1
|
Bhattacharya S, Deka J, Avallone T, Todd L. The neuroimmune interface in retinal regeneration. Prog Retin Eye Res 2025; 106:101361. [PMID: 40287050 DOI: 10.1016/j.preteyeres.2025.101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/12/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Retinal neurodegeneration leads to irreversible blindness due to the mammalian nervous system's inability to regenerate lost neurons. Efforts to regenerate retina involve two main strategies: stimulating endogenous cells to reprogram into neurons or transplanting stem-cell derived neurons into the degenerated retina. However, both approaches must overcome a major barrier in getting new neurons to grow back down the optic nerve and connect to appropriate visual targets in environments that differ significantly from developmental conditions. While immune privilege has historically been associated with the central nervous system, an emerging literature highlights the active role of immune cells in shaping neurodegeneration and regeneration. This review explores the neuroimmune interface in retinal repair, dissecting how immune interactions influence glial reprogramming, transplantation outcomes, and axonal regeneration. By integrating insights from regenerative species with mammalian models, we highlight novel immunomodulatory strategies to optimize retinal regeneration.
Collapse
Affiliation(s)
- Sucheta Bhattacharya
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Jugasmita Deka
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Thomas Avallone
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Levi Todd
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, 13210, USA; Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA.
| |
Collapse
|
2
|
Morino K, Miyake M, Nagasaki M, Kawaguchi T, Numa S, Mori Y, Yasukura S, Akada M, Nakao SY, Nakata A, Hashimoto H, Otokozawa R, Kamoi K, Takahashi H, Tabara Y, Matsuda F, Ohno-Matsui K, Tsujikawa A. Genome-wide Meta-analysis for Myopic Macular Neovascularization Identified a Novel Susceptibility Locus and Revealed a Shared Genetic Susceptibility with Age-Related Macular Degeneration. Ophthalmol Retina 2025; 9:367-377. [PMID: 39489378 DOI: 10.1016/j.oret.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 11/05/2024]
Abstract
PURPOSE To identify the susceptibility loci for myopic macular neovascularization (mMNV) in patients with high myopia. DESIGN A genome-wide association study (GWAS) meta-analysis (meta-GWAS). PARTICIPANTS We included 2783 highly myopic individuals, including 608 patients with mMNV and 2175 control participants without mMNV. METHODS We performed a meta-analysis of 3 independent GWASs conducted according to the genotyping platform (Illumina Asian Screening Array [ASA] data set, Illumina Human610 BeadChip [610K] data set, and whole genome sequencing [WGS] data set), adjusted for age, sex, axial length, and the first to third principal components. We used DeltaSVM to evaluate the binding affinity of transcription factors (TFs) to DNA sequences around the susceptibility of single nucleotide polymorphisms (SNPs). In addition, we evaluated the contribution of previously reported age-related macular degeneration (AMD) susceptibility loci. MAIN OUTCOME MEASURES The association between SNPs and mMNV in patients with high myopia. RESULTS The meta-GWAS identified rs56257842 at TEX29- LINC02337 as a novel susceptibility SNP for mMNV (odds ratio [OR]meta = 0.62, Pmeta = 4.63 × 10-8, I2 = 0.00), which was consistently associated with mMNV in all data sets (ORASA = 0.59, PASA = 1.71 × 10-4; OR610K = 0.63, P610K = 5.53 × 10-4; ORWGS = 0.66, PWGS = 4.38 × 10-2). Transcription factor-wide analysis showed that the TFs ZNF740 and EGR1 lost their binding affinity to this locus when rs56257842 had the C allele (alternative allele), and the WNT signaling-related TF ZBTB33 gained binding affinity when rs56257842 had the C allele. When we examined the associations of AMD susceptibility loci, rs12720922 at CETP showed a statistically significant association with mMNV (ORmeta = 0.52, Pmeta = 1.55 × 10-5), whereas rs61871745 near ARMS2 showed a marginal association (ORmeta = 1.25, Pmeta = 7.79 × 10-3). CONCLUSIONS Our study identified a novel locus associated with mMNV in high myopia. Subsequent analyses offered important insights into the molecular biology of mMNV, providing the potential therapeutic targets for mMNV. Furthermore, our findings imply shared genetic susceptibility between mMNV and AMD. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Kazuya Morino
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Miyake
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Masao Nagasaki
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takahisa Kawaguchi
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shogo Numa
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Mori
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shota Yasukura
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masahiro Akada
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shin-Ya Nakao
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ai Nakata
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroki Hashimoto
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan; Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryoko Otokozawa
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koju Kamoi
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Takahashi
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuharu Tabara
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Shizuoka, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyoko Ohno-Matsui
- Department of Ophthalmology and Visual Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akitaka Tsujikawa
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
3
|
Xie J, Bui BV, Goodbourn PT, Jusuf PR. EFEMP1 contributes to light-dependent ocular growth in zebrafish. Biol Open 2024; 13:bio061741. [PMID: 39607017 PMCID: PMC11625888 DOI: 10.1242/bio.061741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 11/29/2024] Open
Abstract
Myopia (short-sightedness) is the most common ocular disorder. It generally develops after over-exposure to aberrant visual environments, disrupting emmetropization mechanisms that should match eye growth with optical power. A pre-screening of strongly associated myopia-risk genes identified through human genome-wide association studies implicates efemp1 in myopia development, but how this gene impacts ocular growth remains unclear. Here, we modify efemp1 expression specifically in the retina of zebrafish. We found that under normal lighting, efemp1 mutants developed axial myopia, enlarged eyes, reduced spatial vision and altered retinal function. However, under myopia-inducing dark-rearing, compared to control fish, mutants remained emmetropic and showed changes in retinal function. Efemp1 modification changed the expression of efemp1, egr1, tgfb1a, vegfab and rbp3 genes in the eye, and changed the inner retinal distributions of myopia-associated EFEMP1, TIMP2 and MMP2 proteins. Efemp1 modification also impacted dark-rearing-induced responses of vegfab and wnt2b genes and above-mentioned myopia-associated proteins. Together, we provided robust evidence that light-dependent ocular growth is regulated by efemp1.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of BioSciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Bang V. Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Patrick T. Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Patricia R. Jusuf
- School of BioSciences, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
4
|
Liu H, Schaeffel F, Feldkaemper MP. Effects of computer-generated patterns with different temporal and spatial frequencies on choroidal thickness, retinal dopamine and candidate genes in chickens wearing lenses. Front Med (Lausanne) 2024; 11:1469275. [PMID: 39720655 PMCID: PMC11666368 DOI: 10.3389/fmed.2024.1469275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/23/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Changes in choroidal thickness (ChT) are proposed to predict myopia development but evidence is mixed. We investigated time courses of choroidal responses, following different types of dynamic artificial stimulation in chicks with and without spectacle lenses, as well as changes in retinal dopamine metabolism and expression of candidate genes. Methods Chicks were kept in an arena surrounded by computer monitors presenting dynamic checkerboard fields of small, medium and large size. Fields were displayed with different cycle frequencies, as ON (rapid rise, slow decay) or OFF (slow rise, rapid decay) temporal luminance profile. Refractive errors, ocular biometry and ChT were assessed. Dopamine metabolism and candidate gene expression levels were also measured. Stimuli were applied for (1) 3 h with no lens, (2) 3 h and monocular treatment with -7D or +7D lenses, (3) 3 or 7 days. Results (1) The smallest fields caused the largest decrease in ChT. (2) Negative lens treatment induced on average 11.7 μm thinner choroids. ChT thinning was enhanced by 10 Hz-ON medium field size flicker which also reduced choroidal thickening with positive lenses. (3) With prolonged treatment, the choroid recovered from initial thinning in all groups although to varying degrees which were dependent on stimulus parameters. Relative ChT changes were positively correlated with the vitreal level of dopamine metabolites. Retinal EGR-1 mRNA level was positively correlated with choroidal thickness. Retinal melanopsin mRNA was increased by 10 Hz-ON stimulation and choroidal BMPR1A mRNA increased with 10 Hz-OFF stimulation. On average, early choroidal thinning did not predict the amount of negative lens-induced eye growth changes after 7 days, whereas later ChT changes showed a weak association. Conclusion Negative lenses caused long-lasting choroidal thinning, with some recovery during lens wear, especially after stimulation with 10 Hz. The dynamic stimuli modulated choroidal thinning but effects were small. There was little difference between ON and OFF stimulation, perhaps because the checkerboard patterns were too coarse. 10 Hz cycle frequency increased dopamine release. Less dopamine was correlated with thinner choroids. Result do not exclude a predictive value of choroidal thickening for future refractive development since we almost exclusively tested choroidal thinning effects.
Collapse
Affiliation(s)
- Hong Liu
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, China
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
| | - Marita Pauline Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
| |
Collapse
|
5
|
Zhang Y, Zhu Q, Song W, Chuang GM, Sun D, Cheung K, Chou A, He A, Shoghi E, Wildsoet CF. Dynamic BMP gene expression regulation in chick RPE during recovery from short term optical defocus and form-deprivation. PLoS One 2024; 19:e0311505. [PMID: 39392817 PMCID: PMC11469538 DOI: 10.1371/journal.pone.0311505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/19/2024] [Indexed: 10/13/2024] Open
Abstract
PURPOSE This study investigated the differential gene expression of BMPs in chick retinal pigment epithelium (RPE) during recovery from short term exposure to optical defocus and form-deprivation (FD) treatments. METHODS 14-day old White-Leghorn chicks wore either monocular +10 or -10 D lenses, or diffusers for 2 or 48 h, after which eyes were allowed unobstructed vision for up to 96 h. Over this recovery period, refractive errors and choroidal thickness (ChT) were tracked using retinoscopy and high-frequency A-scan ultrasonography. Real-time PCR was used to examine the expression of BMP2, 4, and 7 genes in RPE samples collected 0, 15 min, 2, 24, 48, and 96 h after the termination of treatments. Expression levels in treated eyes and their contralateral control eyes were compared. RESULTS After the termination of the lens and diffuser treatments, eyes gradually recovered from induced shifts in refractive error. With all three treatments, ChT changes reached statistical significance after 48 h of treatment, be it thinning with the -10 D lens and diffuser treatments (-0.06 ± 0.03mm, p < 0.05; -0.11 ± 0.04 mm, p < 0.05, resp.), or thickening with the +10 D lens (0.31 ± 0.04 mm, p < 0.001). BMP2 gene expression was rapidly upregulated in eyes wearing the +10 D lens, being statistical significance after 2 h, as well as 48 h of treatment. With the 2 h treatment, the latter gene expression pattern persisted for 15 min into the recovery period, before decreasing to the same level as that of contralateral control eyes, with a short-lived rebound, i.e., upregulation, 24 h into the recovery period. With the longer, 48 h treatment, BMP2 gene expression decreased more gradually, from 739 ± 121% at the end of the treatment period, to 72 ± 14% after 48 h of recovery. Two and 48 h of both -10 D and FD treatments resulted in BMP2 gene expression downregulation, with the time taken for gene expression levels to fully recover varying with the duration of initial treatments. In both cases, BMP2 gene expression downregulation persisted for 15 min into the recovery period, but reversed to upregulation by 2 h. Similar gene expression patterns were also observed for BMP4, although the changes were smaller. CONCLUSIONS The observed changes in BMP gene expression in chick RPE imply dynamic, albeit complex regulation, with the duration of exposure and recovery being critical variables for all three types of visual manipulations. This study provides further evidence for a role of the RPE as an important signal relay linking the retina to the choroid and sclera in eye growth regulation.
Collapse
Affiliation(s)
- Yan Zhang
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Qiurong Zhu
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wulian Song
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
- Department of Ophthalmology, the 2nd Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Grace May Chuang
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Daniel Sun
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Kiana Cheung
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Andreana Chou
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Andrea He
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Elham Shoghi
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| | - Christine F. Wildsoet
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States of America
| |
Collapse
|
6
|
Xie J, Goodbourn PT, Bui BV, Jusuf PR. Establishment and comprehensive characterization of a novel dark-reared zebrafish model for myopia studies. Exp Eye Res 2024; 246:110009. [PMID: 39067805 DOI: 10.1016/j.exer.2024.110009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/09/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Myopia is predicted to impact approximately 5 billion people by 2050, necessitating mechanistic understanding of its development. Myopia results from dysregulated genetic mechanisms of emmetropization, caused by over-exposure to aberrant visual environments; however, these genetic mechanisms remain unclear. Recent human genome-wide association studies have identified a range of novel myopia-risk genes. To facilitate large-scale in vivo mechanistic examination of gene-environment interactions, this study aims to establish a myopia model platform that allows efficient environmental and genetic manipulations. We established an environmental zebrafish myopia model by dark-rearing. Ocular biometrics including relative ocular refraction were quantified using optical coherence tomography images. Spatial vision was assessed using optomotor response (OMR). Retinal function was analyzed via electroretinography (ERG). Myopia-associated molecular contents or distributions were examined using RT-qPCR or immunohistochemistry. Our model produces robust phenotypic changes, showing myopia after 2 weeks of dark-rearing, which were recoverable within 2 weeks after returning animals to normal lighting. 2-week dark-reared zebrafish have reduced spatial-frequency tuning function. ERG showed reduced photoreceptor and bipolar cell function (a- and b-waves) after only 2 days of dark-rearing, which worsened after 2 weeks of dark-rearing. We also found dark-rearing-induced changes to expression of myopia-risk genes, including egr1, vegfaa, vegfab, rbp3, gjd2a and gjd2b, inner retinal distribution of EFEMP1, TIMP2 and MMP2, as well as transiently reduced PSD95 density in the inner plexiform layer. Coupled with the gene editing tools available for zebrafish, our environmental myopia model provides an excellent platform for large-scale investigation of gene-environment interactions in myopia development.
Collapse
Affiliation(s)
- Jiaheng Xie
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Patrick T Goodbourn
- Melbourne School of Psychological Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| | - Patricia Regina Jusuf
- School of Biosciences, The University of Melbourne, Parkville, 3010, Victoria, Australia.
| |
Collapse
|
7
|
Schaeffel F, Swiatczak B. Mechanisms of emmetropization and what might go wrong in myopia. Vision Res 2024; 220:108402. [PMID: 38705024 DOI: 10.1016/j.visres.2024.108402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
Studies in animal models and humans have shown that refractive state is optimized during postnatal development by a closed-loop negative feedback system that uses retinal image defocus as an error signal, a mechanism called emmetropization. The sensor to detect defocus and its sign resides in the retina itself. The retina and/or the retinal pigment epithelium (RPE) presumably releases biochemical messengers to change choroidal thickness and modulate the growth rates of the underlying sclera. A central question arises: if emmetropization operates as a closed-loop system, why does it not stop myopia development? Recent experiments in young human subjects have shown that (1) the emmetropic retina can perfectly distinguish between real positive defocus and simulated defocus, and trigger transient axial eye shortening or elongation, respectively. (2) Strikingly, the myopic retina has reduced ability to inhibit eye growth when positive defocus is imposed. (3) The bi-directional response of the emmetropic retina is elicited with low spatial frequency information below 8 cyc/deg, which makes it unlikely that optical higher-order aberrations play a role. (4) The retinal mechanism for the detection of the sign of defocus involves a comparison of defocus blur in the blue (S-cone) and red end of the spectrum (L + M-cones) but, again, the myopic retina is not responsive, at least not in short-term experiments. This suggests that it cannot fully trigger the inhibitory arm of the emmetropization feedback loop. As a result, with an open feedback loop, myopia development becomes "open-loop".
Collapse
Affiliation(s)
- Frank Schaeffel
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland; Section Neurobiology of the Eye, Institute of Ophthalmic Research, University of Tübingen, Germany; Zeiss Vision Lab, Institute of Ophthalmic Research, University of Tübingen, Germany.
| | - Barbara Swiatczak
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland
| |
Collapse
|
8
|
Chen YY, Tsai TH, Liu YL, Lin HJ, Wang IJ. The impact of light properties on ocular growth and myopia development. Taiwan J Ophthalmol 2024; 14:143-150. [PMID: 39027063 PMCID: PMC11253990 DOI: 10.4103/tjo.tjo-d-24-00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 07/20/2024] Open
Abstract
The objective of this article is to comprehensively review the effect of environmental lighting on ocular growth and refractive status in both animal and clinical studies, with an emphasis on the underlying mechanisms. This review was performed by searching research articles and reviews utilizing the terms "myopia," "light therapy," "axial length," "refractive error," and "emmetropization" in PubMed datasets. The review was finalized in December 2023. In the animal studies, high lighting brightness, illumination periods aligning with circadian rhythm, and color contrast signals including multiple wavelengths all help regulate ocular growth against myopia. Long wavelengths have been found to induce myopia in chicks, mice, fish, and guinea pigs, whereas shorter wavelengths lead to hyperopia. In contrast, red light has been observed to have a protective effect against myopia in tree shrews and rhesus monkeys. Apart from wavelength, flicker status also showed inconsistent effects on ocular growth, which could be attributed to differences in ocular refractive status, evolutionary disparities in retinal cone cells across species, and the selection of myopia induction models in experiments. In the clinical studies, current evidence suggests a control effect with red light therapy. Although the lighting conditions diverge from those in animal experiments, further reports are needed to assess the long-term effects. In conclusion, this review encompasses research related to the impact of light exposure on myopia and further explores the retinoscleral signaling pathway in refractive development. The aim is to establish a theoretical foundation for optimizing environmental factors in lighting design to address the epidemic of childhood myopia.
Collapse
Affiliation(s)
- Ying-Yi Chen
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Tzu-Hsun Tsai
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yao-Lin Liu
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hui-Ju Lin
- Department of Ophthalmology, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
9
|
Kang BS, Leung TW, Vyas SA, Ayerakwah PA, Lin J, Liang Y, Stell WK, Kee CS. Synchronous myopia development induced by bilateral form deprivation in chicks. Exp Eye Res 2024; 239:109783. [PMID: 38199262 DOI: 10.1016/j.exer.2024.109783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/10/2023] [Accepted: 01/04/2024] [Indexed: 01/12/2024]
Abstract
Form deprivation (FD) is a widely employed experimental paradigm, typically used to induce unilateral myopia in animal models. This model is weakened by potential influence upon the FD eye from vision in the freely-viewing contralateral eye, which could be eliminated by imposing FD in both eyes; but while a few previous studies have explored the feasibility of inducing bilateral FD in chicks, substantial discrepancies in treatment outcomes were noted. Consequently, this study aimed to establish a bilateral FD myopia model in chicks, with validation by investigating the associated ocular growth patterns, feeding, and social behavior. Six-day-old chicks were treated with bilateral (n = 21) or unilateral (n = 10) FD for 12 days; the fellow untreated eyes in the unilateral FD group served as controls. Refractive error, corneal power, and ocular axial dimensions were measured at 4-day intervals after the onset of form deprivation, with a Hartinger refractometer, a custom-made videokeratography system, and a high-resolution A-scan ultrasonographer, respectively. Body weight was monitored to assess the chick's physical development. Our results showed that birds treated with bilateral FD grew as robustly as the unilaterally form-deprived chicks, with similar or slightly heavier body weights and mortalities. Unilateral FD induced significantly higher myopia in the treated eye, with stronger corneal power, deeper anterior and vitreous chambers, and longer axial length. Moreover, either bilaterally or unilaterally FD eyes developed similar refractive error (bilateral FD, left: -28.03 ± 9.06 D, right: -28.44 ± 9.45 D; unilateral FD: -29.48 ± 8.26 D) and ocular biometric changes; but choroidal thickness was thicker in bilaterally FD eyes, rather than thinner as in unilaterally FD eyes. In addition to the highly synchronized (symmetrical, parallel) development reported previously in bilateral FD, we found in this study that the correlations between bilaterally form-deprived eyes were highest for ocular biometric parameters directly contributing to myopia development, including corneal power (r = 0.74 to 0.93), anterior chamber depth (r = 0.60 to 0.85), vitreous chamber depth (r = 0.92 to 0.94), and axial length (r = 0.90 to 0.96). The remarkably synchronized growth pattern confirmed the feasibility of the bilateral FD paradigm for future research on myopia.
Collapse
Affiliation(s)
- Byung Soo Kang
- Centre for Eye and Vision Research Ltd, Hong Kong, China; School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Tsz-Wing Leung
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; Research Centre for Sharp Vision, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Sonal Aswin Vyas
- School of Optometry, University of California, Berkeley, CA, United States
| | | | - Jiachun Lin
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Yuanyuan Liang
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - William K Stell
- Department of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, Alberta, Canada
| | - Chea-Su Kee
- Centre for Eye and Vision Research Ltd, Hong Kong, China; School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China; Research Centre for Sharp Vision, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
10
|
Peng S, Guo M, Wu C, Liu J, Zou B, Chen Y, Su Y, Shi L, Zhu S, Xu S, Guo D, Ju R, Wei L, Wei Y, Liu C. Age and light damage influence Fzd5 regulation of ocular growth-related genes. Exp Eye Res 2024; 239:109769. [PMID: 38154732 DOI: 10.1016/j.exer.2023.109769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/19/2023] [Accepted: 12/20/2023] [Indexed: 12/30/2023]
Abstract
Genetic and environmental factors can independently or coordinatively drive ocular axis growth. Mutations in FRIZZLED5 (FZD5) have been associated with microphthalmia, coloboma, and, more recently, high myopia. The molecular mechanism of how Fzd5 participates in ocular growth remains unknown. In this study, we compiled a list of human genes associated with ocular growth abnormalities based on public databases and a literature search. We identified a set of ocular growth-related genes from the list that was altered in the Fzd5 mutant mice by RNAseq analysis at different time points. The Fzd5 regulation of this set of genes appeared to be impacted by age and light damage. Further bioinformatical analysis indicated that these genes are extracellular matrix (ECM)-related; and meanwhile an altered Wnt signaling was detected. Altogether, the data suggest that Fzd5 may regulate ocular growth through regulating ECM remodeling, hinting at a genetic-environmental interaction in gene regulation of ocular axis control.
Collapse
Affiliation(s)
- Shanzhen Peng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Mingzhu Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Cheng Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Jinsong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Bin Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yuanyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Yingchun Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Lei Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Shiyong Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Shujuan Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Dianlei Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Rong Ju
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Lai Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| | - Yanhong Wei
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Chunqiao Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
11
|
Tapia F, Peñaloza V, Silva-Olivares F, Sotomayor-Zárate R, Schmachtenberg O, Vielma AH. Glucagon Increases Retinal Rod Bipolar Cell Inhibition Through a D1 Dopamine Receptor-Dependent Pathway That Is Altered After Lens-Defocus Treatment in Mice. Invest Ophthalmol Vis Sci 2024; 65:46. [PMID: 38289613 PMCID: PMC10840015 DOI: 10.1167/iovs.65.1.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/05/2024] [Indexed: 02/01/2024] Open
Abstract
Purpose Members of the secretin/glucagon family have diverse roles in retinal physiological and pathological conditions. Out of them, glucagon has been associated with eye growth regulation and image defocus signaling in the eye, both processes central in myopia induction. On the other hand, dopamine is perhaps the most studied molecule in myopia and has been proposed as fundamental in myopia pathogenesis. However, glucagonergic activity in the mammalian retina and its possible link with dopaminergic signaling remain unknown. Methods To corroborate whether glucagon and dopamine participate together in the modulation of synaptic activity in the retina, inhibitory post-synaptic currents were measured in rod bipolar cells from retinal slices of wild type and negative lens-exposed mice, using whole cell patch-clamp recordings and selective pharmacology. Results Glucagon produced an increase of inhibitory post-synaptic current frequency in rod bipolar cells, which was also dependent on dopaminergic activity, as it was abolished by dopamine type 1 receptor antagonism and under scotopic conditions. The effect was also abolished after 3-week negative lens-exposure but could be recovered using dopamine type 1 receptor agonism. Conclusions Altogether, these results support a possible neuromodulatory role of glucagon in the retina of mammals as part of a dopaminergic activity-dependent synaptic pathway that is affected under myopia-inducing conditions.
Collapse
Affiliation(s)
- Felipe Tapia
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Valentín Peñaloza
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Francisco Silva-Olivares
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
| | - Ramón Sotomayor-Zárate
- Laboratorio de Neuroquímica y Neurofarmacología, Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Universidad de Valparaíso, Valparaíso, Chile
| | - Oliver Schmachtenberg
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
- Instituto de Biología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Alex H. Vielma
- Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
12
|
Summers JA, Jones KL. Single Cell Transcriptomics Identifies Distinct Choroid Cell Populations Involved in Visually Guided Eye Growth. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1245891. [PMID: 38390290 PMCID: PMC10883300 DOI: 10.3389/fopht.2023.1245891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/06/2023] [Indexed: 02/24/2024]
Abstract
Postnatal ocular growth is regulated by a vision-dependent mechanism, termed emmetropization, which acts to minimize refractive error through coordinated growth of the ocular tissues. Many studies suggest that the ocular choroid participates in the emmetropization process via the production of scleral growth regulators that control ocular elongation and refractive development. To elucidate the role of the choroid in emmetropization, we used single-cell RNA sequencing (scRNA-seq) to characterize the cell populations in the chick choroid and compare gene expression changes in these cell populations during conditions in which the eye is undergoing emmetropization. UMAP clustering analysis identified 24 distinct cell clusters in all chick choroids. 7 clusters were identified as fibroblast subpopulations; 5 clusters represented different populations of endothelial cells; 4 clusters were CD45+ macrophages, T cells and B cells; 3 clusters were Schwann cell subpopulations; and 2 clusters were identified as melanocytes. Additionally, single populations of RBCs, plasma cells and neuronal cells were identified. Significant changes in gene expression between control and treated choroids were identified in 17 cell clusters, representing 95% of total choroidal cells. The majority of significant gene expression changes were relatively small (< 2 fold). The highest changes in gene expression were identified in a rare cell population (0.11% - 0.49% of total choroidal cells). This cell population expressed high levels of neuron-specific genes as well as several opsin genes suggestive of a rare neuronal cell population that is potentially light sensitive. Our results, for the first time, provide a comprehensive profile of the major choroidal cell types and their gene expression changes during the process of emmetropization as well as insights into the canonical pathways and upstream regulators that coordinate postnatal ocular growth.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| | - Kenneth L Jones
- Bioinformatic Solutions LLC, Sheridan, Wyoming, 82801, United States
| |
Collapse
|
13
|
Huang Y, Chen X, Zhuang J, Yu K. The Role of Retinal Dysfunction in Myopia Development. Cell Mol Neurobiol 2023; 43:1905-1930. [PMID: 36427109 PMCID: PMC11412200 DOI: 10.1007/s10571-022-01309-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
Abstract
Myopia is a refractive disorder arising from a mismatch between refractive power and relatively long axial length of the eye. With its dramatically increasing prevalence, myopia has become a pervasive social problem. It is commonly accepted that abnormal visual input acts as an initiating factor of myopia. As the first station to perceive visual signals, the retina plays an important role in myopia etiology. The retina is a fine-layered structure with multitudinous cells, processing intricate visual signals via numerous molecular pathways. Accordingly, dopaminergic mechanisms, contributions of rod and cone photoreceptors, myopic structural changes of retinal pigment epithelium (RPE) and neuro-retinal layers have all suggested a vital role of retinal dysfunction in myopia development. Herein, we separately discuss myopia-related retinal dysfunction and current dilemmas by different levels, from molecules to cells, with the hope that the comprehensive delineation could contribute to a better understanding of myopia etiology, indicate novel therapeutic targets, and inspire future studies.
Collapse
Affiliation(s)
- Yuke Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guang-Dong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No.7 Jinsui Road, Tianhe District, Guangzhou City, China
| | - Xi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guang-Dong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No.7 Jinsui Road, Tianhe District, Guangzhou City, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guang-Dong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No.7 Jinsui Road, Tianhe District, Guangzhou City, China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guang-Dong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No.7 Jinsui Road, Tianhe District, Guangzhou City, China.
| |
Collapse
|
14
|
Rucker F, Taylor C, Kaser-Eichberger A, Schroedl F. Parasympathetic and sympathetic control of emmetropization in chick. Exp Eye Res 2023; 232:109508. [PMID: 37230289 PMCID: PMC10452042 DOI: 10.1016/j.exer.2023.109508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023]
Abstract
Emmetropization can be altered by temporal visual stimulation and the spectral properties of the visual environment. The goal of the current experiment is to test the hypothesis that there is an interaction between these properties and autonomic innervation. For that purpose, selective lesions of the autonomic nervous system were performed in chickens followed by temporal stimulation. Parasympathetic lesioning involved transection of both the ciliary ganglion and the pterygopalatine ganglion (PPG_CGX; n = 38), while sympathetic lesioning involved transection of the superior cervical ganglion (SCGX; n = 49). After one week of recovery, chicks were then exposed to temporally modulated light (3 days, 2 Hz, Mean: 680 lux) that was either achromatic (with blue [RGB], or without blue [RG]), or chromatic (with blue [B/Y] or without blue [R/G]). Control birds with lesions, or unlesioned, were exposed to white [RGB] or yellow [RG] light. Ocular biometry and refraction (Lenstar and a Hartinger refractometer) was measured before and after exposure to light stimulation. Measurements were statistically analyzed for the effects of a lack of autonomic input and the type of temporal stimulation. In PPG_CGX lesioned eyes, there was no effect of the lesions one-week post-surgery. However, after exposure to achromatic modulation, the lens thickened (with blue) and the choroid thickened (without blue) but there was no effect on axial growth. Chromatic modulation thinned the choroid with R/G. In the SGX lesioned eye, there was no effect of the lesion 1-week post-surgery. However, after exposure to achromatic modulation (without blue), the lens thickened and there was a reduction in vitreous chamber depth and axial length. Chromatic modulation caused a small increase in vitreous chamber depth with R/G. Both autonomic lesion and visual stimulation were necessary to affect the growth of ocular components. The bidirectional responses observed in axial growth and in choroidal changes suggest that autonomic innervation combined with spectral cues from longitudinal chromatic aberration may provide a mechanism for homeostatic control of emmetropization.
Collapse
Affiliation(s)
- Frances Rucker
- New England College of Optometry, 424 Beacon St., Boston, MA, 02115, USA.
| | - Chris Taylor
- New England College of Optometry, 424 Beacon St., Boston, MA, 02115, USA
| | - Alexandra Kaser-Eichberger
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg Paracelsus Medical University, Salzburg, Austria
| | - Falk Schroedl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology - Salzburg Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
15
|
Summers JA, Jones KL. Single Cell Transcriptomics Identifies Distinct Choroid Cell Populations Involved in Visually Guided Eye Growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.30.542876. [PMID: 37398381 PMCID: PMC10312561 DOI: 10.1101/2023.05.30.542876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Postnatal ocular growth is regulated by a vision-dependent mechanism, termed emmetropization, which acts to minimize refractive error through coordinated growth of the ocular tissues. Many studies suggest that the ocular choroid participates in the emmetropization process via the production of scleral growth regulators that control ocular elongation and refractive development. To elucidate the role of the choroid in emmetropization, we used single-cell RNA sequencing (scRNA-seq) to characterize the cell populations in the chick choroid and compare gene expression changes in these cell populations during conditions in which the eye is undergoing emmetropization. UMAP clustering analysis identified 24 distinct cell clusters in all chick choroids. 7 clusters were identified as fibroblast subpopulations; 5 clusters represented different populations of endothelial cells; 4 clusters were CD45+ macrophages, T cells and B cells; 3 clusters were Schwann cell subpopulations; and 2 clusters were identified as melanocytes. Additionally, single populations of RBCs, plasma cells and neuronal cells were identified. Significant changes in gene expression between control and treated choroids were identified in 17 cell clusters, representing 95% of total choroidal cells. The majority of significant gene expression changes were relatively small (< 2 fold). The highest changes in gene expression were identified in a rare cell population (0.11% - 0.49% of total choroidal cells). This cell population expressed high levels of neuron-specific genes as well as several opsin genes suggestive of a rare neuronal cell population that is potentially light sensitive. Our results, for the first time, provide a comprehensive profile of the major choroidal cell types and their gene expression changes during the process of emmetropization as well as insights into the canonical pathways and upstream regulators that coordinate postnatal ocular growth.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| | - Kenneth L Jones
- Bioinformatic Solutions LLC, Sheridan, Wyoming, 82801, United States
| |
Collapse
|
16
|
Uchiyama H, Matsutani S, Ohno H, Yamaoka S, Mizokami T, Sugimoto S, Hirashima Y. Bipolar cells containing protein kinase Cα mediate attentional facilitation of the avian retinal ganglion cells by the retinopetal signal. J Comp Neurol 2023. [PMID: 37130818 DOI: 10.1002/cne.25491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 03/27/2023] [Accepted: 04/05/2023] [Indexed: 05/04/2023]
Abstract
Birds have a well-developed retinopetal system projecting from the midbrain to the contralateral retina. The signal sent to the retina through the retinopetal system facilitates visual responses of the retinal ganglion cells (RGCs), and the retinopetal signals function as attentional signals during visual search. Thus, the retinopetal signal somehow reaches and facilitates visual responses of the RGCs. However, the tertiary neuron of the retinopetal system, the isthmo-optic target cell (IOTC), is unlikely to contact most RGCs directly, because the IOTCs form axon terminals localized in the outermost sublayer (lamina 1) of the inner plexiform layer (IPL) where few RGC dendrites terminate. Therefore, some other intrinsic retinal neurons must be involved in the centrifugal attentional enhancement of visual responses of the RGCs. We investigated connections of the target cells of the IOTCs in chicken and quail, using light and electron microscopic immunohistochemistry. We show that axon terminals of the IOTC make synaptic contacts with protein kinase Cα (PKCα)-immunoreactive (ir) bipolar cells (PKCα-BCs) in lamina 1 of the IPL. Furthermore, with prolonged electrical stimulation of the isthmo-optic nucleus (ION) on one side, whose neurons send their axons to the contralateral retina and make synaptic contacts there with IOTCs, phosphorylation of cAMP response element-binding protein was observed in the PKCα-BCs in the contralateral retina, but not in the ipsilateral retina. This suggests that electrical stimulation of ION activated PKCα-BCs through synapses from IOTCs to PKCα-BCs, thus stimulating transcription in PKCα-BCs. Thus, centrifugal attentional signals may facilitate visual responses of RGCs via the PKCα-BCs.
Collapse
Affiliation(s)
- Hiroyuki Uchiyama
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Shinji Matsutani
- Department of Functional Morphology, School of Nursing, Kitasato University, Sagamihara, Japan
| | - Hiroshi Ohno
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Seiya Yamaoka
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Takuya Mizokami
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Shiho Sugimoto
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| | - Yasuhiro Hirashima
- Department of Information Science and Biomedical Engineering, Graduate School of Science and Engineering, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
17
|
Poudel S, Rahimi-Nasrabadi H, Jin J, Najafian S, Alonso JM. Differences in visual stimulation between reading and walking and implications for myopia development. J Vis 2023; 23:3. [PMID: 37014657 PMCID: PMC10080958 DOI: 10.1167/jov.23.4.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/11/2023] [Indexed: 04/05/2023] Open
Abstract
Visual input plays an important role in the development of myopia (nearsightedness), a visual disorder that blurs vision at far distances. The risk of myopia progression increases with the time spent reading and decreases with outdoor activity for reasons that remain poorly understood. To investigate the stimulus parameters driving this disorder, we compared the visual input to the retina of humans performing two tasks associated with different risks of myopia progression, reading and walking. Human subjects performed the two tasks while wearing glasses with cameras and sensors that recorded visual scenes and visuomotor activity. When compared with walking, reading black text in white background reduced spatiotemporal contrast in central vision and increased it in peripheral vision, leading to a pronounced reduction in the ratio of central/peripheral strength of visual stimulation. It also made the luminance distribution heavily skewed toward negative dark contrast in central vision and positive light contrast in peripheral vision, decreasing the central/peripheral stimulation ratio of ON visual pathways. It also decreased fixation distance, blink rate, pupil size, and head-eye coordination reflexes dominated by ON pathways. Taken together with previous work, these results support the hypothesis that reading drives myopia progression by understimulating ON visual pathways.
Collapse
Affiliation(s)
- Sabina Poudel
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Hamed Rahimi-Nasrabadi
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jianzhong Jin
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Sohrab Najafian
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jose-Manuel Alonso
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| |
Collapse
|
18
|
Ginkgo biloba extracts improve choroidal circulation leading to suppression of myopia in mice. Sci Rep 2023; 13:3772. [PMID: 36882511 PMCID: PMC9989591 DOI: 10.1038/s41598-023-30908-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 03/03/2023] [Indexed: 03/09/2023] Open
Abstract
Myopia is becoming more common across the world, necessitating the development of preventive methods. We investigated the activity of early growth response 1 (EGR-1) protein and discovered that Ginkgo biloba extracts (GBEs) activated EGR-1 in vitro. In vivo, C57BL/6 J mice were fed either normal or 0.0667% GBEs (200 mg/kg) mixed chow (n = 6 each), and myopia was induced with - 30 diopter (D) lenses from 3 to 6 weeks of age. Refraction and axial length were measured by an infrared photorefractor and an SD-OCT system, respectively. In lens-induced myopia mice, oral GBEs significantly improved refractive errors (- 9.92 ± 1.53 D vs. - 1.67 ± 3.51 D, p < 0.001) and axial elongation (0.22 ± 0.02 mm vs. 0.19 ± 0.02 mm, p < 0.05). To confirm the mechanism of GBEs in preventing myopia progression, the 3-week-old mice were divided into normally fed with either myopic-induced or non-myopic-induced groups and GBEs fed with either myopic-induced or non-myopic-induced groups (n = 10 each). Choroidal blood perfusion was measured with optical coherence tomography angiography (OCTA). In both non-myopic induced groups, compared to normal chow, oral GBEs significantly improved choroidal blood perfusion (8.48 ± 15.75%Area vs. 21.74 ± 10.54%Area, p < 0.05) and expression of Egr-1 and endothelial nitric oxide synthase (eNOS) in the choroid. In both myopic-induced groups, compared to normal chow, oral GBEs also improved choroidal blood perfusion (- 9.82 ± 9.47%Area vs. 2.29 ± 11.84%Area, p < 0.05) and was positively correlated with the change in choroidal thickness. These findings suggest that GBEs may inhibit the progression of myopia by improving choroidal blood perfusion.
Collapse
|
19
|
Vyas SA, Lakshmanan Y, Chan HHL, Leung TW, Kee CS. Experimentally induced myopia and myopic astigmatism alter retinal electrophysiology in chickens. Sci Rep 2022; 12:21180. [PMID: 36477183 PMCID: PMC9729572 DOI: 10.1038/s41598-022-25075-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Myopia (or "short-sightedness") and astigmatism are major causes of visual impairment worldwide. Significant amounts of astigmatism are frequently observed in infants and have been associated with myopia development. Although it is well established that both myopia and astigmatism are associated with ocular structural changes from anterior to posterior segments, very little is known on how these refractive errors alter retinal functions. This study investigated the effects of experimentally induced myopia and myopic-astigmatism on retinal electrophysiology by using an image-guided, multifocal global flash stimulation in chickens, a widely used animal model for refractive error development. Myopia and myopic-astigmatism were experimentally induced, respectively, by wearing spherical (- 10 D, n = 12) and sphero-cylindrical lenses (- 6.00 DS/- 8.00 DCx90: Hyperopic With-The Rule, H-WTR, n = 15; - 6.00 DS/- 8.00 DCx180: Hyperopic Against-The-Rule, H-ATR, n = 11) monocularly for a week (post-hatching day 5 to 12). An aged-matched control group without any lens treatment provided normal data (n = 12). Multifocal electrophysiological results revealed significant regional variation in the amplitude of induced component (IC) (central greater than peripheral; both p < 0.05) in the normal and H-ATR groups, but not in the - 10 D and H-WTR groups. Most importantly, for the first time, our results showed that both H-WTR and H-ATR groups exhibited a significantly longer implicit time of the inner retinal response at the central region when compared to the normal and - 10 D groups, highlighting a significant role of astigmatism in retinal physiology.
Collapse
Affiliation(s)
- Sonal Aswin Vyas
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Yamunadevi Lakshmanan
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China.,Laboratory of Experimental Optometry (Neuroscience), School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Henry Ho-Lung Chan
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China.,Laboratory of Experimental Optometry (Neuroscience), School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China.,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China
| | - Tsz-Wing Leung
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China.,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China.,Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chea-Su Kee
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China. .,Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, China. .,Research Centre for SHARP Vision (RCSV), The Hong Kong Polytechnic University, Kowloon, Hong Kong.
| |
Collapse
|
20
|
Ku H, Chen JJY, Hu M, Tien PT, Lin HJ, Xu G, Wan L, Gan D. Myopia Development in Tree Shrew Is Associated with Chronic Inflammatory Reactions. Curr Issues Mol Biol 2022; 44:4303-4313. [PMID: 36135208 PMCID: PMC9498061 DOI: 10.3390/cimb44090296] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/03/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
In this study, we aimed to investigate whether chronic retinal inflammation is involved in the pathogenesis of form-deprivation myopia (FDM) using tree shrews as an animal model. Twenty-one tree shrews were randomly divided into 7-day/14-day FDM (FDM7/FDM14) groups and their corresponding 7-day/14-day control groups. Refraction and axial length were measured. To determine the effects of form deprivation on inflammation, we used real-time polymerase chain reaction (PCR) and immunohistochemistry to assess the expression levels of several proinflammatory cytokines. At day 0, the eyes in the FDM and control groups were hyperopic. However, after 7 and 14 days of form deprivation, the refractive error of the eyes in the FDM7 and FDM14 groups shifted from +6.6 ± 0.3 diopters (D) to +4.0 ± 0.5 D and from +6.4 ± 0.3 D to +5.0 ± 0.3 D, respectively. The levels of tumor necrosis factor-α, interleukin (IL)-6, IL-8, monocyte chemoattractant protein-1, and nuclear factor κB were increased in the FDM eyes, compared with those in the control eyes. The increase in matrix metalloproteinase-2 expression was greater in the FDM eyes than in the contralateral and control eyes, whereas collagen type I expression was downregulated. In conclusion, chronic inflammation may play a crucial pathogenic role in form-deprivation myopia in tree shrews.
Collapse
Affiliation(s)
- Hsiangyu Ku
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
| | | | - Min Hu
- Department of Pediatric Ophthalmology, Hospital of Yunnan University, Kunming 650091, China
| | - Peng-Tai Tien
- Eye Center, China Medical University Hospital, Taichung 404333, Taiwan
- Graduate Institute of Clinical Medical Science, College of Medicine, China Medical University, Taichung 404333, Taiwan
| | - Hui-Ju Lin
- Eye Center, China Medical University Hospital, Taichung 404333, Taiwan
- School of Chinese Medicine, China Medical University, Taichung 404333, Taiwan
| | - Gezhi Xu
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, China
- Key Laboratory of Myopia of State Health Ministry, Shanghai 200031, China
| | - Lei Wan
- School of Chinese Medicine, China Medical University, Taichung 404333, Taiwan
- Department of Biotechnology, Asia University, Taichung 404333, Taiwan
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 404333, Taiwan
- Correspondence: (L.W.); (D.G.)
| | - Dekang Gan
- Department of Ophthalmology and Visual Science, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China
- Correspondence: (L.W.); (D.G.)
| |
Collapse
|
21
|
Goto S, Muroy SE, Zhang Y, Saijo K, Kolora SRR, Zhu Q, Wildsoet CF. Gene Expression Signatures of Contact Lens-Induced Myopia in Guinea Pig Retinal Pigment Epithelium. Invest Ophthalmol Vis Sci 2022; 63:25. [PMID: 36006019 PMCID: PMC9424971 DOI: 10.1167/iovs.63.9.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Purpose To identify key retinal pigment epithelium (RPE) genes linked to the induction of myopia in guinea pigs. Methods To induce myopia, two-week-old pigmented guinea pigs (New Zealand strain, n = 5) wore −10 diopter (D) rigid gas-permeable contact lenses (CLs), for one day; fellow eyes were left without CLs and served as controls. Spherical equivalent refractive errors (SE) and axial length (AL) were measured at baseline and one day after initiation of CL wear. RNA sequencing was applied to RPE collected from both treated and fellow (control) eyes after one day of CL-wear to identify related gene expression changes. Additional RPE-RNA samples from treated and fellow eyes were subjected to quantitative real-time PCR (qRT-PCR) analysis for validation purposes. Results The CLs induced myopia. The change from baseline values in SE was significantly different (P = 0.016), whereas there was no significant difference in the change in AL (P = 0.10). RNA sequencing revealed significant interocular differences in the expression in RPE of 13 genes: eight genes were significantly upregulated in treated eyes relative to their fellows, and five genes, including bone morphogenetic protein 2 (Bmp2), were significantly downregulated. The latter result was also confirmed by qRT-PCR. Additional analysis of differentially expressed genes revealed significant enrichment for bone morphogenetic protein (BMP) and TGF-β signaling pathways. Conclusions The results of this RPE gene expression study provide further supporting evidence for an important role of BMP2 in eye growth regulation, here from a guinea pig myopia model.
Collapse
Affiliation(s)
- So Goto
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States.,Department of Ophthalmology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Ophthalmology, National Hospital Organization, Tokyo Medical Center, Meguro-ku, Tokyo, Japan
| | - Sandra E Muroy
- Department of Integrative Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States
| | - Yan Zhang
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States
| | - Kaoru Saijo
- Department of Molecular & Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, California, United States
| | - Sree Rohit Raj Kolora
- Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Qiurong Zhu
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States.,Department of Optometry and Visual Science, West China Hospital of Sichuan University, China
| | - Christine F Wildsoet
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, United States
| |
Collapse
|
22
|
Li Q, Zhu H, Fan M, Sun J, Reinach PS, Wang Y, Qu J, Zhou X, Zhao F. Form-deprivation myopia downregulates calcium levels in retinal horizontal cells in mice. Exp Eye Res 2022; 218:109018. [PMID: 35240197 DOI: 10.1016/j.exer.2022.109018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/14/2022] [Accepted: 02/25/2022] [Indexed: 11/25/2022]
Abstract
The process of eye axis lengthening in myopic eyes is regulated by multiple mechanisms in the retina, and horizontal cells (HCs) are an essential interneuron in the visual regulatory system. Wherein intracellular Ca2+ plays an important role in the events involved in the regulatory role of HCs in the retinal neural network. It is unknown if intracellular Ca2+ regulation in HCs mediates changes in the retinal neural network during myopia progression. We describe here a novel calcium fluorescence indicator system that monitors HCs' intracellular Ca2+ levels during form-deprivation myopia (FDM) in mice. AAV injection of GCaMP6s, as a protein calcium sensor, into a Gja10-Cre mouse monitored the changes in Ca2+signaling in HC that accompany FDM progression in mice. An alternative Gja10-Cre/Ai96-GCaMP6s mouse model was created by cross mating Gja10-Cre with Ai96 mice. Immunofluorescence imaging and live imaging of the retinal cells verified the identity of these animal models. Changes in retinal horizontal cellular Ca2+ levels were resolved during FDM development. The numbers of GCaMP6s and the proportion of HCs were tracked based on profiling changes in GCaMP6s+calbindin+/calbindin+ coimmunostaining patterns. They significantly decreased more after either two days (P < 0.01) or two weeks (P < 0.001) in form deprived eyes than in the untreated fellow eyes. These decreases in their proportion reached significance only in the retinal central region rather than also in the retinal periphery. A novel approach employing a GCaMP6s mouse model was developed that may ultimately clarify if HCs mediate Ca2+ signals that contribute to controlling FDM progression in mice. The results indicate so far that FDM progression is associated with declines in HC Ca2+ signaling activity.
Collapse
Affiliation(s)
- Qihang Li
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - He Zhu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Miaomiao Fan
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jing Sun
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Peter S Reinach
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Yuhan Wang
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China; Research Unit of Myopia Basic Research and Clinical Prevention and Control, Chinese Academy of Medical Sciences (2019RU025), Wenzhou, Zhejiang, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China; Research Unit of Myopia Basic Research and Clinical Prevention and Control, Chinese Academy of Medical Sciences (2019RU025), Wenzhou, Zhejiang, China; Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
| | - Fuxin Zhao
- School of Optometry and Ophthalmology and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China; State Key Laboratory of Optometry, Ophthalmology and Vision Science, Wenzhou, Zhejiang, China.
| |
Collapse
|
23
|
Thomson K, Game J, Karouta C, Morgan IG, Ashby R. Correlation between small-scale methylation changes and gene expression during the development of myopia. FASEB J 2021; 36:e22129. [PMID: 34958689 DOI: 10.1096/fj.202101487r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/07/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022]
Abstract
Visually induced changes in the expression of early growth response-1 (EGR1), FBJ osteosarcoma oncogene (FOS), and NGFI-A binding protein-2 (NAB2) appear to form a part of a retinal network fundamental to ocular growth regulation, and thus, the development of myopia (short-sightedness). However, it is unclear how environmental (visual) cues are translated into these molecular changes. One possibility is through epigenetic modifications such as DNA methylation, a known regulator of such processes. By sequencing bisulfite-converted DNA amplicons, this study examined whether changes in DNA methylation occur within specific regulatory and promoter regions of EGR1, FOS, and NAB2 during the periods of increased and decreased ocular growth in chicks. Visually induced changes in ocular growth rates were associated with single-point, but not large-scale, shifts in methylation levels within the investigated regions. Analysis of methylation pattern variability (entropy) demonstrated that the observed methylation changes are occurring within small subpopulations of retinal cells. This concurs with previous observations that EGR1 and FOS are differentially regulated at the peptide level within specific retinal cell types. Together, the findings of this study support a potential role for DNA methylation in the translation of external visual cues into molecular changes critical for ocular growth regulation and myopia development.
Collapse
Affiliation(s)
- Kate Thomson
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Jeremy Game
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Cindy Karouta
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Ian G Morgan
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Regan Ashby
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
24
|
Karouta C, Kucharski R, Hardy K, Thomson K, Maleszka R, Morgan I, Ashby R. Transcriptome-based insights into gene networks controlling myopia prevention. FASEB J 2021; 35:e21846. [PMID: 34405458 DOI: 10.1096/fj.202100350rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Myopia (short-sightedness), usually caused by excessive elongation of the eye during development, has reached epidemic proportions worldwide. In animal systems including the chicken model, several treatments have been shown to inhibit ocular elongation and experimental myopia. Although diverse in their apparent mechanism of action, each one leads to a reduction in the rate of ocular growth. We hypothesize that a defined set of retinal molecular changes may underlie growth inhibition, irrespective of the treatment agent used. Accordingly, across five well-established but diverse methods of inhibiting myopia, significant overlap is seen in the retinal transcriptome profile (transcript levels and alternative splicing events) in chicks when analyzed by RNA-seq. Within the two major pathway networks enriched during growth inhibition, that of cell signaling and circadian entrainment, transcription factors form the largest functional grouping. Importantly, a large percentage of those genes forming the defined retinal response are downstream targets of the transcription factor EGR1 which itself shows a universal response to all five growth-inhibitory treatments. This supports EGR1's previously implicated role in ocular growth regulation. Finally, by contrasting our data with human linkage and GWAS studies on refractive error, we confirm the applicability of our study to the human condition. Together, these findings suggest that a universal set of transcriptome changes, which sit within a well-defined retinal network that cannot be bypassed, is fundamental to growth regulation, thus paving a way for designing novel targets for myopia therapies.
Collapse
Affiliation(s)
- Cindy Karouta
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Robert Kucharski
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Kristine Hardy
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Kate Thomson
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Ryszard Maleszka
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Ian Morgan
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Regan Ashby
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
25
|
Summers JA, Schaeffel F, Marcos S, Wu H, Tkatchenko AV. Functional integration of eye tissues and refractive eye development: Mechanisms and pathways. Exp Eye Res 2021; 209:108693. [PMID: 34228967 PMCID: PMC11697408 DOI: 10.1016/j.exer.2021.108693] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022]
Abstract
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.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany; Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - Susana Marcos
- Instituto de Óptica "Daza de Valdés", Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Hao Wu
- Department of Ophthalmology, Columbia University, New York, USA
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, USA; Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
26
|
Tkatchenko TV, Tkatchenko AV. Genome-wide analysis of retinal transcriptome reveals common genetic network underlying perception of contrast and optical defocus detection. BMC Med Genomics 2021; 14:153. [PMID: 34107987 PMCID: PMC8190860 DOI: 10.1186/s12920-021-01005-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Refractive eye development is regulated by optical defocus in a process of emmetropization. Excessive exposure to negative optical defocus often leads to the development of myopia. However, it is still largely unknown how optical defocus is detected by the retina. METHODS Here, we used genome-wide RNA-sequencing to conduct analysis of the retinal gene expression network underlying contrast perception and refractive eye development. RESULTS We report that the genetic network subserving contrast perception plays an important role in optical defocus detection and emmetropization. Our results demonstrate an interaction between contrast perception, the retinal circadian clock pathway and the signaling pathway underlying optical defocus detection. We also observe that the relative majority of genes causing human myopia are involved in the processing of optical defocus. CONCLUSIONS Together, our results support the hypothesis that optical defocus is perceived by the retina using contrast as a proxy and provide new insights into molecular signaling underlying refractive eye development.
Collapse
Affiliation(s)
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
- Edward S. Harkness Eye Institute, Research Annex Room 415, 635 W. 165th Street, New York, NY 10032 USA
| |
Collapse
|
27
|
Morgan IG. The biological basis of myopic refractive error. Clin Exp Optom 2021; 86:276-88. [PMID: 14558849 DOI: 10.1111/j.1444-0938.2003.tb03123.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2003] [Revised: 08/04/2003] [Accepted: 09/02/2003] [Indexed: 11/28/2022] Open
Abstract
Myopia is among the most common refractive errors and is associated with the greatest risk of pathological outcomes. Most animals, including humans, are born with hyperopic errors. During development, axial elongation of the eye occurs and is regulated through a vision-dependent process, known as emmetropisation The extremely rapid changes in the prevalence of myopia and the dependence of myopia on the level of education indicate that there are very strong environmental impacts on the development of myopia. This conflicts with the common occurrence of familial patterns of inheritance of myopia, which suggests a role for genetic determination. There are more than 150 defined genetic syndromes in which familial high myopia is one of the features, including some that are not associated with other syndromes. The evidence for the roles of both nature and nurture in the aetiology of myopia is discussed. This review also examines the experimentally induced refractive errors associated with form-deprivation, recovery from form deprivation and the effects of both negative and positive lenses. In addition, it looks at the local and optical control of eye growth. Finally, the various control pathways for growth are considered. These include dopamine, ZENK-glucagon, retinoic acid and retinoic acid receptors, crystallin, seratonin and melatonin, vasoactive intestinal peptide and enkephalins, nitric oxide and various growth factors.
Collapse
Affiliation(s)
- Ian G Morgan
- Visual Sciences Group, Research School of Biological Science and Centre for VIsual Science, Australian National University, GPO Box 475, Canberra, ACT, 2601, Australia
| |
Collapse
|
28
|
Zhang H, Wong CL, Shan SW, Li KK, Cheng AK, Lee KL, Ge J, To CH, Do CW. Characterisation of Cl‐ transporter and channels in experimentally induced myopic chick eyes. Clin Exp Optom 2021; 94:528-35. [DOI: 10.1111/j.1444-0938.2011.00611.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Affiliation(s)
- Hengli Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‐Sen University, China
- The Centre for Myopia Research, School of Optometry and the
| | - Chun Lung Wong
- The Centre for Myopia Research, School of Optometry and the
| | - Sze Wan Shan
- The Centre for Myopia Research, School of Optometry and the
| | - King Kit Li
- The Centre for Myopia Research, School of Optometry and the
| | - Angela K Cheng
- The Centre for Myopia Research, School of Optometry and the
| | - Kam Len Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong SAR, China, E‐mail:
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat‐Sen University, China
| | - Chi Ho To
- The Centre for Myopia Research, School of Optometry and the
| | - Chi Wai Do
- The Centre for Myopia Research, School of Optometry and the
| |
Collapse
|
29
|
RNA-seq and GSEA identifies suppression of ligand-gated chloride efflux channels as the major gene pathway contributing to form deprivation myopia. Sci Rep 2021; 11:5280. [PMID: 33674625 PMCID: PMC7935918 DOI: 10.1038/s41598-021-84338-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Currently there is no consensus regarding the aetiology of the excessive ocular volume that characterizes high myopia. Thus, we aimed to test whether the gene pathways identified by gene set enrichment analysis of RNA-seq transcriptomics refutes the predictions of the Retinal Ion Driven Efflux (RIDE) hypothesis when applied to the induction of form-deprivation myopia (FDM) and subsequent recovery (post-occluder removal). We found that the induction of profound FDM led to significant suppression in the ligand-gated chloride ion channel transport pathway via suppression of glycine, GABAA and GABAC ionotropic receptors. Post-occluder removal for short term recovery from FDM of 6 h and 24 h, induced significant upregulation of the gene families linked to cone receptor phototransduction, mitochondrial energy, and complement pathways. These findings support a model of form deprivation myopia as a Cl− ion driven adaptive fluid response to the modulation of the visual signal cascade by form deprivation that in turn affects the resultant ionic environment of the outer and inner retinal tissues, axial and vitreal elongation as predicted by the RIDE model. Occluder removal and return to normal light conditions led to return to more normal upregulation of phototransduction, slowed growth rate, refractive recovery and apparent return towards physiological homeostasis.
Collapse
|
30
|
Wang WY, Chen C, Chang J, Chien L, Shih YF, Lin LLK, Pang CP, Wang IJ. Pharmacotherapeutic candidates for myopia: A review. Biomed Pharmacother 2021; 133:111092. [PMID: 33378986 DOI: 10.1016/j.biopha.2020.111092] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/24/2020] [Accepted: 11/28/2020] [Indexed: 01/11/2023] Open
Abstract
This review provides insights into the mechanism underlying the pathogenesis of myopia and potential targets for clinical intervention. Although the etiology of myopia involves both environmental and genetic factors, recent evidence has suggested that the prevalence and severity of myopia appears to be affected more by environmental factors. Current pharmacotherapeutics are aimed at inhibiting environmentally induced changes in visual input and subsequent changes in signaling pathways during myopia pathogenesis and progression. Recent studies on animal models of myopia have revealed specific molecules potentially involved in the regulation of eye development. Among them, the dopamine receptor plays a critical role in controlling myopia. Subsequent studies have reported pharmacotherapeutic treatments to control myopia progression. In particular, atropine treatment yielded favorable outcomes and has been extensively used; however, current studies are aimed at optimizing its efficacy and confirming its safety. Furthermore, future studies are required to assess the efficacy of combinatorial use of low-dose atropine and contact lenses or orthokeratology.
Collapse
Affiliation(s)
- Wen-Yi Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Camille Chen
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Justine Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Lillian Chien
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Feng Shih
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Luke L K Lin
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong Eye Hospital, 147K Argyle Street, KLN, Hong Kong, China.
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan; Graduate Institute of Biomedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
31
|
Muralidharan AR, Lança C, Biswas S, Barathi VA, Wan Yu Shermaine L, Seang-Mei S, Milea D, Najjar RP. Light and myopia: from epidemiological studies to neurobiological mechanisms. Ther Adv Ophthalmol 2021; 13:25158414211059246. [PMID: 34988370 PMCID: PMC8721425 DOI: 10.1177/25158414211059246] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Myopia is far beyond its inconvenience and represents a true, highly prevalent, sight-threatening ocular condition, especially in Asia. Without adequate interventions, the current epidemic of myopia is projected to affect 50% of the world population by 2050, becoming the leading cause of irreversible blindness. Although blurred vision, the predominant symptom of myopia, can be improved by contact lenses, glasses or refractive surgery, corrected myopia, particularly high myopia, still carries the risk of secondary blinding complications such as glaucoma, myopic maculopathy and retinal detachment, prompting the need for prevention. Epidemiological studies have reported an association between outdoor time and myopia prevention in children. The protective effect of time spent outdoors could be due to the unique characteristics (intensity, spectral distribution, temporal pattern, etc.) of sunlight that are lacking in artificial lighting. Concomitantly, studies in animal models have highlighted the efficacy of light and its components in delaying or even stopping the development of myopia and endeavoured to elucidate possible mechanisms involved in this process. In this narrative review, we (1) summarize the current knowledge concerning light modulation of ocular growth and refractive error development based on studies in human and animal models, (2) summarize potential neurobiological mechanisms involved in the effects of light on ocular growth and emmetropization and (3) highlight a potential pathway for the translational development of noninvasive light-therapy strategies for myopia prevention in children.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dan Milea
- Singapore Eye Research Institute, Singapore
| | - Raymond P Najjar
- Visual Neurosciences Group, Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore 169856
| |
Collapse
|
32
|
Abstract
Myopia, also known as short-sightedness or near-sightedness, is a very common condition that typically starts in childhood. Severe forms of myopia (pathologic myopia) are associated with a risk of other associated ophthalmic problems. This disorder affects all populations and is reaching epidemic proportions in East Asia, although there are differences in prevalence between countries. Myopia is caused by both environmental and genetic risk factors. A range of myopia management and control strategies are available that can treat this condition, but it is clear that understanding the factors involved in delaying myopia onset and slowing its progression will be key to reducing the rapid rise in its global prevalence. To achieve this goal, improved data collection using wearable technology, in combination with collection and assessment of data on demographic, genetic and environmental risk factors and with artificial intelligence are needed. Improved public health strategies focusing on early detection or prevention combined with additional effective therapeutic interventions to limit myopia progression are also needed.
Collapse
|
33
|
Khanal S, Rathod SN, Phillips JR. The acute effect of atropine eye drops on the human full-field electroretinogram. Doc Ophthalmol 2020; 142:315-328. [PMID: 33231734 DOI: 10.1007/s10633-020-09806-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/17/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Atropine eye drops are a common and effective treatment for slowing myopia progression, but the site and mode of action of atropine in controlling myopia are unclear. We investigated the early retinal sites of action of atropine by examining its effects on the human full-field electroretinogram (ffERG). METHOD Baseline ffERGs were recorded in both eyes of 24 healthy subjects (mean ± SD: 21.0 ± 2.3 years; spherical equivalent refraction, range: + 1.63 to - 0.75 D) using 6 standard ISCEV protocols, 30 min after bilateral pupil dilation with 1% Tropicamide. Atropine (1 drop, 0.1%) was then instilled into the non-dominant eye. 24 h later, ffERGs were again recorded in both eyes. Ratios (post-atropine: pre-atropine) of dark-adapted (DA) and light-adapted (LA) ffERGs were compared between atropine-treated and control eyes using multivariate repeated measures general linear models. RESULTS Atropine-treated eyes responded with 14% lower DA3.0 OP (oscillatory potential) amplitude (p = 0.003) and 4% delay in the DA10.0 a-wave peak time (p = 0.00099) compared with control eyes. Amplitudes and peak times were not different between atropine-treated and control eyes for DA0.01, LA3.0, and LA3.0 flicker ERGs. While atropine caused a small (1.26 mm2, p = 0.03) extra increase in pupil area in the treated eye, atropine-induced changes in ffERG responses bore no relationship with changes in pupil area (R2 = 2-5%, p > 0.05). CONCLUSIONS The observed changes in oscillatory potentials corroborate previous findings that atropine affects neural activity in the inner retina. However, observed changes to the a-wave suggest that atropine also affects activity in photoreceptors.
Collapse
Affiliation(s)
- Safal Khanal
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, Auckland, New Zealand.,School of Optometry, The University of Alabama At Birmingham, Birmingham, AL, 35294, USA
| | - Sachi Nitinkumar Rathod
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, Auckland, New Zealand
| | - John R Phillips
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, Auckland, New Zealand. .,Department of Optometry, Asia University, Taichung, Taiwan.
| |
Collapse
|
34
|
Seifert M, Baden T, Osorio D. The retinal basis of vision in chicken. Semin Cell Dev Biol 2020; 106:106-115. [PMID: 32295724 DOI: 10.1016/j.semcdb.2020.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/31/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022]
Abstract
The Avian retina is far less known than that of mammals such as mouse and macaque, and detailed study is overdue. The chicken (Gallus gallus) has potential as a model, in part because research can build on developmental studies of the eye and nervous system. One can expect differences between bird and mammal retinas simply because whereas most mammals have three types of visual photoreceptor birds normally have six. Spectral pathways and colour vision are of particular interest, because filtering by oil droplets narrows cone spectral sensitivities and birds are probably tetrachromatic. The number of receptor inputs is reflected in the retinal circuitry. The chicken probably has four types of horizontal cell, there are at least 11 types of bipolar cell, often with bi- or tri-stratified axon terminals, and there is a high density of ganglion cells, which make complex connections in the inner plexiform layer. In addition, there is likely to be retinal specialisation, for example chicken photoreceptors and ganglion cells have separate peaks of cell density in the central and dorsal retina, which probably serve different types of behaviour.
Collapse
Affiliation(s)
- M Seifert
- Sussex Neuroscience, School of Life Sciences, University of Sussex, UK.
| | - T Baden
- Sussex Neuroscience, School of Life Sciences, University of Sussex, UK; Institute for Ophthalmic Research, University of Tuebingen, Germany
| | - D Osorio
- Sussex Neuroscience, School of Life Sciences, University of Sussex, UK
| |
Collapse
|
35
|
Labelle-Dumais C, Pyatla G, Paylakhi S, Tolman NG, Hameed S, Seymens Y, Dang E, Mandal AK, Senthil S, Khanna RC, Kabra M, Kaur I, John SWM, Chakrabarti S, Nair KS. Loss of PRSS56 function leads to ocular angle defects and increased susceptibility to high intraocular pressure. Dis Model Mech 2020; 13:dmm042853. [PMID: 32152063 PMCID: PMC7272341 DOI: 10.1242/dmm.042853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 02/26/2020] [Indexed: 12/31/2022] Open
Abstract
Glaucoma is a leading cause of blindness, affecting up to 70 million people worldwide. High intraocular pressure (IOP) is a major risk factor for glaucoma. It is well established that inefficient aqueous humor (AqH) outflow resulting from structural or functional alterations in ocular drainage tissues causes high IOP, but the genes and pathways involved are poorly understood. We previously demonstrated that mutations in the gene encoding the serine protease PRSS56 induces ocular angle closure and high IOP in mice and identified reduced ocular axial length as a potential contributing factor. Here, we show that Prss56-/- mice also exhibit an abnormal iridocorneal angle configuration characterized by a posterior shift of ocular drainage structures relative to the ciliary body and iris. Notably, we show that retina-derived PRSS56 is required between postnatal days 13 and 18 for proper iridocorneal configuration and that abnormal positioning of the ocular drainage tissues is not dependent on ocular size reduction in Prss56-/- mice. Furthermore, we demonstrate that the genetic context modulates the severity of IOP elevation in Prss56 mutant mice and describe a progressive degeneration of ocular drainage tissues that likely contributes to the exacerbation of the high IOP phenotype observed on the C3H/HeJ genetic background. Finally, we identify five rare PRSS56 variants associated with human primary congenital glaucoma, a condition characterized by abnormal development of the ocular drainage structures. Collectively, our findings point to a role for PRSS56 in the development and maintenance of ocular drainage tissues and IOP homeostasis, and provide new insights into glaucoma pathogenesis.
Collapse
Affiliation(s)
| | - Goutham Pyatla
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
- Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | | | - Nicholas G Tolman
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Syed Hameed
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Yusef Seymens
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA
| | - Eric Dang
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA
| | - Anil K Mandal
- Jasti V. Ramanamma Children's Eye Care Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Sirisha Senthil
- Jasti V. Ramanamma Children's Eye Care Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Rohit C Khanna
- Gullapalli Pratibha Rao International Centre for Advancement of Rural Eye Care, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Meha Kabra
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Inderjeet Kaur
- Brien Holden Eye Research Centre, L V Prasad Eye Institute, Hyderabad 500034, India
| | - Simon W M John
- Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | - K Saidas Nair
- Department of Ophthalmology, University of California, San Francisco, CA 94143, USA
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
36
|
Light adaptation in the chick retina: Dopamine, nitric oxide, and gap-junction coupling modulate spatiotemporal contrast sensitivity. Exp Eye Res 2020; 195:108026. [PMID: 32246982 DOI: 10.1016/j.exer.2020.108026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 11/20/2022]
Abstract
Adaptation to changes in ambient light intensity, in retinal cells and circuits, optimizes visual functions. In the retina, light-adaptation results in changes in light-sensitivity and spatiotemporal tuning of ganglion cells. Under light-adapted conditions, contrast sensitivity (CS) of ganglion cells is a bandpass function of spatial frequency; in contrast, dark-adaptation reduces CS, especially at higher spatial frequencies. In this work, we aimed to understand intrinsic neuromodulatory mechanisms that underlie retinal adaptation to changes in ambient light level. Specifically, we investigated how CS is affected by dopamine (DA), nitric oxide (NO), and modifiers of electrical coupling through gap junctions, under different conditions of adapting illumination. Using the optokinetic response as a behavioral readout of direction-selective ganglion cell activity, we characterized the spatial CS of chicks under high- and low-photopic conditions and how it was regulated by DA, NO, and gap-junction uncouplers. We observed that: (1) DA D2R-family agonists and a donor of NO increased CS tested in low-photopic illumination, as if observed in the high-photopic light; whereas (2) removing their effects using either DA antagonists or NO- synthase inhibitors mimicked low-photopic CS; (3) simulation of high-photopic CS by DA agonists was abolished by NO-synthase inhibitors; and (4) selectively blocking coupling via connexin 35/36-containing gap junctions, using a "designer" mimetic peptide, increased CS, as does strong illumination. We conclude that, in the chicken retina: (1) DA and NO induce changes in spatiotemporal processing, similar to those driven by increasing illumination, (2) DA possibly acts through stimulating NO synthesis, and (3) blockade of coupling via gap junctions containing connexin 35/36 also drives a change in retinal CS functions. As a noninvasive method, the optokinetic response can provide rapid, conditional, and reversible assessment of retinal functions when pharmacological reagents are injected into the vitreous humor. Finally, the chick's large eyes, and the many similarities between their adaptational circuit functions and those in mammals such as the mouse, make them a promising model for future retinal research.
Collapse
|
37
|
Mathis U, Feldkaemper M, Wang M, Schaeffel F. Studies on retinal mechanisms possibly related to myopia inhibition by atropine in the chicken. Graefes Arch Clin Exp Ophthalmol 2019; 258:319-333. [PMID: 31879820 DOI: 10.1007/s00417-019-04573-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 11/28/2019] [Accepted: 12/13/2019] [Indexed: 10/25/2022] Open
Abstract
PURPOSE While low-dose atropine eye drops are currently widely used to inhibit myopia development in children, the underlying mechanisms are poorly understood. Therefore, we studied possible retinal mechanisms and receptors that are potentially involved in myopia inhibition by atropine. METHODS A total of 250 μg atropine were intravitreally injected into one eye of 19 chickens, while the fellow eyes received saline and served as controls. After 1 h, 1.5 h, 2 h, 3 h, and 4 h, eyes were prepared for vitreal dopamine (DA) measurements, using high-pressure liquid chromatography with electrochemical detection. Twenty-four animals were kept either in bright light (8500 lx) or standard light (500 lx) after atropine injection for 1.5 h before DA was measured. In 10 chickens, the α2A-adrenoreceptor (α2A-ADR) agonists brimonidine and clonidine were intravitreally injected into one eye, the fellow eye served as control, and vitreal DA content was measured after 1.5 h. In 6 chickens, immunohistochemical analyses were performed 1.5 h after atropine injection. RESULTS Vitreal DA levels increased after a single intravitreal atropine injection, with a peak difference between both eyes after 1.97 h. DA was also enhanced in fellow eyes, suggesting a systemic action of intravitreally administered atropine. Bright light and atropine (which both inhibit myopia) had additive effects on DA release. Quantitative immunolabelling showed that atropine heavily stimulated retinal activity markers ZENK and c-Fos in cells of the inner nuclear layer. Since atropine was recently found to also bind to α2A-ADRs at doses where it can inhibit myopia, their retinal localization was studied. In amacrine cells, α2A-ADRs were colocalized with tyrosine hydroxylase (TH), glucagon, and nitric oxide synthase, peptides known to play a role in myopia development in chickens. Intravitreal atropine injection reduced the number of neurons that were double-labelled for TH and α2A-ADR. α2A-ADR agonists clonidine and brimonidine (which were also found by other authors to inhibit myopia) severely reduced vitreal DA content in both injected and fellow eyes, compared to eyes of untreated chicks. CONCLUSIONS Merging our results with published data, it can be concluded that both muscarinic and α2A-adrenergic receptors are expressed on dopaminergic neurons and both atropine and α2A-ADR antagonists stimulate DA release whereas α2A-ADR agonists strongly suppress its release. Stimulation of DA by atropine was enhanced by bright light. Results are in line with the hypothesis that inhibition of deprivation myopia is correlated with DA stimulation, as long as no toxicity is involved.
Collapse
Affiliation(s)
- Ute Mathis
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany
| | - Marita Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany
| | - Min Wang
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany.
| |
Collapse
|
38
|
Mori K, Kurihara T, Jiang X, Ikeda SI, Ishida A, Torii H, Tsubota K. Effects of Hyperoxia on the Refraction in Murine Neonatal and Adult Models. Int J Mol Sci 2019; 20:ijms20236014. [PMID: 31795325 PMCID: PMC6928741 DOI: 10.3390/ijms20236014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/22/2019] [Accepted: 11/27/2019] [Indexed: 02/01/2023] Open
Abstract
Whether hyperoxia affects the refraction in neonatal and adult mice is unknown. The mice exposed to 85% oxygen at postnatal 8 days (P8d) for 3 days and the mice exposed to normal air were assigned to the neonatal hyperoxia and normoxia groups, respectively. The refraction, the corneal curvature radius (CR) and the axial length (AL) were measured at P30d and P47d. Postnatal 6 weeks (P6w) adult mice were divided into the adult hyperoxia and normoxia groups. These parameters were measured before oxygen exposure, after 1 and 6 weeks, and every 7 weeks. The lens elasticity was measured at P7w and P26w by enucleation. The neonatal hyperoxia group showed a significantly larger myopic change than the neonatal normoxia group (P47d −6.56 ± 5.89 D, +4.11 ± 2.02 D, p < 0.001), whereas the changes in AL were not significantly different (P47d, 3.31 ± 0.04 mm, 3.31 ± 0.05 mm, p = 0.852). The adult hyperoxia group also showed a significantly larger myopic change (P12w, −7.20 ± 4.09 D, +7.52 ± 2.54 D, p < 0.001). The AL did not show significant difference (P12w, 3.44 ± 0.03 mm, 3.43 ± 0.01 mm, p = 0.545); however, the CR in the adult hyperoxia group was significantly smaller than the adult normoxia group (P12w, 1.44 ± 0.03 mm, 1.50 ± 0.03 mm, p = 0.003). In conclusion, hyperoxia was demonstrated to induce myopic shift both in neonatal and adult mice, which was attributed to the change in the CR rather than the AL. Elucidation of the mechanisms of hyperoxia and the application of this result to humans should be carried out in future studies.
Collapse
Affiliation(s)
- Kiwako Mori
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Correspondence: (T.K.); (K.T.); Tel.: +81-3-5363-3204 (T.K.); +81-3-5363-3269 (K.T.)
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shin-ichi Ikeda
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ayako Ishida
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hidemasa Torii
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.M.); (X.J.); (S.-i.I.); (A.I.); (H.T.)
- Tsubota Laboratory, Inc., Keio University Shinanomachi Campus 2-5F, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Correspondence: (T.K.); (K.T.); Tel.: +81-3-5363-3204 (T.K.); +81-3-5363-3269 (K.T.)
| |
Collapse
|
39
|
Tkatchenko TV, Tkatchenko AV. Pharmacogenomic Approach to Antimyopia Drug Development: Pathways Lead the Way. Trends Pharmacol Sci 2019; 40:833-852. [PMID: 31676152 DOI: 10.1016/j.tips.2019.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022]
Abstract
Myopia is the most common eye disorder in the world which is caused by a mismatch between the optical power of the eye and its excessively long axial length. Recent studies revealed that the regulation of the axial length of the eye occurs via a complex signaling cascade, which originates in the retina and propagates across all ocular tissues to the sclera. The complexity of this regulatory cascade has made it particularly difficult to develop effective antimyopia drugs. The current pharmacological treatment options for myopia are limited to atropine and 7-methylxanthine, which have either significant adverse effects or low efficacy. In this review, we focus on the recent advances in genome-wide studies of the signaling pathways underlying myopia development and discuss the potential of systems genetics and pharmacogenomic approaches for the development of antimyopia drugs.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
40
|
Mori K, Torii H, Fujimoto S, Jiang X, Ikeda SI, Yotsukura E, Koh S, Kurihara T, Nishida K, Tsubota K. The Effect of Dietary Supplementation of Crocetin for Myopia Control in Children: A Randomized Clinical Trial. J Clin Med 2019; 8:jcm8081179. [PMID: 31394821 PMCID: PMC6724222 DOI: 10.3390/jcm8081179] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
The prevalence of myopia has been increasing in recent years. The natural carotenoid crocetin has been reported to suppress experimental myopia in mice. We evaluated the effects of crocetin on myopia suppression in children. A multicenter randomized double-blind placebo-controlled clinical trial was performed with 69 participants aged 6 to 12 years, whose cycloplegic spherical equivalent refractions (SER) were between -1.5 and -4.5 diopter (D). The participants were randomized to receive either a placebo or crocetin and followed up for 24 weeks. Axial length (AL) elongation and changes in SER were evaluated for 24 weeks. Both written informed assent from the participants and written informed consent from legal guardians were obtained in this study because the selection criteria of this trial included children aged between 6 and 12 years old. This trial was approved by the institutional review boards. A mixed-effects model was used for analysis, using both eyes. Two participants dropped out and 67 children completed this trial. The change in SER in the placebo group, -0.41 ± 0.05 D (mean ± standard deviation), was significantly more myopic compared to that in the crocetin group, -0.33 ± 0.05 D (p = 0.049). The AL elongation in the placebo group, 0.21 ± 0.02 mm, was significantly bigger than that in the crocetin group, 0.18 ± 0.02 mm (p = 0.046). In conclusion, dietary crocetin may have a suppressive effect on myopia progression in children, but large-scale studies are required in order to confirm this effect.
Collapse
Affiliation(s)
- Kiwako Mori
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hidemasa Torii
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Satoko Fujimoto
- Department of Ophthalmology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shin-Ichi Ikeda
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Erisa Yotsukura
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shizuka Koh
- Department of Ophthalmology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
- Laboratory of Photobiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
41
|
Kato M, Sato K, Habuta M, Fujita H, Bando T, Morizane Y, Shiraga F, Miyaishi S, Ohuchi H. Localization of the ultraviolet-sensor Opn5m and its effect on myopia-related gene expression in the late-embryonic chick eye. Biochem Biophys Rep 2019; 19:100665. [PMID: 31463372 PMCID: PMC6709407 DOI: 10.1016/j.bbrep.2019.100665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/24/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Abstract
Recent studies show that exposure to ultraviolet (UV) light suppresses ocular elongation, which causes myopia development. However, the specific mechanisms of this process have not been elucidated. A UV-sensor, Opsin 5 (Opn5) mRNA was shown to be present in extraretinal tissues. To test the possibility that UV-signals mediated by Opn5 would have a direct effect on the outer connective tissues of the eye, we first examined the expression patterns of a mammalian type Opn5 (Opn5m) in the late-embryonic chicken eye. Quantitative PCR showed Opn5m mRNA expression in the cornea and sclera. The anti-Opn5m antibody stained a small subset of cells in the corneal stroma and fibrous sclera. We next assessed the effect of UV-A (375 nm) irradiation on the chicken fibroblast cell line DF-1 overexpressing chicken Opn5m. UV-A irradiation for 30 min significantly increased the expression of Early growth response 1 (Egr1), known as an immediate early responsive gene, and of Matrix metalloproteinase 2 (Mmp2) in the presence of retinal chromophore 11-cis-retinal. In contrast, expression of Transforming growth factor beta 2 and Tissue inhibitor of metalloproteinase 2 was not significantly altered. These results indicate that UV-A absorption by Opn5m can upregulate the expression levels of Egr1 and Mmp2 in non-neuronal, fibroblasts. Taken together with the presence of Opn5m in the cornea and sclera, it is suggested that UV-A signaling mediated by Opn5 in the extraretinal ocular tissues could influence directly the outer connective tissues of the chicken late-embryonic eye. Opsin 5 (Opn5) is a non-visual ultraviolet-A (UV-A) absorbing photopigment. We found an Opn5 (Opn5m) is present in cornea and sclera of late-embryonic chick. UV-A absorption by Opn5m upregulated Egr1 and Mmp2 expression in chick fibroblasts. UV-A signaling via Opn5m may have a direct effect on the ocular fibroblasts.
Collapse
Key Words
- Chicken
- Egr1
- Egr1, Early growth response 1
- Fibroblasts
- Gapdh, Glyceraldehyde-3-phosphate dehydrogenase
- MAP kinase, mitogen-activated protein kinase
- Mmp2
- Mmp2, Matrix metalloproteinase 2
- Opn5, Opsin 5
- Opn5m, mammalian type Opn5
- Opsin 5
- Tgfb2, Transforming growth factor beta 2
- Timp2, Tissue inhibitor of metalloproteinase 2
- UV, ultraviolet
- UV-A, ultraviolet-A
- UV-Absorbing pigment
- cAMP, cyclic adenosine monophosphate
- qPCR, quantitative polymerase chain reaction
Collapse
Affiliation(s)
- Mutsuko Kato
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Keita Sato
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Munenori Habuta
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hirofumi Fujita
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Tetsuya Bando
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Yuki Morizane
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Fumio Shiraga
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Satoru Miyaishi
- Department of Legal Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Hideyo Ohuchi
- Department of Cytology and Histology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| |
Collapse
|
42
|
Tang SM, Li FF, Lu SY, Kam KW, Tam POS, Tham CC, Pang CP, Yam JCS, Chen LJ. Association of the ZC3H11B, ZFHX1B and SNTB1 genes with myopia of different severities. Br J Ophthalmol 2019; 104:1472-1476. [PMID: 31300455 DOI: 10.1136/bjophthalmol-2019-314203] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/30/2019] [Accepted: 06/27/2019] [Indexed: 11/03/2022]
Abstract
OBJECTIVE To investigate the associations of single-nucleotide polymorphisms (SNPs) in the ZC3H11B, ZFHX1B, VIPR2, SNTB1 and MIPEP genes with severities of myopia in Chinese populations. METHODS Based on previous myopia genome-wide association studies, five SNPs (ZC3H11B rs4373767, ZFHX1B rs13382811, VIPR2 rs2730260, SNTB1 rs7839488 and MIPEP rs9318086) were selected for genotyping in a Chinese cohort of 2079 subjects: 252 extreme myopia, 277 high myopia, 393 moderate myopia, 366 mild myopia and 791 non-myopic controls. Genotyping was performed by TaqMan assays. Allelic frequencies of the SNPs were compared with myopia severities and ophthalmic biometric measurements. RESULTS The risk allele T of ZC3H11B SNP rs4373767 was significantly associated with high myopia (OR=1.39, p=0.007) and extreme myopia (OR=1.34, p=0.013) when compared with controls, whereas ZFHX1B rs13382811 (allele T, OR=1.33, p=0.018) and SNTB1 rs7839488 (allele G, OR=1.71, p=8.44E-05) were significantly associated with extreme myopia only. In contrast, there was no significant association of these SNPs with moderate or mild myopia. When compared with mild myopia, subjects carrying T allele of rs4373767 had a risk of progressing to high myopia (spherical equivalent ≤-6 dioptres) (OR=1.29, p=0.017). Similarly, the T allele of rs13382811 also imposed a significant risk to high myopia (OR=1.36, p=0.007). In quantitative traits analysis, SNPs rs4373767, rs13382811 and rs7839488 were correlated with axial length and refractive errors. CONCLUSIONS We confirmed ZC3H11B as a susceptibility gene for high and extreme myopia, and ZFHX1B and SNTB for extreme myopia in Chinese populations. Instead of myopia onset, these three genes were more likely to impose risks of progressing to high and extreme myopia.
Collapse
Affiliation(s)
- Shu Min Tang
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong.,Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, New Territories, Hong Kong.,Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Fen Fen Li
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Shi Yao Lu
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Ka Wai Kam
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong.,Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, New Territories, Hong Kong
| | - Pancy O S Tam
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Clement C Tham
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Chi Pui Pang
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Jason C S Yam
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong
| | - Li Jia Chen
- Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, New Territories, Hong Kong .,Department of Ophthalmology and Visual Sciences, Prince of Wales Hospital, New Territories, Hong Kong
| |
Collapse
|
43
|
Khanal S, Turnbull PRK, Lee N, Phillips JR. The Effect of Atropine on Human Global Flash mfERG Responses to Retinal Defocus. Invest Ophthalmol Vis Sci 2019; 60:218-225. [PMID: 30641550 DOI: 10.1167/iovs.18-24600] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the action of atropine on global flash multifocal electroretinogram (gmfERG) responses to retinal defocus. Method gmfERG recordings were made monocularly in 19 healthy adults under three lens-imposed defocus conditions (2 diopters myopic, 2 diopters hyperopic, and no defocus) before and 24 hours after instillation of 1 drop of 0.1% atropine. Signals reflecting activity from the outer and inner retina (direct [DC] and induced [IC] components respectively) were analyzed. Responses were grouped into either a central (0°-6°) or peripheral (6°-24°) retinal zone. The gmfERG responses were compared relative to the no defocus, no atropine condition. Results Within the central zone, atropine had no effect on the amplitudes and peak times of DC or IC responses to defocus. For IC responses in the peripheral zone, there was a significant interaction effect of atropine and defocus (F2,36 = 6.050, P = 0.011) with greater post-atropine amplitudes under myopic defocus (mean increase = 15.5%, 95% confidence interval [CI] = 5.6%-25.4%, P = 0.004). Atropine also had a significant main effect of increasing IC peak times (F1,18 = 9.722, P = 0.006). For DC responses, atropine had a significant main effect of increasing DC amplitudes (F1,18 = 7.821, P = 0.012) and peak times (F1,18 = 15.406, P = 0.001) regardless of sign of defocus. Conclusions Our results imply that atropine acts in the inner layers of the peripheral retina to affect neuronal responses to myopic defocus, raising the possibility that atropine may potentiate the effects of myopic defocus in inhibiting eye growth.
Collapse
Affiliation(s)
- Safal Khanal
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, New Zealand
| | - Philip R K Turnbull
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, New Zealand
| | - Nicholas Lee
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, New Zealand
| | - John R Phillips
- Myopia Laboratory, School of Optometry and Vision Science, The University of Auckland, New Zealand.,Department of Optometry, Asia University, Taichung, Taiwan
| |
Collapse
|
44
|
Vutipongsatorn K, Yokoi T, Ohno-Matsui K. Current and emerging pharmaceutical interventions for myopia. Br J Ophthalmol 2019; 103:1539-1548. [DOI: 10.1136/bjophthalmol-2018-313798] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/22/2019] [Accepted: 04/27/2019] [Indexed: 01/09/2023]
Abstract
Myopia is a major cause of visual impairment. Its prevalence is growing steadily, especially in East Asia. Despite the immense disease and economic burden, there are currently no Food and Drug Administration-approved drugs for myopia. This review aims to summarise pharmaceutical interventions of myopia at clinical and preclinical stages in the last decade and discuss challenges for preclinical myopia drugs to progress to clinical trials. Atropine and oral 7-methylxanthine are shown to reduce myopia progression in human studies. The former has been extensively studied and is arguably the most successful medication. However, it has side effects and trials on low-dose atropine are ongoing. Other pharmaceutical agents being investigated at a clinical trial level include ketorolac tromethamine, oral riboflavin and BHVI2 (an experimental drug). Since the pathophysiology of myopia is not fully elucidated, numerous drugs have been tested at the preclinical stage and can be broadly categorised based on the proposed mechanisms of myopisation, namely antimuscarinic, dopaminergic, anti-inflammatory and more. However, several agents were injected intravitreally or subconjunctivally, hindering their progress to human trials. Furthermore, with atropine being the most successful medication available, future preclinical interventions should be studied in combination with atropine to optimise the treatment of myopia.
Collapse
|
45
|
Zhang Y, Phan E, Wildsoet CF. Retinal Defocus and Form-Deprivation Exposure Duration Affects RPE BMP Gene Expression. Sci Rep 2019; 9:7332. [PMID: 31089149 PMCID: PMC6517395 DOI: 10.1038/s41598-019-43574-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 04/23/2019] [Indexed: 11/09/2022] Open
Abstract
In the context of ocular development and eye growth regulation, retinal defocus and/or image contrast appear key variables although the nature of the signal(s) relayed from the retina to the sclera remains poorly understood. Nonetheless, under optimal visual conditions, eye length is brought into alignment with its optical power to achieve approximate emmetropia, through appropriate adjustment to eye growth. The retinal pigment epithelium (RPE), which lies between the retina and choroid/sclera, appears to play a crucial role in this process. In the investigations reported here, we used a chick model system to assess the threshold duration of exposure to lens-imposed defocus and form-deprivation necessary for conversion of evoked retinal signals into changes in BMP gene expression in the RPE. Our study provides evidence for the following: 1) close-loop, optical defocus-guided (negative and positive lenses) bidirectional BMP gene expression regulation, 2) open-loop, form-deprivation (diffusers)-induced down-regulation of BMP gene expression, and 3) early, transient up-regulation of BMP gene expression in response to both types of lens and diffuser applications. The critical exposure for accurately encoding retinal images as biological signals at the level of the RPE is in the order of minutes to hours, depending on the nature of the visual manipulations.
Collapse
Affiliation(s)
- Yan Zhang
- School of Optometry, University of California, Berkeley, Berkeley, CA, USA.
| | - Eileen Phan
- School of Optometry, University of California, Berkeley, Berkeley, CA, USA
| | | |
Collapse
|
46
|
Comparative transcriptome analysis reveals potential evolutionary differences in adaptation of temperature and body shape among four Percidae species. PLoS One 2019; 14:e0215933. [PMID: 31063465 PMCID: PMC6504104 DOI: 10.1371/journal.pone.0215933] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 04/10/2019] [Indexed: 12/18/2022] Open
Abstract
Considering the divergent temperature habitats and morphological traits of four Percidae species: yellow perch (Perca flavescens), Eurasian perch (Perca fluviatilis), pike perch (Sander lucioperca), and ruffe (Gymnocephalus cernua), we stepped into the transcriptome level to discover genes and mechanisms that drive adaptation to different temperature environments and evolution in body shape. Based on 93,566 to 181,246 annotated unigenes of the four species, we identified 1,117 one-to-one orthologous genes and subsequently constructed the phylogenetic trees that are consistent with previous studies. Together with the tree, the ratios of nonsynonymous to synonymous substitutions presented decreased evolutionary rates from the D. rerio branch to the sub-branch clustered by P. flavescens and P. fluviatilis. The specific 93 fast-evolving genes and 57 positively selected genes in P. flavescens, compared with 22 shared fast-evolving genes among P. fluviatilis, G. cernua, and S. lucioperca, showed an intrinsic foundation that ensure its adaptation to the warmer Great Lakes and farther south, especially in functional terms like “Cul4-RING E3 ubiquitin ligase complex.” Meanwhile, the specific 78 fast-evolving genes and 41 positively selected genes in S. lucioperca drew a clear picture of how it evolved to a large and elongated body with camera-type eyes and muscle strength so that it could occupy the highest position in the food web. Overall, our results uncover genetic basis that support evolutionary adaptation of temperature and body shape in four Percid species, and could furthermore assist studies on environmental adaptation in fishes.
Collapse
|
47
|
Kraft C, Leube A, Ohlendorf A, Wahl S. Contrast adaptation appears independent of the longitudinal chromatic aberration of the human eye. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2019; 36:B77-B84. [PMID: 31044963 DOI: 10.1364/josaa.36.000b77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/06/2019] [Indexed: 06/09/2023]
Abstract
As ocular chromatic aberration was suspected to cue contrast adaptation in human vision, the purpose of this study was to investigate contrast adaptation under monochromatic light conditions. Single and complex frequency adaptation stimuli were used, and monochromatic conditions were achieved using band pass filters with short (470±2 nm), medium (530±2 nm), and long (630±2 nm) transmission wavelengths. Post-adaptational contrast sensitivity was shown to be significantly decreased for all wavelength conditions for the single frequency stimulus. A significant difference of contrast adaptation between short and long wavelengths was found. Consistently, adaptation led to a significant decrease in contrast sensitivity for the complex frequency stimulus. To conclude, contrast adaptation under mesopic illumination occurs independently of the longitudinal chromatic aberration of the eye; it can be inferred that this mechanism can be used to distinguish between the sign of optical defocus in poly- and monochromatic light conditions.
Collapse
|
48
|
Troilo D, Smith EL, Nickla DL, Ashby R, Tkatchenko AV, Ostrin LA, Gawne TJ, Pardue MT, Summers JA, Kee CS, Schroedl F, Wahl S, Jones L. IMI - Report on Experimental Models of Emmetropization and Myopia. Invest Ophthalmol Vis Sci 2019; 60:M31-M88. [PMID: 30817827 PMCID: PMC6738517 DOI: 10.1167/iovs.18-25967] [Citation(s) in RCA: 269] [Impact Index Per Article: 44.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 10/20/2018] [Indexed: 11/24/2022] Open
Abstract
The results of many studies in a variety of species have significantly advanced our understanding of the role of visual experience and the mechanisms of postnatal eye growth, and the development of myopia. This paper surveys and reviews the major contributions that experimental studies using animal models have made to our thinking about emmetropization and development of myopia. These studies established important concepts informing our knowledge of the visual regulation of eye growth and refractive development and have transformed treatment strategies for myopia. Several major findings have come from studies of experimental animal models. These include the eye's ability to detect the sign of retinal defocus and undergo compensatory growth, the local retinal control of eye growth, regulatory changes in choroidal thickness, and the identification of components in the biochemistry of eye growth leading to the characterization of signal cascades regulating eye growth and refractive state. Several of these findings provided the proofs of concepts that form the scientific basis of new and effective clinical treatments for controlling myopia progression in humans. Experimental animal models continue to provide new insights into the cellular and molecular mechanisms of eye growth control, including the identification of potential new targets for drug development and future treatments needed to stem the increasing prevalence of myopia and the vision-threatening conditions associated with this disease.
Collapse
Affiliation(s)
- David Troilo
- SUNY College of Optometry, State University of New York, New York, New York, United States
| | - Earl L. Smith
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Debora L. Nickla
- Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States
| | - Regan Ashby
- Health Research Institute, University of Canberra, Canberra, Australia
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Department of Pathology and Cell Biology, Columbia University, New York, New York, United States
| | - Lisa A. Ostrin
- College of Optometry, University of Houston, Houston, Texas, United States
| | - Timothy J. Gawne
- School of Optometry, University of Alabama Birmingham, Birmingham, Alabama, United States
| | - Machelle T. Pardue
- Biomedical Engineering, Georgia Tech College of Engineering, Atlanta, Georgia, United States31
| | - Jody A. Summers
- College of Medicine, University of Oklahoma, Oklahoma City, Oklahoma, United States
| | - Chea-su Kee
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, SAR, China
| | - Falk Schroedl
- Departments of Ophthalmology and Anatomy, Paracelsus Medical University, Salzburg, Austria
| | - Siegfried Wahl
- Institute for Ophthalmic Research, University of Tuebingen, Zeiss Vision Science Laboratory, Tuebingen, Germany
| | - Lyndon Jones
- CORE, School of Optometry and Vision Science, University of Waterloo, Ontario, Canada
| |
Collapse
|
49
|
Mori K, Kurihara T, Miyauchi M, Ishida A, Jiang X, Ikeda SI, Torii H, Tsubota K. Oral crocetin administration suppressed refractive shift and axial elongation in a murine model of lens-induced myopia. Sci Rep 2019; 9:295. [PMID: 30670743 PMCID: PMC6343000 DOI: 10.1038/s41598-018-36576-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 11/23/2018] [Indexed: 11/09/2022] Open
Abstract
Increased global incidence of myopia necessitates establishment of therapeutic approaches against its progression. To explore agents which may control myopia, we screened 207 types of natural compounds and chemical reagents based on an activity of a myopia suppressive factor, early growth response protein 1 (Egr-1) in vitro. Among the candidates, crocetin showed the highest and dose-dependent activation of Egr-1. For in vivo analysis, experimental myopia was induced in 3-week-old C57BL/6 J mice with −30 diopter (D) lenses for 3 weeks. Animals were fed with normal or mixed chow containing 0.003% (n = 19) and 0.03% (n = 7) of crocetin during myopia induction. Refraction and axial length were measured at 3-week-old and the 6-week-old with an infrared photorefractor and a SD-OCT system. Compared to controls (n = 14), crocetin administration showed a significant smaller change of refractive errors (−13.62 ± 8.14 vs +0.82 ± 5.81 D for 0.003%, p < 0.01, −2.00 ± 4.52 D for 0.03%, p < 0.01) and axial elongation (0.27 ± 0.03 vs 0.22 ± 0.04 mm for 0.003%, p < 0.01, 0.23 ± 0.05 mm for 0.03%, p < 0.05). These results suggest that a dietary factor crocetin may have a preventive effect against myopia progression.
Collapse
Affiliation(s)
- Kiwako Mori
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Toshihide Kurihara
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan. .,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Maki Miyauchi
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ayako Ishida
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Xiaoyan Jiang
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Shin-Ichi Ikeda
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hidemasa Torii
- Laboratory of Photobiology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.,Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
50
|
Tkatchenko TV, Troilo D, Benavente-Perez A, Tkatchenko AV. Gene expression in response to optical defocus of opposite signs reveals bidirectional mechanism of visually guided eye growth. PLoS Biol 2018; 16:e2006021. [PMID: 30300342 PMCID: PMC6177118 DOI: 10.1371/journal.pbio.2006021] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/30/2018] [Indexed: 01/14/2023] Open
Abstract
Myopia (nearsightedness) is the most common eye disorder, which is rapidly becoming one of the leading causes of vision loss in several parts of the world because of a recent sharp increase in prevalence. Nearwork, which produces hyperopic optical defocus on the retina, has been implicated as one of the environmental risk factors causing myopia in humans. Experimental studies have shown that hyperopic defocus imposed by negative power lenses placed in front of the eye accelerates eye growth and causes myopia, whereas myopic defocus imposed by positive lenses slows eye growth and produces a compensatory hyperopic shift in refractive state. The balance between these two optical signals is thought to regulate refractive eye development; however, the ability of the retina to recognize the sign of optical defocus and the composition of molecular signaling pathways guiding emmetropization are the subjects of intense investigation and debate. We found that the retina can readily distinguish between imposed myopic and hyperopic defocus, and identified key signaling pathways underlying retinal response to the defocus of different signs. Comparison of retinal transcriptomes in common marmosets exposed to either myopic or hyperopic defocus for 10 days or 5 weeks revealed that the primate retina responds to defocus of different signs by activation or suppression of largely distinct pathways. We also found that 29 genes differentially expressed in the marmoset retina in response to imposed defocus are localized within human myopia quantitative trait loci (QTLs), suggesting functional overlap between genes differentially expressed in the marmoset retina upon exposure to optical defocus and genes causing myopia in humans. These findings identify retinal pathways involved in the development of myopia, as well as potential new strategies for its treatment. The worldwide prevalence of myopia is predicted to increase from the current 23% to about 50% in the next three decades. Although much effort has been directed towards elucidating the mechanisms underlying refractive eye development and myopia, treatment options for myopia are mostly limited to optical correction, which does not prevent progression of myopia nor the pathological blinding complications often associated with the disease. Several experimental optics-based treatments have had only limited effect on myopia progression, and currently available drug treatments are limited and the mechanisms of action are not well understood. The development of safe and effective pharmacological treatments for myopia is urgently needed to prevent the impending myopia epidemic. The main obstacles that prevent the development of anti-myopia drugs are the uncertainties regarding the mechanisms controlling eye growth and optical development, including the molecular signaling pathways underlying it. In this study, we show that, contrary to the conventional thinking that myopic and hyperopic defocus trigger opposite changes in the same genes and pathways to guide postnatal eye growth, defocus of opposite signs affect eye growth via largely distinct retinal pathways. Knowing that myopic and hyperopic defocus signals drive eye growth in opposite directions and propagate via different pathways provides a framework for the development of new anti-myopia drugs. Myopia can be controlled pharmacologically by stimulating pathways underlying the retinal response to positive lenses and/or by suppressing pathways underlying the retinal response to negative lenses.
Collapse
Affiliation(s)
- Tatiana V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, New York, United States of America
| | - David Troilo
- College of Optometry, State University of New York, New York, New York, United States of America
| | | | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|