Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 385
Filter
1.
Nat Commun ; 15(1): 3773, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710738

ABSTRACT

Bietti crystalline corneoretinal dystrophy (BCD) is an autosomal recessive chorioretinal degenerative disease without approved therapeutic drugs. It is caused by mutations in CYP4V2 gene, and about 80% of BCD patients carry mutations in exon 7 to 11. Here, we apply CRISPR/Cas9 mediated homology-independent targeted integration (HITI)-based gene editing therapy in HEK293T cells, BCD patient derived iPSCs, and humanized Cyp4v3 mouse model (h-Cyp4v3mut/mut) using two rAAV2/8 vectors via sub-retinal administration. We find that sgRNA-guided Cas9 generates double-strand cleavage on intron 6 of the CYP4V2 gene, and the HITI donor inserts the carried sequence, part of intron 6, exon 7-11, and a stop codon into the DNA break, achieving precise integration, effective transcription and translation both in vitro and in vivo. HITI-based editing restores the viability of iPSC-RPE cells from BCD patient, improves the morphology, number and metabolism of RPE and photoreceptors in h-Cyp4v3mut/mut mice. These results suggest that HITI-based editing could be a promising therapeutic strategy for those BCD patients carrying mutations in exon 7 to 11, and one injection will achieve lifelong effectiveness.


Subject(s)
CRISPR-Cas Systems , Corneal Dystrophies, Hereditary , Cytochrome P450 Family 4 , Gene Editing , Genetic Therapy , Induced Pluripotent Stem Cells , Retinal Diseases , Humans , Gene Editing/methods , Animals , HEK293 Cells , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/therapy , Corneal Dystrophies, Hereditary/pathology , Corneal Dystrophies, Hereditary/metabolism , Mice , Induced Pluripotent Stem Cells/metabolism , Genetic Therapy/methods , Cytochrome P450 Family 4/genetics , Cytochrome P450 Family 4/metabolism , Disease Models, Animal , Mutation , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Genetic Vectors/genetics , Introns/genetics , Exons/genetics
2.
Int J Mol Sci ; 25(3)2024 Feb 04.
Article in English | MEDLINE | ID: mdl-38339172

ABSTRACT

The study investigated a profile of chemokines and growth factors in the aqueous humor (AH) of eyes with Fuch's endothelial corneal dystrophy (FECD) and cataracts in comparison with cataract patients as a control group. A total of 52 AH samples (26 FECD + cataract and 26 cataract/control) were collected before cataract surgery. None of the patients had any clinically apparent inflammation at the time of AH collection. The AH levels of MCP-1 (CCL2), MIP-1α (CCL3), MIP-1ß(CCL4), RANTES (CCL5), eotaxin (CCL11), IP-10 (CXCL10), FGF basic, G-CSF, GM-CSF, PDGF-bb, and VEGF were compared between the groups. The analyses were performed using the Bio-Plex 200 System from Bio-Rad. Among the studied parameters, the AH levels of RANTES, eotaxin, and IP-10 significantly increased in the FECD + cataract eyes, compared with the cataract controls (p < 0.05). Elevated levels of the RANTES, Eotaxin, and IP-10 indicate more intense inflammation in the eyes of patients in the FECD + cataract group. Moreover, these factors exhibit potential as predictive biomarkers for early detection of FECD in cataract patients. The discovery of elevated concentrations of biochemical markers in a patient, who has not yet received a clinical diagnosis, may suggest the need for heightened observation of the other eye to monitor the potential development of FECD.


Subject(s)
Cataract , Corneal Dystrophies, Hereditary , Humans , Cytokines/metabolism , Aqueous Humor/metabolism , Chemokine CXCL10/metabolism , Chemokines/metabolism , Cataract/metabolism , Inflammation/metabolism , Corneal Dystrophies, Hereditary/metabolism
3.
Cornea ; 43(4): 466-527, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38359414

ABSTRACT

PURPOSE: The International Committee for the Classification of Corneal Dystrophies (IC3D) was created in 2005 to develop a new classification system integrating current information on phenotype, histopathology, and genetic analysis. This update is the third edition of the IC3D nomenclature. METHODS: Peer-reviewed publications from 2014 to 2023 were evaluated. The new information was used to update the anatomic classification and each of the 22 standardized templates including the level of evidence for being a corneal dystrophy [from category 1 (most evidence) to category 4 (least evidence)]. RESULTS: Epithelial recurrent erosion dystrophies now include epithelial recurrent erosion dystrophy, category 1 ( COL17A1 mutations, chromosome 10). Signs and symptoms are similar to Franceschetti corneal dystrophy, dystrophia Smolandiensis, and dystrophia Helsinglandica, category 4. Lisch epithelial corneal dystrophy, previously reported as X-linked, has been discovered to be autosomal dominant ( MCOLN1 mutations, chromosome 19). Classic lattice corneal dystrophy (LCD) results from TGFBI R124C mutation. The LCD variant group has over 80 dystrophies with non-R124C TGFBI mutations, amyloid deposition, and often similar phenotypes to classic LCD. We propose a new nomenclature for specific LCD pathogenic variants by appending the mutation using 1-letter amino acid abbreviations to LCD. Pre-Descemet corneal dystrophies include category 1, autosomal dominant, punctiform and polychromatic pre-Descemet corneal dystrophy (PPPCD) ( PRDX3 mutations, chromosome 10). Typically asymptomatic, it can be distinguished phenotypically from pre-Descemet corneal dystrophy, category 4. We include a corneal dystrophy management table. CONCLUSIONS: The IC3D third edition provides a current summary of corneal dystrophy information. The article is available online at https://corneasociety.org/publications/ic3d .


Subject(s)
Corneal Dystrophies, Hereditary , Epithelium, Corneal/pathology , Humans , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Mutation , Transforming Growth Factor beta/genetics , Phenotype , Extracellular Matrix Proteins/genetics , Pedigree , DNA Mutational Analysis
5.
Commun Biol ; 6(1): 348, 2023 03 30.
Article in English | MEDLINE | ID: mdl-36997596

ABSTRACT

TGFBI-related corneal dystrophy (CD) is characterized by the accumulation of insoluble protein deposits in the corneal tissues, eventually leading to progressive corneal opacity. Here we show that ATP-independent amyloid-ß chaperone L-PGDS can effectively disaggregate corneal amyloids in surgically excised human cornea of TGFBI-CD patients and release trapped amyloid hallmark proteins. Since the mechanism of amyloid disassembly by ATP-independent chaperones is unknown, we reconstructed atomic models of the amyloids self-assembled from TGFBIp-derived peptides and their complex with L-PGDS using cryo-EM and NMR. We show that L-PGDS specifically recognizes structurally frustrated regions in the amyloids and releases those frustrations. The released free energy increases the chaperone's binding affinity to amyloids, resulting in local restructuring and breakage of amyloids to protofibrils. Our mechanistic model provides insights into the alternative source of energy utilized by ATP-independent disaggregases and highlights the possibility of using these chaperones as treatment strategies for different types of amyloid-related diseases.


Subject(s)
Corneal Dystrophies, Hereditary , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/metabolism , Cornea/metabolism , Corneal Dystrophies, Hereditary/metabolism , Amyloid/metabolism , Molecular Chaperones/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Adenosine Triphosphate/metabolism
6.
Expert Rev Clin Pharmacol ; 15(12): 1423-1442, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36308770

ABSTRACT

INTRODUCTION: Transforming growth factor beta induced (TGFBI) gene mutations have been reported as the cause of a group of genetically inherited, visually debilitating, corneal dystrophies (CD). A scoping literature review to identify and categorize compounds that inhibit corneal TGFBI expression and/or promote TGFBIp degradation was performed. Emphasis was given to their potential to be used as a cost-effective approach via drug repurposing. AREAS COVERED: We performed a thorough search of original peer-reviewed literature using electronic bibliographic databases and selected articles according to a set of criteria. The total number of articles retrieved from the search terms applied to the databases was 2344. The number of relevant full-text articles included added up to 19. We identified 16 compounds that can theoretically reduce the levels of mutant TGFBIp in human corneal cells. EXPERT OPINION: Currently, the only temporary treatments available for this condition are lubricant drops and surgery. Here, we explored the crosstalk between cascades that regulate TGFBI expression and identified compounds that target these pathways. Compounds that inhibit DNA synthesis and function, increase elimination of TGFBIp or bind to mutant TGFBIp were also explored with the aim of highlighting promising compounds that can be used in future cost-effective drug-repurposing studies.


Subject(s)
Corneal Dystrophies, Hereditary , Transforming Growth Factor beta , Humans , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/therapy , Cornea/metabolism , Mutation
7.
Orphanet J Rare Dis ; 17(1): 361, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115991

ABSTRACT

BACKGROUND: Congenital hereditary endothelial dystrophy (CHED) is a rare form of corneal dystrophy caused by SLC4A11 gene variations. This study aims to find the genetic alterations in SLC4A11, in two Indian familial CHED cases with affected members n = 3 and n = 2 respectively and five sporadic CHED cases using direct sequencing, followed by in silico analysis and characterization of the identified variants. RESULTS: All three affected members of the first CHED family were identified with a novel homozygous c.1514C > G (p.Ser489Trp) variation while second family showed presence of a compound heterozygous variation c.529A > C (p.Arg161Arg) + c.2461insT (p.Val805fs). Among five sporadic cases, two showed novel changes, homozygous c.1487G > T (p.Ser480Ile) and c.620-2A > G, while the other one had previously reported homozygous c.2653C > T (p.Arg869Cys) variation. The remaining two cases did not reveal the presence of SLC4A11-related pathogenic variations. The identified variations were excluded from the Indian control (n = 80). In silico analysis using homology-based protein modeling and pathogenicity prediction tools, which revealed these alterations as pathogenic, changing their protein stability, local flexibility, residue contact clashes, and the hydrogen bond interactions. CONCLUSIONS: This study contributed to the CHED mutational spectrum, adding four novel variations and confirming a previously reported one. It demonstrates different type of variations in CHED cases, including coding, non-coding, homozygous, synonymous, and compound heterozygous variations. The identified variations revealed different degrees of pathogenic effects in silico. Moreover, two sporadic cases could not be identified with pathogenic variation emphasizing the involvement of other genes or genetic mechanisms.


Subject(s)
Anion Transport Proteins , Antiporters , Corneal Dystrophies, Hereditary , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antiporters/genetics , Antiporters/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Homozygote , Humans , India , Mutation/genetics
8.
Indian J Ophthalmol ; 70(7): 2239-2248, 2022 07.
Article in English | MEDLINE | ID: mdl-35791103

ABSTRACT

Corneal endothelial dystrophies are a heterogeneous group of diseases with different modes of inheritance and genetic basis for each dystrophy. The genes associated with these diseases encode transcription factors, structural components of the stroma and Descemet membrane, cell transport proteins, and others. Congenital hereditary endothelial dystrophy (CHED) is associated with mutations in two genes, OVOL2 and SLC4A11, for dominant and recessive forms of CHED, respectively. Mutations in three genes are known to cause posterior polymorphous corneal dystrophy (PPCD). They are OVOL2 (PPCD1), ZEB1 (PPCD3), and GRHL1 (PPCD4). The PPCD2 locus involving the collagen gene COL8A2 on chromosome 1 is disputed due to insufficient evidence. Mutations in the COL8A2 gene are associated with early-onset Fuchs' endothelial corneal dystrophy (FECD). Several genes have been associated with the more common, late-onset FECD. Alterations in each of these genes occur in a fraction of patients, and the most prevalent genetic alteration in FECD patients across the world is a triplet repeat expansion in the TCF4 gene. Knowledge of the genetics of corneal endothelial dystrophies has considerably advanced within the last decade and has contributed to better diagnosis of these dystrophies as well as opened up the possibility of novel therapeutic approaches based on the molecular mechanisms involved. The functions of genes identified to date provide insights into the pathogenic mechanisms involved in each disorder.


Subject(s)
Corneal Dystrophies, Hereditary , Fuchs' Endothelial Dystrophy , Anion Transport Proteins/genetics , Anion Transport Proteins/metabolism , Antiporters/genetics , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Fuchs' Endothelial Dystrophy/genetics , Fuchs' Endothelial Dystrophy/pathology , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Int J Mol Sci ; 24(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36613994

ABSTRACT

We determined the efficacy and safety of 0.1% RGN-259 ophthalmic solution (containing the regenerative protein thymosin ß4) in promoting the healing of persistent epithelial defects in patients with Stages 2 and 3 neurotrophic keratopathy. Complete healing occurred after 4 weeks in 6 of the 10 RGN-259-treated subjects and in 1 of the 8 placebo-treated subjects (p = 0.0656), indicating a strong efficacy trend. Additional efficacy was seen in the significant healing (p = 0.0359) with no recurrent defects observed at day 43, two weeks after cessation of treatment, while the one healed placebo-treated subject at day 28 suffered a recurrence at day 43. The Mackie classification disease stage improved in the RGN-259-treated group at Days 29, 36, and 43 (p = 0.0818, 0.0625, and 0.0467, respectively). Time to complete healing also showed a trend towards efficacy (p = 0.0829, Kaplan-Meier) with 0.1% RGN-259. RGN-259-treated subjects had significant improvements at multiple time points in ocular discomfort, foreign body sensation, and dryness which were not seen in the placebo group. No significant adverse effects were observed. In summary, the use of 0.1% RGN-259 promotes rapid healing of epithelial defects in neurotrophic keratopathy, improves ocular comfort, and is safe for treating this challenging population of patients.


Subject(s)
Corneal Dystrophies, Hereditary , Keratitis , Thymosin , Trigeminal Nerve Diseases , Humans , Cornea/metabolism , Ophthalmic Solutions/pharmacology , Thymosin/metabolism , Treatment Outcome , Keratitis/drug therapy , Trigeminal Nerve Diseases/metabolism , Corneal Dystrophies, Hereditary/metabolism , Double-Blind Method
10.
Autophagy ; 18(4): 765-782, 2022 04.
Article in English | MEDLINE | ID: mdl-34403298

ABSTRACT

Thiel-Behnke corneal dystrophy (TBCD) is an epithelial-stromal TGFBI dystrophy caused by mutations in the TGFBI (transforming growth factor beta induced) gene, though the underlying mechanisms and pathogenesis of TBCD are still obscure. The study identifies a novel mutation in the TGFBI gene (p.Gly623_His626del) in a TBCD pedigree. Characteristics of the typical vacuole formation, irregular corneal epithelial thickening and thinning, deposition of eosinophilic substances beneath the epithelium, and involvement of the anterior stroma were observed in this pedigree via transmission electron microscopy (TEM) and histological staining. Tgfbi-p.Gly623_Tyr626del mouse models of TBCD were subsequently generated via CRISPR/Cas9 technology, and the above characteristics were further verified via TEM and histological staining. Lysosomal dysfunction and downregulation of differential expression protein CTSD (cathepsin D) were observed using LysoTracker Green DND-26 and proteomic analysis, respectively. Hence, lysosomal dysfunction probably leads to autophagic flux obstruction in TBCD; this was supported by enhanced LC3-II and SQSTM1 levels and decreased CTSD. TFEB (transcription factor EB) was prominently decreased in TBCD corneal fibroblasts and administration of ATP-competitive MTOR inhibitor torin 1 reversed this decline, resulting in the degradation of accumulated mut-TGFBI (mutant TGFBI protein) via the ameliorative lysosomal function and autophagic flux owing to elevated TFEB activity as measured by western blot, confocal microscopy, and flow cytometry. Transfected HEK 293 cells overexpressing human full-length WT-TGFBI and mut-TGFBI were generated to further verify the results obtained in human corneal fibroblasts. Amelioration of lysosome dysfunction may therefore have therapeutic efficacy in the treatment of TBCD.Abbreviations AS-OCT: anterior segment optical coherence tomography; ATP: adenosine triphosphate; Cas9: CRISPR-associated protein 9; CLEAR: coordinated lysosomal expression and regulation; CRISPR: clustered regularly interspaced short palindromic repeats; CTSB: cathepsin B; CTSD: cathepsin D; CTSF: cathepsin F; CTSL: cathepsin L; DNA: deoxyribonucleic acid; ECM: extracellular matrix; Fas1: fasciclin 1; FC: flow cytometry; GAPDH: glyceraldeyde-3-phosphate dehydrogenase; GCD2: granular corneal dystrophy type 2; HE: hematoxylin and eosin; LAMP2: lysosomal-associated membrane protein; MT: mutation type; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; mut-TGFBI: mutant TGFBI protein; SD: standard deviation; TBCD: Thiel-Behnke corneal dystrophy; TEM: transmission electron microscopy; TFEB: transcription factor EB; TGFBI: transforming growth factor beta induced; WT: wild type.


Subject(s)
Cathepsin D , Corneal Dystrophies, Hereditary , Adenosine Triphosphate/metabolism , Animals , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Blood Proteins , Cathepsin D/metabolism , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , HEK293 Cells , Humans , Lysosomes/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Mutant Proteins/metabolism , Proteomics , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism
11.
Cornea ; 41(6): 779-781, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-34369396

ABSTRACT

PURPOSE: The aim of this study was to report the results of screening peroxiredoxin 3 (PRDX3) and PDZ domain-containing protein 8 (PDZD8) in a previously unreported pedigree with punctiform and polychromatic pre-Descemet corneal dystrophy (PPPCD) to confirm that the PRDX3 mutation c.568G>C is the genetic basis of PPPCD. METHODS: Ophthalmologic examination of the proband and her affected father was performed with slit lamp biomicroscopy. Saliva was collected from the proband as a source of DNA, after which screening for PRDX3 and PDZD8 was performed. RESULTS: Slit lamp examination of the proband revealed polychromatic deposits diffusely distributed at the pre-Descemet level in both corneas and anterior subcapsular in the crystalline lens of both eyes. The proband's father also demonstrated diffuse pre-Descemetic polychromatic deposits in both eyes but no lenticular deposits. Screening of PRDX3 in the proband demonstrated the c.568G>C (p.Asp190His) variant previously associated with PPPCD and failed to identify any variants in PDZD8. CONCLUSIONS: We report the initial confirmation of PRDX3 as the genetic basis of PPPCD in a previously unreported pedigree and expand the phenotype of PPPCD to include polychromatic lenticular deposits.


Subject(s)
Corneal Dystrophies, Hereditary , Peroxiredoxin III , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cornea/metabolism , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Female , Humans , Male , Mutation , Pedigree , Peroxiredoxin III/genetics , Peroxiredoxin III/metabolism
12.
Biochim Biophys Acta Proteins Proteom ; 1870(3): 140746, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34942360

ABSTRACT

Mutations in the fasciclin 1 domain 4 (FAS1-4) of transforming growth factor ß-induced protein (TGFBIp) are associated with insoluble extracellular deposits and corneal dystrophies (CDs). The decrease in solubility upon mutation has been implicated in CD; however, the exact molecular mechanisms are not well understood. Here, we performed molecular dynamics simulations followed by solvation thermodynamic analyses of the FAS1-4 domain and its three mutants-R555W, R555Q, and A546T-linked to granular corneal dystrophy type 1, Thiel-Behnke corneal dystrophy and lattice corneal dystrophy, respectively. We found that both R555W and R555Q mutants have less affinity toward solvent water relative to the wild-type protein. In the R555W mutant, a remarkable increase in solvation free energy was observed because of the structural changes near the mutation site. The mutation site W555 is buried in other hydrophobic residues, and R557 simultaneously forms salt bridges with E554 and D561. In the R555Q mutant, the increase in solvation free energy is caused by structural rearrangements far from the mutation site. R558 separately forms salt bridges with D575, E576, and E598. Thus, we thus identified the relationship between the decrease in solubility and conformational changes caused by mutations, which may be useful in designing potential therapeutics and in blocking FAS1 aggregation related to CD.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Mutation , Transforming Growth Factor beta/genetics , Amyloid/chemistry , Amyloid/metabolism , Cell Adhesion Molecules, Neuronal/chemistry , Corneal Dystrophies, Hereditary/metabolism , Extracellular Matrix Proteins/chemistry , Humans , Molecular Dynamics Simulation , Molecular Structure , Protein Aggregation, Pathological/metabolism , Solubility , Transforming Growth Factor beta/chemistry
13.
Biochem Pharmacol ; 195: 114841, 2022 01.
Article in English | MEDLINE | ID: mdl-34798124

ABSTRACT

Fatty acids are essential in maintaining cellular homeostasis by providing lipids for energy production, cell membrane integrity, protein modification, and the structural demands of proliferating cells. Fatty acids and their derivatives are critical bioactive signaling molecules that influence many cellular processes, including metabolism, cell survival, proliferation, migration, angiogenesis, and cell barrier function. The CYP4 Omega hydroxylase gene family hydroxylate various short, medium, long, and very-long-chain saturated, unsaturated and polyunsaturated fatty acids. Selective members of the CYP4 family metabolize vitamins and biochemicals with long alkyl side chains and bioactive prostaglandins, leukotrienes, and arachidonic acids. It is uncertain of the physiological role of different members of the CYP4 omega hydroxylase gene family in the metabolic control of physiological and pathological processes in the liver. CYP4V2 is a unique member of the CYP4 family. CYP4V2 inactivation in retinal pigment epithelial cells leads to cholesterol accumulation and Bietti's Crystalline Dystrophy (BCD) pathogenesis. This commentary provides information on the role CYP4V2 has in metabolic syndrome and nonalcoholic fatty liver disease progression. This is accomplished by identifying its role in BCD, its control of cholesterol synthesis and lipid droplet formation in C. elegans, and the putative function in cardiovascular disease and gastrointestinal/hepatic pathologies.


Subject(s)
Cytochrome P-450 CYP4A/metabolism , Cytochrome P450 Family 4/metabolism , Fatty Acids/metabolism , Fatty Liver/metabolism , Liver/metabolism , Amino Acid Sequence , Animals , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Cytochrome P-450 CYP4A/genetics , Cytochrome P450 Family 4/genetics , Fatty Liver/genetics , Humans , Liver/pathology , Mutation , Retinal Diseases/genetics , Retinal Diseases/metabolism
14.
Cornea ; 41(1): 89-94, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34469340

ABSTRACT

PURPOSE: The purpose of this study was to investigate the clinical and genetic features of a man and his daughter with posterior polymorphous corneal dystrophy (PPCD), referred to our clinic for Descemet membrane endothelial keratoplasty. No other known relatives were affected. METHODS: Ophthalmic examination and histology, including electron microscopy, were performed. Genetic testing was conducted by means of whole exome sequencing, and variant analysis was achieved by using an internal in silico pipeline. Molecular tests included a dual-luciferase assay. RESULTS: Slowly progressive blurred vision was reported from childhood by the daughter. The father's symptoms started at age 55. Best-corrected visual acuity was reduced in both patients (0.2-0.4). Slit-lamp examination in both patients revealed bilateral corneal clouding with gray endothelial lesions; other family members had no ophthalmological signs. Descemet membrane endothelial keratoplasty was performed uneventfully in both patients. Histology showed thickened Descemet membrane and abnormal endothelium resembling epithelial-like cells. Both patients carried the OVOL2 5' untranslated region NM_021220.4.c.-61G>A variant in the heterozygous state. This change was associated with increased promoter activity and was not present in the unaffected members of the family. CONCLUSIONS: The 5' untranslated region mutation c.-61G>A in OVOL2 has been previously found in 1 individual with PPCD1 and reported as a variant of unknown significance because of insufficient evidence supporting its pathogenicity. Identification of the second family with 2 individuals affected by PPCD1 carrying this change, together with functional data, provides further proofs that it is disease-causing.


Subject(s)
5' Untranslated Regions/genetics , Corneal Dystrophies, Hereditary/genetics , Endothelium, Corneal/ultrastructure , Mutation , Transcription Factors/genetics , Adult , Aged , Corneal Dystrophies, Hereditary/diagnosis , Corneal Dystrophies, Hereditary/metabolism , DNA Mutational Analysis , Endothelium, Corneal/pathology , Female , Humans , Male , Microscopy, Electron , Pedigree , Promoter Regions, Genetic , Slit Lamp Microscopy , Zinc Fingers
15.
Exp Eye Res ; 213: 108804, 2021 12.
Article in English | MEDLINE | ID: mdl-34756941

ABSTRACT

PURPOSE: Alike keratoconus (KC), keratoglobus (KG) and pellucid marginal degeneration (PMD) belong to ectatic corneal diseases. While there are numerous studies on keratoconus pathophysiology, there is no exact knowledge on genetic and pathophysiological background of KG and PMD, so far. It is not yet clarified, whether KG and PMD are independent clinical entities or represent different stages of the same disease. Our purpose was to investigate key parameters concerning collagen synthesis, intracellular LOX expression and inflammation in corneal stromal cells of KG and PMD subjects, in vitro. METHODS: Normal human keratocytes of corneas from the LIONS Cornea Bank Saar-Lor-Lux, Trier/Westpfalz and human keratocytes of KG and PMD patients were isolated and cultured as keratocytes. To examine Collagen I and V (Col I, Col V), heat shock protein 47 (Hsp47), Lysyl Oxidase (LOX), nuclear factor kappa B (NF-κB) mRNA and protein expression in all cell types, quantitative PCR and Western blot analysis has been performed. RESULTS: Col5A1 mRNA expression was significantly lower in KG and PMD keratocytes and LOX mRNA expression was significantly higher in KG-keratocytes, compared to controls. Col1A1, Hsp47 and NF-κB mRNA expression and the analyzed protein expressions did not differ from controls, in KG or PMD. CONCLUSIONS: Col5A1 mRNA expression is decreased in KG and PMD and LOX mRNA expression is increased in KG. Therefore, the pathophysiology of KG and PMD differs from KC and these seem to be from KC independent entities. The explanation of the peripheral corneal thinning in KG and PMD must be investigated in further studies.


Subject(s)
Collagen Type V/genetics , Corneal Dystrophies, Hereditary/genetics , Corneal Keratocytes/metabolism , Gene Expression Regulation/physiology , Keratoconus/genetics , Protein-Lysine 6-Oxidase/genetics , RNA, Messenger/genetics , Adult , Aged , Aged, 80 and over , Blotting, Western , Cells, Cultured , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/physiopathology , Corneal Dystrophies, Hereditary/surgery , Corneal Stroma/cytology , Female , Healthy Volunteers , Humans , Keratoconus/metabolism , Keratoconus/physiopathology , Keratoconus/surgery , Keratoplasty, Penetrating , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Tissue Donors
16.
Invest Ophthalmol Vis Sci ; 62(12): 4, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34499705

ABSTRACT

Purpose: SLC4A11, an electrogenic H+ transporter, is found in the plasma membrane and mitochondria of corneal endothelium. However, the underlying mechanism of SLC4A11 targeting to mitochondria is unknown. Methods: The presence of mitochondrial targeting sequences was examined using in silico mitochondrial proteomic analyses. Thiol crosslinked peptide binding to SLC4A11 was screened by untargeted liquid chromatography/tandem mass spectrometry (LC-MS/MS) analysis. Direct protein interactions between SLC4A11 and chaperones were examined using coimmunoprecipitation analysis and proximity ligation assay. Knockdown or pharmacologic inhibition of chaperones in human corneal endothelial cells (HCECs) or mouse corneal endothelial cells (MCECs), ex vivo kidney, or HA-SLC4A11-transfected fibroblasts was performed to investigate the functional consequences of interfering with mitochondrial SLC4A11 trafficking. Results: SLC4A11 does not contain canonical N-terminal mitochondrial targeting sequences. LC-MS/MS analysis showed that HSC70 and/or HSP90 are bound to HA-SLC4A11-transfected PS120 fibroblast whole-cell lysates or isolated mitochondria, suggesting trafficking through the chaperone-mediated carrier pathway. SLC4A11 and either HSP90 or HSC70 complexes are directly bound to the mitochondrial surface receptor, TOM70. Interference with this trafficking leads to dysfunctional mitochondrial glutamine catabolism and increased reactive oxygen species production. In addition, glutamine (Gln) use upregulated SLC4A11, HSP70, and HSP90 expression in whole-cell lysates or purified mitochondria of HCECs and HA-SLC4A11-transfected fibroblasts. Conclusions: HSP90 and HSC70 are critical in mediating mitochondrial SLC4A11 translocation in corneal endothelial cells and kidney. Gln promotes SLC4A11 import to the mitochondria, and the continuous oxidative stress derived from Gln catabolism induced HSP70 and HSP90, protecting cells against oxidative stress.


Subject(s)
Ammonia/pharmacology , Anion Transport Proteins/genetics , Corneal Dystrophies, Hereditary/genetics , Endothelium, Corneal/metabolism , Mitochondria/pathology , Proteomics/methods , Symporters/genetics , Animals , Anion Transport Proteins/metabolism , Cells, Cultured , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , Endothelium, Corneal/drug effects , Endothelium, Corneal/pathology , Mice , Mice, Knockout , Mitochondria/metabolism , Molecular Chaperones/drug effects , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Protein Transport , Symporters/metabolism
17.
Invest Ophthalmol Vis Sci ; 62(12): 15, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34533563

ABSTRACT

Purpose: The Slc4a11 knock out (KO) mouse model recapitulates the human disease phenotype associated with congenital hereditary endothelial dystrophy (CHED). Increased mitochondrial reactive oxygen species (ROS) in the Slc4a11 KO mouse model is a major cause of edema and endothelial cell loss. Here, we asked if autophagy was activated by ROS in the KO mice. Methods: Immortalized cell lines and mouse corneal endothelia were used to measure autophagy and lysosome associated protein expressions using Protein Simple Wes immunoassay. Autophagy and lysosome functions were examined in wild type (WT) and KO cells as well as animals treated with the mitochondrial ROS quencher MitoQ. Results: Even though autophagy activation was evident, autophagy flux was aberrant in Slc4a11 KO cells and corneal endothelium. Expression of lysosomal proteins and lysosomal mass were decreased along with reduced nuclear translocation of lysosomal master regulator, transcription factor EB (TFEB). MitoQ reversed aberrant lysosomal functions and TFEB nuclear localization in KO cells. MitoQ injections in KO animals reduced corneal edema and decreased the rate of endothelial cell loss. Conclusions: Mitochondrial ROS disrupts TFEB signaling causing lysosomal dysfunction with impairment of autophagy in Slc4a11 KO corneal endothelium. Our study is the first to identify the presence as well as cause of lysosomal dysfunction in an animal model of CHED, and to identify a potential therapeutic approach.


Subject(s)
Autophagy/physiology , Corneal Dystrophies, Hereditary/metabolism , Disease Models, Animal , Lysosomes/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Anion Transport Proteins/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Blotting, Western , Cathepsin L/metabolism , Cells, Cultured , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/pathology , Endothelium, Corneal/drug effects , Endothelium, Corneal/metabolism , Gene Expression Regulation , Immunohistochemistry , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Organophosphorus Compounds/pharmacology , Real-Time Polymerase Chain Reaction , Symporters/genetics , Transfection , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
18.
Cell Signal ; 84: 110028, 2021 08.
Article in English | MEDLINE | ID: mdl-33940163

ABSTRACT

The secreted extracellular protein, transforming growth factor beta induced (TGFBI or ßIGH3), has roles in regulating numerous biological functions, including cell adhesion and bone formation, both during embryonic development and during the pathogenesis of human disease. TGFBI has been most studied in the context of hereditary corneal dystrophies, where mutations in TGFBI result in accumulation of TGFBI in the cornea. In cancer, early studies focused on TGFBI as a tumor suppressor, in part by promoting chemotherapy sensitivity. However, in established tumors, TGFBI largely has a role in promoting tumor progression, with elevated levels correlating to poorer clinical outcomes. As an important regulator of cancer progression, TGFBI expression and function is tightly regulated by numerous mechanisms including epigenetic silencing through promoter methylation and microRNAs. Mechanisms to target TGFBI have potential clinical utility in treating advanced cancers, while assessing TGFBI levels could be a biomarker for chemotherapy resistance and tumor progression.


Subject(s)
Corneal Dystrophies, Hereditary , Neoplasms , Corneal Dystrophies, Hereditary/genetics , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/therapy , Epigenesis, Genetic , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Transforming Growth Factor beta/metabolism
19.
Eye (Lond) ; 35(12): 3397-3403, 2021 12.
Article in English | MEDLINE | ID: mdl-33608638

ABSTRACT

OBJECTIVE: To evaluate longitudinal endothelial cell characteristics of children with posterior polymorphous corneal dystrophy (PPCD). METHODS: In this prospective case-control study, children with PPCD were followed with slit-lamp photography and non-contact specular microscopy. Patient's eyes were subdivided according to the clinical subtypes of PPCD (vesicular, band, diffuse, and unaffected) and the number of lesions present on the posterior corneal surface. Findings were then compared with age-matched controls. RESULTS: Thirty eyes of 15 patients with PPCD with a mean age 10.5 ± 3.1 years were analysed. Mean follow-up was 3.0 ± 1.0 years. PPCD morphology was vesicular in 40%, diffuse in 37%, band type in 10% and 13% had no detectable lesions despite contralateral involvement. Fourteen eyes (47%) had ≥5 endothelial lesions. Patients with PPCD had significantly lower endothelial cell densities (ECD) at recruitment (1918.9 ± 666.3 vs. 3340.1 ± 286.5 cells/mm2, p < 0.007) and at final follow-up (1793.1 ± 684.6 vs. 3265.2 ± 304.3 cells/mm2, p < 0.007) compared to age-matched controls. The lowest ECDs were found in eyes with diffuse type PPCD and those with ≥5 posterior corneal lesions, while clinically unaffected eyes in patients with confirmed PPCD in fellow eye had a normal ECD. However, the rates of annual ECD decline were not significantly different between eyes with PPCD in general, between the subgroups of PPCD and the normative groups. CONCLUSION: Endothelial cell density is significantly reduced among children with PPCD and depends on the clinical subtype and the number of posterior corneal lesions present. However, annual ECD loss is similar between normal eyes and those with PPCD.


Subject(s)
Corneal Dystrophies, Hereditary , Adolescent , Case-Control Studies , Cell Count , Child , Cornea/pathology , Corneal Dystrophies, Hereditary/metabolism , Endothelial Cells/pathology , Endothelium, Corneal/pathology , Humans
20.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513810

ABSTRACT

Background: To evaluate the distribution of the transforming growth factor-beta induced (TGFBI) corneal dystrophies in a multi-ethnic population in Singapore, and to present the different phenotypes with the same genotype. Methods: This study included 32 patients. Slit lamp biomicroscopy was performed for each patient to determine the disease phenotype. Genomic DNA was extracted from the blood samples and the 17 exons of the TGFBI gene were amplified by PCR and sequenced bi-directionally for genotype analysis. Results: Regarding phenotypes, the study patients comprised 11 (34.4%; 8 with R555W and 3 with R124H mutation) patients with granular corneal dystrophy type 1 (GCD1), 6 (18.8%; 5 with R124H and 1 with R124C mutation) patients with GCD2, 13 (40.6%; 7 with R124C, 2 with H626R, 2 with L550P, 1 with A620D and 1 with H572R) patients with lattice corneal dystrophy (LCD) and 2 (6.3%; 1 with R124L and 1 with R124C) patients with Reis-Bückler corneal dystrophy. Regarding genotype, R124H mutation was associated with GCD2 (5 cases; 62.5%) and GCD1 (3 cases; 37.5%). R124C mutation was associated with LCD (7 cases; 87.5%) and GCD2 (1 case; 12.5%). All the 8 cases (100%) of R555W mutation were associated with GCD1. Conclusions: Although the association between genotype and phenotype was good in most cases (65.7%; 21 of 32 patients), genotype/phenotype discrepancy was observed in a significant number.


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Transforming Growth Factor beta/genetics , Adult , Aged , Aged, 80 and over , Asian People/genetics , Corneal Dystrophies, Hereditary/blood , Corneal Dystrophies, Hereditary/metabolism , Corneal Dystrophies, Hereditary/pathology , DNA Mutational Analysis , Exons , Extracellular Matrix Proteins/blood , Extracellular Matrix Proteins/metabolism , Genetic Association Studies , Genotype , Humans , Middle Aged , Mutation , Phenotype , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...