Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Commun ; 15(1): 3562, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670966

ABSTRACT

The diagnosis of inherited retinal degeneration (IRD) is challenging owing to its phenotypic and genotypic complexity. Clinical information is important before a genetic diagnosis is made. Metabolomics studies the entire picture of bioproducts, which are determined using genetic codes and biological reactions. We demonstrated that the common diagnoses of IRD, including retinitis pigmentosa (RP), cone-rod dystrophy (CRD), Stargardt disease (STGD), and Bietti's crystalline dystrophy (BCD), could be differentiated based on their metabolite heatmaps. Hundreds of metabolites were identified in the volcano plot compared with that of the control group in every IRD except BCD, considered as potential diagnosing markers. The phenotypes of CRD and STGD overlapped but could be differentiated by their metabolomic features with the assistance of a machine learning model with 100% accuracy. Moreover, EYS-, USH2A-associated, and other RP, sharing considerable similar characteristics in clinical findings, could also be diagnosed using the machine learning model with 85.7% accuracy. Further study would be needed to validate the results in an external dataset. By incorporating mass spectrometry and machine learning, a metabolomics-based diagnostic workflow for the clinical and molecular diagnoses of IRD was proposed in our study.


Subject(s)
Machine Learning , Metabolomics , Retinal Degeneration , Retinitis Pigmentosa , Stargardt Disease , Humans , Metabolomics/methods , Diagnosis, Differential , Retinal Degeneration/diagnosis , Retinal Degeneration/blood , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Male , Female , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/blood , Retinitis Pigmentosa/metabolism , Stargardt Disease/genetics , Adult , Middle Aged , Adolescent , Young Adult , Biomarkers/blood , Metabolome , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/blood , Cone-Rod Dystrophies/metabolism , Mass Spectrometry , Macular Degeneration/blood , Macular Degeneration/diagnosis , Macular Degeneration/genetics
2.
Hum Mol Genet ; 33(9): 802-817, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38297980

ABSTRACT

Mutations in Cytosolic Carboxypeptidase-like Protein 5 (CCP5) are associated with vision loss in humans. To decipher the mechanisms behind CCP5-associated blindness, we generated a novel mouse model lacking CCP5. In this model, we found that increased tubulin glutamylation led to progressive cone-rod dystrophy, with cones showing a more pronounced and earlier functional loss than rod photoreceptors. The observed functional reduction was not due to cell death, levels, or the mislocalization of major phototransduction proteins. Instead, the increased tubulin glutamylation caused shortened photoreceptor axonemes and the formation of numerous abnormal membranous whorls that disrupted the integrity of photoreceptor outer segments (OS). Ultimately, excessive tubulin glutamylation led to the progressive loss of photoreceptors, affecting cones more severely than rods. Our results highlight the importance of maintaining tubulin glutamylation for normal photoreceptor function. Furthermore, we demonstrate that murine cone photoreceptors are more sensitive to disrupted tubulin glutamylation levels than rods, suggesting an essential role for axoneme in the structural integrity of the cone outer segment. This study provides valuable insights into the mechanisms of photoreceptor diseases linked to excessive tubulin glutamylation.


Subject(s)
Cone-Rod Dystrophies , Tubulin , Humans , Mice , Animals , Tubulin/genetics , Tubulin/metabolism , Cone-Rod Dystrophies/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Mutation
3.
Int J Mol Sci ; 24(13)2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37445847

ABSTRACT

Mutations in the photoreceptor-specific C2orf71 gene (also known as photoreceptor cilium actin regulator protein PCARE) cause autosomal recessive retinitis pigmentosa type 54 and cone-rod dystrophy. No treatments are available for patients with C2orf71 retinal ciliopathies exhibiting a severe clinical phenotype. Our understanding of the disease process and the role of PCARE in the healthy retina significantly limits our capacity to transfer recent technical developments into viable therapy choices. This study summarizes the current understanding of C2orf71-related retinal diseases, including their clinical manifestations and an unclear genotype-phenotype correlation. It discusses molecular and functional studies on the photoreceptor-specific ciliary PCARE, focusing on the photoreceptor cell and its ciliary axoneme. It is proposed that PCARE is an actin-associated protein that interacts with WASF3 to regulate the actin-driven expansion of the ciliary membrane during the development of a new outer segment disk in photoreceptor cells. This review also introduces various cellular and animal models used to model these diseases and provides an overview of potential treatments.


Subject(s)
Cone-Rod Dystrophies , Retinitis Pigmentosa , Animals , Actins/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/therapy , Retinitis Pigmentosa/metabolism , Retina/metabolism , Cone-Rod Dystrophies/metabolism , Mutation
4.
Int J Mol Sci ; 24(8)2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37108442

ABSTRACT

The retina-specific ATP-binding cassette transporter protein ABCA4 is responsible for properly continuing the visual cycle by removing toxic retinoid byproducts of phototransduction. Functional impairment caused by ABCA4 sequence variations is the leading cause of autosomal recessive inherited retinal disorders, including Stargardt disease, retinitis pigmentosa, and cone-rod dystrophy. To date, more than 3000 ABCA4 genetic variants have been identified, approximately 40 percent of which have not been able to be classified for pathogenicity assessments. This study examined 30 missense ABCA4 variants using AlphaFold2 protein modeling and computational structure analysis for pathogenicity prediction. All variants classified as pathogenic (n = 10) were found to have deleterious structural consequences. Eight of the ten benign variants were structurally neutral, while the remaining two resulted in mild structural changes. This study's results provided multiple lines of computational pathogenicity evidence for eight ABCA4 variants of uncertain clinical significance. Overall, in silico analyses of ABCA4 can provide a valuable tool for understanding the molecular mechanisms of retinal degeneration and their pathogenic impact.


Subject(s)
Cone-Rod Dystrophies , Retinal Degeneration , Retinitis Pigmentosa , Humans , Retinal Degeneration/genetics , Retinal Degeneration/metabolism , Retina/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Stargardt Disease/genetics , Stargardt Disease/metabolism , Cone-Rod Dystrophies/metabolism , Mutation , Pedigree , ATP-Binding Cassette Transporters/metabolism
5.
Proc Natl Acad Sci U S A ; 119(49): e2208707119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36445968

ABSTRACT

Pathogenic variants in the Retinitis pigmentosa GTPase regulator (RPGR) gene lead to a clinically severe form of X-linked retinal dystrophy. However, it remains unclear why some variants cause a predominant rod, while others result in a cone-dominated phenotype. Post-translational glutamylation of the photoreceptor-specific RPGRORF15 isoform by the TTLL5 enzyme is essential for its optimal function in photoreceptors, and loss of TTLL5 leads to retinal dystrophy with a cone phenotype. Here we show that RPGR retinal disease, studied in a single cohort of 116 male patients, leads to a clear progressive shift from rod- to cone-dominating phenotype as the RPGRORF15 variant location approaches the distal part of the Open Reading Frame 15 (ORF15) region. The rod photoreceptor involvement on the contrary diminishes along the RGPR sequence, and the variants associated with the cone only phenotype are located predominantly in the very distal part, including the C-terminal basic domain. Moreover, these distal truncating RPGRORF15 variants disrupt the interaction with TTLL5 and lead to a significant impairment of RPGR glutamylation. Thus, consistent with the phenotype of TTLL5 pathogenic variants, our study shows that RPGRORF15 variants, which disrupt its basic domain and the interaction with TTLL5, also impair RPGR glutamylation and lead to the cone phenotype. This has implications for ongoing gene therapy clinical trials where the application of RPGR with impaired glutamylation may be less effective in treating RGPR dystrophies and may even convert a rod-cone dystrophy into a cone dystrophy phenotype.


Subject(s)
Cone-Rod Dystrophies , Retinal Dystrophies , Humans , Male , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Open Reading Frames/genetics , Open Reading Frames/physiology , Phenotype , Retinal Cone Photoreceptor Cells/metabolism , Retinal Dystrophies/genetics , Retinal Dystrophies/metabolism , Glutamic Acid/metabolism
6.
Int J Mol Sci ; 22(15)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34360642

ABSTRACT

The purpose of this work was to identify the gene defect underlying a relatively mild rod-cone dystrophy (RCD), lacking disease-causing variants in known genes implicated in inherited retinal disorders (IRD), and provide transcriptomic and immunolocalization data to highlight the best candidate. The DNA of the female patient originating from a consanguineous family revealed no large duplication or deletion, but several large homozygous regions. In one of these, a homozygous frameshift variant, c.244_246delins17 p.(Trp82Valfs*4); predicted to lead to a nonfunctional protein, was identified in CCDC51. CCDC51 encodes the mitochondrial coiled-coil domain containing 51 protein, also called MITOK. MITOK ablation causes mitochondrial dysfunction. Here we show for the first time that CCDC51/MITOK localizes in the retina and more specifically in the inner segments of the photoreceptors, well known to contain mitochondria. Mitochondrial proteins have previously been implicated in IRD, although usually in association with syndromic disease, unlike our present case. Together, our findings add another ultra-rare mutation implicated in non-syndromic IRD, whose pathogenic mechanism in the retina needs to be further elucidated.


Subject(s)
Cone-Rod Dystrophies/pathology , Genes, Recessive , Mitochondrial Proteins/genetics , Mutation , Potassium Channels/genetics , Adult , Cone-Rod Dystrophies/etiology , Cone-Rod Dystrophies/metabolism , Female , Humans , Male , Pedigree , Phenotype
7.
Retina ; 41(6): 1346-1355, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-34001834

ABSTRACT

PURPOSE: To describe an isolated maculopathy and an intermediate rod-cone dystrophy phenotype as the milder end of the RDH12-related retinal dystrophy spectrum. METHODS: Seven patients (17-34 years of age) underwent an extensive ophthalmic workup including psychophysical and electrophysiological testing and multimodal imaging. RESULTS: Three patients have isolated macular disease. Best-corrected visual acuity (BCVA) ranges from 20/125 to 20/40 with normal visual fields or only limited central, relative scotomata, and normal full-field ERGs. Both optical coherence tomography scans and autofluorescent imaging hint at relatively better-preserved foveal quality initially. An intermediate rod-cone phenotype in four patients is characterized by a central retinal dystrophy extending just beyond the vascular arcades, characteristic peripapillary sparing, and additional scattered atrophic patches. Again, foveal quality is initially better on optical coherence tomography scans. Best-corrected visual acuity ranges from counting fingers to 20/32. Goldmann visual fields vary from central scotomata to severe generalized abnormalities. ERGs range between mild and severe rod-cone dysfunction. Nine distinct RDH12 pathogenic variants, two of which are novel, are identified. CONCLUSION: The classic phenotype of RDH12-related early-onset retinal dystrophy is expanded to include an isolated maculopathy and intermediate dystrophy phenotype, characterized by its later onset and milder course with a fair visual potential until much later in life, emphasizing the phenotypic heterogeneity of RDH12-related retinopathy.


Subject(s)
Alcohol Oxidoreductases/genetics , Cone-Rod Dystrophies/genetics , Macular Degeneration/etiology , Mutation , Photoreceptor Cells, Vertebrate/pathology , Visual Acuity , Visual Fields/physiology , Adolescent , Adult , Alcohol Oxidoreductases/metabolism , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Electroretinography/methods , Female , Humans , Macular Degeneration/diagnosis , Macular Degeneration/genetics , Male , Pedigree , Phenotype , Tomography, Optical Coherence/methods , Young Adult
8.
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
10.
Ophthalmol Retina ; 4(7): 737-749, 2020 07.
Article in English | MEDLINE | ID: mdl-32646556

ABSTRACT

PURPOSE: To investigate quantitatively lipofuscin-associated fundus autofluorescence in patients with macular and cone/cone-rod dystrophies (MD/CCRDs). DESIGN: Prospective, single-center, case-control study. PARTICIPANTS: Two hundred thirty patients with MD/CCRDs who had undergone genetic testing and 110 control participants without any eye disease. METHODS: Participants were examined using quantitative fundus autofluorescence (qAF) imaging with a modified confocal scanning laser ophthalmoscope equipped with an internal fluorescent reference (modified Spectralis HRA-OCT; Heidelberg Engineering, Heidelberg, Germany). Mean qAF values were obtained by averaging measurements from an 8-segment ring centered on the fovea (qAF8) and compared with controls. MAIN OUTCOME MEASURES: The qAF8 levels. RESULTS: Elevated qAF8 values were a frequent finding (n = 105 [45%]) and associated with ABCA4 (n = 73 [70%]), PRPH2 (n = 9 [9%]), CERKL (n = 3 [3%]), PROM1 (n = 2 [2%]), CRX (n = 1 [1%]), and CDHR1 (n = 1 [1%]) mutations. Reduced qAF8 values were rare (n = 15 [7%]) and found predominantly among patients with MERTK (n = 3 [20%]) and RDH5 (n = 2 [13%]) mutations. Patients with normal qAF8 values (n = 110 [48%]) showed high genotypic heterogeneity. For various genes including ABCA4, PRPH2, CDHR1, and PROM1, higher qAF8 measures were associated with specific phenotypes and genotypes. For instance, qAF8 values were normal in PRPH2-related central areolar chorioretinal dystrophy but increased in PRPH2-related Stargardt-like retinopathy. Accordingly, high qAF8 levels were associated with specific genetic causes and mutation detection rates in characteristic but genetically heterogenous clinical phenotypes, such as a Stargardt-like flecked fundus, bull's eye maculopathy, or pattern dystrophy. In genetically unsolved cases (16 with elevated, 35 with normal, 7 with reduced qAF values), qAF8 was used to support or reject ambiguous results of genetic testing, to suggest underlying pathogenic pathways, and to predict disease in otherwise healthy participants. CONCLUSIONS: Quantitative fundus autofluorescence imaging revealed characteristic qAF levels in association with certain gene mutations and in participants without detected mutations. These findings indicate that qAF may facilitate differential diagnostics of MD/CCRDs and may offer novel pathogenetic insights that may be of particular value for the assessment of future treatment approaches.


Subject(s)
Cone-Rod Dystrophies/pathology , DNA/genetics , Eye Proteins/genetics , Fluorescein Angiography/methods , Mutation , Retinal Cone Photoreceptor Cells/pathology , Retinal Pigment Epithelium/pathology , Adolescent , Adult , Case-Control Studies , Child , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/metabolism , Cross-Sectional Studies , DNA Mutational Analysis , Eye Proteins/metabolism , Female , Fundus Oculi , Humans , Male , Middle Aged , Ophthalmoscopy/methods , Pedigree , Prospective Studies , Tomography, Optical Coherence/methods , Young Adult
11.
Invest Ophthalmol Vis Sci ; 61(6): 17, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32516403

ABSTRACT

Purpose: Riboflavin and its cofactors are essential for cellular energy generation, responses to oxidative stress, and overall homeostasis. Retbindin is a novel retina-specific riboflavin binding protein essential for the maintenance of retinal flavin levels, but its function remains poorly understood. To further elucidate the function of retbindin in retinal health and disease, we evaluated its role in retinal degeneration in a cone-rod dystrophy model associated with the R172W mutation in the photoreceptor tetraspanin Prph2. Methods: We performed structural, functional, and biochemical characterization of R172W-Prph2 mice with and without retbindin (Rtbdn-/-/Prph2R172W). Results: Retbindin is significantly upregulated during degeneration in the R172W model, suggesting that retbindin plays a protective role in retinal degenerative diseases. This hypothesis was supported by our findings that R172W mice lacking retbindin (Rtbdn-/-/Prph2R172W) exhibit functional and structural defects in rods and cones that are significantly worse than in controls. Retinal flavin levels were also altered in the Rtbdn-/-/Prph2R172W retina. However, in contrast to the Rtbdn-/- retina which has reduced flavin levels compared to wild-type, Rtbdn-/-/Prph2R172W retinas exhibited elevated levels of riboflavin and the flavin cofactor FMN. Conclusions: These results indicate that retbindin plays a protective role during retinal degeneration, but that its function is more complex than previously thought, and suggest a possible role for retbindin in protecting the retina from phototoxicity associated with unbound flavins. This study highlights the essential role of precisely regulated homeostatic mechanisms in photoreceptors, and shows that disruption of this metabolic balance can contribute to the degenerative process associated with other cellular defects.


Subject(s)
Cone-Rod Dystrophies/genetics , Disease Models, Animal , Eye Proteins/genetics , Gene Expression Regulation/physiology , Membrane Transport Proteins/genetics , Retina/pathology , Adenosine Triphosphate/metabolism , Animals , Chromatography, High Pressure Liquid , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/physiopathology , Electroretinography , Female , Fluorescein Angiography , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Retina/physiopathology
12.
Retin Cases Brief Rep ; 14(2): 203-210, 2020.
Article in English | MEDLINE | ID: mdl-29200130

ABSTRACT

PURPOSE: To describe a specific cone-rod dystrophy phenotype in a family with the homozygous c.1429G>A; p.Gly477Arg mutation in CRB1. The detailed phenotype of subjects with this specific mutation has not been described previously. METHODS: Clinical examination included full-field electroretinography and high-resolution and widefield retinal imaging and uveitis workup. Molecular genetic analysis included next-generation sequencing of known retinal dystrophy genes and Sanger sequencing for segregation analysis. RESULTS: Three affected male siblings (26, 16, and 8 years old) were diagnosed with cone-rod dystrophy, featuring bilateral macular hypoautofluorescent lesions. In addition, the eldest brother was found to have retinal vascular leakage throughout the retina without telangiectasia. Uveitis laboratory workup was unremarkable. The homozygous c.1429G>A; p.Gly477Arg mutation in CRB1 was found to segregate with disease in this family. CONCLUSION: To the best of our knowledge, diffuse vascular leakage without telangiectasia or exudation, with bull's eye maculopathy, has not been reported previously in CRB1-cone rod dystrophy. This expands the phenotype complexity associated with CRB1 mutations and confirms that dystrophies associated with mutations in this gene may appear with features of uveitis.


Subject(s)
Cone-Rod Dystrophies/genetics , DNA/genetics , Eye Proteins/genetics , Homozygote , Membrane Proteins/genetics , Mutation, Missense , Nerve Tissue Proteins/genetics , Retina/pathology , Adolescent , Adult , Child , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Eye Proteins/metabolism , Follow-Up Studies , Humans , Male , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Pedigree , Phenotype , Retina/metabolism , Retrospective Studies , Tomography, Optical Coherence
13.
Hum Mol Genet ; 28(24): 4078-4088, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31628458

ABSTRACT

Mutations of the photoreceptor disc component (PRCD) gene are associated with rod-cone degeneration in both dogs and humans. Prcd is expressed in the mouse eye as early as embryonic day 14. In the adult mouse retina, PRCD is expressed in the outer segments of both rod and cone photoreceptors. Immunoelectron microscopy revealed that PRCD is located at the outer segment rim and that it is highly concentrated at the base of the outer segment. Prcd-knockout mice present with progressive retinal degeneration, starting at 20 weeks of age and onwards. This process is reflected by a significant and progressive reduction of both scotopic and photopic electroretinographic responses and by thinning of the retina, and specifically of the outer nuclear layer, indicating photoreceptor loss. Electron microscopy revealed severe damage to photoreceptor outer segments, which is associated with immigration of microglia cells to the Prcd-knockout retina and accumulation of vesicles in the inter-photoreceptor space. Phagocytosis of photoreceptor outer segment discs by the retinal pigmented epithelium is severely reduced. Our data show that Prcd-knockout mice serve as a good model for retinal degeneration caused by PRCD mutations in humans. Our findings in these mice support the involvement of PRCD in outer segment disc formation of both rod and cone photoreceptors. Furthermore, they suggest a feedback mechanism which coordinates the rate of photoreceptor outer segment disc formation, shedding and phagocytosis. This study has important implications for understanding the function of PRCD in the retina, as well as for future development of treatment modalities for PRCD deficiency in humans.


Subject(s)
Cone-Rod Dystrophies/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Degeneration/genetics , Rod Cell Outer Segment/pathology , Animals , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/pathology , Eye Proteins/genetics , Female , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Rod Cell Outer Segment/metabolism , Signal Transduction
14.
Exp Eye Res ; 189: 107846, 2019 12.
Article in English | MEDLINE | ID: mdl-31626798

ABSTRACT

The cone-rod homeobox (CRX) gene is specifically expressed in developing and mature photoreceptors and is relatively conserved, with limited polymorphisms in coding regions. Rare variants in CRX are usually considered causative for different forms of retinal degeneration, but this might be problematic based on recent data. This study aimed to classify CRX variants based on a genotype-phenotype analysis of our data and the literature. Twenty-four CRX variants, including 14 novel variants, were detected in 37 Chinese families based on exome sequencing data obtained from 4971 Chinese probands with different forms of eye diseases. After detailed phenotypic analysis and cosegregation analysis in families with CRX variants, the 24 variants could be classified into three groups: benign (six), likely benign (six), and pathogenic (12). Somatic mosaicism was identified in a family with unaffected parents (the father had a mutant allele that was detected in approximately 17% of his leukocyte DNA) and two affected sons. Furthermore, a thorough reassessment was systematically performed for all 113 heterozygous variants as well as for their associated phenotypes from our cohort and patients previously reported. Two critical findings on the pathogenicity of CRX variants were obtained based on the genotype-phenotype correlation, family segregation and ensemble predicting methods: 1) approximately half of heterozygous missense variants are likely benign, and 2) heterozygous truncating variants affecting the homeodomain are likely benign. Truncating mutations after the homeodomain are likely associated with a more severe phenotype. Although most heterozygous pathogenic variants in CRX are associated with autosomal dominant retinal degeneration, a homozygous c.268C> T (p.Arg90Trp) substitution and homozygous complete deletion of CRX have been reported to cause Leber congenital amaurosis. In conclusion, many rare missense variants and some truncating variants in CRX are likely benign, although previously, they might have been predicted to be damaging by some online tools. Evaluation of the pathogenicity of a CRX variant should consider both its nature and location. The information obtained in this study is critical in the era of routine clinical genetic test, not only for CRX but also for many other genes with many more variants. Functional studies and additional genotype-phenotype analyses are expected to confirm these associations.


Subject(s)
Cone-Rod Dystrophies/genetics , DNA/genetics , Homeodomain Proteins/genetics , Leber Congenital Amaurosis/genetics , Mutation , Trans-Activators/genetics , Adult , Alleles , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Female , Genetic Association Studies , Genetic Testing , Homeodomain Proteins/metabolism , Humans , Leber Congenital Amaurosis/diagnosis , Leber Congenital Amaurosis/metabolism , Male , Pedigree , Phenotype , Trans-Activators/metabolism
15.
Stem Cell Res ; 38: 101478, 2019 07.
Article in English | MEDLINE | ID: mdl-31203166

ABSTRACT

We generated an induced pluripotent stem cell (iPSC) line using dermal fibroblasts from a 53 year-old patient with autosomal dominant cone-rod dystrophy (CRD) caused by a missense mutation, c.121C > T, in the CRX gene. Patient fibroblasts were reprogrammed using the non-integrative Sendai virus reprogramming system and the human OSKM transcription factor cocktail. The generated iPSCs contained the congenital mutation in exon 3 of CRX and were pluripotent and genetically stable. This iPSC line will be an important tool for retinal differentiation studies to better understand the CRD phenotype caused by the mutant p.Arg41Trp CRX protein.


Subject(s)
Cellular Reprogramming Techniques , Cone-Rod Dystrophies , Fibroblasts/metabolism , Homeodomain Proteins , Induced Pluripotent Stem Cells/metabolism , Mutation, Missense , Trans-Activators , Amino Acid Substitution , Cell Line , Cone-Rod Dystrophies/genetics , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/pathology , Fibroblasts/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/pathology , Male , Middle Aged , Trans-Activators/genetics , Trans-Activators/metabolism
16.
Can J Ophthalmol ; 54(1): 40-50, 2019 02.
Article in English | MEDLINE | ID: mdl-30851773

ABSTRACT

OBJECTIVE: To identify the underlying genetic defect of childhood-onset severe rod-cone dystrophy (RCD) in a consanguineous family from North India with autosomal recessive retinitis pigmentosa. METHODS: A detailed family history, clinical data, and blood samples were collected from 11 members of the family, including 4 affected by an autosomal recessive rod-cone dystrophy (arRCD), and DNA was extracted. Whole-exome sequencing (WES) was performed on DNA samples of proband and her unaffected maternal uncle. Ion Reporter software (ver. 4.4) was used for the annotation of variants obtained by WES. The variants detected in proband were tested for validation in all other affected and unaffected family members using Sanger sequencing technique. RESULTS: We have identified a novel nonsense mutation-c.1647T>G (p.Tyr549Ter)-in the exon 11 of MERTK that co-segregated completely with the disease phenotype in all the 4 affected members and was not observed in the 7 unaffected members of the family. This mutation was also not detected in 120 ethnically matched controls (240 chromosomes), hence excluding it as a polymorphism. CONCLUSIONS: MERTK has a role in retinal pigment epithelium as a regulator of rod outer segments' phagocytosis. Due to c.1647T > G substitution, the stop codon (p.Tyr549Ter) appears early in the transcript. It seems that either the altered transcript would degenerate through nonsense-mediated decay (NMD) or potentially form truncated protein lacking a functionally important domain (i.e., tyrosine kinase domain). These findings thus further expand the mutation spectrum in MERTK and substantiate its role in the pathogenesis of retinal dystrophy.


Subject(s)
Cone-Rod Dystrophies/genetics , DNA/metabolism , Mutation , c-Mer Tyrosine Kinase/genetics , Adolescent , Adult , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Electroretinography , Female , Humans , India , Male , Middle Aged , Retinal Pigment Epithelium/pathology , Visual Acuity , Young Adult , c-Mer Tyrosine Kinase/metabolism
17.
Mol Vis ; 25: 106-117, 2019.
Article in English | MEDLINE | ID: mdl-30820146

ABSTRACT

Purpose: Inherited retinal diseases (IRDs) are clinically and genetically heterogeneous showing progressive retinal cell death which results in vision loss. IRDs include a wide spectrum of disorders, such as retinitis pigmentosa (RP), Leber congenital amaurosis (LCA), cone-rod dystrophy (CRD), and Stargardt disease (STGD1). Methods: In this study, we performed targeted next-generation sequencing based on molecular inversion probes (MIPs) that allowed the sequence analysis of 108 IRD-associated genes in 50 Iranian IRD probands. Results: The sequencing and variant filtering led to the identification of putative pathogenic variants in 36 out of 50 (72%) probands. Among 36 unique variants, we identified 20 novel variants in 15 genes. Four out of 36 probands carry compound heterozygous variants, and 32 probands carry homozygous variants. Conclusions: Employing a cost-effective targeted next-generation sequencing procedure, we identified the genetic causes of different retinal disorders in the majority of Iranian families in this study.


Subject(s)
Cone-Rod Dystrophies/genetics , Eye Proteins/genetics , Leber Congenital Amaurosis/genetics , Macular Degeneration/congenital , Mutation , Retinitis Pigmentosa/genetics , Adolescent , Adult , Child , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/pathology , Eye Proteins/metabolism , Female , Gene Expression , Genetic Association Studies , Genotype , Heterozygote , High-Throughput Nucleotide Sequencing/economics , High-Throughput Nucleotide Sequencing/methods , Homozygote , Humans , Iran , Leber Congenital Amaurosis/metabolism , Leber Congenital Amaurosis/pathology , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Pedigree , Phenotype , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/congenital , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Stargardt Disease
18.
J Med Genet ; 56(6): 358-369, 2019 06.
Article in English | MEDLINE | ID: mdl-30705057

ABSTRACT

Jalili syndrome is a rare genetic disorder first identified by Jalili in Gaza. Amelogenesis imperfecta and cone-rode dystrophy are simultaneously seen in Jalili syndrome patients as the main and primary manifestations. Molecular analysis has revealed that the CNNM4 gene is responsible for this rare syndrome. Jalili syndrome has been observed in many countries around the world, especially in the Middle East and North Africa. In the current scoping systematic review we searched electronic databases to find studies related to Jalili syndrome. In this review we summarise the reported clinical symptoms, CNNM4 gene and protein structure, CNNM4 mutations, attempts to reach a genotype-phenotype correlation, the functional role of CNNM4 mutations, and epidemiological aspects of Jalili syndrome. In addition, we have analysed the reported mutations in mutation effect prediction databases in order to gain a better understanding of the mutation's outcomes.


Subject(s)
Amelogenesis Imperfecta/diagnosis , Amelogenesis Imperfecta/genetics , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Phenotype , Amelogenesis Imperfecta/epidemiology , Amelogenesis Imperfecta/metabolism , Biomarkers , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cone-Rod Dystrophies/epidemiology , Cone-Rod Dystrophies/metabolism , Genetic Association Studies/methods , Humans , Mutation
19.
J Biol Chem ; 294(10): 3476-3488, 2019 03 08.
Article in English | MEDLINE | ID: mdl-30622141

ABSTRACT

The guanylyl cyclase-activating protein, GCAP1, activates photoreceptor membrane guanylyl cyclase (RetGC) in the light, when free Ca2+ concentrations decline, and decelerates the cyclase in the dark, when Ca2+ concentrations rise. Here, we report a novel mutation, G86R, in the GCAP1 (GUCA1A) gene in a family with a dominant retinopathy. The G86R substitution in a "hinge" region connecting EF-hand domains 2 and 3 in GCAP1 strongly interfered with its Ca2+-dependent activator-to-inhibitor conformational transition. The G86R-GCAP1 variant activated RetGC at low Ca2+ concentrations with higher affinity than did the WT GCAP1, but failed to decelerate the cyclase at the Ca2+ concentrations characteristic of dark-adapted photoreceptors. Ca2+-dependent increase in Trp94 fluorescence, indicative of the GCAP1 transition to its RetGC inhibiting state, was suppressed and shifted to a higher Ca2+ range. Conformational changes in G86R GCAP1 detectable by isothermal titration calorimetry (ITC) also became less sensitive to Ca2+, and the dose dependence of the G86R GCAP1-RetGC1 complex inhibition by retinal degeneration 3 (RD3) protein was shifted toward higher than normal concentrations. Our results indicate that the flexibility of the hinge region between EF-hands 2 and 3 is required for placing GCAP1-regulated Ca2+ sensitivity of the cyclase within the physiological range of intracellular Ca2+ at the expense of reducing GCAP1 affinity for the target enzyme. The disease-linked mutation of the hinge Gly86, leading to abnormally high affinity for the target enzyme and reduced Ca2+ sensitivity of GCAP1, is predicted to abnormally elevate cGMP production and Ca2+ influx in photoreceptors in the dark.


Subject(s)
Calcium/metabolism , Cone-Rod Dystrophies/genetics , Guanylate Cyclase-Activating Proteins/genetics , Guanylate Cyclase-Activating Proteins/metabolism , Guanylate Cyclase/metabolism , Mutation , Retina/enzymology , Cell Death/genetics , Cone-Rod Dystrophies/enzymology , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/pathology , Guanylate Cyclase-Activating Proteins/chemistry , Humans , Models, Molecular , Protein Conformation, alpha-Helical , Retina/pathology , Retinal Cone Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/pathology
20.
Graefes Arch Clin Exp Ophthalmol ; 257(3): 619-628, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30588538

ABSTRACT

PURPOSE: To characterize two patients with macular and rod-cone dystrophy and identify the genetic basis for disease. METHOD: Ophthalmic examinations were performed for the family and the peripheral blood samples were collected for whole exome sequencing. The mutated sequences of PROM1 gene were cloned and expressed in cultured cell lines after transient transfection followed by analysis with confocal microscopy and bridge-PCR. RESULT: We reported that two patients, brothers in a family, were diagnosed with macular and rod-cone dystrophy. Phenotypically, both patients experience progressive visual impairment and nyctalopia. The fundus examination showed macular and choroid dystrophy with pigment deposits in the macular region. Functionally, photoreceptor response to electrophysiological stimulation was significantly compromised with more severe decline in rods. Genetic analysis by whole exome sequencing revealed two novel compound heterogeneous point mutations in PROM1 gene that co-segregate with patients in an autosomal recessive manner. Specifically, the c.C1902G(p.Y634X) nonsense mutation results in a truncated, labile, and mislocalized protein, while the c.C1682+3A>G intronic mutation disrupts messenger RNA splicing. CONCLUSION: Our findings have identified two novel deleterious mutations in PROM1 gene that are associated with hereditary macular and rod-cone dystrophy in human.


Subject(s)
AC133 Antigen/genetics , Cone-Rod Dystrophies/genetics , DNA/genetics , Mutation , Retinal Rod Photoreceptor Cells/pathology , Tomography, Optical Coherence/methods , AC133 Antigen/metabolism , Adult , Blotting, Western , Cells, Cultured , Cone-Rod Dystrophies/diagnosis , Cone-Rod Dystrophies/metabolism , DNA Mutational Analysis , Electroretinography , Female , Genes, Recessive , Humans , Male , Microscopy, Confocal , Middle Aged , Pedigree , Phenotype , Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...