Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Exp Eye Res ; 241: 109850, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38423204

RESUMO

This study aims to determine the risk associated with early age-related macular degeneration (AMD) due to refractive errors (RE) using an analysis of genome-wide association study (GWAS) data through the two-sample Mendelian randomization approach. Single-nucleotide polymorphisms (SNPs) linked to refractive errors (RE) were obtained from numerous GWAS studies involving individuals of European descent. The data for early AMD was obtained from a diverse, multiethnic GWAS meta-analysis that included 105,248 participants (14,034 cases and 91,214 controls). The primary outcome measure focused on the rise in early AMD risk corresponding to a 1-diopter alteration in spherical power and cylindrical power. In the main Mendelian randomization analysis, inverse-variance weighting (IVW) methods were applied for the evaluation. Mendelian Randomization (MR) study revealed a substantial impact of refractive error (RE) on early AMD risk, with a 1-diopter increase in hypermetropia being related to a 1.16 odds ratio (OR) for a greater risk of early AMD (95% CI, 1.10-1.23; P < 0.01). This conclusion was further supported by four supplementary approaches, namely, Weighted mode, Weighted-median, Simple mode, and MR-Egger. The results suggest a heightened risk of early AMD correlated with hyperopia, necessitating further research to thoroughly elucidate this potential causal relationship.


Assuntos
Hiperopia , Degeneração Macular , Erros de Refração , Humanos , Estudo de Associação Genômica Ampla , Degeneração Macular/genética , Análise da Randomização Mendeliana , Polimorfismo de Nucleotídeo Único , Erros de Refração/genética , Metanálise como Assunto
2.
Invest Ophthalmol Vis Sci ; 64(14): 28, 2023 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-37982764

RESUMO

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.


Assuntos
Miopia , Erros de Refração , Adulto , Humanos , Adulto Jovem , Predisposição Genética para Doença , Tempo de Tela , Luz Solar/efeitos adversos , Erros de Refração/genética , Miopia/genética , Túnica Conjuntiva
3.
Sci Rep ; 13(1): 2017, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737489

RESUMO

Genome-wide association studies (GWAS) have dissected numerous genetic factors underlying refractive errors (RE) such as myopia. Despite significant insights into understanding the genetic architecture of RE, few studies have validated and explored the functional role of candidate genes within these loci. To functionally follow-up on GWAS and characterize the potential role of candidate genes on the development of RE, we prioritized nine genes (TJP2, PDE11A, SHISA6, LAMA2, LRRC4C, KCNQ5, GNB3, RBFOX1, and GRIA4) based on biological and statistical evidence; and used CRISPR/cas9 to generate knock-out zebrafish mutants. These mutant fish were screened for abnormalities in axial length by spectral-domain optical coherence tomography and refractive status by eccentric photorefraction at the juvenile (2 months) and adult (4 months) developmental stage. We found a significantly increased axial length and myopic shift in refractive status in three of our studied mutants, indicating a potential involvement of the human orthologs (LAMA2, LRRC4C, and KCNQ5) in myopia development. Further, in-situ hybridization studies showed that all three genes are expressed throughout the zebrafish retina. Our zebrafish models provide evidence of a functional role of these three genes in refractive error development and offer opportunities to elucidate pathways driving the retina-to-sclera signaling cascade that leads to myopia.


Assuntos
Miopia , Erros de Refração , Animais , Humanos , Estudo de Associação Genômica Ampla , Miopia/genética , Erros de Refração/genética , Retina , Peixe-Zebra/genética
4.
Commun Biol ; 6(1): 6, 2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36596879

RESUMO

Refractive error, measured here as mean spherical equivalent (SER), is a complex eye condition caused by both genetic and environmental factors. Individuals with strong positive or negative values of SER require spectacles or other approaches for vision correction. Common genetic risk factors have been identified by genome-wide association studies (GWAS), but a great part of the refractive error heritability is still missing. Some of this heritability may be explained by rare variants (minor allele frequency [MAF] ≤ 0.01.). We performed multiple gene-based association tests of mean Spherical Equivalent with rare variants in exome array data from the Consortium for Refractive Error and Myopia (CREAM). The dataset consisted of over 27,000 total subjects from five cohorts of Indo-European and Eastern Asian ethnicity. We identified 129 unique genes associated with refractive error, many of which were replicated in multiple cohorts. Our best novel candidates included the retina expressed PDCD6IP, the circadian rhythm gene PER3, and P4HTM, which affects eye morphology. Future work will include functional studies and validation. Identification of genes contributing to refractive error and future understanding of their function may lead to better treatment and prevention of refractive errors, which themselves are important risk factors for various blinding conditions.


Assuntos
Miopia , Erros de Refração , Humanos , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Miopia/genética , Erros de Refração/genética , População Branca , População do Leste Asiático
5.
PLoS Genet ; 18(11): e1010478, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36395078

RESUMO

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.


Assuntos
Miopia , Erros de Refração , Humanos , Pessoa de Meia-Idade , Adolescente , Estudo de Associação Genômica Ampla , Miopia/genética , Escolaridade , Erros de Refração/genética , Alelos , Fatores de Processamento de RNA/genética
6.
PLoS One ; 17(9): e0272379, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36137074

RESUMO

PURPOSE: Genetic variants identified through population-based genome-wide studies are generally of high frequency, exerting their action in the central part of the refractive error spectrum. However, the power to identify associations with variants of lower minor allele frequency is greatly reduced, requiring considerable sample sizes. Here we aim to assess the impact of rare variants on genetic variation of refractive errors in a very large general population cohort. METHODS: Genetic association analyses of non-cyclopaedic autorefraction calculated as mean spherical equivalent (SPHE) used whole-exome sequence genotypic information from 50,893 unrelated participants in the UK Biobank of European ancestry. Gene-based analyses tested for association with SPHE using an optimised SNP-set kernel association test (SKAT-O) restricted to rare variants (minor allele frequency < 1%) within protein-coding regions of the genome. All models were adjusted for age, sex and common lead variants within the same locus reported by previous genome-wide association studies. Potentially causal markers driving association at significant loci were elucidated using sensitivity analyses by sequentially dropping the most associated variants from gene-based analyses. RESULTS: We found strong statistical evidence for association of SPHE with the SIX6 (p-value = 2.15 x 10-10, or Bonferroni-Corrected p = 4.41x10-06) and the CRX gene (p-value = 6.65 x 10-08, or Bonferroni-Corrected p = 0.001). The SIX6 gene codes for a transcription factor believed to be critical to the eye, retina and optic disc development and morphology, while CRX regulates photoreceptor specification and expression of over 700 genes in the retina. These novel associations suggest an important role of genes involved in eye morphogenesis in refractive error. CONCLUSION: The results of our study support previous research highlighting the importance of rare variants to the genetic risk of refractive error. We explain some of the origins of the genetic signals seen in GWAS but also report for the first time a completely novel association with the CRX gene.


Assuntos
Estudo de Associação Genômica Ampla , Erros de Refração , Frequência do Gene , Predisposição Genética para Doença , Humanos , Polimorfismo de Nucleotídeo Único , Erros de Refração/genética , Fatores de Transcrição/genética
7.
Genes (Basel) ; 13(7)2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35885949

RESUMO

Purpose: This study aimed to evaluate the associations of GJD2 (rs634990, rs524952) and RASGRF1 (rs8027411, rs4778879, rs28412916) gene polymorphisms with refractive errors. Methods: The study included 373 subjects with refractive errors (48 myopia, 239 myopia with astigmatism, 14 hyperopia, and 72 hyperopia with astigmatism patients) and 104 ophthalmologically healthy subjects in the control group. A quantitative real-time polymerase chain reaction (qPCR) method was chosen for genotyping. Statistical calculations and analysis of results were performed with IBM SPSS Statistics 27 software. Results: The correlations in monozygotic (MZ) twin pairs were higher compared to DZ pairs, indicating genetic effects on hyperopia and astigmatism. The heritability (h2) of hyperopia and astigmatism was 0.654 for the right eye and 0.492 for the left eye. The GJD2 rs634990 TT genotype increased the incidence of hyperopia with astigmatism by 2.4-fold and the CT genotype decreased the incidence of hyperopia with astigmatism by 0.51-fold (p < 0.05). The GJD2 rs524952 AT genotype reduced the incidence of hyperopia with astigmatism by 0.53-fold (p < 0.05). Haplotype analysis of SNPs in the GJD2 gene revealed two statistically significant haplotypes: ACTAGG for rs634990 and TTTAGA for rs524952, which statistically significantly reduced the incidence of hyperopia and hyperopia with astigmatism by 0.41-fold (95% CI: 0.220−0.765) and 0.383-fold (95% CI: 0.199−0.737), respectively (p < 0.05). It was also found that, in the presence of haplotypes ACTAGG for rs634990 and TATAGA for rs524952, the possibility of hyperopia was reduced by 0.4-fold (p < 0.05). Conclusions: the heritability of hyperopia and hyperopia with astigmatism was 0.654−0.492, according to different eyes in patients between 20 and 40 years. The GJD2 rs634990 was identified as an SNP, which has significant associations with the co-occurrence of hyperopia and astigmatism. Patients with the GJD2 gene rs634990 TT genotype were found to have a 2.4-fold higher risk of develop hyperopia with astigmatism.


Assuntos
Astigmatismo , Hiperopia , Miopia , Erros de Refração , Astigmatismo/epidemiologia , Conexinas , Humanos , Hiperopia/epidemiologia , Hiperopia/genética , Miopia/genética , Erros de Refração/genética
8.
Eur J Hum Genet ; 30(11): 1226-1232, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35618892

RESUMO

Parents pass on both their genes and environment to offspring, prompting debate about the relative importance of nature versus nurture in the inheritance of complex traits. Advances in molecular genetics now make it possible to quantify an individual's genetic predisposition to a trait via his or her 'polygenic score'. However, part of the risk captured by an individual's polygenic score may actually be attributed to the genotype of their parents. In the most well-studied example of this indirect 'genetic nurture' effect, about half the genetic contribution to educational attainment was found to be attributed to parental alleles, even if those alleles were not inherited by the child. Refractive errors, such as myopia, are a common cause of visual impairment and pose high economic and quality-of-life costs. Despite strong evidence that refractive errors are highly heritable, the extent to which genetic risk is conferred directly via transmitted risk alleles or indirectly via the environment that parents create for their children is entirely unknown. Here, an instrumental variable analysis in 1944 pairs of adult siblings from the United Kingdom was used to quantify the proportion of the genetic risk ('single nucleotide polymorphism (SNP) heritability') of refractive error contributed by genetic nurture. We found no evidence of a contribution from genetic nurture: non-within-family SNP-heritability estimate = 0.213 (95% confidence interval 0.134-0.310) and within-family SNP-heritability estimate = 0.250 (0.152-0.372). Our findings imply the genetic contribution to refractive error is principally an intrinsic effect from alleles transmitted from parents to offspring.


Assuntos
Herança Multifatorial , Erros de Refração , Adulto , Criança , Humanos , Predisposição Genética para Doença , Genótipo , Polimorfismo de Nucleotídeo Único , Erros de Refração/genética
9.
Invest Ophthalmol Vis Sci ; 63(3): 5, 2022 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-35262731

RESUMO

Refractive errors are common eye disorders characterized by a mismatch between the focal power of the eye and its axial length. An increased axial length is a common cause of the refractive error myopia (nearsightedness). The substantial increase in myopia prevalence over the last decades has raised public health concerns because myopia can lead to severe ocular complications later in life. Genomewide association studies (GWAS) have made considerable contributions to the understanding of the genetic architecture of refractive errors. Among the hundreds of genetic variants identified, common variants near the gap junction delta-2 (GJD2) gene have consistently been reported as one of the top hits. GJD2 encodes the connexin 36 (Cx36) protein, which forms gap junction channels and is highly expressed in the neural retina. In this review, we provide current evidence that links GJD2(Cx36) to the development of myopia. We summarize the gap junctional communication in the eye and the specific role of GJD2(Cx36) in retinal processing of visual signals. Finally, we discuss the pathways involving dopamine and gap junction phosphorylation and coupling as potential mechanisms that may explain the role of GJD2(Cx36) in refractive error development.


Assuntos
Conexinas , Miopia , Erros de Refração , Conexinas/genética , Conexinas/metabolismo , Junções Comunicantes/metabolismo , Humanos , Miopia/genética , Miopia/metabolismo , Erros de Refração/genética , Erros de Refração/metabolismo , Retina/metabolismo
10.
Hum Mol Genet ; 31(17): 3012-3019, 2022 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-35220419

RESUMO

Refractive errors, particularly myopia, are the most common eye conditions, often leading to serious visual impairment. The age of onset is correlated with the severity of refractive error in adulthood observed in epidemiological and genetic studies and can be used as a proxy in refractive error genetic studies. To further elucidate genetic factors that influence refractive error, we analysed self-reported age of refractive error correction data from the UK Biobank European and perform genome-wide time-to-event analyses on the age of first spectacle wear (AFSW). Genome-wide proportional hazards ratio analyses were conducted in 340 318 European subjects. We subsequently assessed the similarities and differences in the genetic architectures of refractive error correction from different causes. All-cause AFSW was genetically strongly correlated (rg = -0.68) with spherical equivalent (the measured strength of spectacle lens required to correct the refractive error) and was used as a proxy for refractive error. Time-to-event analyses found genome-wide significant associations at 44 independent genomic loci, many of which (GJD2, LAMA2, etc.) were previously associated with refractive error. We also identified six novel regions associated with AFSW, the most significant of which was on chromosome 17q (P = 3.06 × 10-09 for rs55882072), replicating in an independent dataset. We found that genes associated with AFSW were significantly enriched for expression in central nervous system tissues and were involved in neurogenesis. This work demonstrates the merits of time-to-event study design in the genetic investigation of refractive error and contributes additional knowledge on its genetic risk factors in the general population.


Assuntos
Miopia , Erros de Refração , Adulto , Óculos , Estudo de Associação Genômica Ampla , Humanos , Miopia/genética , Erros de Refração/genética
11.
Hum Mol Genet ; 31(11): 1909-1919, 2022 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-35022715

RESUMO

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.


Assuntos
Miopia , Erros de Refração , Adulto , Exoma/genética , Estudo de Associação Genômica Ampla/métodos , Humanos , Miopia/genética , Proteínas de Neoplasias/genética , Proteínas Citotóxicas Formadoras de Poros , Erros de Refração/genética , Sequenciamento do Exoma
12.
Exp Eye Res ; 209: 108693, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34228967

RESUMO

Refractive eye development is a tightly coordinated developmental process. The general layout of the eye and its various components are established during embryonic development, which involves a complex cross-tissue signaling. The eye then undergoes a refinement process during the postnatal emmetropization process, which relies heavily on the integration of environmental and genetic factors and is controlled by an elaborate genetic network. This genetic network encodes a multilayered signaling cascade, which converts visual stimuli into molecular signals that guide the postnatal growth of the eye. The signaling cascade underlying refractive eye development spans across all ocular tissues and comprises multiple signaling pathways. Notably, tissue-tissue interaction plays a key role in both embryonic eye development and postnatal eye emmetropization. Recent advances in eye biometry, physiological optics and systems genetics of refractive error have significantly advanced our understanding of the biological processes involved in refractive eye development and provided a framework for the development of new treatment options for myopia. In this review, we summarize the recent data on the mechanisms and signaling pathways underlying refractive eye development and discuss new evidence suggesting a wide-spread signal integration across different tissues and ocular components involved in visually guided eye growth.


Assuntos
Refração Ocular/fisiologia , Erros de Refração/fisiopatologia , Animais , Redes Reguladoras de Genes , Humanos , Erros de Refração/diagnóstico , Erros de Refração/genética
13.
Commun Biol ; 4(1): 676, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083742

RESUMO

Myopia is the most common developmental disorder of juvenile eyes, and it has become an increasing cause of severe visual impairment. The GJD2 locus has been consistently associated with myopia in multiple independent genome-wide association studies. However, despite the strong genetic evidence, little is known about the functional role of GJD2 in refractive error development. Here, we find that depletion of gjd2a (Cx35.5) or gjd2b (Cx35.1) orthologs in zebrafish, cause changes in the biometry and refractive status of the eye. Our immunohistological and scRNA sequencing studies show that Cx35.5 (gjd2a) is a retinal connexin and its depletion leads to hyperopia and electrophysiological changes in the retina. These findings support a role for Cx35.5 (gjd2a) in the regulation of ocular biometry. Cx35.1 (gjd2b) has previously been identified in the retina, however, we found an additional lenticular role. Lack of Cx35.1 (gjd2b) led to a nuclear cataract that triggered axial elongation. Our results provide functional evidence of a link between gjd2 and refractive error.


Assuntos
Conexinas/genética , Modelos Animais de Doenças , Proteínas do Olho/genética , Mutação , Erros de Refração/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Catarata/genética , Conexinas/metabolismo , Proteínas do Olho/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Miopia/genética , RNA-Seq/métodos , Retina/metabolismo , Retina/patologia , Epitélio Pigmentado da Retina/metabolismo , Epitélio Pigmentado da Retina/patologia , Análise de Célula Única/métodos , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
14.
Exp Eye Res ; 209: 108669, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34126082

RESUMO

M-opsin, encoded by opn1mw gene, is involved in green-light perception of mice. The role of M-opsin in emmetropization of mice remains uncertain. To answer the above question, 4-week-old wild-type (WT) mice were exposed to white light or green light (460-600 nm, a peak at 510 nm) for 12 weeks. Refractive development was estimated biweekly. After treatment, retinal function was assessed using electroretinogram (ERG). Dopamine (DA) in the retina was evaluated by high-performance liquid chromatography, M-opsin and S-opsin protein levels by Western blot and ELISA, and mRNA expressions of opn1mw and opn1sw by RT-PCR. Effects of M-opsin were further verified in Opn1mw-/- and WT mice raised in white light for 4 weeks. Refractive development was examined at 4, 6, and 8 weeks after birth. The retinal structure was estimated through hematoxylin and eosin staining (H&E) and transmission electron microscopy (TEM). Retinal wholemounts from WT and Opn1mw-/- mice were co-immunolabeled with M-opsin and S-opsin, their distribution and quantity were then assayed by immunofluorescence staining (IF). Expression of S-opsin protein and opn1sw mRNA were determined by Western blot, ELISA, or RT-PCR. Retinal function and DA content were analyzed by ERG and liquid chromatography tandem-mass spectrometry (LC-MS/MS), respectively. Lastly, visual cliff test was used to evaluate the depth perception of the Opn1mw-/- mice. We found that green light-treated WT mice were more myopic with increased M-opsin expression and decreased DA content than white light-treated WT mice after 12-week illumination. No electrophysiologic abnormalities were recorded in mice exposed to green light compared to those exposed to white light. A more hyperopic shift was further observed in 8-week-old Opn1mw-/- mice in white light with lower DA level and weakened cone function than the WT mice under white light. Neither obvious structural disruption of the retina nor abnormal depth perception was found in Opn1mw-/- mice. Together, these results suggested that the M-opsin-based color vision participated in the refractive development of mice. Overexposure to green light caused myopia, but less perception of the middle-wavelength components in white light promoted hyperopia in mice. Furthermore, possible dopaminergic signaling pathway was suggested in myopia induced by green light.


Assuntos
Visão de Cores/genética , Regulação da Expressão Gênica , Opsinas/genética , Refração Ocular/genética , Erros de Refração/genética , Retina/metabolismo , Animais , Modelos Animais de Doenças , Eletrorretinografia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Opsinas/biossíntese , RNA/genética , Erros de Refração/diagnóstico , Erros de Refração/metabolismo , Retina/ultraestrutura , Tomografia Óptica
15.
Clin Exp Optom ; 104(5): 595-601, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33689658

RESUMO

Clinical relevance: Investigation of refractive errors amongst Swedish schoolchildren will help identify risk factors associated with myopia development.Background: Genetic and hereditary aspects have been linked with the development of myopia. Nevertheless, in the case of 'school myopia' some authors suggest that environmental factors may affect gene expression, causing school myopia to soar. Additional understanding about which environmental factors play a relevant role can be gained by studying refractive errors in countries like Sweden, where prevalence of myopia is expected to be low.Methods: Swedish schoolchildren aged 8-16 years were invited to participate. Participants underwent an eye examination, including cycloplegic refraction and axial length (AL) measurements. Predictors such as time spent in near work, outdoor activities and parental myopia were obtained using a questionnaire. Myopia was defined as spherical equivalent refraction (SER) ≤ -0.50D and hyperopia as SER ≥ +0.75D.Results: A total of 128 children (70 females and 58 males) participated in this study with mean age of 12.0 years (SD = 2.4). Based on cycloplegic SER of the right eye, the distribution of refractive errors was: hyperopia 48.0% (CI95 = 38.8-56.7), emmetropia 42.0% (CI95 = 33.5-51.2) and myopia 10.0%. (CI95 = 4.4-14.9). The mean AL was 23.1 mm (SD = 0.86), there was a correlation between SER and AL, r = -0.65 (p < 0.001). Participants with two myopic parents had higher myopia and increased axial length than those with one or no myopic parents. The mean time spent in near work, outside of school, was 5.3 hours-per-day (SD = 3.1), and mean outdoor time reported was 2.6 hours-per-day (SD = 2.2) for all the participants. The time spent in near work and outdoor time were different for different refractive error categories.Conclusion: The prevalence of myopia amongst Swedish schoolchildren is low. Hereditary and environmental factors are associated with refractive error categories. Further studies with this sample are warranted to investigate how refractive errors and environmental factors interact over time.


Assuntos
Hiperopia , Miopia , Erros de Refração , Comprimento Axial do Olho , Criança , Feminino , Humanos , Hiperopia/epidemiologia , Hiperopia/genética , Recém-Nascido , Masculino , Miopia/epidemiologia , Miopia/genética , Refração Ocular , Erros de Refração/epidemiologia , Erros de Refração/genética , Suécia/epidemiologia
16.
Biochem Biophys Res Commun ; 549: 14-20, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33652205

RESUMO

Ametropia is one of the most common ocular disorders worldwide, to which almost half of visual impairments are attributed. Growing evidence has linked the development of ametropia with ambient light, including blue light, which is ubiquitous in our surroundings and has the highest photonic energy among the visible spectrum. However, the underlying mechanism of blue light-mediated ametropia remains controversial and unclear. In the present study, our data demonstrated that exposure of the retinal pigment epithelium (RPE) to blue light elevated the levels of the vital ametropia-related factor type Ⅰ collagen (COL1) via ß-catenin inhibition in scleral fibroblasts, leading to axial ametropia (hyperopic shift). Herein, our study provides evidence for the vital role of blue light-induced RPE dysfunction in the process of blue light-mediated ametropia, providing intriguing insights into ametropic aetiology and pathology by proposing a link among blue light, RPE dysfunction and ametropia.


Assuntos
Luz , Erros de Refração/patologia , Epitélio Pigmentado da Retina/patologia , Epitélio Pigmentado da Retina/efeitos da radiação , Animais , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , Colágeno Tipo I/metabolismo , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Camundongos Endogâmicos C57BL , Erros de Refração/genética , Refratometria , Esclera/patologia , Regulação para Cima/efeitos da radiação , beta Catenina/metabolismo
17.
Genes (Basel) ; 12(2)2021 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-33562844

RESUMO

Dual-specificity tyrosine phosphorylation-regulated kinase 1A or DYRK1A, contributes to central nervous system development in a dose-sensitive manner. Triallelic DYRK1A is implicated in the neuropathology of Down syndrome, whereas haploinsufficiency causes the rare DYRK1A-related intellectual disability syndrome (also known as mental retardation 7). It is characterised by intellectual disability, autism spectrum disorder and microcephaly with a typical facial gestalt. Preclinical studies elucidate a role for DYRK1A in eye development and case studies have reported associated ocular pathology. In this study families of the DYRK1A Syndrome International Association were asked to self-report any co-existing ocular abnormalities. Twenty-six patients responded but only 14 had molecular confirmation of a DYRK1A pathogenic variant. A further nineteen patients from the UK Genomics England 100,000 Genomes Project were identified and combined with 112 patients reported in the literature for further analysis. Ninety out of 145 patients (62.1%) with heterozygous DYRK1A variants revealed ocular features, these ranged from optic nerve hypoplasia (13%, 12/90), refractive error (35.6%, 32/90) and strabismus (21.1%, 19/90). Patients with DYRK1A variants should be referred to ophthalmology as part of their management care pathway to prevent amblyopia in children and reduce visual comorbidity, which may further impact on learning, behaviour, and quality of life.


Assuntos
Sistema Nervoso Central/anormalidades , Sistema Nervoso Central/patologia , Deficiência Intelectual/genética , Doenças do Nervo Óptico/genética , Nervo Óptico/anormalidades , Proteínas Serina-Treonina Quinases/genética , Proteínas Tirosina Quinases/genética , Sistema Nervoso Central/crescimento & desenvolvimento , Sistema Nervoso Central/metabolismo , Criança , Pré-Escolar , Síndrome de Down/genética , Síndrome de Down/patologia , Olho/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Feminino , Haploinsuficiência/genética , Humanos , Lactente , Deficiência Intelectual/patologia , Masculino , Pessoa de Meia-Idade , Nervo Óptico/patologia , Doenças do Nervo Óptico/patologia , Erros de Refração/genética , Erros de Refração/patologia , Estrabismo/genética , Estrabismo/patologia
18.
Exp Eye Res ; 200: 108205, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32866531

RESUMO

We had previously found that M to L cone abundancy ratios in the chicken retina are correlated with vitreous chamber depth and refractive state in chickens eyes, when they have normal visual exposure but not when they develop deprivation myopia. The finding suggests an interaction between cone abundancies and emmetropization. In the current study, we analyzed how stable this correlation was against changes in environmental variables and strain differences. We found that the correlation was preserved in two chicken strains, as long as they were raised in the laboratory facilities and not in the animal facilities of the institute. To determine the reasons for this difference, spectral and temporal lighting parameters were better adjusted in both places, whereas temperature, humidity, food, diurnal lighting cycles and illuminance were already matched. It was also verified that both strains of chickens had the same cone opsin amino acid sequences. The correlation between M to L cone abundancy and ocular biometry is highly susceptible to changes in environmental variables. Yet undetermined differences in lighting parameters were the most likely reasons. Other striking findings were that green cone opsin mRNA expression was downregulated when deprivation myopia developed. Similarly, red opsin mRNA was downregulated when chicks wore red spectacles, which made them more hyperopic. In summary, our experiments show that photoreceptor abundancies, opsin expression, and the responses to deprivation, and therefore emmetropization, are surprisingly dependent on subtle differences in lighting parameters.


Assuntos
Opsinas dos Cones/genética , Regulação da Expressão Gênica , Iluminação , RNA/genética , Refração Ocular/fisiologia , Erros de Refração/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Biometria , Galinhas , Opsinas dos Cones/biossíntese , Opsinas dos Cones/efeitos da radiação , Modelos Animais de Doenças , Erros de Refração/metabolismo , Erros de Refração/fisiopatologia , Células Fotorreceptoras Retinianas Cones/efeitos da radiação
19.
Nat Genet ; 52(4): 401-407, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32231278

RESUMO

Refractive errors, in particular myopia, are a leading cause of morbidity and disability worldwide. Genetic investigation can improve understanding of the molecular mechanisms that underlie abnormal eye development and impaired vision. We conducted a meta-analysis of genome-wide association studies (GWAS) that involved 542,934 European participants and identified 336 novel genetic loci associated with refractive error. Collectively, all associated genetic variants explain 18.4% of heritability and improve the accuracy of myopia prediction (area under the curve (AUC) = 0.75). Our results suggest that refractive error is genetically heterogeneous, driven by genes that participate in the development of every anatomical component of the eye. In addition, our analyses suggest that genetic factors controlling circadian rhythm and pigmentation are also involved in the development of myopia and refractive error. These results may enable the prediction of refractive error and the development of personalized myopia prevention strategies in the future.


Assuntos
Predisposição Genética para Doença/genética , Miopia/genética , Erros de Refração/genética , Adulto , Idoso , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , População Branca/genética
20.
Mol Genet Genomics ; 295(4): 843-853, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32227305

RESUMO

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.


Assuntos
Estudo de Associação Genômica Ampla , Miopia/genética , Característica Quantitativa Herdável , Erros de Refração/genética , Adulto , Idoso , Bancos de Espécimes Biológicos , Feminino , Genes Dominantes/genética , Predisposição Genética para Doença , Variação Genética/genética , Humanos , Laminina/genética , Masculino , Pessoa de Meia-Idade , Herança Multifatorial/genética , Miopia/patologia , Polimorfismo de Nucleotídeo Único/genética , Erros de Refração/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...