1
|
Li Y, Wong D, Sreng S, Chung J, Toh A, Yuan H, Eppenberger LS, Leow C, Ting D, Liu N, Schmetterer L, Saw SM, Jonas JB, Chia A, Ang M. Effect of childhood atropine treatment on adult choroidal thickness using sequential deep learning-enabled segmentation. Asia Pac J Ophthalmol (Phila) 2024; 13:100107. [PMID: 39378966 DOI: 10.1016/j.apjo.2024.100107] [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: 07/23/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024] Open
Abstract
PURPOSE To describe choroidal thickness measurements using a sequential deep learning segmentation in adults who received childhood atropine treatment for myopia control. DESIGN Prospective, observational study. METHODS Choroidal thickness was measured by swept-source optical coherence tomography in adults who received childhood atropine, segmented using a sequential deep learning approach. RESULTS Of 422 eyes, 94 (22.3 %) had no previous exposure to atropine treatment, while 328 (77.7 %) had received topical atropine during childhood. After adjusting for age, sex, and axial length, childhood atropine exposure was associated with a thicker choroid by 32.1 μm (95 % CI, 9.2-55.0; P = 0.006) in the inner inferior, 23.5 μm (95 % CI, 1.9-45.1; P = 0.03) in the outer inferior, 21.8 μm (95 % CI, 0.76-42.9; P = 0.04) in the inner nasal, and 21.8 μm (95 % CI, 2.6-41.0; P = 0.03) in the outer nasal. Multivariable analysis, adjusted for age, sex, atropine use, and axial length, showed an independent association between central subfield choroidal thickness and the incidence of tessellated fundus (P < 0.001; OR, 0.97; 95 % CI, 0.96-0.98). CONCLUSIONS This study demonstrated that short-term (2-4 years) atropine treatment during childhood was associated with an increase in choroidal thickness of 20-40 μm in adulthood (10-20 years later), after adjusting for age, sex, and axial length. We also observed an independent association between eyes with thicker central choroidal measurements and reduced incidence of tessellated fundus. Our study suggests that childhood exposure to atropine treatment may affect choroidal thickness in adulthood.
Collapse
Affiliation(s)
- Yong Li
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Damon Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Syna Sreng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore
| | - Joey Chung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Angeline Toh
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Han Yuan
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Leila Sara Eppenberger
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Cheryl Leow
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Daniel Ting
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore; Byers Eye Institute, Sandford University, Palo Alto, CA, USA
| | - Nan Liu
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore; SERI-NTU Advanced Ocular Engineering (STANCE), Singapore, Singapore; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore; Department of Clinical Pharmacology, Medical University Vienna, Vienna, Austria; Center for Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Seang-Mei Saw
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Jost B Jonas
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland; Privatpraxis Prof Jonas and Dr Panda-Jonas, Heidelberg, Germany
| | - Audrey Chia
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Marcus Ang
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
2
|
Zhang C, Jiao Q, Wang M, Zhu Z, Zhao J, Zheng Y. High correlated color temperature artificial lighting impairs retinal pigment epithelium integrity and chloride ion transport: A potential mechanism for choroidal thinning. Biochem Biophys Res Commun 2024; 718:150078. [PMID: 38735140 DOI: 10.1016/j.bbrc.2024.150078] [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: 03/21/2024] [Revised: 04/04/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
Among the environmental factors contributing to myopia, the role of correlated color temperature (CCT) of ambient light emerges as a key element warranting in-depth investigation. The choroid, a highly vascularized and dynamic structure, often undergoes thinning during the progression of myopia, though the precise mechanism remains elusive. The retinal pigment epithelium (RPE), the outermost layer of the retina, plays a pivotal role in regulating the transport of ion and fluid between the subretinal space and the choroid. A hypothesis suggests that variations in choroidal thickness (ChT) may be modulated by transepithelial fluid movement across the RPE. Our experimental results demonstrate that high CCT illumination significantly compromised the integrity of tight junctions in the RPE and disrupted chloride ion transport. This functional impairment of the RPE may lead to a reduction in fluid transfer across the RPE, consequently resulting in choroidal thinning and potentially accelerating axial elongation. Our findings provide support for the crucial role of the RPE in regulating ChT. Furthermore, we emphasize the potential hazards posed by high CCT artificial illumination on the RPE, the choroid, and refractive development, underscoring the importance of developing eye-friendly artificial light sources to aid in the prevention and control of myopia.
Collapse
Affiliation(s)
- Chenchen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Qing Jiao
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Mingxuan Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Zhe Zhu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, 266000, PR China
| | - Jing Zhao
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Yajuan Zheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, PR China.
| |
Collapse
|
3
|
Schaeffel F, Wildsoet CF. Red light therapy for myopia: Merits, risks and questions. Ophthalmic Physiol Opt 2024; 44:801-807. [PMID: 38563650 DOI: 10.1111/opo.13306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Affiliation(s)
- Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany
| | - Christine F Wildsoet
- Herbert Wertheim School of Optometry and Vision Science, University California Berkeley, Berkeley, California, USA
- School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
4
|
Thomson K, Karouta C, Weber D, Hoffmann N, Morgan I, Kelly T, Ashby R. The role of the serotonergic system in atropine's anti-myopic effects. Biomed Pharmacother 2023; 167:115542. [PMID: 37742601 DOI: 10.1016/j.biopha.2023.115542] [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: 07/13/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
The muscarinic cholinergic antagonist atropine is the most widely used pharmacological treatment for the visual disorder myopia (short-sightedness), the leading cause of low-vision worldwide. This study sought to better define the mechanism by which atropine inhibits myopic growth. Although classified as a muscarinic-cholinergic antagonist, atropine has been found to bind and modulate the activity of several non-cholinergic systems (e.g., serotonin). Thus, this study investigated whether the serotonergic system could underly atropine's anti-myopic effects. Using a chick model of myopia, we report that atropine's growth-inhibitory effects can be attenuated by pharmacological stimulation of the serotonin system. This may suggest that atropine can slow the development of myopia through inhibiting serotonergic receptor activity. We also observed that pharmacological antagonism of serotonergic receptors inhibits the development of experimental myopia in a dose-dependent manner, further demonstrating that modulation of serotonergic receptor activity can alter ocular growth rates. Finally, we found that neither experimental myopia, nor atropine treatment, induced a significant change in retinal serotonergic output (i.e., synthesis, transport, release and catabolism). This may suggest that, although myopic growth can be inhibited through modulation of serotonergic receptor activity (by atropine or serotonergic antagonists), this does not require a change in serotonin levels. These findings regarding a serotonergic mechanism for atropine may have significant ramifications for the treatment of human myopia. This includes assessing the use of atropine in patients who are also undergoing treatment to upregulate serotonergic signaling (e.g., serotonergic anti-depressants).
Collapse
Affiliation(s)
- Kate Thomson
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Australia.
| | - Cindy Karouta
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Australia
| | - Daniel Weber
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Australia
| | - Nichola Hoffmann
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Australia
| | - Ian Morgan
- Research School of Biology, Australian National University, Australia
| | - Tamsin Kelly
- Faculty of Science and Technology, University of Canberra, Australia
| | - Regan Ashby
- Centre for Research in Therapeutic Solutions, Faculty of Science and Technology, University of Canberra, Australia; Research School of Biology, Australian National University, Australia
| |
Collapse
|
5
|
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
|
6
|
Merkley MB, Soriano D, Jones KL, Summers JA. The Effects of Nitric Oxide on Choroidal Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545343. [PMID: 37398322 PMCID: PMC10312785 DOI: 10.1101/2023.06.16.545343] [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
Purpose Nitric oxide (NO) is recognized as an important biological mediator that controls several physiological functions, and evidence is now emerging that this molecule may play a significant role in the postnatal control of ocular growth and myopia development. We therefore sought to understand the role that nitric oxide plays in visually-guided ocular growth in order to gain insight into the underlying mechanisms of this process. Methods Choroids were incubated in organ culture in the presence of the NO donor, PAPA-NONOate (1.5 mM). Following RNA extraction, bulk RNA-seq was used to quantify and compare choroidal gene expression in the presence and absence of PAPA-NONOate. We used bioinformatics to identify enriched canonical pathways, predicted diseases and functions, and regulatory effects of NO in the choroid. Results Upon treatment of normal chick choroids with the NO donor, PAPA-NONOate, we identified a total of 837 differentially expressed genes (259 upregulated genes, 578 down-regulated genes) compared with untreated controls. Among these, the top five upregulated genes were LSMEM1, STEAP4, HSPB9, and CCL19, and the top five down-regulated genes were CDCA3, SMC2, a novel gene (ENSALGALG00000050836), an uncharacterized gene (LOC107054158), and SPAG5. Bioinformatics predicted that NO treatment will activate pathways involved in cell and organismal death, necrosis, and cardiovascular system development, and inhibit pathways involved in cell proliferation, cell movement, and gene expression. Conclusions The findings reported herein may provide insight into possible effects of NO in the choroid during visually regulated eye growth, and help to identify targeted therapies for the treatment of myopia and other ocular diseases.
Collapse
Affiliation(s)
- Makenzie B Merkley
- Department of Biology, University of Oklahoma, Norman, Oklahoma, 73019, United States
| | - Diana Soriano
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| | | | - Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, United States
| |
Collapse
|
7
|
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
|
8
|
Ostrin LA, Harb E, Nickla DL, Read SA, Alonso-Caneiro D, Schroedl F, Kaser-Eichberger A, Zhou X, Wildsoet CF. IMI-The Dynamic Choroid: New Insights, Challenges, and Potential Significance for Human Myopia. Invest Ophthalmol Vis Sci 2023; 64:4. [PMID: 37126359 PMCID: PMC10153586 DOI: 10.1167/iovs.64.6.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 02/07/2023] [Indexed: 05/02/2023] Open
Abstract
The choroid is the richly vascular layer of the eye located between the sclera and Bruch's membrane. Early studies in animals, as well as more recent studies in humans, have demonstrated that the choroid is a dynamic, multifunctional structure, with its thickness directly and indirectly subject to modulation by a variety of physiologic and visual stimuli. In this review, the anatomy and function of the choroid are summarized and links between the choroid, eye growth regulation, and myopia, as demonstrated in animal models, discussed. Methods for quantifying choroidal thickness in the human eye and associated challenges are described, the literature examining choroidal changes in response to various visual stimuli and refractive error-related differences are summarized, and the potential implications of the latter for myopia are considered. This review also allowed for the reexamination of the hypothesis that short-term changes in choroidal thickness induced by pharmacologic, optical, or environmental stimuli are predictive of future long-term changes in axial elongation, and the speculation that short-term choroidal thickening can be used as a biomarker of treatment efficacy for myopia control therapies, with the general conclusion that current evidence is not sufficient.
Collapse
Affiliation(s)
- Lisa A Ostrin
- University of Houston College of Optometry, Houston, Texas, United States
| | - Elise Harb
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, California, United States
| | - Debora L Nickla
- Department of Biomedical Sciences and Disease, New England College of Optometry, Boston, Massachusetts, United States
| | - Scott A Read
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - David Alonso-Caneiro
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Falk Schroedl
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology-Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Alexandra Kaser-Eichberger
- Center for Anatomy and Cell Biology, Institute of Anatomy and Cell Biology-Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Xiangtian Zhou
- Eye Hospital and School of Optometry and Ophthalmology, National Clinical Research Center for Ocular Diseases, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Christine F Wildsoet
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, California, United States
- Centre for Vision and Eye Research, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
9
|
Jiang L, Liu X, Zhou L, Busoy JMF, Khine MT, Dan YS, Ke M, Brennan NA, Catbagan KJV, Schmetterer L, Barathi VA, Hoang QV. Choroidal Thickness in Early Postnatal Guinea Pigs Predicts Subsequent Naturally Occurring and Form-Deprivation Myopia. Invest Ophthalmol Vis Sci 2022; 63:10. [PMID: 36239975 PMCID: PMC9586133 DOI: 10.1167/iovs.63.11.10] [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: 11/24/2022] Open
Abstract
Purpose To identify choroidal characteristics associated with susceptibility to development of naturally occurring and experimentally induced myopia. Methods We compared choroidal properties between pigmented and albino guinea pig (GP) strains. Biometry, cycloplegic refractive error (RE), and eye wall sublayer thickness were measured from 171 GPs at postnatal day (P)6, 14, and 28. Forty-three P14 GPs underwent two-week monocular form-deprivation myopia (FDM). En face images of choroidal vasculature were obtained with a customized swept-source optical coherence tomography. Multivariate regression analyses were performed, with P28 RE as the outcome and P14 choroidal thickness (ChT) as the main predictor variable. Proteomic analysis was performed on choroidal tissue from P14 albino and pigmented GPs. Results At P14, RE was correlated with thickness of the choroid (β = 0.06), sclera (β = 0.12), and retina (β = 0.27; all P < 0.001). P14 ChT was correlated with P28 RE both with (β = 0.06, P = 0.0007) and without FDM (β = 0.05, P = 0.008). Multivariate regression analysis, taking into account FDM (versus physiological growth) and strain, revealed that for every 10-µm greater ChT at P14, P28 RE was 0.50D more positive (P = 0.005, n = 70). En face images of choroidal sublayers showed that albino choroids were relatively underdeveloped, with frequent avascular regions. Consistent with this finding, proteomic analysis suggested abnormalities of the nitric oxide system in the albino GP choroid. Conclusions Current results are consistent with the notion that greater ChT could protect from or delay the onset of myopia, while lower ChT is associated with greater susceptibility to myopia development. The underlying mechanism could be related to dysfunction of the choroidal vascular system.
Collapse
Affiliation(s)
- Liqin Jiang
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Xinyu Liu
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Lei Zhou
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Joanna M Fianza Busoy
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Myo Thu Khine
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Yee Shan Dan
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Mengyuan Ke
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Noel A Brennan
- Johnson & Johnson Vision, Jacksonville, Florida, United States
| | - Karen J V Catbagan
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore
| | - Leopold Schmetterer
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Veluchamy A Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Quan V Hoang
- Singapore Eye Research Institute, Singapore National Eye Centre, Duke-NUS Medical School, Singapore.,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Ophthalmology, Columbia University, New York, New York, United States
| |
Collapse
|
10
|
Chen W, Li L, Feng Q, Li CX, Zhang Y, Li ZW. Quantitative Assessment of the Choroidal Vessel Diameter during the Recovery of Form-Deprivation Myopia in Guinea Pigs. Curr Eye Res 2022; 47:1329-1338. [PMID: 35775095 DOI: 10.1080/02713683.2022.2072897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
PURPOSE The development and recovery (REC) of myopia is associated with changing of choroidal thickness (CT) in the model of guinea pigs. Nitric oxide synthase (NOS) is an enzyme which can affect choroidal vasodilatation. This study wants to investigate the changes of choroidal vessel diameter (CVD) and NOS during the REC of form-deprivation (FD) myopia in guinea pigs. METHODS Forty-eight guinea pigs were randomly assigned to the normal control (NC) group, FD group (FD for 21 d), and four REC groups: REC1/2 group (removal the deprivation and re-exposure to the normal environment for 1/2 d), REC1 group (1 d), REC 2 group (2 d), and REC7 group (7 d). CT was measured by optical coherence tomography (OCT), and CVD of foveal choroid was quantitatively assessed on OCT angiography images using MATLAB software at each time point. NOS in choroid was measured using enzyme-linked immunosorbent assays. Measurements were compared between groups and correlations between CT, CVD, and NOS were assessed using regression analyses. RESULTS CVD and CT in FD group were significantly smaller than in NC group (both p < .05), while the NOS significantly larger (p < .001). When deprivation was removed, CVD and NOS were significantly larger and reached a peak in the REC1 group, while CT reached the peak in the REC2 group, then all gradually decreased, and no significant differences were observed in NC and REC7 group (all p > .05). In the REC and NC groups, there was a significant positive correlation between CVD and NOS (p < .001), CVD and CT (p = .0092), but no correlation was found between NOS and CT (p > .05). CONCLUSIONS This study indicated that the CVD in guinea pigs could be significantly dilated following myopia REC, and this change coincides with changes in NOS and CT.
Collapse
Affiliation(s)
- Wei Chen
- Department of Ophthalmology, Xinjiang Hotan District People's Hospital, Ho tian, PR China.,Department of Ophthalmology, Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, PR China
| | - Li Li
- Department of Ophthalmology, Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, PR China
| | - Qiang Feng
- Department of Ophthalmology, Xinjiang Hotan District People's Hospital, Ho tian, PR China
| | - Chen Xi Li
- School of Precision Instruments and Optoelectronics Engineering, Tianjin University, Tianjin, PR China
| | - Yue Zhang
- Department of Ophthalmology, Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, PR China
| | - Zhi Wei Li
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, PR China
| |
Collapse
|
11
|
Abstract
INTRODUCTION The aim of this article was to comprehensively review the relationship between light exposure and myopia with a focus on the effects of the light wavelength, illuminance, and contrast on the occurrence and progression of myopia. METHODS This review was performed by searching PubMed data sets including research articles and reviews utilizing the terms "light", "myopia", "refractive error", and "illuminance", and the review was concluded in November 2021. Myopia onset and progression were closely linked with emmetropization and hyperopia. To better elucidate the mechanism of myopia, some of the articles that focused on this topic were included. This article is based on previously conducted studies and does not contain any new studies with human participants or animals performed by any of the authors. RESULTS The pathogenesis and prevention of myopia are not completely clear. Studies have provided evidence supporting the idea that light could affect eye growth in three ways. Changing the corresponding conditions will cause changes in the growth rate and mode of the eyes, and preliminary results have shown that FR/NIR (far red/near-infrared) light is effective for myopia in juveniles. CONCLUSION This review discusses the results of studies on the effects of light exposure on myopia with the aims of providing clues and a theoretical basis for the use of light to control the development of myopia and offering new ideas for subsequent studies.
Collapse
|
12
|
Zhu Q, Goto S, Singh S, Torres JA, Wildsoet CF. Daily or Less Frequent Topical 1% Atropine Slows Defocus-Induced Myopia Progression in Contact Lens-Wearing Guinea Pigs. Transl Vis Sci Technol 2022; 11:26. [PMID: 35323888 PMCID: PMC8963669 DOI: 10.1167/tvst.11.3.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Purpose This study compared the efficacy of topical 1% atropine applied daily versus every 3 days for controlling myopia progression in guinea pigs. Methods To induce myopia, pigmented guinea pigs (New Zealand strain, n = 38) wore monocular −10 D rigid gas-permeable (RGP) contact lenses, which were replaced after 3 weeks with −15 diopter (D) contact lenses. Animals were treated with 1% atropine either daily (Atr-QD; n = 12), or every 3 days (Atr-Q3D; n = 11), or with artificial tears (control group; n = 15). Spherical equivalent refractive error (SER) and axial length (AL) data, as well as retinal and choroidal thickness data were collected weekly. Results Whereas mean (±SEM) interocular differences (treated - fellow) in both SER and AL at week 0 (baseline) were similar for all groups, significant differences between the atropine-treated and control groups were evident by week 6 (SER and AL, P < 0.001). The treated eyes of the control group showed relatively more axial elongation and myopia progression than both the Atr-QD and Atr-Q3D groups. Choroidal blood vessel area also decreased over time in the treated eyes of the control group, coupled with choroidal thinning overall, with these changes being attenuated by atropine. Retinal thickness showed a developmental decrease over the treatment period but was unaffected by atropine. Conclusions For this defocus-induced guinea pig model of myopia, application of 1% topical atropine slows myopia progression, even when applied every 3 days. Translational Relevance The results from this study suggest that the frequency of dosing for topical atropine may be reduced from the widely used daily dosing regimen without loss of myopia control efficacy.
Collapse
Affiliation(s)
- Qiurong Zhu
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, USA.,Department of Optometry and Visual Science, West China Hospital of Sichuan University, Sichuan, China
| | - So Goto
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, USA.,Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.,Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Sarah Singh
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, USA
| | - Josue A Torres
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, USA
| | - Christine F Wildsoet
- Herbert Wertheim School Optometry and Vision Science, University of California, Berkeley, California, USA
| |
Collapse
|
13
|
Summers JA, Martinez E. Visually induced changes in cytokine production in the chick choroid. eLife 2021; 10:70608. [PMID: 34608867 PMCID: PMC8612705 DOI: 10.7554/elife.70608] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 10/04/2021] [Indexed: 12/18/2022] Open
Abstract
Postnatal ocular growth is regulated by a vision-dependent mechanism that acts to minimize refractive error through coordinated growth of the ocular tissues. Of great interest is the identification of the chemical signals that control visually guided ocular growth. Here, we provide evidence that the pro-inflammatory cytokine, interleukin-6 (IL-6), may play a pivotal role in the control of ocular growth using a chicken model of myopia. Microarray, real-time RT-qPCR, and ELISA analyses identified IL-6 upregulation in the choroids of chick eyes under two visual conditions that introduce myopic defocus and slow the rate of ocular elongation (recovery from induced myopia and compensation for positive lenses). Intraocular administration of atropine, an agent known to slow ocular elongation, also resulted in an increase in choroidal IL-6 gene expression. Nitric oxide appears to directly or indirectly upregulate choroidal IL-6 gene expression, as administration of the non-specific nitric oxide synthase inhibitor, L-NAME, inhibited choroidal IL-6 gene expression, and application of a nitric oxide donor stimulated IL-6 gene and protein expression in isolated chick choroids. Considering the pleiotropic nature of IL-6 and its involvement in many biological processes, these results suggest that IL-6 may mediate many aspects of the choroidal response in the control of ocular growth.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| | - Elizabeth Martinez
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, United States
| |
Collapse
|
14
|
Sander BP. Prevention of Choroidal Thinning by 0.01% Atropine Administered 24 h Before Exposure to Hyperopic Blur in Young Myopes. J Ocul Pharmacol Ther 2021; 37:510-517. [PMID: 34491840 DOI: 10.1089/jop.2021.0006] [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] [Indexed: 02/03/2023] Open
Abstract
Purpose: To evaluate the persistence of atropine's effect upon choroidal thickness and ocular biometrics and its interaction with hyperopic blur in a population of young adult myopes. Methods: Twenty young (aged 18-35 years) myopic participants with spherical equivalent refractive error of -0.75 to -6.00 D (mean ± SD -2.85 ± 1.64 D) had subfoveal choroidal thickness (SFCT) measurements derived from scans collected from the right eye only with a SD-OCT instrument (Copernicus SOCT-HR) before, as well as 60 min following the introduction of 3 testing conditions: (1) placebo/hyperopic (-3 D) blur, (2) placebo/hyperopic blur one day after administration of 0.01% atropine, and (3) placebo/no blur. Each combination of blur and pharmacological agent was tested on a separate day at approximately the same time of day between 9 am and 2 pm. Results: Repeated measures ANOVA revealed that hyperopic blur and placebo were associated with a decrease in choroidal thickness (mean change: -10.7 ± 2.7 μm, P < 0.001 after 60 min), whereas administration of 0.01% atropine one day before the introduction of hyperopic blur prevented the thinning of the choroid (mean change of +1.1 ± 3.7 μm after 60 min) compared to baseline (both, P > 0.05). There was also no significant difference between the baseline choroidal thickness measurements for any of the conditions tested. Conclusion: Low dose atropine can inhibit signals associated with hyperopic defocus that cause thinning of the choroid for at least 24 h after initial instillation.
Collapse
Affiliation(s)
- Beata P Sander
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia.,Lees and Henschell Optometrists, Kenmore, Australia
| |
Collapse
|
15
|
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 DOI: 10.1016/j.exer.2021.108693] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [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
|
16
|
Tkatchenko TV, Tkatchenko AV. Genetic network regulating visual acuity makes limited contribution to visually guided eye emmetropization. Genomics 2021; 113:2780-2792. [PMID: 34147636 DOI: 10.1016/j.ygeno.2021.06.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 12/13/2022]
Abstract
During postnatal development, the eye undergoes a refinement process whereby optical defocus guides eye growth towards sharp vision in a process of emmetropization. Optical defocus activates a signaling cascade originating in the retina and propagating across the back of the eye to the sclera. Several observations suggest that visual acuity might be important for optical defocus detection and processing in the retina; however, direct experimental evidence supporting or refuting the role of visual acuity in refractive eye development is lacking. Here, we used genome-wide transcriptomics to determine the relative contribution of the retinal genetic network regulating visual acuity to the signaling cascade underlying visually guided eye emmetropization. Our results provide evidence that visual acuity is regulated at the level of molecular signaling in the retina by an extensive genetic network. The genetic network regulating visual acuity makes relatively small contribution to the signaling cascade underlying refractive eye development. This genetic network primarily affects baseline refractive eye development and this influence is primarily facilitated by the biological processes related to melatonin signaling, nitric oxide signaling, phototransduction, synaptic transmission, and dopamine signaling. We also observed that the visual-acuity-related genes associated with the development of human myopia are chiefly involved in light perception and phototransduction. Our results suggest that the visual-acuity-related genetic network primarily contributes to the signaling underlying baseline refractive eye development, whereas its impact on visually guided eye emmetropization is modest.
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
|
17
|
Erdinest N, London N, Ovadia H, Levinger N. Nitric Oxide Interaction with the Eye. Vision (Basel) 2021; 5:29. [PMID: 34207828 PMCID: PMC8293394 DOI: 10.3390/vision5020029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/03/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Nitric oxide (NO) is acknowledged as a vital intercellular messenger in multiple systems in the body. Medicine has focused on its functions and therapeutic applications for decades, especially in cardiovascular and nervous systems, and its role in immunological responses. This review was composed to demonstrate the prevalence of NO in components of the ocular system, including corneal cells and multiple cells in the retina. It discussed NO's assistance during the immune, inflammation and wound-healing processes. NO is identified as a vascular endothelial relaxant that can alter the choroidal blood flow and prompt or suppress vascular changes in age-related macular degeneration and diabetes, as well as the blood supply to the optic nerve, possibly influencing the progression of glaucoma. It will provide a deeper understanding of the role of NO in ocular homeostasis, the delicate balance between overproduction or underproduction and the effect on the processes from aqueous outflow and subsequent intraocular pressure to axial elongation and the development of myopia. This review also recognized the research and investigation of therapies being developed to target the NO complex and treat various ocular diseases.
Collapse
Affiliation(s)
- Nir Erdinest
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (N.E.); (N.L.)
| | | | - Haim Ovadia
- Agnes Ginges, Center for Human Neurogenetics, Department of Neurology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel;
| | - Nadav Levinger
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel; (N.E.); (N.L.)
- Enaim Refractive Surgery Center, Jerusalem 9438307, Israel
| |
Collapse
|
18
|
Ye L, Shi Y, Yin Y, Li S, He J, Zhu J, Xu X. Effects of Atropine Treatment on Choroidal Thickness in Myopic Children. Invest Ophthalmol Vis Sci 2021; 61:15. [PMID: 33320168 PMCID: PMC7745623 DOI: 10.1167/iovs.61.14.15] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To examine the changes in choroidal thickness (ChT) after 6 months of 1% or 0.01% atropine treatment and the independent factors associated with eye elongation. Methods A total of 207 myopic children aged 6 to 12 years were recruited and randomly assigned to groups A and B in a ratio of 1:1. Participants in group A received 1% atropine once a day for 1 week, and then once a week for 23 weeks. Participants in group B received 0.01% atropine once a day for 6 months. ChT and internal axial length (IAL) were measured at baseline, 1 week, 3 months, and 6 months. Results In group A, the ChT significantly increased after a 1-week loading dose of 1% atropine (26 ± 14 µm; P < 0.001) and the magnitude of increase stabilized throughout the following weekly treatment. The internal axial length did not significantly change at the 6-month visit (−0.01 ± 0.11 mm; P = 0.74). In contrast, a decreased ChT (−5 ± 17 µm; P < 0.001) and pronounced eye elongation (0.19 ± 0.12 mm; P < 0.001) were observed in group B after 6 months. Multivariable regression analysis showed that less increase in ChT at the 1-week visit (P = 0.03), younger age (P < 0.001), and presence of peripapillary atrophy (P = 0.001) were significantly associated with greater internal axial length increase over 6 months in group A. Conclusions One percent atropine could increase the ChT, whereas 0.01% atropine caused a decrease in ChT after 6 months of treatment. For participants receiving 1% atropine, the short-term increase in ChT was negatively associated with long-term eye elongation. Younger age and the presence of peripapillary atrophy were found to be risk factors for greater eye elongation.
Collapse
Affiliation(s)
- Luyao Ye
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Ya Shi
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yao Yin
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China
| | - Shanshan Li
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China
| | - Jiangnan He
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China
| | - Jianfeng Zhu
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China
| | - Xun Xu
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai Eye Hospital, Shanghai, China.,Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
19
|
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: 6.0] [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
|
20
|
Najjar RP, Chao De La Barca JM, Barathi VA, Ho CEH, Lock JZ, Muralidharan AR, Tan RKY, Dhand C, Lakshminarayanan R, Reynier P, Milea D. Ocular growth and metabolomics are dependent upon the spectral content of ambient white light. Sci Rep 2021; 11:7586. [PMID: 33828194 PMCID: PMC8026599 DOI: 10.1038/s41598-021-87201-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Myopia results from an excessive axial growth of the eye, causing abnormal projection of remote images in front of the retina. Without adequate interventions, myopia is forecasted to affect 50% of the world population by 2050. Exposure to outdoor light plays a critical role in preventing myopia in children, possibly through the brightness and blue-shifted spectral composition of sunlight, which lacks in artificial indoor lighting. Here, we evaluated the impact of moderate levels of ambient standard white (SW: 233.1 lux, 3900 K) and blue-enriched white (BEW: 223.8 lux, 9700 K) lights on ocular growth and metabolomics in a chicken-model of form-deprivation myopia. Compared to SW light, BEW light decreased aberrant ocular axial elongation and accelerated recovery from form-deprivation. Furthermore, the metabolomic profiles in the vitreous and retinas of recovering form-deprived eyes were distinct from control eyes and were dependent on the spectral content of ambient light. For instance, exposure to BEW light was associated with deep lipid remodeling and metabolic changes related to energy production, cell proliferation, collagen turnover and nitric oxide metabolism. This study provides new insight on light-dependent modulations in ocular growth and metabolomics. If replicable in humans, our findings open new potential avenues for spectrally-tailored light-therapy strategies for myopia.
Collapse
Affiliation(s)
- Raymond P Najjar
- Singapore Eye Research Institute, Singapore, Singapore.
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore.
| | - Juan Manuel Chao De La Barca
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire d'Angers, Angers, France
- Unité Mixte de Recherche MITOVASC, CNRS 6015, INSERM U1083, Université d'Angers, Angers, France
| | - Veluchamy A Barathi
- Singapore Eye Research Institute, Singapore, Singapore
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | | - Royston K Y Tan
- Department of Ocular Bio-Engineering, National University of Singapore, Singapore, Singapore
| | - Chetna Dhand
- Singapore Eye Research Institute, Singapore, Singapore
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal, 462026, India
| | | | - Pascal Reynier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire d'Angers, Angers, France
- Unité Mixte de Recherche MITOVASC, CNRS 6015, INSERM U1083, Université d'Angers, Angers, France
| | - Dan Milea
- Singapore Eye Research Institute, Singapore, Singapore.
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore.
- Singapore National Eye Center, Singapore, Singapore.
| |
Collapse
|
21
|
Ye L, Li S, Shi Y, Yin Y, He J, Zhu J, Xu X. Comparisons of atropine versus cyclopentolate cycloplegia in myopic children. Clin Exp Optom 2021; 104:143-150. [PMID: 32844483 DOI: 10.1111/cxo.13128] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
CLINICAL RELEVANCE In clinical practice, 1% atropine and 1% cyclopentolate are used as cycloplegia agents to diagnose refractive error. The influence of 1% atropine on ocular biometry is obscure, and the impact of 1% cyclopentolate remains controversial. BACKGROUND This study aims to compare the effects of atropine versus cyclopentolate cycloplegia on ocular biometry in myopic children and to determine the sites of action for atropine. METHODS A total of 207 myopic children aged 6-12-years were included in the analysis. All participants underwent comprehensive eye examinations before and after cyclopentolate cycloplegia, after which they were randomly assigned into two groups, A and B, in a ratio of 1:1, to receive 1% or 0.01% atropine, respectively. The treatment was administered once every night for a week. Participants were re-examined one week later. RESULTS Cyclopentolate cycloplegia caused a decrease in choroidal thickness (-3 ± 9 μm, p = 0.001), elongation of axial length (9 ± 16 μm, p < 0.001), loss of lens power (-0.14 ± 0.37 dioptre, p < 0.001), and a hyperopic shift (0.14 ± 0.22 dioptre, p < 0.001) in both groups. However, ocular biometry showed different changes after one-week use of 1% or 0.01% atropine (all p < 0.001). In Group A, choroid thickening (24 ± 13 μm, p < 0.001) and reduced axial length (-30 ± 27 μm, p < 0.001) were observed after atropine cycloplegia, with greater changes in lens power (0.50 ± 0.37 dioptre, p < 0.001) and spherical equivalent (0.52 ± 0.23 dioptre, p < 0.001). Group B showed a slight increase in choroidal thickness following one-week use of 0.01% atropine (6 ± 9 μm, p < 0.001), but other biometric measures showed no significant changes. CONCLUSION Cyclopentolate and atropine cycloplegia have different effects on ocular biometry. Both 1% cyclopentolate cycloplegia and 0.01% atropine resulted in choroidal thickening, indicating that the choroid may be a site of action for atropine.
Collapse
Affiliation(s)
- Luyao Ye
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China.,National Clinical Research Centre for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Centre for Visual Science and Photomedicine, Shanghai Engineering Centre for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shanshan Li
- Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China
| | - Ya Shi
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China.,National Clinical Research Centre for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Centre for Visual Science and Photomedicine, Shanghai Engineering Centre for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Yao Yin
- Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China
| | - Jiangnan He
- Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China
| | - Jianfeng Zhu
- Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Eye Disease Prevention and Treatment Centre, Shanghai Eye Hospital, Shanghai, China.,National Clinical Research Centre for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Centre for Visual Science and Photomedicine, Shanghai Engineering Centre for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
22
|
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: 26] [Impact Index Per Article: 8.7] [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
|
23
|
Prousali E, Dastiridou A, Ziakas N, Androudi S, Mataftsi A. Choroidal thickness and ocular growth in childhood. Surv Ophthalmol 2020; 66:261-275. [PMID: 32634443 DOI: 10.1016/j.survophthal.2020.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
The involvement of the choroid in ocular growth regulation has been postulated in studies showing that refractive errors correlate with alterations in choroidal thickness (ChT). The advent of optical coherence tomography imaging has enabled qualitative and quantitative assessment of the choroid. In children, ChT changes correlate with a number of ocular pathologies, including myopia, retinopathy of prematurity, and amblyopia. We synthesize mechanisms and evidence regarding choroidal thickness variation during childhood. Subfoveal ChT is influenced by a number of factors including age, ethnicity, gender, axial length, and intraocular pressure. Myopic eyes have thinner choroids compared to emmetropic and hyperopic eyes. ChT may in fact serve as a marker of myopic progression, as ChT thinning occurs early during myopic development, but this association has not been established quantitatively. In addition, subfoveal ChT appears thicker in amblyopic eyes, while prematurity and retinopathy of prematurity may be associated with thinner ChT. Overall, both animal models and clinical research indicate that ChT induces or reflects physiological changes in the eye pertaining to ocular growth or maturation.
Collapse
Affiliation(s)
- Efthymia Prousali
- 2(nd) Department of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anna Dastiridou
- 2(nd) Department of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Ziakas
- 2(nd) Department of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sofia Androudi
- Department of Ophthalmology, University of Thessaly, Larissa, Greece
| | - Asimina Mataftsi
- 2(nd) Department of Ophthalmology, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
24
|
Ulaganathan S, Read SA, Collins MJ, Vincent SJ. Daily axial length and choroidal thickness variations in young adults: Associations with light exposure and longitudinal axial length and choroid changes. Exp Eye Res 2019; 189:107850. [PMID: 31639338 DOI: 10.1016/j.exer.2019.107850] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/11/2019] [Accepted: 10/18/2019] [Indexed: 12/24/2022]
Abstract
Evidence from animal studies suggests that the eye's natural diurnal rhythms can be disrupted by altering the light/dark cycle or during refractive error development. Although diurnal variations in axial length (AL) and choroidal thickness are well documented in human eyes, the relationship between ambient light exposure, refractive error progression and diurnal AL and choroidal thickness variations is not well understood. Therefore we examined the association between objective ambient light exposure and daily variations in AL and subfoveal choroidal thickness (SFCT), along with longer-term changes in AL and SFCT over 12 months. Thirty-four young adult emmetropes and myopes had their daily variations (measurements ~ every 3 h from 9 a.m. to 9 p.m.) in AL and SFCT assessed on a weekday and weekend in winter and then six months later in summer. Participants then returned six months later for a single measurement session to determine the longer-term change in AL and SFCT. Personal ambient light exposure was captured in winter and summer using wrist-worn light sensors (Actiwatch-2) worn for 14 days over the same period of time when the diurnal measurements were collected. Linear mixed model analyses revealed significant daily variations in AL and SFCT (each p < 0.05). The mean daily peak to trough difference (amplitude) in AL was significantly greater in myopes (0.020 mm; 95% CI: 0.014-0.026 mm) compared to emmetropes (0.010 mm; 95% CI: 0.005-0.015 mm) (p < 0.01), but the SFCT variations were not significantly different between the refractive groups (p = 0.45). Daily variations in AL were negatively associated with the daily SFCT variations (r = -0.603, p < 0.001). Correlation analyses indicated that the amplitude of daily AL variations was negatively associated with the daily time exposed to bright light (r = -0.511, p = 0.002) and positively associated with the longitudinal AL changes over 12 months (r = 0.381, p = 0.04). There was an inverse association between the longer-term changes in AL and SFCT (r = -0.352, p = 0.002). The daily ocular diurnal variations were not significantly different between weekdays and weekends, or between summer and winter (each p > 0.05). In summary, diurnal variations in AL were higher in amplitude in myopes compared to emmetropes and were also associated with longitudinal changes in AL. These findings suggest that diurnal variations may be associated with longer-term axial eye growth. Time spent in bright light also significantly influenced the amplitude of daily AL variations, with more time exposed to bright light associated with a smaller amplitude of diurnal AL change. Choroidal thickness exhibited an inverse association with the AL changes, implying a potential role for the choroid in eye growth.
Collapse
Affiliation(s)
- Sekar Ulaganathan
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia.
| | - Scott A Read
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia.
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia.
| | - Stephen J Vincent
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia.
| |
Collapse
|
25
|
Zheng H, Tse DY, Tang X, To C, Lam TC. The Interactions Between Bright Light and Competing Defocus During Emmetropization in Chicks. Invest Ophthalmol Vis Sci 2019; 59:2932-2943. [PMID: 30025112 DOI: 10.1167/iovs.17-22973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The environment comprises multiple optical signals that affect eye growth. We aimed to determine if the inhibitory effects of myopic defocus and bright light (BL) against myopia are additive in the presence of the myopia-genic hyperopic defocus. Methods In experiment 1, three groups of 24 chicks each were fitted with the following multizone dual-power lenses (pl): pl/-10 D (50:50 area), +10/-10 D (50:50 area), and +10/-10 D (33:67 area) monocularly for 6 days. Half of each group were raised under normal illumination of 500 lux, 12/12-hour light/dark cycle, whereas the remainder were exposed to 6-hour BL of 40 klx and 6-hour 500 lux during the light cycle. In experiment 2, 38 chicks wore +10/-10 D (33:67 area) lenses monocularly for 8 days and were exposed to one of four light intensities for 6 hours per day-500 lux, 10 klx, 20 klx, or 40 klx-and received 500 lux for the remainder of the light cycle. Results In experiment 1, interocular difference in refractions after 6 days for the three groups were -3.6 D, +2.0 D, and -4.2 D, respectively, under normal light and were -0.9 D, +4.2 D, and +0.67 D under BL, manifesting as a shorter anterior segment and vitreous chamber. In experiment 2, the effect of BL increased with light intensity in the +10/-10 D (33:67) group, with a significant difference in refraction between the 10 klx and 20 klx groups (interocular difference -2.75 ± 2.76 D vs. 1.70 ± 2.40 D, P < 0.01), but plateaued between 20 klx and 40 klx (1.70 ± 2.40 D vs. 1.70 ± 0.35 D, P > 0.05). Conclusions The protective effects of myopic defocus and BL against experimental myopia were additive. The inhibitory effect of BL against myopia was dose dependent at 10 klx and above but plateaued at 20 klx.
Collapse
Affiliation(s)
- Hui Zheng
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China.,School of Medicine, Nankai University, Tianjin, China.,Tianjin Eye Hospital, Tianjin, China
| | - Dennis Y Tse
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xin Tang
- School of Medicine, Nankai University, Tianjin, China.,Tianjin Eye Hospital, Tianjin, China
| | - Chiho To
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| | - Thomas Chuen Lam
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
26
|
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: 231] [Impact Index Per Article: 46.2] [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
|
27
|
Chakraborty R, Yang V, Park HN, Landis EG, Dhakal S, Motz CT, Bergen MA, Iuvone PM, Pardue MT. Lack of cone mediated retinal function increases susceptibility to form-deprivation myopia in mice. Exp Eye Res 2018; 180:226-230. [PMID: 30605665 DOI: 10.1016/j.exer.2018.12.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/18/2018] [Accepted: 12/29/2018] [Indexed: 11/30/2022]
Abstract
Retinal photoreceptors are important in visual signaling for normal eye growth in animals. We used Gnat2cplf3/cplf3 (Gnat2-/-) mice, a genetic mouse model of cone dysfunction to investigate the influence of cone signaling in ocular refractive development and myopia susceptibility in mice. Refractive development under normal visual conditions was measured for Gnat2-/- and age-matched Gnat2+/+ mice, every 2 weeks from 4 to 14 weeks of age. Weekly measurements were performed on a separate cohort of mice that underwent monocular form-deprivation (FD) in the right eye from 4 weeks of age using head-mounted diffusers. Refraction, corneal curvature, and ocular biometrics were obtained using photorefraction, keratometry and optical coherence tomography, respectively. Retinas from FD mice were harvested, and analyzed for dopamine (DA) and 3,4-dihydroxyphenylacetate (DOPAC) using high-performance liquid chromatography. Under normal visual conditions, Gnat2+/+ and Gnat2-/- mice showed similar refractive error, axial length, and corneal radii across development (p > 0.05), indicating no significant effects of the Gnat2 mutation on normal ocular refractive development in mice. Three weeks of FD produced a significantly greater myopic shift in Gnat2-/- mice compared to Gnat2+/+ controls (-5.40 ± 1.33 D vs -2.28 ± 0.28 D, p = 0.042). Neither the Gnat2 mutation nor FD altered retinal levels of DA or DOPAC. Our results indicate that cone pathways needed for high acuity vision in primates are not as critical for normal refractive development in mice, and that both rods and cones contribute to visual signalling pathways needed to respond to FD in mammalian eyes.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, 5001, Adelaide, Australia
| | - Victoria Yang
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA
| | - Han Na Park
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA
| | - Erica G Landis
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA
| | - Susov Dhakal
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA
| | - Cara T Motz
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA
| | - Michael A Bergen
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA; Department of Pharmacology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr, Atlanta, GA, 30332, USA.
| |
Collapse
|
28
|
Li Z, Cui D, Hu Y, Ao S, Zeng J, Yang X. Choroidal thickness and axial length changes in myopic children treated with orthokeratology. Cont Lens Anterior Eye 2017; 40:417-423. [PMID: 28935528 DOI: 10.1016/j.clae.2017.09.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 09/01/2017] [Accepted: 09/11/2017] [Indexed: 12/22/2022]
Abstract
PURPOSE To analyze the change in subfoveal choroidal thickness (SFChT) and its relationship with changes in axial length (AL) in myopic children treated with Orthokeratology (Ortho-k). METHODS Fifty myopic children participated in this study: 29 subjects were treated with Ortho-k lenses and 21 with single vision distance spectacles. The SFChT and ocular biometrics, including AL, were measured at baseline, one month, and six months after lens wear in both groups. RESULTS AL significantly increased in both groups over time. In the Ortho-k group, SFChT also increased; however, there was no significant change in SFChT in the control group over time. At the six-month visit, the magnitude of eye growth was significantly reduced in the Ortho-k group compared to the control group (0.06±0.10mm vs. 0.17±0.10mm, P<0.001). SFChT was significantly thicker in the Ortho-k group compared to the control group at the one-month and six-month visits (15.78±11.37μm vs. -2.98±8.96μm, P<0.001 (one-month visit); 21.03±12.74μm vs. -2.50±14.43μm, P<0.001 (six-month visit)), although there was no significant difference between the two follow-up visits (P=0.102 for the Ortho-k group; P=0.898 for the control group). Changes in the large choroidal vascular layer (LCVL) accounted for the majority of subfoveal choroidal thickening (approximately 77% and 80% at one-month and six-month visits, respectively). CONCLUSION Ortho-k treatment induced significant choroidal thickening and a slowing of eye growth. LCVL thickening accounted for the majority of SFChT thickening. However, its potential mechanism in myopia control requires further investigation.
Collapse
Affiliation(s)
- Zhouyue Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Dongmei Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Sichun Ao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Junwen Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiao Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
29
|
Zhou X, Pardue MT, Iuvone PM, Qu J. Dopamine signaling and myopia development: What are the key challenges. Prog Retin Eye Res 2017; 61:60-71. [PMID: 28602573 DOI: 10.1016/j.preteyeres.2017.06.003] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 01/11/2023]
Abstract
In the face of an "epidemic" increase in myopia over the last decades and myopia prevalence predicted to reach 2.5 billion people by the end of this decade, there is an urgent need to develop effective and safe therapeutic interventions to slow down this "myopia booming" and prevent myopia-related complications and vision loss. Dopamine (DA) is an important neurotransmitter in the retina and mediates diverse functions including retina development, visual signaling, and refractive development. Inspired by the convergence of epidemiological and animal studies in support of the inverse relationship between outdoor activity and risk of developing myopia and by the close biological relationship between light exposure and dopamine release/signaling, we felt it is timely and important to critically review the role of DA in myopia development. This review will revisit several key points of evidence for and against DA mediating light control of myopia: 1) the causal role of extracellular retinal DA levels, 2) the mechanism and action of dopamine D1 and D2 receptors and 3) the roles of cellular/circuit retinal pathways. We examine the experiments that show causation by altering DA, DA receptors and visual pathways using pharmacological, transgenic, or visual environment approaches. Furthermore, we critically evaluate the safety issues of a DA-based treatment strategy and some approaches to address these issues. The review identifies the key questions and challenges in translating basic knowledge on DA signaling and myopia from animal studies into effective pharmacological treatments for myopia in children.
Collapse
Affiliation(s)
- Xiangtian Zhou
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; State Key Laboratory of Ophthalmology, Optometry and Vision Science. 270 Xueyuan Road, Wenzhou, Zhejiang, 325003, China
| | - Machelle T Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA 30332, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Rd, Decatur, GA 30033, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA 30322, United States; Department of Pharmacology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA 30322, United States
| | - Jia Qu
- School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou, Zhejiang 325003, China; State Key Laboratory of Ophthalmology, Optometry and Vision Science. 270 Xueyuan Road, Wenzhou, Zhejiang, 325003, China.
| |
Collapse
|
30
|
Carr BJ, Stell WK. Nitric Oxide (NO) Mediates the Inhibition of Form-Deprivation Myopia by Atropine in Chicks. Sci Rep 2016; 6:9. [PMID: 28442706 PMCID: PMC5431363 DOI: 10.1038/s41598-016-0002-7] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 08/22/2016] [Indexed: 11/30/2022] Open
Abstract
Myopia is the most common childhood refractive disorder. Atropine inhibits myopia progression, but its mechanism is unknown. Here, we show that myopia-prevention by atropine requires production of nitric oxide (NO). Form-deprivation myopia (FDM) was induced in week-old chicks by diffusers over the right eye (OD); the left eye (OS) remained ungoggled. On post-goggling days 1, 3, and 5, OD received intravitreally 20 µL of phosphate-buffered saline (vehicle), or vehicle plus: NO source: L-arginine (L-Arg, 60–6,000 nmol) or sodium nitroprusside (SNP, 10–1,000 nmol); atropine (240 nmol); NO inhibitors: L-NIO or L-NMMA (6 nmol); negative controls: D-Arg (10 µmol) or D-NMMA (6 nmol); or atropine plus L-NIO, L-NMMA, or D-NMMA; OS received vehicle. On day 6 post-goggling, refractive error, axial length, equatorial diameter, and wet weight were measured. Vehicle-injected goggled eyes developed significant FDM. This was inhibited by L-Arg (ED50 = 400 nmol) or SNP (ED50 = 20 nmol), but not D-Arg. Higher-dose SNP, but not L-Arg, was toxic to retina/RPE. Atropine inhibited FDM as expected; adding NOS-inhibitors (L-NIO, L-NMMA) to atropine inhibited this effect dose-dependently, but adding D-NMMA did not. Equatorial diameter, wet weight, and metrics of control eyes were not affected by any treatment. In summary, intraocular NO inhibits myopia dose-dependently and is obligatory for inhibition of myopia by atropine.
Collapse
Affiliation(s)
- Brittany J Carr
- Neuroscience Graduate Program, Snyder Institute for Chronic Diseases, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Calgary, Alberta, Canada
| | - William K Stell
- Department of Cell Biology and Anatomy and Department of Surgery; Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
31
|
Schaeffel F, Feldkaemper M. Animal models in myopia research. Clin Exp Optom 2016; 98:507-17. [PMID: 26769177 DOI: 10.1111/cxo.12312] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/20/2015] [Accepted: 04/26/2015] [Indexed: 12/18/2022] Open
Abstract
Our current understanding of the development of refractive errors, in particular myopia, would be substantially limited had Wiesel and Raviola not discovered by accident that monkeys develop axial myopia as a result of deprivation of form vision. Similarly, if Josh Wallman and colleagues had not found that simple plastic goggles attached to the chicken eye generate large amounts of myopia, the chicken model would perhaps not have become such an important animal model. Contrary to previous assumptions about the mechanisms of myopia, these animal models suggested that eye growth is visually controlled locally by the retina, that an afferent connection to the brain is not essential and that emmetropisation uses more sophisticated cues than just the magnitude of retinal blur. While animal models have shown that the retina can determine the sign of defocus, the underlying mechanism is still not entirely clear. Animal models have also provided knowledge about the biochemical nature of the signal cascade converting the output of retinal image processing to changes in choroidal thickness and scleral growth; however, a critical question was, and still is, can the results from animal models be applied to myopia in children? While the basic findings from chickens appear applicable to monkeys, some fundamental questions remain. If eye growth is guided by visual feedback, why is myopic development not self-limiting? Why does undercorrection not arrest myopic progression even though positive lenses induce myopic defocus, which leads to the development of hyperopia in emmetropic animals? Why do some spectacle or contact lens designs reduce myopic progression and others not? It appears that some major differences exist between animals reared with imposed defocus and children treated with various optical corrections, although without the basic knowledge obtained from animal models, we would be lost in an abundance of untestable hypotheses concerning human myopia.
Collapse
Affiliation(s)
- Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, Tuebingen, Germany.
| | - Marita Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, Tuebingen, Germany
| |
Collapse
|
32
|
Goldberg LA, Rucker FJ. Opposing effects of atropine and timolol on the color and luminance emmetropization mechanisms in chicks. Vision Res 2016; 122:1-11. [PMID: 26971621 PMCID: PMC4861675 DOI: 10.1016/j.visres.2016.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 03/04/2016] [Accepted: 03/08/2016] [Indexed: 11/29/2022]
Abstract
This study analyzed the luminance and color emmetropization response in chicks treated with the nonselective parasympathetic antagonist atropine and the sympathetic β-receptor blocker timolol. Chicks were binocularly exposed (8h/day) for 4days to one of three illumination conditions: 2Hz sinusoidal luminance flicker, 2Hz sinusoidal blue/yellow color flicker, or steady light (mean 680lux). Atropine experiments involved monocular daily injections of either 20μl of atropine (18nmol) or 20μl of phosphate-buffered saline. Timolol experiments involved monocular daily applications of 2 drops of 0.5% timolol or 2 drops of distilled H2O. Changes in the experimental eye were compared with those in the fellow eye after correction for the effects of saline/water treatments. Atropine caused a reduction in axial length with both luminance flicker (-0.078±0.021mm) and color flicker (-0.054±0.017mm), and a reduction in vitreous chamber depth with luminance flicker (-0.095±0.023mm), evoking a hyperopic shift in refraction (3.40±1.77D). Timolol produced an increase in axial length with luminance flicker (0.045±0.030mm) and a myopic shift in refraction (-4.07±0.92D), while color flicker caused a significant decrease in axial length (-0.046±0.017mm) that was associated with choroidal thinning (-0.046±0.015mm). The opposing effects on growth and refraction seen with atropine and timolol suggest a balancing mechanism between the parasympathetic and β-receptor mediated sympathetic system through stimulation of the retina with luminance and color contrast.
Collapse
Affiliation(s)
- Laura A Goldberg
- New England College of Optometry, 424 Beacon Street, Boston, MA 02115, United States.
| | - Frances J Rucker
- New England College of Optometry, 424 Beacon Street, Boston, MA 02115, United States
| |
Collapse
|
33
|
Chakraborty R, Pardue MT. Molecular and Biochemical Aspects of the Retina on Refraction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 134:249-67. [PMID: 26310159 DOI: 10.1016/bs.pmbts.2015.06.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mutant mouse models with specific visual pathway defects offer an advantage to comprehensively investigate the role of specific pathways/neurons involved in refractive development. In this review, we will focus on recent studies using mouse models that have provided insight into retinal pathways and neurotransmitters controlling refractive development. Specifically, we will examine the contributions of rod and cone photoreceptors and the ON and OFF retinal pathways to visually driven eye growth with emphasis on dopaminergic mechanisms.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
34
|
Mathis U, Ziemssen F, Schaeffel F. Effects of a human VEGF antibody (Bevacizumab) on deprivation myopia and choroidal thickness in the chicken. Exp Eye Res 2014; 127:161-9. [PMID: 25094067 DOI: 10.1016/j.exer.2014.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 07/02/2014] [Accepted: 07/26/2014] [Indexed: 12/11/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a dimeric glycoprotein which is responsible for neovascularization and fenestrations of the choriocapillaris. In neovascular maculopathies secondary to age-related degeneration (nAMD) or pathologic myopia (PM-CNV), its inhibition by humanized antibodies is currently the most successful therapy. The choroid has an important role in maintaining retinal health and its thickness declines with age and with myopia. Since choroidal thickness depends on its perfusion rate, one would expect that anti-VEGF agents can also change choroidal thickness. We have tested the hypothesis in the chicken model, using a humanized antibody, Bevacizumab, and also studied the distribution of VEGF-A in the chicken fundal layers by immunohistochemical techniques. Even though it was raised against human VEGF, Bevacizumab had several long lasting effects in the chicken eye (1) after a single unilateral intravitreal injection of 0.5 mg, it partially suppressed the development of deprivation myopia, similarly in both eyes, (2) it completely suppressed choroidal thickening that normally occurs when eyes recover from induced myopia over a time period of about 10 days, (3) it had little effect on the choroidal thickness in eyes that had normal visual experience, (4) VEGF-A was absent in sclera, but highly expressed in the walls of choroidal blood vessels and presumed nerve fiber bundles, as well as in retinal photoreceptors and cells of the inner and outer nuclear layer. One day after the injection of Bevacizumab, the immunoreactivity against VEGF-A had largely disappeared. In conclusion, Bevacizumab is similary effective in human and chicken tissue, has similar time constants (few days), has almost symmetrical effects on myopia in both eyes even after monocular application, and fully suppresses choroidal thickening that normally occurs during recovery from deprivation myopia. The mechanisms by which Bevacizumab acts on the choroidal thickness are perhaps most interesting, both to better understand the role of the choroid in myopia development but also to clarify its potential side effects during nAMD and PM-CNV treatment in the clinics.
Collapse
Affiliation(s)
- Ute Mathis
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Calwerstrasse 7/1, 72076 Tübingen, Germany
| | - Focke Ziemssen
- University Eye Hospital, Department of Ophthalmology, Schleichstrasse 12-16, 72076 Tuebingen, Germany
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Calwerstrasse 7/1, 72076 Tübingen, Germany.
| |
Collapse
|
35
|
He L, Frost MR, Siegwart JT, Norton TT. Gene expression signatures in tree shrew choroid during lens-induced myopia and recovery. Exp Eye Res 2014; 123:56-71. [PMID: 24742494 PMCID: PMC4155741 DOI: 10.1016/j.exer.2014.04.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 03/20/2014] [Accepted: 04/03/2014] [Indexed: 01/19/2023]
Abstract
Gene expression in tree shrew choroid was examined during the development of minus-lens induced myopia (LIM, a GO condition), after completion of minus-lens compensation (a STAY condition), and early in recovery (REC) from induced myopia (a STOP condition). Five groups of tree shrews (n = 7 per group) were used. Starting 24 days after normal eye-opening (days of visual experience [DVE]), one minus-lens group wore a monocular -5 D lens for 2 days (LIM-2), another minus-lens group achieved stable lens compensation while wearing a monocular -5 D lens for 11 days (LIM-11); a recovery group also wore a -5 D lens for 11 days and then received 2 days of recovery starting at 35 DVE (REC-2). Two age-matched normal groups were examined at 26 DVE and 37 DVE. Quantitative PCR was used to measure the relative differences in mRNA levels in the choroid for 77 candidate genes that were selected based on previous studies or because a whole-transcriptome analysis suggested their expression would change during myopia development or recovery. Small myopic changes were observed in the treated eyes of the LIM-2 group (-1.0 ± 0.2 D; mean ± SEM) indicating eyes were early in the process of developing LIM. The LIM-11 group exhibited complete refractive compensation (-5.1 ± 0.2 D) that was stable for five days. The REC-2 group recovered by 1.3 ± 0.3 D from full refractive compensation. Sixty genes showed significant mRNA expression differences during normal development, LIM, or REC conditions. In LIM-2 choroid (GO), 18 genes were significantly down-regulated in the treated eyes relative to the fellow control eyes and 10 genes were significantly up-regulated. In LIM-11 choroid (STAY), 10 genes were significantly down-regulated and 12 genes were significantly up-regulated. Expression patterns in GO and STAY were similar, but not identical. All genes that showed differential expression in GO and STAY were regulated in the same direction in both conditions. In REC-2 choroid (STOP), 4 genes were significantly down-regulated and 18 genes were significantly up-regulated. Thirteen genes showed bi-directional regulation in GO vs. STOP. The pattern of differential gene expression in STOP was very different from that in GO or in STAY. Significant regulation was observed in genes involved in signaling as well as extracellular matrix turnover. These data support an active role for the choroid in the signaling cascade from retina to sclera. Distinctly different treated eye vs. control eye mRNA signatures are present in the choroid in the GO, STAY, and STOP conditions. The STAY signature, present after full compensation has occurred and the GO visual stimulus is no longer present, may participate in maintaining an elongated globe. The 13 genes with bi-directional expression differences in GO and STOP responded in a sign of defocus-dependent manner. Taken together, these data further suggest that a network of choroidal gene expression changes generate the signal that alters scleral fibroblast gene expression and axial elongation rate.
Collapse
Affiliation(s)
- Li He
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, USA.
| | - Michael R Frost
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, USA
| | - John T Siegwart
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, USA
| | - Thomas T Norton
- Department of Vision Sciences, School of Optometry, University of Alabama at Birmingham, USA
| |
Collapse
|
36
|
Nitric oxide synthase inhibitors prevent the growth-inhibiting effects of quinpirole. Optom Vis Sci 2014; 90:1167-75. [PMID: 24061155 DOI: 10.1097/opx.0000000000000041] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Both dopamine and nitric oxide (NO) have been implicated in the signal cascade mediating ocular growth inhibition. If both are part of the same pathway, which precedes the other? We tested the hypothesis that dopamine acts upstream of NO, by using two NOS inhibitors in combination with the dopamine agonist quinpirole, and measured the effects on ocular growth rate. METHODS Chicks wore -10 D lenses or diffusers (FD) for 4 days starting at age 13 days. Experimental eyes received daily 20 μL injections of the following: quinpirole-lens: n = 12, FD: n = 20; n-ω-propyl-L-arginine (NPA)-lens: n = 6, FD: n = 4; quinpirole + NPA-lens: n = 17, FD: n = 19; and quinpirole + L-NIO-lens: n = 12, FD: n = 12. Saline injections were done as controls. High-frequency ultrasonography was done at the start, and on day 5, prior to injections and 3 hours later. Refractions were measured on day 5. RESULTS As expected, quinpirole prevented the development of axial myopia in both paradigms. When quinpirole was combined with either NOS inhibitor, however, eyes became myopic compared to quinpirole (FD: NPA: -5.9 D vs. -3.4 D; L-NIO: -5.8 D vs. -3.4 D; lens: NPA: -3.5 D vs. -0.4 D; p < 0.05 for all; L-NIO was not significant). This was the result of a disinhibition of vitreous chamber growth versus quinpirole (FD: NPA: 401 vs. 275 μm/4 d; L-NIO: 440 vs. 275 μm/4 d; LENS: NPA: 407 vs. 253µm/4 d; L-NIO: 403 vs. 253 μm/4 d; p < 0.05). Only NPA prevented the quinpirole-induced choroidal thickening in lens-wearing eyes (0 vs. 31 μm/3 h; p < 0.05). Choroidal thickening was not inhibited by either drug in FD eyes. CONCLUSIONS Dopamine acts upstream of NO and the choroidal response in the signal cascade mediating ocular growth inhibition in both form deprivation and negative lens wear. That neither NOS inhibitor inhibits choroidal thickening in FD eyes suggests that the choroidal mechanisms differ in the two paradigms.
Collapse
|
37
|
Abstract
PURPOSE Bright light is a potent inhibitor of myopia development in animal models. Because development of refractive errors has been linked to changes in choroidal thickness, we have studied in chickens whether bright light may exert its effects on myopia also through changes in choroidal thickness. METHODS Three-day-old chickens were exposed to "bright light" (15,000 lux; n = 14) from 10 AM to 4 PM but kept under "normal light" (500 lux) during the remaining time of the light phase for 5 days (total duration of light phase 8 AM to 6 PM). A control group (n = 14) was kept under normal light during the entire light phase. Choroidal thickness was measured in alert, hand-held animals with optical coherence tomography at 10 AM, 4 PM, and 8 PM every day. RESULTS Complete data sets were available for 12 chicks in bright light group and nine in normal light group. The striking inter-individual variability in choroidal thickness (coefficient of variance: 23%) made it necessary to normalize changes to the individual baseline thickness of the choroid. During the 6 hours of exposure to bright light, choroidal thickness decreased by -5.2 ± 4.0% (mean ± SEM). By contrast, in the group kept under normal light, choroidal thickness increased by +15.4 ± 4.7% (difference between both groups p = 0.003). After an additional 4 hours, choroidal thickness increased also in the "bright light group" by +17.8 ± 3.5%, while there was little further change (+0.6 ± 4.0%) in the "normal light group" (difference p = 0.004). Finally, the choroid was thicker in the "bright light group" (+7.6 ± 26.0%) than in the "normal light group" (day 5: -18.6 ± 26.9%; difference p = 0.036). CONCLUSIONS Bright light stimulates choroidal thickening in chickens, although the response is smaller than with experimentally imposed myopic defocus, and it occurs with some time delay. It nevertheless suggests that choroidal thickening is also involved in myopia inhibition by bright light.
Collapse
|
38
|
Abstract
Myopia is the result of a mismatch between the optical power and the length of the eye, with the latter being too long. Driving the research in this field is the need to develop myopia treatments that can limit axial elongation. When axial elongation is excessive, as in high myopia, there is an increased risk of visual impairment and blindness due to ensuing pathologies such as retinal detachments. This article covers both clinical studies involving myopic children, and studies involving animal models for myopia. Atropine, a nonselective muscarinic antagonist, has been studied most extensively in both contexts. Because it remains the only drug used in a clinical setting, it is a major focus of the first part of this article, which also covers the many shortcomings of topical ophthalmic atropine. The second part of this article focuses on in vitro and animal-based drug studies, which encompass a range of drug targets including the retina, retinal pigment epithelium and sclera. While the latter studies have contributed to a better understanding of how eye growth is regulated, no new antimyopia drug treatments have reached the clinical setting. Less conservative approaches in research, and in particular, the exploration of new bioengineering approaches for drug delivery, are needed to advance this field.
Collapse
Affiliation(s)
- Prema Ganesan
- School of Optometry, University of California, Berkeley, CA 94720-2020, USA
| | | |
Collapse
|
39
|
Nickla DL, Zhu X, Wallman J. Effects of muscarinic agents on chick choroids in intact eyes and eyecups: evidence for a muscarinic mechanism in choroidal thinning. Ophthalmic Physiol Opt 2013; 33:245-56. [PMID: 23662958 DOI: 10.1111/opo.12054] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 03/06/2013] [Indexed: 12/31/2022]
Abstract
PURPOSE In chicks, ocular growth inhibition is associated with choroidal thickening and growth stimulation with choroidal thinning, suggesting a mechanistic link between the two responses. Because muscarinic antagonists inhibit the development of myopia in animal models by a non-accommodative mechanism, we tested the hypothesis that agonists would stimulate eye growth and thin the choroid. We also hypothesized that the effective growth-inhibiting antagonists would thicken the choroid. METHODS Chicks, age 12-16 days, were used. In vivo: Agonists: Single intravitreal injections (20 μL) of oxotremorine (oxo), pilocarpine (pilo), carbachol (carb), or arecaidine (arec) were given to otherwise untreated eyes. A-scan ultrasonography was done prior to injections, and at 3, 24, 48 and 72 h. Antagonists: -10D lenses were worn on one eye for 4 days. Atropine (atro), pirenzepine (pirz), oxyphenonium (oxy) or dicyclomine (dicy) were injected (20 μL) daily into lens-wearing eyes; saline injections were done as controls. Ultrasonography was done on d1 and on d4; on d4 measurements were done before and 3 h after injections. In vitro: Paired eyecups of retinal pigment epithelium (RPE), choroid and sclera were made from 1-week old chicks. All drugs except atropine were tested on one eyecup, its pair in plain medium. Choroidal thickness was measured at various times over 48 h. RESULTS Agonists: In vivo, oxotremorine caused an increase in the rate of axial elongation (drug vs saline: 24-72 h: 338 μm vs 250 μm; p < 0.001). All except pilocarpine caused choroidal thinning by 24 h (oxo, carb and arec vs saline: -25, -35 and -46 μm vs 3 μm). In vitro, all agonists thinned choroids by 24 h (oxo: -6 vs 111 μm; pilo: 45 vs 212 μm; carb: -58 vs 65 μm; arec: 47 vs 139 μm; p < 0.05). Antagonists: Atropine, pirenzepine and oxyphenonium inhibited the development of myopia in negative lens-wearing eyes, and also caused choroidal thickening (drug vs saline: 42, 80, 88 vs 10 μm per 3 h). In vitro, pirenzepine thickened choroids by 3 h (77 vs 2 μm, p < 0.01). CONCLUSIONS Muscarinic agonists caused choroidal thinning in intact eyes and eyecups, supporting a role for acetylcholine in the choroidal response to hyperopic defocus or form deprivation. Only oxotremorine stimulated eye growth, which is inconsistent with a muscarinic receptor mechanism for antagonist-induced eye growth inhibition. The dissociation between choroidal thinning and ocular growth stimulation for the other agonists in vivo suggest separate pathways for the two.
Collapse
Affiliation(s)
- Debora L Nickla
- Department of Biomedical Sciences and Disease, The New England College of Optometry, Boston, MA, USA.
| | | | | |
Collapse
|
40
|
The effects and interactions of GABAergic and dopaminergic agents in the prevention of form deprivation myopia by brief periods of normal vision. Exp Eye Res 2013; 110:88-95. [PMID: 23474145 DOI: 10.1016/j.exer.2013.02.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/18/2013] [Accepted: 02/20/2013] [Indexed: 11/22/2022]
Abstract
Intravitreal injections of GABA antagonists, dopamine agonists and brief periods of normal vision have been shown separately to inhibit form-deprivation myopia (FDM). Our study had three aims: (i) establish whether GABAergic agents modify the myopia protective effect of normal vision, (ii) investigate the receptor sub-type specificity of any observed effect, and (iii) consider an interaction with the dopamine (DA) system. Prior to the period of normal vision GABAergic agents were applied either (i) individually, (ii) in combination with other GABAergic agents (an agonist with an antagonist), or (iii) in combination with DA agonists and antagonists. Water injections were given to groups not receiving drug treatments so that all experimental eyes received intravitreal injections. As shown previously, constant form-deprivation resulted in high myopia and when diffusers were removed for 2 h per day the period of normal vision greatly reduced the FDM that developed. GABA agonists inhibited the protective effect of normal vision whereas antagonists had the opposite effect. GABAA/C agonists and D2 DA antagonists when used in combination were additive in suppressing the protective effect of normal vision. A D2 DA agonist restored some of the protective effect of normal vision that was inhibited by a GABA agonist (muscimol). The protective effect of normal vision against form-deprivation is modifiable by both the GABAergic and DAergic pathways.
Collapse
|
41
|
Murphy MJ, Crewther DP, Goodyear MJ, Crewther SG. Light modulation, not choroidal vasomotor action, is a regulator of refractive compensation to signed optical blur. Br J Pharmacol 2011; 164:1614-26. [PMID: 21418189 PMCID: PMC3230809 DOI: 10.1111/j.1476-5381.2011.01347.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 01/19/2011] [Accepted: 02/02/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE The nitric oxide system has two proposed sites and mechanisms of action within the ocular growth/refractive compensation platform-neuromodulatory effects on retinal physiology, and vascular/smooth muscle effects in the choroid. The relative contribution of these mechanisms are tested here with drugs that perturb the nitric oxide system and with slow flicker modulation of the ON and OFF pathways of the retina. EXPERIMENTAL APPROACH Intravitreal injection of saline or 900 nmol N(G) -nitro-L-arginine methyl ester or L-arginine in saline was followed by monocular defocus with ±10 D lens (or no lens), from days 5-9 under standard diurnal (SD) or daytime 1 Hz ramped flicker conditions. Biometric, electrophysiological and histological analyses were conducted. KEY RESULTS After 4 days of SD conditions, both drugs enhanced electroretinogram (ERG) b-wave cf. d-wave amplitudes compared with saline and reduced refractive compensation to -10 D lenses. Under flicker conditions compensation to +10 D lenses was suppressed. Choroidal thinning was observed in the drug, no lens groups under SD conditions, whereas choroidal thickening was seen in most groups under flicker conditions, irrespective of refractive outcomes. CONCLUSIONS AND IMPLICATIONS As choroidal thickness was not predictive of final refractive compensation across any of the variables of drug, defocus sign or light condition, it is unlikely that choroidal thickness is a primary mechanism underlying refractive compensation across the range of parameters of this study. Rather, the changes in refractive compensation observed under these particular drug and light conditions are more likely due to a neuromodulatory action on retinal ON and OFF pathways.
Collapse
Affiliation(s)
- Melanie J Murphy
- School of Psychological Science, La Trobe University, Melbourne, Victoria, Australia
| | | | | | | |
Collapse
|
42
|
Nickla DL, Totonelly K. Dopamine antagonists and brief vision distinguish lens-induced- and form-deprivation-induced myopia. Exp Eye Res 2011; 93:782-5. [PMID: 21872586 DOI: 10.1016/j.exer.2011.08.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 08/03/2011] [Accepted: 08/03/2011] [Indexed: 11/25/2022]
Abstract
In eyes wearing negative lenses, the D2 dopamine antagonist spiperone was only partly effective in preventing the ameliorative effects of brief periods of vision (Nickla et al., 2010), in contrast to reports from studies using form-deprivation. The present study was done to directly compare the effects of spiperone, and the D1 antagonist SCH-23390, on the two different myopiagenic paradigms. 12-day old chickens wore monocular diffusers (form-deprivation) or -10 D lenses attached to the feathers with matching rings of Velcro. Each day for 4 days, 10 μl intravitreal injections of the dopamine D2/D4 antagonist spiperone (5 nmoles) or the D1 antagonist SCH-23390, were given under isoflurane anesthesia, and the diffusers (n = 16; n = 5, respectively) or lenses (n = 20; n = 6) were removed for 2 h immediately after. Saline injections prior to vision were done as controls (form-deprivation: n = 11; lenses: n = 10). Two other saline-injected groups wore the lenses (n = 12) or diffusers (n = 4) continuously. Axial dimensions were measured by high frequency A-scan ultrasonography at the start, and on the last day immediately prior to, and 3 h after the injection. Refractive errors were measured at the end of the experiment using a Hartinger's refractometer. In form-deprived eyes, spiperone, but not SCH-23390, prevented the ocular growth inhibition normally effected by the brief periods of vision (change in vitreous chamber depth, spiperone vs saline: 322 vs 211 μm; p = 0.01). By contrast, neither had any effect on negative lens-wearing eyes given similar unrestricted vision (210 and 234 μm respectively, vs 264 μm). The increased elongation in the spiperone-injected form-deprived eyes did not, however, result in a myopic shift, probably due to the inhibitory effect of the drug on anterior chamber growth (drug vs saline: 96 vs 160 μm; p < 0.01). Finally, spiperone inhibited the vision-induced transient choroidal thickening in form-deprived eyes, while SCH-23390 did not. These results indicate that the dopaminergic mechanisms mediating the protective effects of brief periods of unrestricted vision differ for form-deprivation versus negative lens-wear, which may imply different growth control mechanisms between the two.
Collapse
|
43
|
Nickla DL, Totonelly K, Dhillon B. Dopaminergic agonists that result in ocular growth inhibition also elicit transient increases in choroidal thickness in chicks. Exp Eye Res 2010; 91:715-20. [PMID: 20801115 PMCID: PMC2962673 DOI: 10.1016/j.exer.2010.08.021] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 08/12/2010] [Accepted: 08/19/2010] [Indexed: 10/19/2022]
Abstract
The dopaminergic system has been implicated in ocular growth regulation in chicks and monkeys. In both, dopamine D2 agonists inhibit the development of myopia in response to form deprivation, and in chicks, to negative lenses as well. Because there is mounting evidence that the choroidal response to defocus plays a role in ocular growth regulation, we asked whether the effective agonists also elicit transient thickening of the choroid concomitant with the growth inhibition. Negative lenses mounted on velcro rings were worn on one eye starting at age 8-12 days. Intravitreal injections (20 μl; dose = 10 nmole) of the agonist (dissolved in saline) or saline, were given through the superior temporal sclera using a 30G needle. Eyes were injected daily at noon, for 4 days, and the lenses immediately replaced. Agonists used were apomorphine (non-specific; n = 17), quinpirole (D2; n = 10), SKF-38393 (D1; n = 9), and saline controls (n = 22). For the antagonists, the same protocol was used, but on each day, the lenses were removed for 2 h. Immediately prior to lens-removal, the antagonist was injected (20 μl; dose = 5 nmole). Antagonists used were methylergonovine (non-specific; n = 12), spiperone (D2; n = 20), SCH-23390 (D1; n = 6) and saline controls (n = 27). Comparisons to saline (continuous lens wear) controls were from the agonist experiment. Axial dimensions were measured using high frequency A-scan ultrasonography at the start of lens wear, and on day 4 prior to the injections, and then again 3 h later. Refractive errors were measured using a Hartinger's refractometer at the end of the experiment. Apomorphine and quinpirole inhibited the refractive response to the hyperopic defocus induced by the negative lenses (drug vs saline controls: -1.3 and 1.2 D vs -5.6 D; p < 0.005 for both). This effect was axial: both drugs prevented the excessive ocular elongation (change in axial length: 233 and 205 μm vs 417 μm; p < 0.01 for both). Both drugs were also associated with a transient thickening of the choroid over 3 h (41 and 32 μm vs -1 μm; p < 0.01; p = 0.059 respectively) that did not summate: choroids thinned significantly over the 4 day period in all lens-wearing eyes. Two daily hours of unrestricted vision during negative lens wear normally prevents the development of myopia. Spiperone and SCH-23390 inhibited the ameliorating effects of periods of vision on lens-induced refractive error (-2.9 and -2.8 D vs 0.6 D; p < 0.0001), however, the effects on neither axial length nor choroidal thickness were significant. These data support a role for both D1 and D2 receptors in the ocular growth responses.
Collapse
Affiliation(s)
- Debora L Nickla
- The New England College of Optometry, Dept. of Biosciences, 424 Beacon St., Boston, MA 02115, USA.
| | | | | |
Collapse
|
44
|
Stübinger K, Brehmer A, Neuhuber WL, Reitsamer H, Nickla D, Schrödl F. Intrinsic choroidal neurons in the chicken eye: chemical coding and synaptic input. Histochem Cell Biol 2010; 134:145-57. [PMID: 20607273 DOI: 10.1007/s00418-010-0723-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2010] [Indexed: 10/19/2022]
Abstract
Intrinsic choroidal neurons (ICNs) exist in some primates and bird species. They may act on both vascular and non-vascular smooth muscle cells, potentially influencing choroidal blood flow. Here, we report on the chemical coding of ICNs and eye-related cranial ganglia in the chicken, an important model in myopia research, and further to determine synaptic input onto ICN. Chicken choroid, ciliary, superior cervical, pterygopalatine, and trigeminal ganglia were prepared for double or triple immunohistochemistry of calcitonin gene-related peptide (CGRP), choline acetyltransferase (ChAT), dopamine-beta-hydroxylase, galanin (GAL), neuronal nitric oxide synthase (nNOS), somatostatin (SOM), tyrosine hydroxylase (TH), vasoactive intestinal polypeptide (VIP), vesicular monoamine-transporter 2 (VMAT2), and alpha-smooth muscle actin. For documentation, light, fluorescence, and confocal laser scanning microscopy were used. Chicken ICNs express nNOS/VIP/GAL and do not express ChAT and SOM. ICNs are approached by TH/VMAT2-, CGRP-, and ChAT-positive nerve fibers. About 50% of the pterygopalatine ganglion neurons and about 9% of the superior cervical ganglion neurons share the same chemical code as ICN. SOM-positive neurons in the ciliary ganglion are GAL/NOS negative. CGRP-positive neurons in the trigeminal ganglion lack GAL/SOM. The neurochemical phenotype and synaptic input of ICNs in chicken resemble that of other bird and primate species. Because ICNs lack cholinergic markers, they cannot be readily incorporated into current concepts of the autonomic nervous system. The data obtained provide the basis for the interpretation of future functional experiments to clarify the role of these cells in achieving ocular homeostasis.
Collapse
Affiliation(s)
- Karin Stübinger
- Institut für Anatomie I, Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Stone RA, Khurana TS. Gene profiling in experimental models of eye growth: clues to myopia pathogenesis. Vision Res 2010; 50:2322-33. [PMID: 20363242 DOI: 10.1016/j.visres.2010.03.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 03/18/2010] [Accepted: 03/22/2010] [Indexed: 12/22/2022]
Abstract
To understand the complex regulatory pathways that underlie the development of refractive errors, expression profiling has evaluated gene expression in ocular tissues of well-characterized experimental models that alter postnatal eye growth and induce refractive errors. Derived from a variety of platforms (e.g. differential display, spotted microarrays or Affymetrix GeneChips), gene expression patterns are now being identified in species that include chicken, mouse and primate. Reconciling available results is hindered by varied experimental designs and analytical/statistical features. Continued application of these methods offers promise to provide the much-needed mechanistic framework to develop therapies to normalize refractive development in children.
Collapse
Affiliation(s)
- Richard A Stone
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Scheie Eye Institute, Philadelphia, PA 19104-6075, USA.
| | | |
Collapse
|
46
|
Abstract
The choroid of the eye is primarily a vascular structure supplying the outer retina. It has several unusual features: It contains large membrane-lined lacunae, which, at least in birds, function as part of the lymphatic drainage of the eye and which can change their volume dramatically, thereby changing the thickness of the choroid as much as four-fold over a few days (much less in primates). It contains non-vascular smooth muscle cells, especially behind the fovea, the contraction of which may thin the choroid, thereby opposing the thickening caused by expansion of the lacunae. It has intrinsic choroidal neurons, also mostly behind the central retina, which may control these muscles and may modulate choroidal blood flow as well. These neurons receive sympathetic, parasympathetic and nitrergic innervation. The choroid has several functions: Its vasculature is the major supply for the outer retina; impairment of the flow of oxygen from choroid to retina may cause Age-Related Macular Degeneration. The choroidal blood flow, which is as great as in any other organ, may also cool and warm the retina. In addition to its vascular functions, the choroid contains secretory cells, probably involved in modulation of vascularization and in growth of the sclera. Finally, the dramatic changes in choroidal thickness move the retina forward and back, bringing the photoreceptors into the plane of focus, a function demonstrated by the thinning of the choroid that occurs when the focal plane is moved back by the wearing of negative lenses, and, conversely, by the thickening that occurs when positive lenses are worn. In addition to focusing the eye, more slowly than accommodation and more quickly than emmetropization, we argue that the choroidal thickness changes also are correlated with changes in the growth of the sclera, and hence of the eye. Because transient increases in choroidal thickness are followed by a prolonged decrease in synthesis of extracellular matrix molecules and a slowing of ocular elongation, and attempts to decouple the choroidal and scleral changes have largely failed, it seems that the thickening of the choroid may be mechanistically linked to the scleral synthesis of macromolecules, and thus may play an important role in the homeostatic control of eye growth, and, consequently, in the etiology of myopia and hyperopia.
Collapse
Affiliation(s)
- Debora L Nickla
- Department of Biosciences, New England College of Optometry, Boston, MA 02115, USA.
| | | |
Collapse
|
47
|
Nickla DL, Damyanova P, Lytle G. Inhibiting the neuronal isoform of nitric oxide synthase has similar effects on the compensatory choroidal and axial responses to myopic defocus in chicks as does the non-specific inhibitor L-NAME. Exp Eye Res 2009; 88:1092-9. [PMID: 19450449 DOI: 10.1016/j.exer.2009.01.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 01/21/2009] [Accepted: 01/23/2009] [Indexed: 10/21/2022]
Abstract
In birds, the choroid plays a role in the visual regulation of eye growth, thickening in response to myopic defocus, and thinning in response to hyperopic defocus, in both cases moving the retina towards the image plane. This response is rapid, occurring within hours of the defocus stimulus. These changes are consistently associated with slower changes in the sclera, that result in the appropriate changes in axial elongation, decreasing growth in response to myopic defocus and increasing it in response to hyperopic defocus. The molecular mechanisms underlying the scleral response involve changes in the synthesis of extracellular matrix molecules, however, those underlying the changes in choroidal thickness are not known. However, evidence suggests that it may involve the gaseous signal molecule nitric oxide, as nitric oxide is a potent smooth muscle relaxant, and injections of the non-specific nitric oxide synthase inhibitor L-NAME transiently inhibits the thickening response. Interestingly, it also dis-inhibits ocular growth, in accordance with a mechanistic link between the two responses. If nitric oxide is part of the signal cascade underlying the visual regulation of eye growth, it would be important to ascertain the source of the molecule. As a first step towards doing so, we used various more specific NOS inhibitors and studied their effects on the choroidal and growth responses. Birds (7-12 days old) were fitted with +10 D lenses on one eye. On that day, single intravitreal injections (30 microl) of the following inhibitors were used: nNOS inhibitor N(omega)-propyl-L-arginine (n=12), iNOS inhibitor L-NIL (n=16), eNOS/iNOS inhibitor L-NIO (n=15), non-specific inhibitor L-NMMA (n=30) or physiological saline (n=18). Ocular dimensions were measured using high-frequency A-scan ultrasonography at the start of the experiment, and at 7, 24 and 48 h after. We found that the nNOS inhibitor N(omega)-propyl-L-arginine had the same inhibitory effects on the choroidal response, and dis-inhibition of the growth response, as did L-NAME; neither of the other inhibitors had any effect except L-NMMA. We conclude that the choroidal compensatory response is influenced by nNOS, possibly from the intrinsic choroidal neurons, or the parasympathetic innervation from the ciliary and/or pterygopalatine ganglia.
Collapse
Affiliation(s)
- Debora L Nickla
- The New England College of Optometry, Bioscience Department, 424 Beacon St., Boston, MA 02115, USA.
| | | | | |
Collapse
|
48
|
Padmanabhan V, Shih J, Wildsoet CF. Patching fellow eyes during subjective night does not prevent disruption to minus lens compensation in constant light-reared chicks. Vision Res 2008; 48:1992-8. [PMID: 18585401 PMCID: PMC2567919 DOI: 10.1016/j.visres.2008.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2008] [Revised: 05/23/2008] [Accepted: 06/02/2008] [Indexed: 11/16/2022]
Abstract
PURPOSE This study re-examined an earlier claim that monocular patching during subjective night (i.e. patched at the usual time that night would occur) in the chicks reared in continuous lighting (CL), offered unpatched eyes some protection from the ocular effects of CL. It also examined whether this monocular patching protected unpatched eyes against the disruptive effect of CL on compensation to minus lenses. METHODS Hatchling White-Leghorn chicks were reared in either constant or diurnal lighting conditions (n=28) for 2 weeks. Some CL chicks had their right eyes patched every night during the entire study. Lenses of either +10 or -10D power were fitted to the unpatched eyes of some patched chicks at the beginning of the second week. Retinoscopy, IR photo-keratometry and high-frequency A-scan ultrasonography were used to track refractions, corneal radius of curvature and ocular axial dimensions respectively; data were collected on experimental days 0, 7, 9 and 14. RESULTS The patched eyes were completely protected from the ocular growth effects of CL, i.e. accelerated posterior segment (vitreous chamber) growth and inhibited anterior segment growth. Although the unpatched eyes showed no protection from the anterior chamber effects of CL, they were completely protected from the effects of CL on vitreous chamber growth. Nonetheless, the response to the -10D lenses was disrupted in unpatched eyes, which responded in the wrong direction for compensation (+5.5+/-0.25D more hyperopic than no lens-unpatched eyes). The response to the +10D lenses was preserved (+9.25+/-0.25D more hyperopic than no lens-unpatched eyes). CONCLUSION These data provide further support for local control of emmetropization, as reflected in compensatory lens responses, but point to additional influences on eye growth as reflected in CL-induced ocular changes.
Collapse
Affiliation(s)
- Varuna Padmanabhan
- School of Optometry, University of California-Berkeley, Berkeley, CA 94720-2020, USA.
| | | | | |
Collapse
|
49
|
Wiechmann AF, Summers JA. Circadian rhythms in the eye: The physiological significance of melatonin receptors in ocular tissues. Prog Retin Eye Res 2008; 27:137-60. [DOI: 10.1016/j.preteyeres.2007.10.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
50
|
Changes of nitric oxide synthase and cyclic guanosine monophosphate in form deprivation myopia in guinea pigs. Chin Med J (Engl) 2007. [DOI: 10.1097/00029330-200712020-00016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|