Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 18(11): e1010478, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36395078

RESUMEN

Myopia most often develops during school age, with the highest incidence in countries with intensive education systems. Interactions between genetic variants and educational exposure are hypothesized to confer susceptibility to myopia, but few such interactions have been identified. Here, we aimed to identify genetic variants that interact with education level to confer susceptibility to myopia. Two groups of unrelated participants of European ancestry from UK Biobank were studied. A 'Stage-I' sample of 88,334 participants whose refractive error (avMSE) was measured by autorefraction and a 'Stage-II' sample of 252,838 participants who self-reported their age-of-onset of spectacle wear (AOSW) but who did not undergo autorefraction. Genetic variants were prioritized via a 2-step screening process in the Stage-I sample: Step 1 was a genome-wide association study for avMSE; Step 2 was a variance heterogeneity analysis for avMSE. Genotype-by-education interaction tests were performed in the Stage-II sample, with University education coded as a binary exposure. On average, participants were 58 years-old and left full-time education when they were 18 years-old; 35% reported University level education. The 2-step screening strategy in the Stage-I sample prioritized 25 genetic variants (GWAS P < 1e-04; variance heterogeneity P < 5e-05). In the Stage-II sample, 19 of the 25 (76%) genetic variants demonstrated evidence of variance heterogeneity, suggesting the majority were true positives. Five genetic variants located near GJD2, RBFOX1, LAMA2, KCNQ5 and LRRC4C had evidence of a genotype-by-education interaction in the Stage-II sample (P < 0.002) and consistent evidence of a genotype-by-education interaction in the Stage-I sample. For all 5 variants, University-level education was associated with an increased effect of the risk allele. In this cohort, additional years of education were associated with an enhanced effect of genetic variants that have roles including axon guidance and the development of neuronal synapses and neural circuits.


Asunto(s)
Miopía , Errores de Refracción , Humanos , Persona de Mediana Edad , Adolescente , Estudio de Asociación del Genoma Completo , Miopía/genética , Escolaridad , Errores de Refracción/genética , Alelos , Factores de Empalme de ARN/genética
2.
Hum Mol Genet ; 31(11): 1909-1919, 2022 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-35022715

RESUMEN

Refractive errors are associated with a range of pathological conditions, such as myopic maculopathy and glaucoma, and are highly heritable. Studies of missense and putative loss of function (pLOF) variants identified via whole exome sequencing (WES) offer the prospect of directly implicating potentially causative disease genes. We performed a genome-wide association study for refractive error in 51 624 unrelated adults, of European ancestry, aged 40-69 years from the UK and genotyped using WES. After testing 29 179 pLOF and 495 263 missense variants, 1 pLOF and 18 missense variants in 14 distinct genomic regions were taken forward for fine-mapping analysis. This yielded 19 putative causal variants of which 18 had a posterior inclusion probability >0.5. Of the 19 putative causal variants, 12 were novel discoveries. Specific variants were associated with a more myopic refractive error, while others were associated with a more hyperopic refractive error. Association with age of onset of spectacle wear (AOSW) was examined in an independent validation sample (38 100 early AOSW cases and 74 243 controls). Of 11 novel variants that could be tested, 8 (73%) showed evidence of association with AOSW status. This work identified COL4A4 and ATM as novel candidate genes associated with refractive error. In addition, novel putative causal variants were identified in the genes RASGEF1, ARMS2, BMP4, SIX6, GSDMA, GNGT2, ZNF652 and CRX. Despite these successes, the study also highlighted the limitations of community-based WES studies compared with high myopia case-control WES studies.


Asunto(s)
Miopía , Errores de Refracción , Adulto , Exoma/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Miopía/genética , Proteínas de Neoplasias/genética , Proteínas Citotóxicas Formadoras de Poros , Errores de Refracción/genética , Secuenciación del Exoma
3.
J Urol ; 206(3): 679-687, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33904754

RESUMEN

PURPOSE: Genome-wide association studies have not identified replicable genetic risk loci for stress or urgency urinary incontinence. MATERIALS AND METHODS: We carried out a discovery stage, case control, genome-wide association study in 3 independent discovery cohorts of European women (8,979) for stress incontinence, urgency incontinence, and any incontinence phenotypes. We conducted replication in 6 additional studies of European ancestry (4,069). We collected bladder biopsies from women with incontinence (50) to further investigate bladder expression of implicated genes and pathways and used symptom questionnaires for phenotyping. We conducted meta-analyses using inverse variance fixed effects models and whole transcriptome analyses using Affymetrix® arrays with replication with TaqMan® polymerase chain reaction. RESULTS: In the discovery stage, we identified 16 single nucleotide polymorphisms genotyped or imputed at 5 loci that reached genome-wide significance (p <5×10-8). In replication, rs138724718 on chromosome 2 near the macrophage receptor with collagenous structure (MARCO) gene (replication p=0.003) was associated with stress incontinence. In addition, rs34998271 on chromosome 6 near the endothelin 1 (EDN1) gene (replication p=0.0008) was associated with urgency incontinence. In combined meta-analyses of discovery and replication cohorts, associations with genome-wide significance for these 2 single nucleotide polymorphisms were confirmed. Transcriptomics analyses showed differential expression of 7 of 19 genes in the endothelin pathway between stress and urgency incontinence (p <0.0001). CONCLUSIONS: We uncovered 2 new risk loci near the genes endothelin 1 (EDN1), associated with urgency incontinence, and macrophage receptor with collagenous structure (MARCO), associated with stress incontinence. These loci are biologically plausible given their roles in smooth muscle contraction and innate host defense, respectively.


Asunto(s)
Sitios Genéticos , Incontinencia Urinaria de Esfuerzo/genética , Estudios de Casos y Controles , Endotelina-1/genética , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Receptores Inmunológicos/genética , Población Blanca/genética
4.
Mol Genet Genomics ; 295(4): 843-853, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32227305

RESUMEN

Genome-wide association studies (GWAS) have revealed that the genetic contribution to certain complex diseases is well-described by Fisher's infinitesimal model in which a vast number of polymorphisms each confer a small effect. Under Fisher's model, variants have additive effects both across loci and within loci. However, the latter assumption is at odds with the common observation of dominant or recessive rare alleles responsible for monogenic disorders. Here, we searched for evidence of non-additive (dominant or recessive) effects for GWAS variants known to confer susceptibility to the highly heritable quantitative trait, refractive error. Of 146 GWAS variants examined in a discovery sample of 228,423 individuals whose refractive error phenotype was inferred from their age-of-onset of spectacle wear, only 8 had even nominal evidence (p < 0.05) of non-additive effects. In a replication sample of 73,577 individuals who underwent direct assessment of refractive error, 1 of these 8 variants had robust independent evidence of non-additive effects (rs7829127 within ZMAT4, p = 4.76E-05) while a further 2 had suggestive evidence (rs35337422 in RD3L, p = 7.21E-03 and rs12193446 in LAMA2, p = 2.57E-02). Accounting for non-additive effects had minimal impact on the accuracy of a polygenic risk score for refractive error (R2 = 6.04% vs. 6.01%). Our findings demonstrate that very few GWAS variants for refractive error show evidence of a departure from an additive mode of action and that accounting for non-additive risk variants offers little scope to improve the accuracy of polygenic risk scores for myopia.


Asunto(s)
Estudio de Asociación del Genoma Completo , Miopía/genética , Carácter Cuantitativo Heredable , Errores de Refracción/genética , Adulto , Anciano , Bancos de Muestras Biológicas , Femenino , Genes Dominantes/genética , Predisposición Genética a la Enfermedad , Variación Genética/genética , Humanos , Laminina/genética , Masculino , Persona de Mediana Edad , Herencia Multifactorial/genética , Miopía/patología , Polimorfismo de Nucleótido Simple/genética , Errores de Refracción/patología
5.
Hum Genet ; 138(7): 723-737, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31073882

RESUMEN

Strabismus refers to an abnormal alignment of the eyes leading to the loss of central binocular vision. Concomitant strabismus occurs when the angle of deviation is constant in all positions of gaze and often manifests in early childhood when it is considered to be a neurodevelopmental disorder of the visual system. As such, it is inherited as a complex genetic trait, affecting 2-4% of the population. A genome-wide association study (GWAS) for self-reported strabismus (1345 cases and 65,349 controls from UK Biobank) revealed a single genome-wide significant locus on chromosome 17q25. Approximately 20 variants across the NPLOC4-TSPAN10-PDE6G gene cluster and in almost perfect linkage disequilibrium (LD) were most strongly associated (lead variant: rs75078292, OR = 1.26, p = 2.24E-08). A recessive model provided a better fit to the data than an additive model. Association with strabismus was independent of refractive error, and the degree of association with strabismus was minimally attenuated after adjustment for amblyopia. The association with strabismus was replicated in an independent cohort of clinician-diagnosed children aged 7 years old (116 cases and 5084 controls; OR = 1.85, p = 0.009). The associated variants included 2 strong candidate causal variants predicted to have functional effects: rs6420484, which substitutes tyrosine for a conserved cysteine (C177Y) in the TSPAN10 gene, and a 4-bp deletion variant, rs397693108, predicted to cause a frameshift in TSPAN10. The population-attributable risk for the locus was approximately 8.4%, indicating an important role in conferring susceptibility to strabismus.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética , Mutación , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Estrabismo/genética , Estrabismo/patología , Tetraspaninas/genética , Adulto , Anciano , Animales , Estudios de Casos y Controles , Niño , Estudios de Cohortes , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/metabolismo , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones , Persona de Mediana Edad , Familia de Multigenes , Proteínas Nucleares/metabolismo , Retina/metabolismo , Factores de Riesgo , Estrabismo/metabolismo , Tetraspaninas/metabolismo , Agudeza Visual
6.
Ophthalmic Physiol Opt ; 39(1): 11-25, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30628743

RESUMEN

PURPOSE: Randomised controlled trials (RCTs) allow reliable causal inferences to be drawn regarding the effectiveness of specific interventions. However, they are expensive to carry out, and not all exposure-outcome relationships can be tested in an RCT framework: for example, it would be unethical to deliberately expose participants to a putative risk factor, or the time-scale involved may be prohibitive. Mendelian randomisation (MR) has been proposed as an alternative approach for drawing causal inferences, with the major advantage that the method can often be applied to existing, cross-sectional study datasets. Therefore, results from an MR study can be obtained much more quickly and cheaply than through an RCT. RECENT FINDINGS: The validity of causal inferences from an MR study are dependent on two key assumptions, neither of which can be tested fully. Nevertheless, several approaches have been proposed in the last 3 years that either highlight questionable results, or provide valid causal inference if the necessary assumptions are met only in part. Compared to certain other areas of clinical practice, the ophthalmic research community has been slow to adopt MR. SUMMARY: An MR study cannot match an RCT in its strength of evidence for a claim of causality. However, MR still has much to offer. In some circumstances, an MR study can provide causal insight into research questions that cannot be addressed by an RCT, while more generally, an MR study can be used to evaluate the supporting evidence before deciding to embark on a lengthy and costly RCT.


Asunto(s)
Oftalmopatías/genética , Análisis de la Aleatorización Mendeliana/métodos , Estudios Observacionales como Asunto/métodos , Genotipo , Humanos , Reproducibilidad de los Resultados
7.
Hum Genet ; 137(11-12): 881-896, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30306274

RESUMEN

Previous studies have suggested that naturally occurring genetic variation contributes to the risk of astigmatism. The purpose of this investigation was to identify genetic markers associated with corneal and refractive astigmatism in a large-scale European ancestry cohort (UK Biobank) who underwent keratometry and autorefraction at an assessment centre. Genome-wide association studies for corneal and refractive astigmatism were performed in individuals of European ancestry (N = 86,335 and 88,005 respectively), with the mean corneal astigmatism or refractive astigmatism in fellow eyes analysed as a quantitative trait (dependent variable). Genetic correlation between the two traits was calculated using LD Score regression. Gene-based and gene-set tests were carried out using MAGMA. Single marker-based association tests for corneal astigmatism identified four genome-wide significant loci (P < 5 × 10-8) near the genes ZC3H11B (1q41), LINC00340 (6p22.3), HERC2/OCA2 (15q13.1) and NPLOC4/TSPAN10 (17q25.3). Three of these loci also demonstrated genome-wide significant association with refractive astigmatism: LINC00340, HERC2/OCA2 and NPLOC4/TSPAN10. The genetic correlation between corneal and refractive astigmatism was 0.85 (standard error = 0.068, P = 1.37 × 10-35). Here, we have undertaken the largest genome-wide association studies for corneal and refractive astigmatism to date and identified four novel loci for corneal astigmatism, two of which were also novel loci for refractive astigmatism. These loci have previously demonstrated association with axial length (ZC3H11B), myopia (NPLOC4), spherical equivalent refractive error (LINC00340) and eye colour (HERC2). The shared role of these novel candidate genes for astigmatism lends further support to the shared genetic susceptibility of myopia and astigmatism.


Asunto(s)
Astigmatismo/genética , Enfermedades de la Córnea/genética , Estudio de Asociación del Genoma Completo , Miopía/genética , Astigmatismo/patología , Bancos de Muestras Biológicas , Córnea/patología , Enfermedades de la Córnea/patología , Femenino , Predisposición Genética a la Enfermedad , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Masculino , Persona de Mediana Edad , Miopía/patología , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple/genética , Ubiquitina-Proteína Ligasas , Reino Unido , Población Blanca/genética
8.
Mol Vis ; 24: 127-142, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29422769

RESUMEN

Purpose: To identify genes and genetic markers associated with corneal astigmatism. Methods: A meta-analysis of genome-wide association studies (GWASs) of corneal astigmatism undertaken for 14 European ancestry (n=22,250) and 8 Asian ancestry (n=9,120) cohorts was performed by the Consortium for Refractive Error and Myopia. Cases were defined as having >0.75 diopters of corneal astigmatism. Subsequent gene-based and gene-set analyses of the meta-analyzed results of European ancestry cohorts were performed using VEGAS2 and MAGMA software. Additionally, estimates of single nucleotide polymorphism (SNP)-based heritability for corneal and refractive astigmatism and the spherical equivalent were calculated for Europeans using LD score regression. Results: The meta-analysis of all cohorts identified a genome-wide significant locus near the platelet-derived growth factor receptor alpha (PDGFRA) gene: top SNP: rs7673984, odds ratio=1.12 (95% CI:1.08-1.16), p=5.55×10-9. No other genome-wide significant loci were identified in the combined analysis or European/Asian ancestry-specific analyses. Gene-based analysis identified three novel candidate genes for corneal astigmatism in Europeans-claudin-7 (CLDN7), acid phosphatase 2, lysosomal (ACP2), and TNF alpha-induced protein 8 like 3 (TNFAIP8L3). Conclusions: In addition to replicating a previously identified genome-wide significant locus for corneal astigmatism near the PDGFRA gene, gene-based analysis identified three novel candidate genes, CLDN7, ACP2, and TNFAIP8L3, that warrant further investigation to understand their role in the pathogenesis of corneal astigmatism. The much lower number of genetic variants and genes demonstrating an association with corneal astigmatism compared to published spherical equivalent GWAS analyses suggest a greater influence of rare genetic variants, non-additive genetic effects, or environmental factors in the development of astigmatism.


Asunto(s)
Fosfatasa Ácida/genética , Astigmatismo/genética , Claudinas/genética , Enfermedades de la Córnea/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Pueblo Asiatico , Astigmatismo/diagnóstico , Astigmatismo/etnología , Astigmatismo/patología , Estudios de Cohortes , Córnea/metabolismo , Córnea/patología , Enfermedades de la Córnea/diagnóstico , Enfermedades de la Córnea/etnología , Enfermedades de la Córnea/patología , Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Programas Informáticos , Población Blanca
10.
Ophthalmic Physiol Opt ; 38(5): 492-502, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30182516

RESUMEN

PURPOSE: To investigate whether a genetic risk score (GRS) improved performance of predicting refractive error compared to knowing a child's number of myopic parents (NMP) alone. METHODS: This was a retrospective analysis of data from the Avon Longitudinal Study of Parents and Children (ALSPAC) birth cohort study. Refractive error was assessed longitudinally between age 7-15 using non-cycloplegic autorefraction. Genetic variants (n = 149) associated with refractive error from a Consortium for Refractive Error And Myopia (CREAM) genome-wide association study were used to calculate a GRS for each child. Using refractive error at ages 7 and 15 years as the outcome variable, coefficient of determination (R2 ) values were calculated via linear regression models for the predictors: NMP, GRS and a combined model. RESULTS: Number of myopic parents was weakly predictive of refractive error in children aged 7 years, R2  = 3.0% (95% CI 1.8-4.1%, p < 0.0001) and aged 15 years, R2  = 4.8% (3.1-6.5%, p < 0.0001). The GRS was also weakly predictive; age 7 years, R2  = 1.1% (0.4-1.9%, p < 0.0001) and 15 years R2  = 2.6% (1.3-3.9%, p < 0.0001). Combining the 2 variables gave larger R2 values at age 7, R2  = 3.7% (2.5-5.0%, p < 0.0001) and 15, R2  = 7.0% (5.0-9.0%, p < 0.0001). The combined model improved performance at both ages (both p < 0.0001). CONCLUSION: A GRS improved the ability to detect children at risk of myopia independently of knowing the NMP. We speculate this may be because NMP captures information concerning environmental risk factors for myopia. Nevertheless, further gains are required to make such predictive tests worthwhile in the clinical environment.


Asunto(s)
Pruebas Genéticas/métodos , Estudio de Asociación del Genoma Completo , Miopía/genética , Refracción Ocular/fisiología , Adolescente , Niño , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Masculino , Miopía/diagnóstico , Miopía/epidemiología , Valor Predictivo de las Pruebas , Estudios Retrospectivos , Factores de Riesgo , Reino Unido/epidemiología
11.
PLoS Genet ; 11(8): e1005432, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26313004

RESUMEN

Myopia is the most common vision disorder and the leading cause of visual impairment worldwide. However, gene variants identified to date explain less than 10% of the variance in refractive error, leaving the majority of heritability unexplained ("missing heritability"). Previously, we reported that expression of APLP2 was strongly associated with myopia in a primate model. Here, we found that low-frequency variants near the 5'-end of APLP2 were associated with refractive error in a prospective UK birth cohort (n = 3,819 children; top SNP rs188663068, p = 5.0 × 10-4) and a CREAM consortium panel (n = 45,756 adults; top SNP rs7127037, p = 6.6 × 10-3). These variants showed evidence of differential effect on childhood longitudinal refractive error trajectories depending on time spent reading (gene x time spent reading x age interaction, p = 4.0 × 10-3). Furthermore, Aplp2 knockout mice developed high degrees of hyperopia (+11.5 ± 2.2 D, p < 1.0 × 10-4) compared to both heterozygous (-0.8 ± 2.0 D, p < 1.0 × 10-4) and wild-type (+0.3 ± 2.2 D, p < 1.0 × 10-4) littermates and exhibited a dose-dependent reduction in susceptibility to environmentally induced myopia (F(2, 33) = 191.0, p < 1.0 × 10-4). This phenotype was associated with reduced contrast sensitivity (F(12, 120) = 3.6, p = 1.5 × 10-4) and changes in the electrophysiological properties of retinal amacrine cells, which expressed Aplp2. This work identifies APLP2 as one of the "missing" myopia genes, demonstrating the importance of a low-frequency gene variant in the development of human myopia. It also demonstrates an important role for APLP2 in refractive development in mice and humans, suggesting a high level of evolutionary conservation of the signaling pathways underlying refractive eye development.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Hiperopía/genética , Miopía/genética , Proteínas del Tejido Nervioso/genética , Agudeza Visual/genética , Adolescente , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Niño , Chlorocebus aethiops , Interacción Gen-Ambiente , Predisposición Genética a la Enfermedad , Variación Genética/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades de los Monos/genética , Proteínas del Tejido Nervioso/metabolismo , Retina/fisiología , Agudeza Visual/fisiología
12.
Genet Epidemiol ; 40(8): 756-766, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27611182

RESUMEN

Previous studies have identified many genetic loci for refractive error and myopia. We aimed to investigate the effect of these loci on ocular biometry as a function of age in children, adolescents, and adults. The study population consisted of three age groups identified from the international CREAM consortium: 5,490 individuals aged <10 years; 5,000 aged 10-25 years; and 16,274 aged >25 years. All participants had undergone standard ophthalmic examination including measurements of axial length (AL) and corneal radius (CR). We examined the lead SNP at all 39 currently known genetic loci for refractive error identified from genome-wide association studies (GWAS), as well as a combined genetic risk score (GRS). The beta coefficient for association between SNP genotype or GRS versus AL/CR was compared across the three age groups, adjusting for age, sex, and principal components. Analyses were Bonferroni-corrected. In the age group <10 years, three loci (GJD2, CHRNG, ZIC2) were associated with AL/CR. In the age group 10-25 years, four loci (BMP2, KCNQ5, A2BP1, CACNA1D) were associated; and in adults 20 loci were associated. Association with GRS increased with age; ß = 0.0016 per risk allele (P = 2 × 10-8 ) in <10 years, 0.0033 (P = 5 × 10-15 ) in 10- to 25-year-olds, and 0.0048 (P = 1 × 10-72 ) in adults. Genes with strongest effects (LAMA2, GJD2) had an early effect that increased with age. Our results provide insights on the age span during which myopia genes exert their effect. These insights form the basis for understanding the mechanisms underlying high and pathological myopia.


Asunto(s)
Conexinas/genética , Estudio de Asociación del Genoma Completo , Laminina/genética , Miopía/genética , Polimorfismo de Nucleótido Simple/genética , Adolescente , Adulto , Alelos , Biometría , Niño , Femenino , Sitios Genéticos , Genotipo , Humanos , Masculino , Factores de Riesgo , Adulto Joven , Proteína delta-6 de Union Comunicante
13.
Am J Hum Genet ; 93(2): 264-77, 2013 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24144296

RESUMEN

Refractive errors are common eye disorders of public health importance worldwide. Ocular axial length (AL) is the major determinant of refraction and thus of myopia and hyperopia. We conducted a meta-analysis of genome-wide association studies for AL, combining 12,531 Europeans and 8,216 Asians. We identified eight genome-wide significant loci for AL (RSPO1, C3orf26, LAMA2, GJD2, ZNRF3, CD55, MIP, and ALPPL2) and confirmed one previously reported AL locus (ZC3H11B). Of the nine loci, five (LAMA2, GJD2, CD55, ALPPL2, and ZC3H11B) were associated with refraction in 18 independent cohorts (n = 23,591). Differential gene expression was observed for these loci in minus-lens-induced myopia mouse experiments and human ocular tissues. Two of the AL genes, RSPO1 and ZNRF3, are involved in Wnt signaling, a pathway playing a major role in the regulation of eyeball size. This study provides evidence of shared genes between AL and refraction, but importantly also suggests that these traits may have unique pathways.


Asunto(s)
Longitud Axial del Ojo/metabolismo , Proteínas del Ojo/genética , Sitios Genéticos , Predisposición Genética a la Enfermedad , Errores de Refracción/genética , Adolescente , Adulto , Anciano , Pueblo Asiatico , Longitud Axial del Ojo/patología , Proteínas del Ojo/metabolismo , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Errores de Refracción/etnología , Errores de Refracción/patología , Transducción de Señal , Población Blanca
14.
Mol Vis ; 21: 621-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26019481

RESUMEN

PURPOSE: Studies in relatives have generally yielded high heritability estimates for refractive error: twins 75-90%, families 15-70%. However, because related individuals often share a common environment, these estimates are inflated (via misallocation of unique/common environment variance). We calculated a lower-bound heritability estimate for refractive error free from such bias. METHODS: Between the ages 7 and 15 years, participants in the Avon Longitudinal Study of Parents and Children (ALSPAC) underwent non-cycloplegic autorefraction at regular research clinics. At each age, an estimate of the variance in refractive error explained by single nucleotide polymorphism (SNP) genetic variants was calculated using genome-wide complex trait analysis (GCTA) using high-density genome-wide SNP genotype information (minimum N at each age=3,404). RESULTS: The variance in refractive error explained by the SNPs ("SNP heritability") was stable over childhood: Across age 7-15 years, SNP heritability averaged 0.28 (SE=0.08, p<0.001). The genetic correlation for refractive error between visits varied from 0.77 to 1.00 (all p<0.001) demonstrating that a common set of SNPs was responsible for the genetic contribution to refractive error across this period of childhood. Simulations suggested lack of cycloplegia during autorefraction led to a small underestimation of SNP heritability (adjusted SNP heritability=0.35; SE=0.09). To put these results in context, the variance in refractive error explained (or predicted) by the time participants spent outdoors was <0.005 and by the time spent reading was <0.01, based on a parental questionnaire completed when the child was aged 8-9 years old. CONCLUSIONS: Genetic variation captured by common SNPs explained approximately 35% of the variation in refractive error between unrelated subjects. This value sets an upper limit for predicting refractive error using existing SNP genotyping arrays, although higher-density genotyping in larger samples and inclusion of interaction effects is expected to raise this figure toward twin- and family-based heritability estimates. The same SNPs influenced refractive error across much of childhood. Notwithstanding the strong evidence of association between time outdoors and myopia, and time reading and myopia, less than 1% of the variance in myopia at age 15 was explained by crude measures of these two risk factors, indicating that their effects may be limited, at least when averaged over the whole population.


Asunto(s)
Polimorfismo de Nucleótido Simple , Errores de Refracción/genética , Adolescente , Niño , Estudios de Cohortes , Inglaterra , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Actividades Recreativas , Masculino , Miopía/genética , Carácter Cuantitativo Heredable , Lectura
16.
Mol Vis ; 19: 243-53, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23401653

RESUMEN

PURPOSE: Corneal curvature is a key determinant of the refractive power of the eye. Variants in two genes, FKBP12-rapamycin complex-associated protein 1 (FRAP1) on chromosome 1p36.2 and platelet-derived growth factor receptor alpha (PDGFRA) on chromosome 4q12, have shown genome-wide significant association with normal variation in corneal curvature in a study of subjects of Asian origin. Variants at the PDGFRA locus have also shown genome-wide significant association with corneal astigmatism. Whether these variants influence other ocular parameters such as axial length has yet to be reported. We performed a genome-wide association study for corneal curvature in white European subjects from a population-based birth cohort, with the aim of replicating and extending the above findings. METHODS: White European children participating in the Avon Longitudinal Study of Parents and Children (ALSPAC) birth cohort were examined at age about 15.5 years (95% confidence interval=15.45 to 15.48 years). Radius of corneal curvature and axial eye length were measured with an IOLmaster. DNA samples were genotyped with Illumina HumanHap550 arrays and untyped variants imputed using MACH, with CEU individuals from HapMap release 22, Phase II NCBI B36, Single Nucleotide Polymorphism database 126 as the reference panel. Association between corneal curvature and single nucleotide polymorphism (SNP) genotype was tested, genome-wide, using mach2qtl, with sex as a covariate (n=2023; 46.6% male). RESULTS: The variant exhibiting the strongest evidence for association with corneal curvature (rs6554163; p=2.8×10(-6)) was located in the same linkage disequilibrium block as the previously discovered PDGFRA variants. Meta-analysis of the current and prior findings enhanced the evidence for association (rs17084051, p=4.5×10(-14)). rs6554163 genotype predicted 1.0% of variation in corneal curvature. In addition, these PDGFRA variants were associated with axial eye length, predicting 0.6% of the normal trait variation (p=5.3×10(-4)). Each copy of the minor allele of variants at the locus also increased the risk of corneal astigmatism in this white European cohort (odds ratio [OR]=1.24, 95% confidence interval=1.07-1.45; p=0.006). CONCLUSION: As in Asians, variants at the PDGFRA locus influence corneal curvature (and corneal astigmatism). However, rather than affecting corneal curvature in isolation, this locus influences the size of the eye while maintaining its scaling.


Asunto(s)
Córnea/anatomía & histología , Córnea/metabolismo , Ojo/anatomía & histología , Sitios de Carácter Cuantitativo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Adolescente , Astigmatismo/genética , Astigmatismo/patología , Estudios de Cohortes , Europa (Continente) , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Desequilibrio de Ligamiento/genética , Masculino , Miopía/genética , Miopía/patología , Tamaño de los Órganos/genética , Polimorfismo de Nucleótido Simple , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Población Blanca/genética
17.
Ophthalmology ; 120(5): 1064-73.e1, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23415774

RESUMEN

PURPOSE: Stature at a particular age can be considered the cumulative result of growth during a number of preceding growth trajectory periods. We investigated whether height and weight growth trajectories from birth to age 10 years were related to refractive error at ages 11 and 15 years, and eye size at age 15 years. DESIGN: Prospective analysis in a birth cohort. PARTICIPANTS: Children participating in the Avon Longitudinal Study of Parents and Children (ALSPAC) U.K. birth cohort (minimum N = 2676). METHODS: Growth trajectories between birth and 10 years were modeled from a series of height and weight measurements (N = 6815). Refractive error was assessed by noncycloplegic autorefraction at ages 11 and 15 years (minimum N = 4737). Axial length (AXL) and radius of corneal curvature were measured with an IOLMaster (Carl Zeiss Meditec, Welwyn Garden City, U.K.) at age 15 years (minimum N = 2676). Growth trajectories and an allelic score for 180 genetic variants associated with adult height were tested for association with refractive error and eye size. MAIN OUTCOME MEASURES: Noncycloplegic autorefraction at ages 11 and 15 years, and AXL and corneal curvature at age 15 years. RESULTS: Height growth trajectory during the linear phase between 2.5 and 10 years was negatively associated with refractive error at 11 and 15 years (P<0.001), but explained <0.5% of intersubject variation. Height and weight growth trajectories, especially shortly after birth, were positively associated with AXL and corneal curvature (P<0.001), predicting 1% to 5% of trait variation. Height growth after 2.5 years was not associated with corneal curvature, whereas the association with AXL continued up to 10 years. The height allelic score was associated with corneal curvature (P = 0.03) but not with refractive error or AXL. CONCLUSIONS: Up to the age of 10 years, shared growth mechanisms contribute to scaling of eye and body size but minimally to the development of myopia. FINANCIAL DISCLOSURE(S): The author(s) have no proprietary or commercial interest in any materials discussed in this article.


Asunto(s)
Estatura/fisiología , Peso Corporal/fisiología , Miopía/etiología , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Miopía/fisiopatología , Estudios Prospectivos , Errores de Refracción/fisiopatología , Análisis de Regresión
18.
Front Genet ; 14: 1113058, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37351342

RESUMEN

Introduction: Long axial length (AL) is a risk factor for myopia. Although family studies indicate that AL has an important genetic component with heritability estimates up to 0.94, there have been few reports of AL-associated loci. Methods: Here, we conducted a multiethnic genome-wide association study (GWAS) of AL in 19,420 adults of European, Latino, Asian, and African ancestry from the Genetic Epidemiology Research on Adult Health and Aging (GERA) cohort, with replication in a subset of the Consortium for Refractive Error and Myopia (CREAM) cohorts of European or Asian ancestry. We further examined the effect of the identified loci on the mean spherical equivalent (MSE) within the GERA cohort. We also performed genome-wide genetic correlation analyses to quantify the genetic overlap between AL and MSE or myopia risk in the GERA European ancestry sample. Results: Our multiethnic GWA analysis of AL identified a total of 16 genomic loci, of which 5 are novel. We found that all AL-associated loci were significantly associated with MSE after Bonferroni correction. We also found that AL was genetically correlated with MSE (rg = -0.83; SE, 0.04; p = 1.95 × 10-89) and myopia (rg = 0.80; SE, 0.05; p = 2.84 × 10-55). Finally, we estimated the array heritability for AL in the GERA European ancestry sample using LD score regression, and found an overall heritability estimate of 0.37 (s.e. = 0.04). Discussion: In this large and multiethnic study, we identified novel loci, associated with AL at a genome-wide significance level, increasing substantially our understanding of the etiology of AL variation. Our results also demonstrate an association between AL-associated loci and MSE and a shared genetic basis between AL and myopia risk.

19.
Invest Ophthalmol Vis Sci ; 64(14): 38, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-38010695

RESUMEN

Purpose: The purpose of this study was to investigate if education contributes to the risk of myopia because educational activities typically occur indoors or because of other factors, such as prolonged near viewing. Methods: This was a two-sample Mendelian randomization study. Participants were from the UK Biobank, Avon Longitudinal Study of Parents and Children, and Generation R. Genetic variants associated with years spent in education or time spent outdoors were used as instrumental variables. The main outcome measures were: (1) spherical equivalent refractive error attained by adulthood, and (2) risk of an early age-of-onset of spectacle wear (EAOSW), defined as an age-of-onset of 15 years or below. Results: Time spent outdoors was found to have a small genetic component (heritability 9.8%) that tracked from childhood to adulthood. A polygenic score for time outdoors was associated with children's time outdoors; a polygenic score for years spent in education was inversely associated with children's time outdoors. Accounting for the relationship between time spent outdoors and myopia in a multivariable Mendelian randomization analysis reduced the size of the causal effect of more years in education on myopia to -0.17 diopters (D) per additional year of formal education (95% confidence interval [CI] = -0.32 to -0.01) compared with the estimate from a univariable Mendelian randomization analysis of -0.27 D per year (95% CI = -0.41 to -0.13). Comparable results were obtained for the outcome EAOSW. Conclusions: Accounting for the effects of time outdoors reduced the estimated causal effect of education on myopia by 40%. These results suggest about half of the relationship between education and myopia may be mediated by children not being outdoors during schooling.


Asunto(s)
Actividades Recreativas , Miopía , Adolescente , Niño , Humanos , Adulto Joven , Escolaridad , Estudios Longitudinales , Miopía/epidemiología , Miopía/genética , Factores de Riesgo , Análisis de la Aleatorización Mendeliana
20.
Invest Ophthalmol Vis Sci ; 64(14): 28, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37982764

RESUMEN

Purpose: Changes in refractive error during young adulthood is common yet risk factors at this age are largely unexplored. This study explored risk factors for these changes, including gene-environmental interactions. Methods: Spherical equivalent refraction (SER) and axial length (AL) for 624 community-based adults were measured at 20 (baseline) and 28 years old. Participants were genotyped and their polygenic scores (PGS) for refractive error calculated. Self-reported screen time (computer, television, and mobile devices) from 20 to 28 years old were collected prospectively and longitudinal trajectories were generated. Past sun exposure was quantified using conjunctival ultraviolet autofluorescence (CUVAF) area. Results: Median change in SER and AL were -0.023 diopters (D)/year (interquartile range [IQR] = -0.062 to -0.008) and +0.01 mm/year (IQR = 0.000 to 0.026), respectively. Sex, baseline myopia, parental myopia, screen time, CUVAF, and PGS were significantly associated with myopic shift. Collectively, these factors accounted for approximately 20% of the variance in refractive error change, with screen time, CUVAF, and PGS each explaining approximately 1% of the variance. Four trajectories for total screen time were found: "consistently low" (n = 148), "consistently high" (n = 250), "consistently very high" (n = 76), and "increasing" (n = 150). Myopic shift was faster in those with "consistently high" or "consistently very high" screen time compared to "consistently-low" (P ≤ 0.031). For each z-score increase in PGS, changes in SER and AL increased by -0.005 D/year and 0.002 mm/year (P ≤ 0.045). Of the three types of screen time, only computer time was associated with myopic shift (P ≤ 0.040). There was no two- or three-way interaction effect between PGS, CUVAF, or screen time (P ≥ 0.26). Conclusions: Higher total or computer screen time, less sun exposure, and genetic predisposition are each independently associated with greater myopic shifts during young adulthood. Given that these factors explained only a small amount of the variance, there are likely other factors driving refractive error change during young adulthood.


Asunto(s)
Miopía , Errores de Refracción , Adulto , Humanos , Adulto Joven , Predisposición Genética a la Enfermedad , Tiempo de Pantalla , Luz Solar/efectos adversos , Errores de Refracción/genética , Miopía/genética , Conjuntiva
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA