Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 135
Filter
1.
2.
Nat Genet ; 55(7): 1116-1125, 2023 07.
Article in English | MEDLINE | ID: mdl-37386247

ABSTRACT

Glaucoma, a leading cause of irreversible blindness, is a highly heritable human disease. Previous genome-wide association studies have identified over 100 loci for the most common form, primary open-angle glaucoma. Two key glaucoma-associated traits also show high heritability: intraocular pressure and optic nerve head excavation damage quantified as the vertical cup-to-disc ratio. Here, since much of glaucoma heritability remains unexplained, we conducted a large-scale multitrait genome-wide association study in participants of European ancestry combining primary open-angle glaucoma and its two associated traits (total sample size over 600,000) to substantially improve genetic discovery power (263 loci). We further increased our power by then employing a multiancestry approach, which increased the number of independent risk loci to 312, with the vast majority replicating in a large independent cohort from 23andMe, Inc. (total sample size over 2.8 million; 296 loci replicated at P < 0.05, 240 after Bonferroni correction). Leveraging multiomics datasets, we identified many potential druggable genes, including neuro-protection targets likely to act via the optic nerve, a key advance for glaucoma because all existing drugs only target intraocular pressure. We further used Mendelian randomization and genetic correlation-based approaches to identify novel links to other complex traits, including immune-related diseases such as multiple sclerosis and systemic lupus erythematosus.


Subject(s)
Glaucoma, Open-Angle , Glaucoma , Humans , Genome-Wide Association Study , Glaucoma, Open-Angle/genetics , Glaucoma/genetics , Intraocular Pressure/genetics , Optic Nerve , Polymorphism, Single Nucleotide/genetics , Genetic Predisposition to Disease
3.
Hum Mutat ; 43(12): 2170-2186, 2022 12.
Article in English | MEDLINE | ID: mdl-36217948

ABSTRACT

The standardization of variant curation criteria is essential for accurate interpretation of genetic results and clinical care of patients. The variant curation guidelines developed by the American College of Medical Genetics and Genomics (ACMG) and the Association for Molecular Pathology (AMP) in 2015 are widely used but are not gene specific. To address this issue, the Clinical Genome Resource (ClinGen) Variant Curation Expert Panels (VCEP) have been tasked with developing gene-specific variant curation guidelines. The Glaucoma VCEP was created to develop rule specifications for genes associated with primary glaucoma, including myocilin (MYOC), the most common cause of Mendelian glaucoma. Of the 28 ACMG/AMP criteria, the Glaucoma VCEP adapted 15 rules to MYOC and determined 13 rules not applicable. Key specifications included determining minor allele frequency thresholds, developing an approach to counting probands and segregations, and reviewing functional assays. The rules were piloted on 81 variants and led to a change in classification in 40% of those that were classified in ClinVar, with functional evidence influencing the classification of 18 variants. The standardized variant curation guidelines for MYOC provide a framework for the consistent application of the rules between laboratories, to improve MYOC genetic testing in the management of glaucoma.


Subject(s)
Genome, Human , Glaucoma , Humans , Genetic Testing/methods , Genetic Variation , Glaucoma/diagnosis , Glaucoma/genetics , Pathology, Molecular , United States
4.
Invest Ophthalmol Vis Sci ; 63(6): 25, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35749127

ABSTRACT

Purpose: To identify the genetic basis of an unusual pediatric cortical cataract demonstrating autosomal dominant inheritance in a large European-Australian pedigree. Methods: DNA from four affected individuals were exome sequenced utilizing a NimbleGen SeqCap EZ Exome V3 kit and HiSeq 2500. DNA from 12 affected and four unaffected individuals were genotyped using Human OmniExpress-24 BeadChips. Multipoint linkage and haplotyping were performed (Superlink-Online SNP). DNA from one affected individual and his unaffected father were whole-genome sequenced on a HiSeq X Ten system. Rare small insertions/deletions and single-nucleotide variants (SNVs) were identified in the disease-linked region (Golden Helix SVS). Combined Annotation Dependent Depletion (CADD) analysis predicted variant deleteriousness. Putative enhancer function and variant effects were determined using the Dual-Glo Luciferase Assay system. Results: Linkage mapping identified a 6.23-centimorgan support interval at chromosome 7q36. A co-segregating haplotype refined the critical region to 6.03 Mbp containing 21 protein-coding genes. Whole-genome sequencing uncovered 114 noncoding variants from which CADD predicted one was highly deleterious, a novel substitution within intron-1 of the sonic hedgehog signaling molecule (SHH) gene. ENCODE data suggested this site was a putative enhancer, subsequently confirmed by luciferase reporter assays with variant-associated gene overexpression. Conclusions: In a large pedigree, we have identified a SHH intron variant that co-segregates with an unusual pediatric cortical cataract phenotype. SHH is important for lens formation, and mutations in its receptor (PTCH1) cause syndromic cataract. Our data implicate increased function of an enhancer important for SHH expression primarily within developing eye tissues.


Subject(s)
Cataract , Hedgehog Proteins , Australia , Cataract/genetics , Child , Genetic Linkage , Hedgehog Proteins/genetics , Humans , Introns/genetics , Mutation , Pedigree
5.
J Glaucoma ; 31(7): 584-589, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35131981

ABSTRACT

PRCIS: Glaucoma cascade screening in first-degree relatives (FDRs) of young Haitian glaucoma patients had high yield for diagnosing manifest and suspected glaucoma in 30.8% of those screened despite modest participation. PURPOSE: To evaluate the outcomes of glaucoma cascade screening in FDRs (parents, siblings, and offspring) of Haitian juvenile open-angle glaucoma (JOAG) patients. PATIENTS AND METHODS: Consecutive index patients (Haitians with JOAG) were identified, and the number/type of FDRs residing in South Florida were recorded. These FDRs were invited for free glaucoma screening, which included a comprehensive ophthalmic exam, gonioscopy, automated visual field testing and optical coherence tomographic analysis of the retinal nerve fiber layers. FDR characteristics and clinical findings from screening are reported. RESULTS: A total of 77 FDRs were invited, 26 (33.8%) agreed to undergo screening (18 females, 9 males), which revealed 2 (7.7%) with manifest glaucoma (mean age 77.5 y; one of whom was previously unaware of his glaucoma diagnosis), 6 (23.1%) with suspected glaucoma (mean age 29.8±18.3 y), and 18 (69.2%) without manifest or suspected glaucoma (mean age 37.2±21.8 y). Siblings of index patients were least likely to participate in cascade glaucoma screening when compared with index patients' parents or offspring. FDR eyes with manifest glaucoma had significantly worse best-corrected visual acuities, higher intraocular pressures, thinner central corneal thicknesses, and thinner circumferential papillary retinal nerve fiber layer thicknesses than those without glaucoma. CONCLUSION: Glaucoma cascade screening of Haitian JOAG patients' FDRs revealed that 30.8% had suspected or manifest glaucoma. Future efforts centered on provider-initiated recruitment and improving public glaucoma awareness and education may increase screening participation.


Subject(s)
Glaucoma, Open-Angle , Glaucoma , Ocular Hypertension , Adolescent , Adult , Aged , Child , Female , Glaucoma/diagnosis , Glaucoma, Open-Angle/diagnosis , Haiti/epidemiology , Humans , Intraocular Pressure , Male , Middle Aged , Pilot Projects , Tomography, Optical Coherence , Young Adult
6.
Ophthalmic Genet ; 43(1): 48-57, 2022 02.
Article in English | MEDLINE | ID: mdl-34612139

ABSTRACT

BACKGROUND: Costello syndrome (CS) is a multisystem developmental disorder caused by germline pathogenic variants in HRAS resulting in dysregulation of the Ras pathway. A systematic characterization of ophthalmic manifestations provides a unique opportunity to understand the role of Ras signal transduction in ocular development and guide optimal ophthalmic care in CS individuals. METHODS: Visual function, ocular features and genotype/phenotype correlations were evaluated in CS individuals harboring HRAS pathogenic variants, by cross-sectional and retrospective studies, and were recruited through the Costello Syndrome Family Network (CSFN) between 2007 and 2020. RESULTS: Fifty-six molecularly diagnosed CS individuals including 34 females and 22 males, ages ranging from 0.5 to 37 years were enrolled. The most common ophthalmic manifestations in the cross-sectional study were lack of stereopsis (96%), refractive errors (83%), strabismus (72%), nystagmus (69%), optic nerve hypoplasia or pallor (55%) and ptosis (13.7%) with higher prevalence than in the retrospective data (refractive errors (41%), strabismus (44%), nystagmus (26%), optic nerve hypoplasia or pallor (7%) and ptosis (11%)). Visual acuities were found to ranged from 20/25 to 20/800 and contrast sensitivity from 1.6% to 44%. HRAS pathogenic variants included p.G12S (84%), p.G13C (7%), p.G12A (5.4%), p.G12C (1.8%) and p.A146V (1.8%). CONCLUSION: Majority of individuals with CS have refractive errors, strabismus, nystagmus, absent stereopsis, and optic nerve abnormalities suggesting that HRAS and the Ras pathway play a vital role in visual system development. Ptosis, refractive errors and strabismus are amenable to treatment and early ophthalmic evaluation is crucial to prevent long-term vision impairment and improve overall quality of life in CS.


Subject(s)
Costello Syndrome , Optic Nerve Hypoplasia , Refractive Errors , Strabismus , Costello Syndrome/diagnosis , Costello Syndrome/genetics , Cross-Sectional Studies , Female , Humans , Male , Pallor , Quality of Life , Retrospective Studies
7.
Nat Commun ; 12(1): 6072, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663817

ABSTRACT

Primary congenital glaucoma (PCG) is a severe disease characterized by developmental defects in the trabecular meshwork (TM) and Schlemm's canal (SC), comprising the conventional aqueous humor outflow pathway of the eye. Recently, heterozygous loss of function variants in TEK and ANGPT1 or compound variants in TEK/SVEP1 were identified in children with PCG. Moreover, common variants in ANGPT1and SVEP1 have been identified as risk alleles for primary open angle glaucoma (POAG) in GWAS studies. Here, we show tissue-specific deletion of Angpt1 or Svep1 from the TM causes PCG in mice with severe defects in the adjacent SC. Single-cell transcriptomic analysis of normal and glaucomatous Angpt1 deficient eyes allowed us to identify distinct TM and SC cell populations and discover additional TM-SC signaling pathways. Furthermore, confirming the importance of angiopoietin signaling in SC, delivery of a recombinant ANGPT1-mimetic promotes developmental SC expansion in healthy and Angpt1 deficient eyes, blunts intraocular pressure (IOP) elevation and RGC loss in a mouse model of PCG and lowers IOP in healthy adult mice. Our data highlight the central role of ANGPT1-TEK signaling and TM-SC crosstalk in IOP homeostasis and provide new candidates for SC-targeted glaucoma therapy.


Subject(s)
Aqueous Humor/metabolism , Cell Communication/physiology , Glaucoma, Open-Angle/pathology , Glaucoma, Open-Angle/therapy , Angiopoietin-1/administration & dosage , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Anterior Chamber/blood supply , Anterior Chamber/cytology , Anterior Chamber/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Communication/drug effects , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression Profiling , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/metabolism , Intraocular Pressure/drug effects , Intraocular Pressure/genetics , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/metabolism , Proteins/genetics , Proteins/metabolism , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Single-Cell Analysis , Trabecular Meshwork/cytology , Trabecular Meshwork/metabolism
8.
Invest Ophthalmol Vis Sci ; 62(13): 24, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34698770

ABSTRACT

Purpose: Emmetropization requires coordinated scaling of the major ocular components, corneal curvature and axial length. This coordination is achieved in part through a shared set of genetic variants that regulate eye size. Poorly coordinated scaling of corneal curvature and axial length results in refractive error. We tested the hypothesis that genetic variants regulating eye size in emmetropic eyes are distinct from those conferring susceptibility to refractive error. Methods: A genome-wide association study (GWAS) for corneal curvature in 22,180 adult emmetropic individuals was performed as a proxy for a GWAS for eye size. A polygenic score created using lead GWAS variants was tested for association with corneal curvature and axial length in an independent sample: 437 classified as emmetropic and 637 as ametropic. The genetic correlation between eye size and refractive error was calculated using linkage disequilibrium score regression for approximately 1 million genetic variants. Results: The GWAS for corneal curvature in emmetropes identified 32 independent genetic variants (P < 5.0e-08). A polygenic score created using these 32 genetic markers explained 3.5% (P < 0.001) and 2.0% (P = 0.001) of the variance in corneal curvature and axial length, respectively, in the independent sample of emmetropic individuals but was not predictive of these traits in ametropic individuals. The genetic correlation between eye size and refractive error was close to zero (rg = 0.00; SE = 0.06; P = 0.95). Conclusions: These results support the hypothesis that genetic variants regulating eye size in emmetropic eyes do not overlap with those conferring susceptibility to myopia. This suggests that distinct biological pathways regulate normal eye growth and myopia development.


Subject(s)
Axial Length, Eye/diagnostic imaging , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Myopia/genetics , Polymorphism, Single Nucleotide , Refraction, Ocular/physiology , Adolescent , Adult , Aged , Child , Female , Humans , Linkage Disequilibrium , Male , Middle Aged , Myopia/diagnosis , Myopia/physiopathology , Young Adult
9.
JAMA Ophthalmol ; 139(6): 601-609, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33830181

ABSTRACT

IMPORTANCE: Uncertainty currently exists about whether the same genetic variants are associated with susceptibility to low myopia (LM) and high myopia (HM) and to myopia and hyperopia. Addressing this question is fundamental to understanding the genetics of refractive error and has clinical relevance for genotype-based prediction of children at risk for HM and for identification of new therapeutic targets. OBJECTIVE: To assess whether a common set of genetic variants are associated with susceptibility to HM, LM, and hyperopia. DESIGN, SETTING, AND PARTICIPANTS: This genetic association study assessed unrelated UK Biobank participants 40 to 69 years of age of European and Asian ancestry. Participants 40 to 69 years of age living in the United Kingdom were recruited from January 1, 2006, to October 31, 2010. Of the total sample of 502 682 participants, 117 279 (23.3%) underwent an ophthalmic assessment. Data analysis was performed from December 12, 2019, to June 23, 2020. EXPOSURES: Four refractive error groups were defined: HM, -6.00 diopters (D) or less; LM, -3.00 to -1.00 D; hyperopia, +2.00 D or greater; and emmetropia, 0.00 to +1.00 D. Four genome-wide association study (GWAS) analyses were performed in participants of European ancestry: (1) HM vs emmetropia, (2) LM vs emmetropia, (3) hyperopia vs emmetropia, and (4) LM vs hyperopia. Polygenic risk scores were generated from GWAS summary statistics, yielding 4 sets of polygenic risk scores. Performance was assessed in independent replication samples of European and Asian ancestry. MAIN OUTCOMES AND MEASURES: Odds ratios (ORs) of polygenic risk scores in replication samples. RESULTS: A total of 51 841 unrelated individuals of European ancestry and 2165 unrelated individuals of Asian ancestry were assigned to a specific refractive error group and included in our analyses. Polygenic risk scores derived from all 4 GWAS analyses were predictive of all categories of refractive error in both European and Asian replication samples. For example, the polygenic risk score derived from the HM vs emmetropia GWAS was predictive in the European sample of HM vs emmetropia (OR, 1.58; 95% CI, 1.41-1.77; P = 1.54 × 10-15) as well as LM vs emmetropia (OR, 1.15; 95% CI, 1.07-1.23; P = 8.14 × 10-5), hyperopia vs emmetropia (OR, 0.83; 95% CI, 0.77-0.89; P = 4.18 × 10-7), and LM vs hyperopia (OR, 1.45; 95% CI, 1.33-1.59; P = 1.43 × 10-16). CONCLUSIONS AND RELEVANCE: Genetic risk variants were shared across HM, LM, and hyperopia and across European and Asian samples. Individuals with HM inherited a higher number of variants from among the same set of myopia-predisposing alleles and not different risk alleles compared with individuals with LM. These findings suggest that treatment interventions targeting common genetic risk variants associated with refractive error could be effective against both LM and HM.


Subject(s)
Hyperopia , Myopia , Refractive Errors , Child , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hyperopia/genetics , Myopia/genetics
10.
Nat Commun ; 12(1): 1258, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627673

ABSTRACT

Primary open-angle glaucoma (POAG), is a heritable common cause of blindness world-wide. To identify risk loci, we conduct a large multi-ethnic meta-analysis of genome-wide association studies on a total of 34,179 cases and 349,321 controls, identifying 44 previously unreported risk loci and confirming 83 loci that were previously known. The majority of loci have broadly consistent effects across European, Asian and African ancestries. Cross-ancestry data improve fine-mapping of causal variants for several loci. Integration of multiple lines of genetic evidence support the functional relevance of the identified POAG risk loci and highlight potential contributions of several genes to POAG pathogenesis, including SVEP1, RERE, VCAM1, ZNF638, CLIC5, SLC2A12, YAP1, MXRA5, and SMAD6. Several drug compounds targeting POAG risk genes may be potential glaucoma therapeutic candidates.


Subject(s)
Genome-Wide Association Study/methods , Glaucoma, Open-Angle/genetics , Asian People , Genetic Loci/genetics , Genetic Predisposition to Disease/genetics , Genotype , Humans , Polymorphism, Single Nucleotide/genetics , White People
11.
Invest Ophthalmol Vis Sci ; 61(12): 6, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33027505

ABSTRACT

Purpose: Affecting children by age 3, primary congenital glaucoma (PCG) can cause debilitating vision loss by the developmental impairment of aqueous drainage resulting in high intraocular pressure (IOP), globe enlargement, and optic neuropathy. TEK haploinsufficiency accounts for 5% of PCG in diverse populations, with low penetrance explained by variable dysgenesis of Schlemm's canal (SC) in mice. We report eight families with TEK-related PCG, and provide evidence for SVEP1 as a disease modifier in family 8 with a higher penetrance and severity. Methods: Exome sequencing identified coding/splice site variants with an allele frequency less than 0.0001 (gnomAD). TEK variant effects were assayed in construct-transfected HEK293 cells via detection of autophosphorylated (active) TEK protein. An enucleated eye from an affected member of family 8 was examined via histology. SVEP1 expression in developing outflow tissues was detected by immunofluorescent staining of 7-day mouse anterior segments. SVEP1 stimulation of TEK expression in human umbilical vascular endothelial cells (HUVECs) was measured by TaqMan quantitative PCR. Results: Heterozygous TEK loss-of-function alleles were identified in eight PCG families, with parent-child disease transmission observed in two pedigrees. Family 8 exhibited greater disease penetrance and severity, histology revealed absence of SC in one eye, and SVEP1:p.R997C was identified in four of the five affected individuals. During SC development, SVEP1 is secreted by surrounding tissues. SVEP1:p.R997C abrogates stimulation of TEK expression by HUVECs. Conclusions: We provide further evidence for PCG caused by TEK haploinsufficiency, affirm autosomal dominant inheritance in two pedigrees, and propose SVEP1 as a modifier of TEK expression during SC development, affecting disease penetrance and severity.


Subject(s)
Cell Adhesion Molecules/genetics , Genes, Modifier/genetics , Hydrophthalmos/genetics , Receptor, TIE-2/genetics , Aged , Animals , Blotting, Western , Child, Preschool , Female , Gene Frequency , Genotyping Techniques , HEK293 Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrophthalmos/diagnosis , Hydrophthalmos/physiopathology , Infant , Infant, Newborn , Intraocular Pressure/physiology , Male , Mice , Middle Aged , Mutation, Missense , Pedigree , Penetrance , Phosphorylation , Protein Isoforms , Receptor, TIE-2/metabolism , Exome Sequencing
12.
Commun Biol ; 3(1): 133, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32193507

ABSTRACT

Corneal curvature, a highly heritable trait, is a key clinical endophenotype for myopia - a major cause of visual impairment and blindness in the world. Here we present a trans-ethnic meta-analysis of corneal curvature GWAS in 44,042 individuals of Caucasian and Asian with replication in 88,218 UK Biobank data. We identified 47 loci (of which 26 are novel), with population-specific signals as well as shared signals across ethnicities. Some identified variants showed precise scaling in corneal curvature and eye elongation (i.e. axial length) to maintain eyes in emmetropia (i.e. HDAC11/FBLN2 rs2630445, RBP3 rs11204213); others exhibited association with myopia with little pleiotropic effects on eye elongation. Implicated genes are involved in extracellular matrix organization, developmental process for body and eye, connective tissue cartilage and glycosylation protein activities. Our study provides insights into population-specific novel genes for corneal curvature, and their pleiotropic effect in regulating eye size or conferring susceptibility to myopia.


Subject(s)
Axial Length, Eye/pathology , Cornea/pathology , Corneal Topography , Genetic Loci , Myopia/genetics , Polymorphism, Single Nucleotide , Asian People/genetics , Databases, Genetic , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Myopia/ethnology , Myopia/pathology , Phenotype , Refractometry , Risk Assessment , Risk Factors , White People/genetics
14.
PLoS One ; 14(8): e0220143, 2019.
Article in English | MEDLINE | ID: mdl-31415580

ABSTRACT

PURPOSE: To evaluate the roles of known myopia-associated genetic variants for development of myopic macular degeneration (MMD) in individuals with high myopia (HM), using case-control studies from the Consortium of Refractive Error and Myopia (CREAM). METHODS: A candidate gene approach tested 50 myopia-associated loci for association with HM and MMD, using meta-analyses of case-control studies comprising subjects of European and Asian ancestry aged 30 to 80 years from 10 studies. Fifty loci with the strongest associations with myopia were chosen from a previous published GWAS study. Highly myopic (spherical equivalent [SE] ≤ -5.0 diopters [D]) cases with MMD (N = 348), and two sets of controls were enrolled: (1) the first set included 16,275 emmetropes (SE ≤ -0.5 D); and (2) second set included 898 highly myopic subjects (SE ≤ -5.0 D) without MMD. MMD was classified based on the International photographic classification for pathologic myopia (META-PM). RESULTS: In the first analysis, comprising highly myopic cases with MMD (N = 348) versus emmetropic controls without MMD (N = 16,275), two SNPs were significantly associated with high myopia in adults with HM and MMD: (1) rs10824518 (P = 6.20E-07) in KCNMA1, which is highly expressed in human retinal and scleral tissues; and (2) rs524952 (P = 2.32E-16) near GJD2. In the second analysis, comprising highly myopic cases with MMD (N = 348) versus highly myopic controls without MMD (N = 898), none of the SNPs studied reached Bonferroni-corrected significance. CONCLUSIONS: Of the 50 myopia-associated loci, we did not find any variant specifically associated with MMD, but the KCNMA1 and GJD2 loci were significantly associated with HM in highly myopic subjects with MMD, compared to emmetropes.


Subject(s)
Genetic Variation , Macular Degeneration/complications , Macular Degeneration/genetics , Myopia/complications , Aged, 80 and over , Case-Control Studies , Female , Gene Expression Profiling , Humans , Phenotype
15.
Invest Ophthalmol Vis Sci ; 60(8): 3142-3149, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31323684

ABSTRACT

Purpose: To determine genetic correlations between common myopia and primary open-angle glaucoma (POAG). Methods: We tested the association of myopia polygenic risk scores (PRSs) with POAG and POAG endophenotypes using two studies: the Australian & New Zealand Registry of Advanced Glaucoma (ANZRAG) study comprising 798 POAG cases with 1992 controls, and the Rotterdam Study (RS), a population-based study with 11,097 participants, in which intraocular pressure (IOP) and optic disc parameter measurements were catalogued. PRSs were derived from genome-wide association study meta-analyses conducted by the Consortium for Refractive Error and Myopia (CREAM) and 23andMe. In total, 12 PRSs were constructed and tested. Further, we explored the genetic correlation between myopia, POAG, and POAG endophenotypes by using the linkage disequilibrium score regression (LDSC) method. Results: We did not find significant evidence for an association between PRS of myopia with POAG (P = 0.81), IOP (P = 0.07), vertical cup-disc ratio (P = 0.42), or cup area (P = 0.25). We observed a nominal association with retinal nerve fiber layer (P = 7.7 × 10-3) and a significant association between PRS for myopia and disc area (P = 1.59 × 10-9). Using the LDSC method, we found a genetic correlation only between myopia and disc area (genetic correlation [RhoG] = -0.12, P = 1.8 × 10-3), supporting the findings of the PRS approach. Conclusions: Using two complementary approaches we found no evidence to support a genetic overlap between myopia and POAG; our results suggest that the comorbidity of these diseases is not influenced by common variants. The association between myopia and optic disc size is well known and validates this methodology.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Glaucoma, Open-Angle/genetics , Intraocular Pressure/physiology , Myopia/genetics , Refraction, Ocular/physiology , Registries , Aged , Australia/epidemiology , Female , Follow-Up Studies , Glaucoma, Open-Angle/epidemiology , Glaucoma, Open-Angle/physiopathology , Humans , Incidence , Male , Middle Aged , Myopia/epidemiology , Myopia/physiopathology , New Zealand/epidemiology , Optic Disk/pathology , Prospective Studies , Risk Factors
16.
PLoS One ; 14(5): e0214791, 2019.
Article in English | MEDLINE | ID: mdl-31100065

ABSTRACT

OBJECTIVES: To assess whether epigenetic mechanisms affecting gene expression may be involved in the pathogenesis of early-onset myopia, we performed genome-wide DNA methylation analyses of umbilical cord tissues, and assessed any associations between CpG site-specific methylation and the development of the disorder when the children were 3 years old. METHODS: Genome-wide DNA methylation profiling of umbilical cord samples from 519 Singaporean infants involved in a prospective birth cohort 'Growing Up in Singapore Towards healthy Outcomes' (GUSTO) was performed using the Illumina Infinium HumanMethylation450K chip microarray. Multivariable logistic regression models were used to assess any associations between site-specific CpG methylation of umbilical cord tissue at birth and myopia risk in 3 year old children, adjusting for potential confounders. Gene expression of genes located near CpG sites that demonstrated statistically significant associations were measured in relevant ocular tissues using human and mouse fetal and adult eye samples. RESULTS: We identified statistically significant associations between DNA methylation levels at five CpG sites and early-onset myopia risk after correcting for multiple comparisons using a false discovery rate of 5%. Two statistically significant CpG sites were identified in intergenic regions: 8p23(p = 1.70×10-7) and 12q23.2(p = 2.53×10-7). The remaining 3 statistically significant CpG sites were identified within the following genes: FGB (4q28, p = 3.60×10-7), PQLC1 (18q23, p = 8.9×10-7) and KRT12 (17q21.2, p = 1.2×10-6). Both PQLC1 and KRT12 were found to be significantly expressed in fetal and adult cornea and sclera tissues in both human and mouse. CONCLUSIONS: We identified five CpG methylation sites that demonstrate a statistically significant association with increased risk of developing early-onset myopia. These findings suggest that variability in the neonatal cord epigenome may influence early-onset myopia risk in children. Further studies of the epigenetic influences on myopia risk in larger study populations, and the associations with adulthood myopia risk are warranted.


Subject(s)
Epigenesis, Genetic , Myopia/diagnosis , Animals , Child, Preschool , CpG Islands , DNA Methylation , Disease Models, Animal , Eye/metabolism , Female , Fibrinogen/genetics , Gene Expression Regulation , Genome-Wide Association Study , Humans , Keratin-12/genetics , Male , Mice , Mice, Inbred C57BL , Myopia/genetics , Risk Factors , Umbilical Cord/metabolism
17.
Ophthalmol Glaucoma ; 2(4): 232-239, 2019.
Article in English | MEDLINE | ID: mdl-32672544

ABSTRACT

PURPOSE: To compare the angle structures during surgery using anterior segment (AS) OCT in children with and without glaucoma. DESIGN: Prospective comparative study, with single masked grader evaluating AS OCT images. PARTICIPANTS: This study included a total of 44 eyes of 8 children with glaucoma and 14 children without glaucoma undergoing ophthalmic surgery with general anesthesia. The median age for the glaucoma group was 6 months (range, 0.5-108 months), and that for the nonglaucoma group was 42 months (range, 14-95 months). METHODS: An Envisu C2300 handheld OCT with a high-resolution AS lens was used to capture videos of temporal and nasal quadrant anterior chamber angles from each eye. Still images of the temporal and nasal angles were selected and then graded by a masked ophthalmologist examiner to determine whether Schlemm's canal (SC) was visible, if abnormal tissue was present in the angle, and whether intrascleral lumens were present. Clinical correlation with severity of glaucoma was performed after AS OCT images were graded. MAIN OUTCOME MEASURES: Presence or absence of SC on AS OCT in glaucomatous and nonglaucomatous eyes. RESULTS: Schlemm's canal was present in 7 of 13 glaucomatous eyes compared with 27 of 27 nonglaucomatous eyes. In patients with unilateral glaucoma, absence of SC was noted only in the affected eye. Eight eyes with glaucoma showed abnormal tissue over the angle and SC. Intrascleral lumen and collector channels were found in both groups. Two patients with severe stenosis of SC during glaucoma angle surgery showed absent SC on AS OCT. CONCLUSIONS: Intraoperative AS OCT identified differences in the presence and characteristics of SC and the anterior chamber angle in pediatric patients with and without glaucoma. Further studies are needed to determine clinical correlation.


Subject(s)
Anterior Eye Segment/diagnostic imaging , Filtering Surgery/methods , Glaucoma/diagnosis , Tomography, Optical Coherence/methods , Adolescent , Child , Child, Preschool , Female , Follow-Up Studies , Glaucoma/physiopathology , Glaucoma/surgery , Humans , Infant , Infant, Newborn , Intraoperative Period , Male , Prospective Studies
18.
J Alzheimers Dis ; 64(1): 49-54, 2018.
Article in English | MEDLINE | ID: mdl-29865051

ABSTRACT

Cohort studies investigating aging and dementia require APOE genotyping. We compared directly measured APOE genotypes to 'hard-call' genotypes derived from imputing genome-wide genotyping data from a range of platforms using several imputation panels. Older GWAS arrays imputed to 1000 Genomes Project (1KGP) phases and the Haplotype Reference Consortium (HRC) reference panels were able to achieve concordance rates of over 98% with stringent quality control (hard-call-threshold 0.8). However, this resulted in high levels of missingness (>12% with 1KGP and 5% with HRC). With recent GWAS arrays, concordance of 99% could be obtained with relatively lenient QC, resulting in no missingness.


Subject(s)
Apolipoproteins E/genetics , Polymorphism, Single Nucleotide/genetics , Alzheimer Disease/genetics , Cohort Studies , Female , Gene Frequency , Genome-Wide Association Study , Genotype , Humans , Male
19.
J Clin Invest ; 127(12): 4421-4436, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29106382

ABSTRACT

Primary congenital glaucoma (PCG) is a leading cause of blindness in children worldwide and is caused by developmental defects in 2 aqueous humor outflow structures, Schlemm's canal (SC) and the trabecular meshwork. We previously identified loss-of-function mutations in the angiopoietin (ANGPT) receptor TEK in families with PCG and showed that ANGPT/TEK signaling is essential for SC development. Here, we describe roles for the major ANGPT ligands in the development of the aqueous outflow pathway. We determined that ANGPT1 is essential for SC development, and that Angpt1-knockout mice form a severely hypomorphic canal with elevated intraocular pressure. By contrast, ANGPT2 was dispensable, although mice deficient in both Angpt1 and Angpt2 completely lacked SC, indicating that ANGPT2 compensates for the loss of ANGPT1. In addition, we identified 3 human subjects with rare ANGPT1 variants within an international cohort of 284 PCG patients. Loss of function in 2 of the 3 patient alleles was observed by functional analysis of ANGPT1 variants in a combined in silico, in vitro, and in vivo approach, supporting a causative role for ANGPT1 in disease. By linking ANGPT1 with PCG, these results highlight the importance of ANGPT/TEK signaling in glaucoma pathogenesis and identify a candidate target for therapeutic development.


Subject(s)
Angiopoietin-1/metabolism , Lymphatic Vessels/embryology , Signal Transduction , Angiopoietin-1/genetics , Animals , Cohort Studies , Female , Genetic Diseases, Inborn/embryology , Genetic Diseases, Inborn/genetics , Glaucoma/embryology , Glaucoma/genetics , Humans , Lymphatic Vessels/pathology , Male , Mice , Mice, Knockout , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Trabecular Meshwork/embryology , Trabecular Meshwork/pathology
20.
Bio Protoc ; 7(10)2017 May 20.
Article in English | MEDLINE | ID: mdl-28758139

ABSTRACT

There are several in silico programs that endeavor to predict the functional impact of an individual's sequence variation at splice donor/acceptor sites, but experimental confirmation is problematic without a source of RNA from the individual that carries the variant. With the aid of an exon trapping vector, such as pSPL3, an investigator can test whether a splice site sequence change leads to altered RNA splicing, through expression of reference and variant mini-genes in mammalian cells and analysis of the resultant RNA products.

SELECTION OF CITATIONS
SEARCH DETAIL
...