.
Am J Hum Genet ; 97(5): 754-60, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26593267

ABSTRACT

Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.


Subject(s)
Carrier Proteins/genetics , Fibroblasts/pathology , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mutation/genetics , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology , Amino Acid Sequence , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Electron Transport Complex I , Female , Fibroblasts/metabolism , Follow-Up Studies , Genes, Recessive , Humans , Male , Mice , Mitochondria/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Nerve Degeneration , Pedigree , Prognosis , Retinal Ganglion Cells/metabolism , Sequence Homology, Amino Acid , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
17.
Mol Vis ; 23: 198-209, 2017.
Article in English | MEDLINE | ID: mdl-28442884

ABSTRACT

PURPOSE: Sixteen different mutations in the guanylate cyclase activator 1A gene (GUCA1A), have been previously identified to cause autosomal dominant cone dystrophy (adCOD), cone-rod dystrophy (adCORD), macular dystrophy (adMD), and in an isolated patient, retinitis pigmentosa (RP). The purpose of this study is to report on two novel mutations and the patients' clinical features. METHODS: Clinical investigations included visual acuity and visual field testing, fundus examination, high-resolution spectral-domain optical coherence tomography (OCT), fundus autofluorescence imaging, and full-field and multifocal electroretinogram (ERG) recordings. GUCA1A was screened by Sanger sequencing in a cohort of 12 French families with adCOD, adCORD, and adMD. RESULTS: We found two novel GUCA1A mutations-one amino acid deletion, c.302_304delTAG (p.Val101del), and one missense mutation, c.444T>A (p.Asp148Glu)-each of which was found in one family. The p.Asp148Glu mutation affected one of the Ca2+-binding amino acids of the EF4 hand, while the p.Val101del mutation resulted in the in-frame deletion of Valine-101, localized between two Ca2+-binding aspartic acid residues at positions 100 and 102 of the EF3 hand. Both families complained of visual acuity loss worsening with age. However, the p.Asp148Glu mutation was present in one family with adCOD involving abnormal cone function and an absence of macular atrophy, whereas p.Val101del mutation was encountered in another family with adMD without a generalized cone defect. CONCLUSIONS: The two novel mutations described in this study are associated with distinct phenotypes, MD for p.Val101del and COD for p.Asp148Glu, with no intrafamilial phenotypic heterogeneity.


Subject(s)
Cone-Rod Dystrophies/genetics , Guanylate Cyclase-Activating Proteins/genetics , Macular Degeneration/genetics , Mutation, Missense , Retinal Cone Photoreceptor Cells/pathology , Retinal Degeneration/genetics , Sequence Deletion , Adult , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/physiopathology , DNA Mutational Analysis , Electroretinography , Female , Genes, Dominant , Humans , Macular Degeneration/diagnosis , Macular Degeneration/physiopathology , Male , Middle Aged , Optical Imaging , Pedigree , Retinal Degeneration/diagnosis , Retinal Degeneration/physiopathology , Tomography, Optical Coherence , Visual Acuity/physiology , Visual Fields/physiology , Young Adult
18.
Am J Hum Genet ; 92(1): 67-75, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23246293

ABSTRACT

Congenital stationary night blindness (CSNB) is a clinically and genetically heterogeneous retinal disorder. Two forms can be distinguished clinically: complete CSNB (cCSNB) and incomplete CSNB. Individuals with cCSNB have visual impairment under low-light conditions and show a characteristic electroretinogram (ERG). The b-wave amplitude is severely reduced in the dark-adapted state of the ERG, representing abnormal function of ON bipolar cells. Furthermore, individuals with cCSNB can show other ocular features such as nystagmus, myopia, and strabismus and can have reduced visual acuity and abnormalities of the cone ERG waveform. The mode of inheritance of this form can be X-linked or autosomal recessive, and the dysfunction of four genes (NYX, GRM6, TRPM1, and GPR179) has been described so far. Whole-exome sequencing in one simplex cCSNB case lacking mutations in the known genes led to the identification of a missense mutation (c.983G>A [p.Cys328Tyr]) and a nonsense mutation (c.1318C>T [p.Arg440(∗)]) in LRIT3, encoding leucine-rich-repeat (LRR), immunoglobulin-like, and transmembrane-domain 3 (LRIT3). Subsequent Sanger sequencing of 89 individuals with CSNB identified another cCSNB case harboring a nonsense mutation (c.1151C>G [p.Ser384(∗)]) and a deletion predicted to lead to a premature stop codon (c.1538_1539del [p.Ser513Cysfs(∗)59]) in the same gene. Human LRIT3 antibody staining revealed in the outer plexiform layer of the human retina a punctate-labeling pattern resembling the dendritic tips of bipolar cells; similar patterns have been observed for other proteins implicated in cCSNB. The exact role of this LRR protein in cCSNB remains to be elucidated.


Subject(s)
Eye Diseases, Hereditary/genetics , Genetic Diseases, X-Linked/genetics , Membrane Proteins/genetics , Myopia/genetics , Night Blindness/genetics , Polymorphism, Genetic , Exome , Female , Humans , Male , Membrane Proteins/analysis , Middle Aged , Mutation , Retina/chemistry
19.
Am J Hum Genet ; 93(3): 571-8, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23993198

ABSTRACT

Vitelliform macular dystrophies (VMD) are inherited retinal dystrophies characterized by yellow, round deposits visible upon fundus examination and encountered in individuals with juvenile Best macular dystrophy (BMD) or adult-onset vitelliform macular dystrophy (AVMD). Although many BMD and some AVMD cases harbor mutations in BEST1 or PRPH2, the underlying genetic cause remains unknown for many affected individuals. In a large family with autosomal-dominant VMD, gene mapping and whole-exome sequencing led to the identification of a c.713T>G (p.Leu238Arg) IMPG1 mutation, which was subsequently found in two other families with autosomal-dominant VMD and the same phenotype. IMPG1 encodes the SPACR protein, a component of the rod and cone photoreceptor extracellular matrix domains. Structural modeling indicates that the p.Leu238Arg substitution destabilizes the conserved SEA1 domain of SPACR. Screening of 144 probands who had various forms of macular dystrophy revealed three other IMPG1 mutations. Two individuals from one family affected by autosomal-recessive VMD were homozygous for the splice-site mutation c.807+1G>T, and two from another family were compound heterozygous for the mutations c.461T>C (p.Leu154Pro) and c.1519C>T (p.Arg507(∗)). Most cases had a normal or moderately decreased electrooculogram Arden ratio. We conclude that IMPG1 mutations cause both autosomal-dominant and -recessive forms of VMD, thus indicating that impairment of the interphotoreceptor matrix might be a general cause of VMD.


Subject(s)
Extracellular Matrix Proteins/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , Mutation/genetics , Proteoglycans/genetics , Vitelliform Macular Dystrophy/genetics , Adult , Amino Acid Sequence , Base Sequence , Chromosomes, Human/genetics , Extracellular Matrix Proteins/chemistry , Eye Proteins/chemistry , Female , Fundus Oculi , Humans , Inheritance Patterns/genetics , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phenotype , Proteoglycans/chemistry , Young Adult
20.
Ophthalmology ; 123(9): 1989-98, 2016 09.
Article in English | MEDLINE | ID: mdl-27395765

ABSTRACT

PURPOSE: To search for WFS1 mutations in patients with optic atrophy (OA) and assess visual impairment. DESIGN: Retrospective molecular genetic and clinical study. PARTICIPANTS: Patients with OA followed at a national referral center specialized in genetic sensory diseases. METHODS: Mutation screening in WFS1 was performed by Sanger sequencing. WFS1-positive patients were evaluated on visual acuity (VA) and retinal nerve fiber layer (RNFL) thickness using time-domain (TD) or spectral-domain (SD) optical coherence tomography (OCT). Statistical analysis was performed. MAIN OUTCOME MEASURES: Mutation identification, VA values, and RNFL thickness in sectors. RESULTS: Biallelic WFS1 mutations were found in 3 of 24 unrelated patients (15%) with autosomal recessive nonsyndromic optic atrophy (arNSOA) and in 8 patients with autosomal recessive Wolfram syndrome (arWS) associated with diabetes mellitus and OA. Heterozygous mutations were found in 4 of 20 unrelated patients (20%) with autosomal dominant OA. The 4 WFS1-mutated patients of this latter group with hearing loss were diagnosed with autosomal dominant Wolfram-like syndrome (adWLS). Most patients had VA decrease, with logarithm of the minimum angle of resolution (logMAR) values lower in arWS than in arNSOA (1.530 vs. 0.440; P = 0.026) or adWLS (0.240; P = 0.006) but not differing between arNSOA and adWLS (P = 0.879). All patients had decreased RNFL thickness that was worse in arWS than in arNSOA (SD OCT, 35.50 vs. 53.80 µm; P = 0.018) or adWLS (TD-OCT, 45.84 vs. 59.33 µm; P = 0.049). The greatest difference was found in the inferior bundle. Visual acuity was negatively correlated with RNFL thickness (r = -0.89; P = 0.003 in SD OCT and r = -0.75; P = 0.01 in TD-OCT). CONCLUSIONS: WFS1 is a gene causing arNSOA. Patients with this condition had significantly less visual impairment than those with arWS. Thus systematic screening of WFS1 must be performed in isolated, sporadic, or familial optic atrophies.


Subject(s)
Membrane Proteins/genetics , Mutation , Optic Atrophy/genetics , Adolescent , Adult , Child , Female , Humans , Male , Middle Aged , Nerve Fibers/pathology , Optic Atrophy/physiopathology , Retinal Ganglion Cells/pathology , Retrospective Studies , Visual Acuity/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL