1
|
Poels MMF, de Wit GC, Bijveld MMC, van Genderen MM. Natural Course of Refractive Error in Congenital Stationary Night Blindness: Implications for Myopia Treatment. Invest Ophthalmol Vis Sci 2024; 65:9. [PMID: 39625438 PMCID: PMC11620013 DOI: 10.1167/iovs.65.14.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/27/2024] [Indexed: 12/08/2024] Open
Abstract
Purpose A range of pharmacological and optical therapies are being studied and implemented in children with myopia to reduce the progression of myopia. At present, the efficacy of these myopia reduction treatments in children with underlying inherited retinal disorders (IRDs) is largely unknown. To evaluate this efficacy, it is essential to first understand the natural progression of myopia within each distinct underlying IRD. We investigated the natural course of refractive error throughout childhood in patients with congenital stationary night blindness (CSNB) of the Schubert-Bornschein type. Methods We retrospectively assessed a total of 295 refraction measurements in 127 patients with CSNB (48 with "complete" CSNB [CSNB1] and 79 with "incomplete" CSNB [CSNB2]) at different ages between 0 and 21 years old. None had a history of myopia control treatment. A linear mixed effects model was fitted on the data to analyze the natural course of refraction in these patients. Results The fitted model showed that refractive error in patients with CSNB increases quickly toward myopia in the first years of life. After the age of 4 years, there was a minimal progression of only -0.12 diopters (D) per year up to 15 years, after which the refraction seemed stable. All (43/43) of the patients with CSNB1 aged > 4 years were myopic and 84% (62/74) of the patients with CSNB2 aged > 4 years were myopic at the last refraction measurement. Conclusions In general, the refractive error of children with CSNB changes minimally after the age of 4 years old. A critical approach to myopia control interventions in these children is warranted.
Collapse
Affiliation(s)
- Mariëlle M. F. Poels
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, The Netherlands
| | - Gerard C. de Wit
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, The Netherlands
| | - Mieke M. C. Bijveld
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, The Netherlands
| | - Maria M. van Genderen
- Bartiméus Diagnostic Center for Complex Visual Disorders, Zeist, The Netherlands
- Department of Ophthalmology University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
2
|
Xu Z, Tan JK, Vetrivel K, Jiang X, Leo SM, Bhatti T, Tariq A, Webster AR, Robson AG, Hammond CJ, Hysi PG, Mahroo OA. The Electroretinogram I-Wave, a Component Originating in the Retinal OFF-Pathway, Associates With a Myopia Genetic Risk Polymorphism. Invest Ophthalmol Vis Sci 2024; 65:21. [PMID: 39530998 PMCID: PMC11562975 DOI: 10.1167/iovs.65.13.21] [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: 07/08/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Purpose One of the strongest genetic associations with myopia is near the GJD2 gene. Recently, this locus was associated with cone-driven electroretinograms (ERGs), with findings highlighting OFF pathway signals specifically. The ERG i-wave is thought to originate in retinal OFF pathways. We explored this component and tested the hypothesis that it would be associated with the myopia risk locus. Methods International standard LA3 ERGs, recorded with conductive fiber electrodes, were analyzed, first from patients with rare monogenic deficits impairing the ON pathway, or both ON and OFF pathways, to explore effects on the i-wave. Responses were then analyzed from adult participants from the TwinsUK cohort: i-wave amplitudes were measured by two investigators independently, blinded to genotype at the GJD2 locus. We investigated the association between i-wave amplitude and allelic identity at this locus, adjusting for age, sex, and familial relatedness. Results Patient recordings showed the i-wave persisted in the absence of ON pathway signals, but was abolished when both ON and OFF pathways were impaired. For TwinsUK participants, recordings and genotypes were available in 184 individuals (95% female participants; mean standard deviation [SD] age, 64.1 [9.7] years). Mean (SD) i-wave amplitude was 14.5 (SD = 6.5) microvolts. Allelic dosage at the risk locus was significantly associated with i-wave amplitude (P = 0.027). Conclusions Patient ERGs were consistent with the i-wave arising from cone-driven OFF pathways. Amplitudes associated significantly with allelic dosage at the myopia risk locus, supporting the importance of cone-driven signaling in myopia development and further highlighting relevance of the OFF pathway in relation to this locus.
Collapse
Affiliation(s)
- Zihe Xu
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
| | - Jit Kai Tan
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Krishnika Vetrivel
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
| | - Xiaofan Jiang
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
| | - Shaun M. Leo
- Institute of Ophthalmology, University College London, London, United Kingdom
- Electrophysiology Service, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Taha Bhatti
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
| | - Ambreen Tariq
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
| | - Andrew R. Webster
- Institute of Ophthalmology, University College London, London, United Kingdom
- Genetics Service, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Anthony G. Robson
- Institute of Ophthalmology, University College London, London, United Kingdom
- Electrophysiology Service, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Christopher J. Hammond
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
| | - Pirro G. Hysi
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Sørlandet Sykehus Arendal, Arendal Hospital, Norway
| | - Omar A. Mahroo
- Section of Ophthalmology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King's College London, St. Thomas’ Hospital Campus, London, United Kingdom
- Institute of Ophthalmology, University College London, London, United Kingdom
- Genetics Service, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
3
|
Wilmet B, Michiels C, Zhang J, Callebert J, Sahel JA, Picaud S, Audo I, Zeitz C. Loss of ON-Pathway Function in Mice Lacking Lrit3 Decreases Recovery From Lens-Induced Myopia. Invest Ophthalmol Vis Sci 2024; 65:18. [PMID: 39250117 PMCID: PMC11385651 DOI: 10.1167/iovs.65.11.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Abstract
Purpose To determine whether the Lrit3-/- mouse model of complete congenital stationary night blindness with an ON-pathway defect harbors myopic features and whether the genetic defect influences the recovery from lens-induced myopia. Methods Retinal levels of dopamine (DA) and 3,4 dihydroxyphenylacetic acid (DOPAC) from adult isolated Lrit3-/- retinas were quantified using ultra performance liquid chromatography after light adaptation. Natural refractive development of Lrit3-/- mice was measured from three weeks to nine weeks of age using an infrared photorefractometer. Susceptibility to myopia induction was assessed using a lens-induced myopia protocol with -25 D lenses placed in front of the right eye of the animals for three weeks; the mean interocular shift was measured with an infrared photorefractometer after two and three weeks of goggling and after one and two weeks after removal of goggles. Results Compared to wild-type littermates (Lrit3+/+), both DA and DOPAC were drastically reduced in Lrit3-/- retinas. Natural refractive development was normal but Lrit3-/- mice showed a higher myopic shift and a lower ability to recover from induced myopia. Conclusions Our data consolidate the link between ON pathway defect altered dopaminergic signaling and myopia. We document for the first time the role of ON pathway on the recovery from myopia induction.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Jingyi Zhang
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburg, PA, United States
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
4
|
Mazade R, Palumaa T, Pardue MT. Insights into Myopia from Mouse Models. Annu Rev Vis Sci 2024; 10:213-238. [PMID: 38635876 PMCID: PMC11615738 DOI: 10.1146/annurev-vision-102122-102059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Animal models are critical for understanding the initiation and progression of myopia, a refractive condition that causes blurred distance vision. The prevalence of myopia is rapidly increasing worldwide, and myopia increases the risk of developing potentially blinding diseases. Current pharmacological, optical, and environmental interventions attenuate myopia progression in children, but it is still unclear how this occurs or how these interventions can be improved to increase their protective effects. To optimize myopia interventions, directed mechanistic studies are needed. The mouse model is well-suited to these studies because of its well-characterized visual system and the genetic experimental tools available, which can be combined with pharmacological and environmental manipulations for powerful investigations of causation. This review describes aspects of the mouse visual system that support its use as a myopia model and presents genetic, pharmacological, and environmental studies that significantly contribute to our understanding of the mechanisms that underlie myopigenesis.
Collapse
Affiliation(s)
- Reece Mazade
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
| | - Teele Palumaa
- Eye Clinic, East Tallinn Central Hospital, Tallinn, Estonia
- Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, USA
- Department of Ophthalmology, Emory University School of Medicine, Atlanta, Georgia, USA; , ,
| |
Collapse
|
5
|
Schaeffel F, Swiatczak B. Mechanisms of emmetropization and what might go wrong in myopia. Vision Res 2024; 220:108402. [PMID: 38705024 DOI: 10.1016/j.visres.2024.108402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
Studies in animal models and humans have shown that refractive state is optimized during postnatal development by a closed-loop negative feedback system that uses retinal image defocus as an error signal, a mechanism called emmetropization. The sensor to detect defocus and its sign resides in the retina itself. The retina and/or the retinal pigment epithelium (RPE) presumably releases biochemical messengers to change choroidal thickness and modulate the growth rates of the underlying sclera. A central question arises: if emmetropization operates as a closed-loop system, why does it not stop myopia development? Recent experiments in young human subjects have shown that (1) the emmetropic retina can perfectly distinguish between real positive defocus and simulated defocus, and trigger transient axial eye shortening or elongation, respectively. (2) Strikingly, the myopic retina has reduced ability to inhibit eye growth when positive defocus is imposed. (3) The bi-directional response of the emmetropic retina is elicited with low spatial frequency information below 8 cyc/deg, which makes it unlikely that optical higher-order aberrations play a role. (4) The retinal mechanism for the detection of the sign of defocus involves a comparison of defocus blur in the blue (S-cone) and red end of the spectrum (L + M-cones) but, again, the myopic retina is not responsive, at least not in short-term experiments. This suggests that it cannot fully trigger the inhibitory arm of the emmetropization feedback loop. As a result, with an open feedback loop, myopia development becomes "open-loop".
Collapse
Affiliation(s)
- Frank Schaeffel
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland; Section Neurobiology of the Eye, Institute of Ophthalmic Research, University of Tübingen, Germany; Zeiss Vision Lab, Institute of Ophthalmic Research, University of Tübingen, Germany.
| | - Barbara Swiatczak
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland
| |
Collapse
|
6
|
Amorim-de-Sousa A, Macedo-de-Araújo RJ, Fernandes P, González-Méijome JM, Queirós A. Enhancement of the Inner Foveal Response of Young Adults with Extended-Depth-of-Focus Contact Lens for Myopia Management. Vision (Basel) 2024; 8:19. [PMID: 38651440 PMCID: PMC11036275 DOI: 10.3390/vision8020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Myopia management contact lenses have been shown to successfully decrease the rate of eye elongation in children by changing the peripheral refractive profile of the retina. Despite the efforts of the scientific community, the retinal response mechanism to defocus is still unknown. The purpose of this study was to evaluate the local electrophysiological response of the retina with a myopia control contact lens (CL) compared to a single-vision CL of the same material. METHODS The retinal electrical activity and peripheral refraction of 16 eyes (16 subjects, 27.5 ± 5.7 years, 13 females and 3 males) with myopia between -0.75 D and -6.00 D (astigmatism < 1.00 D) were assessed with two CLs (Filcon 5B): a single-vision (SV) CL and an extended-depth-of-focus (EDOF) CL used for myopia management. The peripheral refraction was assessed with an open-field WAM-5500 auto-refractometer/keratometer in four meridians separated by 45° at 2.50 m distance. The global-flash multifocal electroretinogram (gf-mfERG) was recorded with the Reti-port/scan21 (Roland Consult) using a stimulus of 61 hexagons. The implicit time (in milliseconds) and response density (RD, in nV/deg2) of the direct (DC) and induced (IC) components were used for comparison between lenses in physiological pupil conditions. RESULTS Although the EDOF decreased both the HCVA and the LCVA (one and two lines, respectively; p < 0.003), it still allowed a good VA. The EDOF lens induced a myopic shift in most retinal areas, with a higher and statistically significant effect on the nasal retina. No differences in the implicit times of the DC and IC components were observed between SV and EDOF. Compared with the SV, the EDOF lens showed a higher RD in the IC component in the foveal region (p = 0.032). In the remaining retinal areas, the EDOF evoked lower, non-statistically significant RD in both the DC and IC components. CONCLUSIONS The EDOF myopia control CL enhanced the response of the inner layers of the fovea. This might suggest that, besides other mechanisms potentially involved, the central foveal retinal activity might be involved in the mechanism of myopia control with these lenses.
Collapse
Affiliation(s)
- Ana Amorim-de-Sousa
- Clinical and Experimental Optometry Research Lab (CEORLab), School of Science, University of Minho, 4710-057 Braga, Portugal
| | - Rute J. Macedo-de-Araújo
- Clinical and Experimental Optometry Research Lab (CEORLab), School of Science, University of Minho, 4710-057 Braga, Portugal
- Physics Center of Minho and Porto Universities (CF-UM-UP), 4710-057 Braga, Portugal
| | - Paulo Fernandes
- Clinical and Experimental Optometry Research Lab (CEORLab), School of Science, University of Minho, 4710-057 Braga, Portugal
- Physics Center of Minho and Porto Universities (CF-UM-UP), 4710-057 Braga, Portugal
| | - José M. González-Méijome
- Clinical and Experimental Optometry Research Lab (CEORLab), School of Science, University of Minho, 4710-057 Braga, Portugal
- Physics Center of Minho and Porto Universities (CF-UM-UP), 4710-057 Braga, Portugal
| | - António Queirós
- Clinical and Experimental Optometry Research Lab (CEORLab), School of Science, University of Minho, 4710-057 Braga, Portugal
- Physics Center of Minho and Porto Universities (CF-UM-UP), 4710-057 Braga, Portugal
| |
Collapse
|
7
|
Ablordeppey RK, Nieu R, Lin CR, Benavente-Perez A. Early Alterations in Inner-Retina Neural and Glial Saturated Responses in Lens-Induced Myopia. Transl Vis Sci Technol 2024; 13:16. [PMID: 38591944 PMCID: PMC11008749 DOI: 10.1167/tvst.13.4.16] [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: 07/19/2023] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Purpose Myopic marmosets are known to exhibit significant inner retinal thinning compared to age-matched controls. The purpose of this study was to assess inner retinal activity in marmosets with lens-induced myopia compared to age-matched controls and evaluate its relationship with induced changes in refractive state and eye growth. Methods Cycloplegic refractive error (Rx), vitreous chamber depth (VCD), and photopic full-field electroretinogram were measured in 14 marmosets treated binocularly with negative contact lenses compared to 9 untreated controls at different stages throughout the experimental period (from 74 to 369 days of age). The implicit times of the a-, b-, d-, and photopic negative response (PhNR) waves, as well as the saturated amplitude (Vmax), semi-saturation constant (K), and slope (n) estimated from intensity-response functions fitted with Naka-Rushton equations were analyzed. Results Compared to controls, treated marmosets exhibited attenuated b-, d-, and PhNR waves Vmax amplitudes 7 to 14 days into treatment before compensatory changes in refraction and eye growth occurred. At later time points, when treated marmosets had developed axial myopia, the amplitudes and implicit times of the b-, d-, and PhNR waves were similar between groups. In controls, the PhNR wave saturated amplitude increased as the b + d-wave Vmax increased. This trend was absent in treated marmosets. Conclusions Marmosets induced with negative defocus exhibit early alterations in inner retinal saturated amplitudes compared to controls, prior to the development of compensatory myopia. These early ERG changes are independent of refraction and eye size and may reflect early changes in bipolar, ganglion, amacrine, or glial cell physiology prior to myopia development. Translational Relevance The early changes in retinal function identified in the negative lens-treated marmosets may serve as clinical biomarkers to help identify children at risk of developing myopia.
Collapse
Affiliation(s)
- Reynolds K. Ablordeppey
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Rita Nieu
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Carol R. Lin
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| | - Alexandra Benavente-Perez
- Department of Biological and Vision Sciences, State University of New York College of Optometry, New York, NY, USA
| |
Collapse
|
8
|
Poudel S, Jin J, Rahimi-Nasrabadi H, Dellostritto S, Dul MW, Viswanathan S, Alonso JM. Contrast Sensitivity of ON and OFF Human Retinal Pathways in Myopia. J Neurosci 2024; 44:e1487232023. [PMID: 38050109 PMCID: PMC10860621 DOI: 10.1523/jneurosci.1487-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 12/06/2023] Open
Abstract
The human visual cortex processes light and dark stimuli with ON and OFF pathways that are differently modulated by luminance contrast. We have previously demonstrated that ON cortical pathways have higher contrast sensitivity than OFF cortical pathways and the difference increases with luminance range (defined as the maximum minus minimum luminance in the scene). Here, we demonstrate that these ON-OFF cortical differences are already present in the human retina and that retinal responses measured with electroretinography are more affected by reductions in luminance range than cortical responses measured with electroencephalography. Moreover, we show that ON-OFF pathway differences measured with electroretinography become more pronounced in myopia, a visual disorder that elongates the eye and blurs vision at far distance. We find that, as the eye axial length increases across subjects, ON retinal pathways become less responsive, slower in response latency, less sensitive, and less effective and slower at driving pupil constriction. Based on these results, we conclude that myopia is associated with a deficit in ON pathway function that decreases the ability of the retina to process low contrast and regulate retinal illuminance in bright environments.
Collapse
Affiliation(s)
- Sabina Poudel
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Jianzhong Jin
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Hamed Rahimi-Nasrabadi
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Stephen Dellostritto
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Mitchell W Dul
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Suresh Viswanathan
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| | - Jose-Manuel Alonso
- Department of Biological and Visual Sciences, State University of New York College of Optometry, New York, New York 10036
| |
Collapse
|
9
|
Wagner S, Strasser T. Impact of text contrast polarity on the retinal activity in myopes and emmetropes using modified pattern ERG. Sci Rep 2023; 13:11101. [PMID: 37423936 DOI: 10.1038/s41598-023-38192-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023] Open
Abstract
Environmental factors favoring myopia development are still being studied and there is accumulating evidence for a significant role of nearwork. Recently, reading standard black-on-white text was found to activate the retinal OFF pathway and induce choroidal thinning, which is associated with myopia onset. Contrarily, reading white-on-black text led to thicker choroids, being protective against myopia. Respective effects on retinal processing are yet unknown. Here, we exploratively assessed the impact of contrast polarity on the retinal activity and possible interactions with eccentricity and refractive error. We recorded pattern electroretinograms in myopic and emmetropic adults while presenting a dead leaves stimulus (DLS), overlaid by masks of different size in ring or circle shape, either filled with uniform gray or text of inverted or standard contrast. In myopes, retinal responses for DLS with standard and inverted contrast were larger when the perifovea was stimulated (6-12 deg), however, including the fovea resulted in smaller amplitudes for inverted contrast than in emmetropes. The retina of emmetropes was more sensitive to inverted contrast than to standard and gray within 12 deg, but most sensitive for gray in the perifovea. This demonstrates that the refractive error influences the sensitivity to text contrast polarity, with a special role of the peripheral retina, which is in line with previous studies about blur sensitivity. Defining whether the differences derive from retinal processing or anatomical features of a myopic eye requires further investigation. Our approach might be a first step to explain how nearwork promotes the eye's elongation.
Collapse
Affiliation(s)
- Sandra Wagner
- Institute for Ophthalmic Research, University of Tuebingen, Elfriede-Aulhorn-Str. 7, 72076, Tuebingen, Germany.
| | - Torsten Strasser
- Institute for Ophthalmic Research, University of Tuebingen, Elfriede-Aulhorn-Str. 7, 72076, Tuebingen, Germany
- University Eye Hospital Tuebingen, Elfriede-Aulhorn-Str. 7, 72076, Tuebingen, Germany
| |
Collapse
|
10
|
Linne C, Mon KY, D’Souza S, Jeong H, Jiang X, Brown DM, Zhang K, Vemaraju S, Tsubota K, Kurihara T, Pardue MT, Lang RA. Encephalopsin (OPN3) is required for normal refractive development and the GO/GROW response to induced myopia. Mol Vis 2023; 29:39-57. [PMID: 37287644 PMCID: PMC10243678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/05/2023] [Indexed: 06/09/2023] Open
Abstract
Purpose Myopia, or nearsightedness, is the most common form of refractive error and is increasing in prevalence. While significant efforts have been made to identify genetic variants that predispose individuals to myopia, these variants are believed to account for only a small portion of the myopia prevalence, leading to a feedback theory of emmetropization, which depends on the active perception of environmental visual cues. Consequently, there has been renewed interest in studying myopia in the context of light perception, beginning with the opsin family of G-protein coupled receptors (GPCRs). Refractive phenotypes have been characterized in every opsin signaling pathway studied, leaving only Opsin 3 (OPN3), the most widely expressed and blue-light sensing noncanonical opsin, to be investigated for function in the eye and refraction. Methods Opn3 expression was assessed in various ocular tissues using an Opn3eGFP reporter. Weekly refractive development in Opn3 retinal and germline mutants from 3 to 9 weeks of age was measured using an infrared photorefractor and spectral domain optical coherence tomography (SD-OCT). Susceptibility to lens-induced myopia was then assessed using skull-mounted goggles with a -30 diopter experimental and a 0 diopter control lens. Mouse eye biometry was similarly tracked from 3 to 6 weeks. A myopia gene expression signature was assessed 24 h after lens induction for germline mutants to further assess myopia-induced changes. Results Opn3 was found to be expressed in a subset of retinal ganglion cells and a limited number of choroidal cells. Based on an assessment of Opn3 mutants, the OPN3 germline, but not retina conditional Opn3 knockout, exhibits a refractive myopia phenotype, which manifests in decreased lens thickness, shallower aqueous compartment depth, and shorter axial length, atypical of traditional axial myopias. Despite the short axial length, Opn3 null eyes demonstrate normal axial elongation in response to myopia induction and mild changes in choroidal thinning and myopic shift, suggesting that susceptibility to lens-induced myopia is largely unchanged. Additionally, the Opn3 null retinal gene expression signature in response to induced myopia after 24 h is distinct, with opposing Ctgf, Cx43, and Egr1 polarity compared to controls. Conclusions The data suggest that an OPN3 expression domain outside the retina can control lens shape and thus the refractive performance of the eye. Prior to this study, the role of Opn3 in the eye had not been investigated. This work adds OPN3 to the list of opsin family GPCRs that are implicated in emmetropization and myopia. Further, the work to exclude retinal OPN3 as the contributing domain in this refractive phenotype is unique and suggests a distinct mechanism when compared to other opsins.
Collapse
Affiliation(s)
- Courtney Linne
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Khine Yin Mon
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Shane D’Souza
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Heonuk Jeong
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaoyan Jiang
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Dillon M. Brown
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Kevin Zhang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Molecular & Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH
- Medical Scientist Training Program, University of Cincinnati, College of Medicine, Cincinnati, OH
| | - Shruti Vemaraju
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Tsubota Laboratory, Inc., Tokyo, Japan
| | - Toshihide Kurihara
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Photobiology, Keio University School of Medicine, Tokyo, Japan
| | - Machelle T. Pardue
- Department of Ophthalmology and Neuroscience Program, Emory University School of Medicine, Atlanta, GA
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, GA
| | - Richard A. Lang
- Visual Systems Group, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH
| |
Collapse
|
11
|
Sankaridurg P, Berntsen DA, Bullimore MA, Cho P, Flitcroft I, Gawne TJ, Gifford KL, Jong M, Kang P, Ostrin LA, Santodomingo-Rubido J, Wildsoet C, Wolffsohn JS. IMI 2023 Digest. Invest Ophthalmol Vis Sci 2023; 64:7. [PMID: 37126356 PMCID: PMC10155872 DOI: 10.1167/iovs.64.6.7] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Myopia is a dynamic and rapidly moving field, with ongoing research providing a better understanding of the etiology leading to novel myopia control strategies. In 2019, the International Myopia Institute (IMI) assembled and published a series of white papers across relevant topics and updated the evidence with a digest in 2021. Here, we summarize findings across key topics from the previous 2 years. Studies in animal models have continued to explore how wavelength and intensity of light influence eye growth and have examined new pharmacologic agents and scleral cross-linking as potential strategies for slowing myopia. In children, the term premyopia is gaining interest with increased attention to early implementation of myopia control. Most studies use the IMI definitions of ≤-0.5 diopters (D) for myopia and ≤-6.0 D for high myopia, although categorization and definitions for structural consequences of high myopia remain an issue. Clinical trials have demonstrated that newer spectacle lens designs incorporating multiple segments, lenslets, or diffusion optics exhibit good efficacy. Clinical considerations and factors influencing efficacy for soft multifocal contact lenses and orthokeratology are discussed. Topical atropine remains the only widely accessible pharmacologic treatment. Rebound observed with higher concentration of atropine is not evident with lower concentrations or optical interventions. Overall, myopia control treatments show little adverse effect on visual function and appear generally safe, with longer wear times and combination therapies maximizing outcomes. An emerging category of light-based therapies for children requires comprehensive safety data to enable risk versus benefit analysis. Given the success of myopia control strategies, the ethics of including a control arm in clinical trials is heavily debated. IMI recommendations for clinical trial protocols are discussed.
Collapse
Affiliation(s)
- Padmaja Sankaridurg
- Brien Holden Vision Institute, Sydney, Australia
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - David A Berntsen
- University of Houston, College of Optometry, Houston, Texas, United States
| | - Mark A Bullimore
- University of Houston, College of Optometry, Houston, Texas, United States
| | - Pauline Cho
- West China Hospital, Sichuan University, Sichuan, China
- Eye & ENT Hospital of Fudan University, Shanghai, China
- Affiliated Eye Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ian Flitcroft
- Centre for Eye Research Ireland, School of Physics and Clinical and Optometric Sciences, Technological University Dublin, Dublin, Ireland
- Department of Ophthalmology, Children's Health Ireland at Temple Street Hospital, Dublin, Ireland
| | - Timothy J Gawne
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Kate L Gifford
- Queensland University of Technology, Brisbane, Australia
| | - Monica Jong
- Johnson & Johnson Vision, Jacksonville, Florida, United States
| | - Pauline Kang
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Lisa A Ostrin
- University of Houston, College of Optometry, Houston, Texas, United States
| | | | - Christine Wildsoet
- UC Berkeley Wertheim School Optometry & Vision Science, Berkeley, California, United States
| | - James S Wolffsohn
- College of Health & Life Sciences, Aston University, Birmingham, United Kingdom
| |
Collapse
|
12
|
Poudel S, Rahimi-Nasrabadi H, Jin J, Najafian S, Alonso JM. Differences in visual stimulation between reading and walking and implications for myopia development. J Vis 2023; 23:3. [PMID: 37014657 PMCID: PMC10080958 DOI: 10.1167/jov.23.4.3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 02/11/2023] [Indexed: 04/05/2023] Open
Abstract
Visual input plays an important role in the development of myopia (nearsightedness), a visual disorder that blurs vision at far distances. The risk of myopia progression increases with the time spent reading and decreases with outdoor activity for reasons that remain poorly understood. To investigate the stimulus parameters driving this disorder, we compared the visual input to the retina of humans performing two tasks associated with different risks of myopia progression, reading and walking. Human subjects performed the two tasks while wearing glasses with cameras and sensors that recorded visual scenes and visuomotor activity. When compared with walking, reading black text in white background reduced spatiotemporal contrast in central vision and increased it in peripheral vision, leading to a pronounced reduction in the ratio of central/peripheral strength of visual stimulation. It also made the luminance distribution heavily skewed toward negative dark contrast in central vision and positive light contrast in peripheral vision, decreasing the central/peripheral stimulation ratio of ON visual pathways. It also decreased fixation distance, blink rate, pupil size, and head-eye coordination reflexes dominated by ON pathways. Taken together with previous work, these results support the hypothesis that reading drives myopia progression by understimulating ON visual pathways.
Collapse
Affiliation(s)
- Sabina Poudel
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Hamed Rahimi-Nasrabadi
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jianzhong Jin
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Sohrab Najafian
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| | - Jose-Manuel Alonso
- Department of Biological and Visual Sciences, SUNY College of Optometry, New York, NY, USA
| |
Collapse
|
13
|
Zeitz C, Roger JE, Audo I, Michiels C, Sánchez-Farías N, Varin J, Frederiksen H, Wilmet B, Callebert J, Gimenez ML, Bouzidi N, Blond F, Guilllonneau X, Fouquet S, Léveillard T, Smirnov V, Vincent A, Héon E, Sahel JA, Kloeckener-Gruissem B, Sennlaub F, Morgans CW, Duvoisin RM, Tkatchenko AV, Picaud S. Shedding light on myopia by studying complete congenital stationary night blindness. Prog Retin Eye Res 2023; 93:101155. [PMID: 36669906 DOI: 10.1016/j.preteyeres.2022.101155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 01/20/2023]
Abstract
Myopia is the most common eye disorder, caused by heterogeneous genetic and environmental factors. Rare progressive and stationary inherited retinal disorders are often associated with high myopia. Genes implicated in myopia encode proteins involved in a variety of biological processes including eye morphogenesis, extracellular matrix organization, visual perception, circadian rhythms, and retinal signaling. Differentially expressed genes (DEGs) identified in animal models mimicking myopia are helpful in suggesting candidate genes implicated in human myopia. Complete congenital stationary night blindness (cCSNB) in humans and animal models represents an ON-bipolar cell signal transmission defect and is also associated with high myopia. Thus, it represents also an interesting model to identify myopia-related genes, as well as disease mechanisms. While the origin of night blindness is molecularly well established, further research is needed to elucidate the mechanisms of myopia development in subjects with cCSNB. Using whole transcriptome analysis on three different mouse models of cCSNB (in Gpr179-/-, Lrit3-/- and Grm6-/-), we identified novel actors of the retinal signaling cascade, which are also novel candidate genes for myopia. Meta-analysis of our transcriptomic data with published transcriptomic databases and genome-wide association studies from myopia cases led us to propose new biological/cellular processes/mechanisms potentially at the origin of myopia in cCSNB subjects. The results provide a foundation to guide the development of pharmacological myopia therapies.
Collapse
Affiliation(s)
- Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France.
| | - Jérome E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, Saclay, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France
| | | | | | - Juliette Varin
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, APHP, Paris, France
| | | | - Nassima Bouzidi
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Frederic Blond
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Stéphane Fouquet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Vasily Smirnov
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Ajoy Vincent
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Elise Héon
- Department of Ophthalmology and Vision Sciences, The Hospital for Sick Children, Toronto, ON, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - José-Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France; CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, Paris, France; Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Catherine W Morgans
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Robert M Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR, USA
| | - Andrei V Tkatchenko
- Oujiang Laboratory, Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health, Wenzhou, China; Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University, New York, NY, USA
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
14
|
Quint WH, Tadema KCD, Kokke NCCJ, Meester-Smoor MA, Miller AC, Willemsen R, Klaver CCW, Iglesias AI. Post-GWAS screening of candidate genes for refractive error in mutant zebrafish models. Sci Rep 2023; 13:2017. [PMID: 36737489 PMCID: PMC9898536 DOI: 10.1038/s41598-023-28944-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Genome-wide association studies (GWAS) have dissected numerous genetic factors underlying refractive errors (RE) such as myopia. Despite significant insights into understanding the genetic architecture of RE, few studies have validated and explored the functional role of candidate genes within these loci. To functionally follow-up on GWAS and characterize the potential role of candidate genes on the development of RE, we prioritized nine genes (TJP2, PDE11A, SHISA6, LAMA2, LRRC4C, KCNQ5, GNB3, RBFOX1, and GRIA4) based on biological and statistical evidence; and used CRISPR/cas9 to generate knock-out zebrafish mutants. These mutant fish were screened for abnormalities in axial length by spectral-domain optical coherence tomography and refractive status by eccentric photorefraction at the juvenile (2 months) and adult (4 months) developmental stage. We found a significantly increased axial length and myopic shift in refractive status in three of our studied mutants, indicating a potential involvement of the human orthologs (LAMA2, LRRC4C, and KCNQ5) in myopia development. Further, in-situ hybridization studies showed that all three genes are expressed throughout the zebrafish retina. Our zebrafish models provide evidence of a functional role of these three genes in refractive error development and offer opportunities to elucidate pathways driving the retina-to-sclera signaling cascade that leads to myopia.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nina C C J Kokke
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adam C Miller
- Institute of Neuroscience, University of Oregon, Eugene, USA
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, Audo I, Picaud S, Zeitz C. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int J Mol Sci 2022; 24:ijms24010219. [PMID: 36613663 PMCID: PMC9820543 DOI: 10.3390/ijms24010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
| | - Robert Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruben Goulet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christophe Tourain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
| | - Christelle Michiels
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
- Académie des Sciences, Institut de France, 75006 Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| |
Collapse
|
16
|
Huang Y, Chen X, Zhuang J, Yu K. The Role of Retinal Dysfunction in Myopia Development. Cell Mol Neurobiol 2022:10.1007/s10571-022-01309-1. [DOI: 10.1007/s10571-022-01309-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 11/16/2022] [Indexed: 11/27/2022]
|
17
|
Gupta SK, Chakraborty R, Verkicharla PK. Electroretinogram responses in myopia: a review. Doc Ophthalmol 2022; 145:77-95. [PMID: 34787722 PMCID: PMC9470726 DOI: 10.1007/s10633-021-09857-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/11/2021] [Indexed: 11/02/2022]
Abstract
The stretching of a myopic eye is associated with several structural and functional changes in the retina and posterior segment of the eye. Recent research highlights the role of retinal signaling in ocular growth. Evidence from studies conducted on animal models and humans suggests that visual mechanisms regulating refractive development are primarily localized at the retina and that the visual signals from the retinal periphery are also critical for visually guided eye growth. Therefore, it is important to study the structural and functional changes in the retina in relation to refractive errors. This review will specifically focus on electroretinogram (ERG) changes in myopia and their implications in understanding the nature of retinal functioning in myopic eyes. Based on the available literature, we will discuss the fundamentals of retinal neurophysiology in the regulation of vision-dependent ocular growth, findings from various studies that investigated global and localized retinal functions in myopia using various types of ERGs.
Collapse
Affiliation(s)
- Satish Kumar Gupta
- Myopia Research Lab, Prof. Brien Holden Eye Research Centre, Brien Holden Institute of Optometry and Vision Sciences, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India
| | - Ranjay Chakraborty
- Caring Futures Institute, College of Nursing and Health Sciences, Optometry and Vision Science, Flinders University, Adelaide, South Australia, Australia
| | - Pavan Kumar Verkicharla
- Myopia Research Lab, Prof. Brien Holden Eye Research Centre, Brien Holden Institute of Optometry and Vision Sciences, Kallam Anji Reddy Campus, L V Prasad Eye Institute, Hyderabad, India.
| |
Collapse
|
18
|
Aung MH, Hogan K, Mazade RE, Park HN, Sidhu CS, Iuvone PM, Pardue MT. ON than OFF pathway disruption leads to greater deficits in visual function and retinal dopamine signaling. Exp Eye Res 2022; 220:109091. [PMID: 35487263 PMCID: PMC9701101 DOI: 10.1016/j.exer.2022.109091] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/15/2022] [Accepted: 04/20/2022] [Indexed: 11/19/2022]
Abstract
The visual system uses ON and OFF pathways to signal luminance increments and decrements. Increasing evidence suggests that ON and OFF pathways have different signaling properties and serve specialized visual functions. However, it is still unclear the contribution of ON and OFF pathways to visual behavior. Therefore, we examined the effects on optomotor response and the retinal dopamine system in nob mice with ON pathway dysfunction and Vsx1-/- mice with partial OFF pathway dysfunction. Spatial frequency and contrast sensitivity thresholds were determined, and values were compared to age-matched wild-type controls. Retinas were collected immediately after visual testing to measure levels of dopamine and its metabolite, DOPAC. At 4 weeks of age, we found that nob mice had significantly reduced spatial frequency (19%) and contrast sensitivity (60%) thresholds compared to wild-type mice. Vsx1-/- mice also exhibited reductions in optomotor responses (3% in spatial frequency; 18% in contrast sensitivity) at 4 weeks, although these changes were significantly smaller than those found in nob mice. Furthermore, nob mice had significantly lower DOPAC levels (53%) and dopamine turnover (41%) compared to controls while Vsx1-/- mice displayed a transient increase in DOPAC levels at 4 weeks of age (55%). Our results show that dysfunction of ON pathways leads to reductions in contrast sensitivity, spatial frequency threshold, and retinal dopamine turnover whereas partial loss of the OFF pathway has minimal effect. We conclude that ON pathways play a critical role in visual reflexes and retinal dopamine signaling, highlighting a potential association for future investigations.
Collapse
Affiliation(s)
- Moe H Aung
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA; Department of Ophthalmology, Dell Medical School, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Kelleigh Hogan
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Reece E Mazade
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, USA
| | - Han Na Park
- Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA; Department of Ophthalmology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 0322, USA
| | - Curran S Sidhu
- Department of Ophthalmology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 0322, USA
| | - P Michael Iuvone
- Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA; Department of Ophthalmology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 0322, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, USA; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, USA; Department of Ophthalmology, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 0322, USA.
| |
Collapse
|
19
|
Brown DM, Mazade R, Clarkson-Townsend D, Hogan K, Datta Roy PM, Pardue MT. Candidate pathways for retina to scleral signaling in refractive eye growth. Exp Eye Res 2022; 219:109071. [PMID: 35447101 PMCID: PMC9701099 DOI: 10.1016/j.exer.2022.109071] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/25/2022] [Accepted: 04/05/2022] [Indexed: 12/22/2022]
Abstract
The global prevalence of myopia, or nearsightedness, has increased at an alarming rate over the last few decades. An eye is myopic if incoming light focuses prior to reaching the retinal photoreceptors, which indicates a mismatch in its shape and optical power. This mismatch commonly results from excessive axial elongation. Important drivers of the myopia epidemic include environmental factors, genetic factors, and their interactions, e.g., genetic factors influencing the effects of environmental factors. One factor often hypothesized to be a driver of the myopia epidemic is environmental light, which has changed drastically and rapidly on a global scale. In support of this, it is well established that eye size is regulated by a homeostatic process that incorporates visual cues (emmetropization). This process allows the eye to detect and minimize refractive errors quite accurately and locally over time by modulating the rate of elongation of the eye via remodeling its outermost coat, the sclera. Critically, emmetropization is not dependent on post-retinal processing. Thus, visual cues appear to influence axial elongation through a retina-to-sclera, or retinoscleral, signaling cascade, capable of transmitting information from the innermost layer of the eye to the outermost layer. Despite significant global research interest, the specifics of retinoscleral signaling pathways remain elusive. While a few pharmacological treatments have proven to be effective in slowing axial elongation (most notably topical atropine), the mechanisms behind these treatments are still not fully understood. Additionally, several retinal neuromodulators, neurotransmitters, and other small molecules have been found to influence axial length and/or refractive error or be influenced by myopigenic cues, yet little progress has been made explaining how the signal that originates in the retina crosses the highly vascular choroid to affect the sclera. Here, we compile and synthesize the evidence surrounding three of the major candidate pathways receiving significant research attention - dopamine, retinoic acid, and adenosine. All three candidates have both correlational and causal evidence backing their involvement in axial elongation and have been implicated by multiple independent research groups across diverse species. Two hypothesized mechanisms are presented for how a retina-originating signal crosses the choroid - via 1) all-trans retinoic acid or 2) choroidal blood flow influencing scleral oxygenation. Evidence of crosstalk between the pathways is discussed in the context of these two mechanisms.
Collapse
Affiliation(s)
- Dillon M Brown
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA
| | - Reece Mazade
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA
| | - Danielle Clarkson-Townsend
- Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA; Division of Sleep and Circadian Disorders, Brigham and Women's Hospital, Harvard Medical School, 221 Longwood Avenue, Boston, MA, 02115, USA; Division of Sleep Medicine, Harvard Medical School, Boston, MA, 02115, USA; Gangarosa Department of Environmental Health, Emory University, 1518 Clifton Rd, Atlanta, GA, 30322, USA
| | - Kelleigh Hogan
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA
| | - Pooja M Datta Roy
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA
| | - Machelle T Pardue
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, 313 Ferst Drive, Atlanta, GA, 30332, USA; Center for Visual and Neurocognitive Rehabilitation, Atlanta Veterans Affairs Healthcare System, 1670 Clairmont Rd, Atlanta, GA, 30033, USA.
| |
Collapse
|
20
|
Jiang X, Xu Z, Soorma T, Tariq A, Bhatti T, Baneke AJ, Pontikos N, Leo SM, Webster AR, Williams KM, Hammond CJ, Hysi PG, Mahroo OA. Electrical responses from human retinal cone pathways associate with a common genetic polymorphism implicated in myopia. Proc Natl Acad Sci U S A 2022; 119:e2119675119. [PMID: 35594404 PMCID: PMC9173800 DOI: 10.1073/pnas.2119675119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/08/2022] [Indexed: 11/25/2022] Open
Abstract
Myopia is the commonest visual impairment. Several genetic loci confer risk, but mechanisms by which they do this are unknown. Retinal signals drive eye growth, and myopia usually results from an excessively long eye. The common variant most strongly associated with myopia is near the GJD2 gene, encoding connexin-36, which forms retinal gap junctions. Light-evoked responses of retinal neurons can be recorded noninvasively as the electroretinogram (ERG). We analyzed these responses from 186 adult twin volunteers who had been genotyped at this locus. Participants underwent detailed ERG recordings incorporating international standard stimuli as well as experimental protocols aiming to separate dark-adapted rod- and cone-driven responses. A mixed linear model was used to explore association between allelic dosage at the locus and international standard ERG parameters after adjustment for age, sex, and family structure. Significant associations were found for parameters of light-adapted, but not dark-adapted, responses. Further investigation of isolated rod- and cone-driven ERGs confirmed associations with cone-driven, but not rod-driven, a-wave amplitudes. Comparison with responses to similar experimental stimuli from a patient with a prior central retinal artery occlusion, and from two patients with selective loss of ON-bipolar cell signals, was consistent with the associated parameters being derived from signals from cone-driven OFF-bipolar cells. Analysis of single-cell transcriptome data revealed strongest GJD2 expression in cone photoreceptors; bipolar cell expression appeared strongest in OFF-bipolar cells and weakest in rod-driven ON-bipolar cells. Our findings support a potential role for altered signaling in cone-driven OFF pathways in myopia development.
Collapse
Affiliation(s)
- Xiaofan Jiang
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Zihe Xu
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Talha Soorma
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
| | - Ambreen Tariq
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Taha Bhatti
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Alexander J. Baneke
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
| | - Nikolas Pontikos
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
| | - Shaun M. Leo
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
- Medical Retina Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
- Inherited Eye Disease Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
| | - Andrew R. Webster
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
- Medical Retina Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
- Inherited Eye Disease Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
| | - Katie M. Williams
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
- Medical Retina Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
- Inherited Eye Disease Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
| | - Christopher J. Hammond
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Pirro G. Hysi
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
| | - Omar A. Mahroo
- Institute of Ophthalmology, University College London, London EC1V 9EL, United Kingdom
- Department of Ophthalmology, King’s College London, London SE1 7EH, United Kingdom
- Department of Twin Research and Genetic Epidemiology, King’s College London, London SE1 7EH, United Kingdom
- Medical Retina Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
- Inherited Eye Disease Service, Moorfields Eye Hospital, London EC1V 2PD, United Kingdom
- Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| |
Collapse
|
21
|
Increase in b-wave amplitude after light stimulation of the blind spot is positively correlated with the axial length of myopic individuals. Sci Rep 2022; 12:4785. [PMID: 35314724 PMCID: PMC8938467 DOI: 10.1038/s41598-022-08319-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/01/2022] [Indexed: 12/02/2022] Open
Abstract
Altered retinal dopamine and ON-pathway activity may underlie myopia development. It has been shown that the stimulation of the blind spot with short-wavelength light increases the electroretinogram (ERG) b-wave amplitude of myopic eyes and may engage the retinal dopaminergic system. This study evaluated the impact of various durations of blind spot stimulation on the electrophysiological response of the myopic retina and their relationship to axial length. Six myopic individuals underwent three short-wavelength blue light blind spot stimulation protocols (10 s, 1 min, 10 min) using a virtual reality headset. As a control condition, no stimulation was shown for 1 min. The b-wave amplitude of the photopic full-field ERG was measured at baseline and 10, 20, 30, 40, 50, and 60 min after each condition. A significant increase in b-wave amplitude was observed for all stimulation protocols compared to the control. The peak b-wave amplitude was observed 20 min after the 1-min stimulation protocol and 60 min after the 10-min stimulation protocol. A significant positive correlation was found between axial length of the eye and percent change in b-wave amplitude for the 10-min stimulation protocol. A rapid and a delayed b-wave time course responses were observed following 1 min and 10 min of blind spot stimulation, respectively. Overall, these results indicate that light stimulation of the blind spot for various durations elevates ON-bipolar cell activity in the retina and as such is assumed to reduce the myopic response. These findings could have implications for future myopia treatment.
Collapse
|
22
|
Quint WH, Tadema KCD, Crins JHC, Kokke NCCJ, Meester-Smoor MA, Willemsen R, Klaver CCW, Iglesias AI. Zebrafish: An In Vivo Screening Model to Study Ocular Phenotypes. Transl Vis Sci Technol 2022; 11:17. [PMID: 35285860 PMCID: PMC8934544 DOI: 10.1167/tvst.11.3.17] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To establish a set of assays that allow the in vivo screening of candidate genes for ocular diseases in zebrafish, with an emphasis on refractive error. Methods Our pipeline includes the most relevant ocular screening measurements to assess (1) ocular biometry using spectral domain optical coherence tomography, (2) refractive status using an eccentric photorefractor, (3) intraocular pressure by tonometry, and (4) optokinetic response to study visual capability in zebrafish. To validate our pipeline and to demonstrate the potential of zebrafish as a valid animal model, we chose two well-characterized genes with an ocular phenotype (PRSS56 and FBN1) and generated two mutant zebrafish lines (prss56 and fbn1). Mutant fish were assessed at 2, 4, and 6 months after fertilization. Results With the proposed phenotyping pipeline, we showed that ocular biometry, refractive status, intraocular pressure, and visual function can be studied in zebrafish. In the prss56 mutant, the pipeline revealed a dramatic decrease in axial length, mainly owing to a decreased vitreous chamber depth, whereas in the fbn1 mutant, ectopia lentis was the most distinctive ocular phenotype observed. Tonometry in both mutant lines showed an increase in intraocular pressure. Conclusions The proposed pipeline was applied successfully in zebrafish and can be used for future genetic screenings of candidate genes. While validating our pipeline, we found a close resemblance between the ocular manifestations in the zebrafish mutants and patients harboring mutations in PRSS56 and FBN1. Our results support the validity of our pipeline and highlight the potential of zebrafish as an animal model for in vivo screening of candidate genes for ocular diseases.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Johan H C Crins
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Nina C C J Kokke
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Magda A Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Ophthalmology, Radboud University Medical Center, Nijmegen, the Netherlands.,Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, the Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
23
|
van der Sande E, Haarman AEG, Quint WH, Tadema KCD, Meester-Smoor MA, Kamermans M, De Zeeuw CI, Klaver CCW, Winkelman BHJ, Iglesias AI. The Role of GJD2(Cx36) in Refractive Error Development. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 35262731 PMCID: PMC8934558 DOI: 10.1167/iovs.63.3.5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
Refractive errors are common eye disorders characterized by a mismatch between the focal power of the eye and its axial length. An increased axial length is a common cause of the refractive error myopia (nearsightedness). The substantial increase in myopia prevalence over the last decades has raised public health concerns because myopia can lead to severe ocular complications later in life. Genomewide association studies (GWAS) have made considerable contributions to the understanding of the genetic architecture of refractive errors. Among the hundreds of genetic variants identified, common variants near the gap junction delta-2 (GJD2) gene have consistently been reported as one of the top hits. GJD2 encodes the connexin 36 (Cx36) protein, which forms gap junction channels and is highly expressed in the neural retina. In this review, we provide current evidence that links GJD2(Cx36) to the development of myopia. We summarize the gap junctional communication in the eye and the specific role of GJD2(Cx36) in retinal processing of visual signals. Finally, we discuss the pathways involving dopamine and gap junction phosphorylation and coupling as potential mechanisms that may explain the role of GJD2(Cx36) in refractive error development.
Collapse
Affiliation(s)
- Emilie van der Sande
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
| | - Annechien E. G. Haarman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim H. Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Kirke C. D. Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Magda A. Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Maarten Kamermans
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Biomedical Physics and Biomedical Photonics, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Chris I. De Zeeuw
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C. W. Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
- Institute of Molecular and Clinical Ophthalmology, Basel, Switzerland
| | - Beerend H. J. Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Netherlands Institute for Neuroscience (NIN), Royal Dutch Academy of Art & Science (KNAW), Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adriana I. Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
24
|
Chakraborty R, Landis EG, Mazade R, Yang V, Strickland R, Hattar S, Stone RA, Iuvone PM, Pardue MT. Melanopsin modulates refractive development and myopia. Exp Eye Res 2022; 214:108866. [PMID: 34838844 PMCID: PMC8792255 DOI: 10.1016/j.exer.2021.108866] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Myopia, or nearsightedness, is the most common form of refractive abnormality and is characterized by excessive ocular elongation in relation to ocular power. Retinal neurotransmitter signaling, including dopamine, is implicated in myopic ocular growth, but the visual pathways that initiate and sustain myopia remain unclear. Melanopsin-expressing retinal ganglion cells (mRGCs), which detect light, are important for visual function, and have connections with retinal dopamine cells. Here, we investigated how mRGCs influence normal and myopic refractive development using two mutant mouse models: Opn4-/- mice that lack functional melanopsin photopigments and intrinsic mRGC responses but still receive other photoreceptor-mediated input to these cells; and Opn4DTA/DTA mice that lack intrinsic and photoreceptor-mediated mRGC responses due to mRGC cell death. In mice with intact vision or form-deprivation, we measured refractive error, ocular properties including axial length and corneal curvature, and the levels of retinal dopamine and its primary metabolite, L-3,4-dihydroxyphenylalanine (DOPAC). Myopia was measured as a myopic shift, or the difference in refractive error between the form-deprived and contralateral eyes. We found that Opn4-/- mice had altered normal refractive development compared to Opn4+/+ wildtype mice, starting ∼4D more myopic but developing ∼2D greater hyperopia by 16 weeks of age. Consistent with hyperopia at older ages, 16 week-old Opn4-/- mice also had shorter eyes compared to Opn4+/+ mice (3.34 vs 3.42 mm). Opn4DTA/DTA mice, however, were more hyperopic than both Opn4+/+ and Opn4-/- mice across development ending with even shorter axial lengths. Despite these differences, both Opn4-/- and Opn4DTA/DTA mice had ∼2D greater myopic shifts in response to form-deprivation compared to Opn4+/+ mice. Furthermore, when vision was intact, dopamine and DOPAC levels were similar between Opn4-/- and Opn4+/+ mice, but higher in Opn4DTA/DTA mice, which differed with age. However, form-deprivation reduced retinal dopamine and DOAPC by ∼20% in Opn4-/- compared to Opn4+/+ mice but did not affect retinal dopamine and DOPAC in Opn4DTA/DTA mice. Lastly, systemically treating Opn4-/- mice with the dopamine precursor L-DOPA reduced their form-deprivation myopia by half compared to non-treated mice. Collectively our findings show that disruption of retinal melanopsin signaling alters the rate and magnitude of normal refractive development, yields greater susceptibility to form-deprivation myopia, and changes dopamine signaling. Our results suggest that mRGCs participate in the eye's response to myopigenic stimuli, acting partly through dopaminergic mechanisms, and provide a potential therapeutic target underling myopia progression. We conclude that proper mRGC function is necessary for correct refractive development and protection from myopia progression.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; College of Nursing and Health Sciences, Optometry and Vision Science, Flinders University, Bedford Park, SA, 5001, Adelaide, Australia; Caring Futures Institute, Flinders University, Bedford Park, SA, 5042, Adelaide, Australia
| | - Erica G Landis
- Department of Ophthalmology, Emory University School of Medicine, 1365B Clifton Rd NE, Atlanta, GA, 30322, United States; Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States
| | - Reece Mazade
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States
| | - Victoria Yang
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States
| | - Ryan Strickland
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States
| | - Samer Hattar
- Section on Light and Circadian Rhythms, NIMH, NIH, 9000 Rockville Pike, Bethesda, MD, USA, 20892
| | - Richard A Stone
- Department of Ophthalmology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 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
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, 1670 Clairmont Rd, Decatur, GA, 30033, United States; Neuroscience Program, Emory University School of Medicine, 1365 Clifton Rd NE, Atlanta, GA, 30322, United States; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr, Atlanta, GA, 30332, United States.
| |
Collapse
|
25
|
Hoseini-Yazdi H, Read SA, Alonso-Caneiro D, Collins MJ. Retinal OFF-Pathway Overstimulation Leads to Greater Accommodation-Induced Choroidal Thinning. Invest Ophthalmol Vis Sci 2021; 62:5. [PMID: 34636878 PMCID: PMC8525845 DOI: 10.1167/iovs.62.13.5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To examine the interactions between accommodation and overstimulation of the retinal ON- and OFF-pathways, and their association with changes in choroidal thickness (ChT) and vascularity. Methods Optical coherence tomography imaging of the choroid of twenty young adults (ages 25 ± 5 years) was performed before and after a series of 30-minute-long viewing tasks, including reading a bright text on dark background (ON-pathway overstimulation) and dark text on bright background (OFF-pathway overstimulation), and a control task of viewing a movie with unbiased ON-/OFF-pathway activation. The viewing tasks were performed with relaxed, and 5 diopter (D) accommodation (induced by soft contact lenses) demands. Both reading texts were matched for the mean luminance (35 cd/m2), luminance contrast (87%), and letter size (approximately 11.8 arc minutes). The change in ChT from baseline associated with contrast polarity and accommodation was examined using linear mixed model analysis. Results The subfoveal ChT decreased significantly by −7 ± 1 µm with 5 D accommodation compared with relaxed accommodation (−3 ± 1 µm; P < 0.001), and by −9 ± 1 µm with OFF-pathway compared with ON-pathway overstimulation (−4 ± 1 µm; P = 0.002) and the control condition (−2 ± 1 µm; P < 0.001). Overstimulation of the OFF-pathway, but not the ON-pathway, resulted in a significantly greater choroidal thinning compared with the control condition, both at relaxed (−7 ± 1 µm; P = 0.003) and 5 D (−11 ± 1 µm; P = 0.005) accommodation levels. Similar changes were also observed for macular total, stromal, and luminal ChT. Conclusions Retinal OFF-pathway stimulation enhanced the choroidal thinning associated with accommodation, thereby providing a potential mechanism that involves accommodation and the retinal OFF-signaling pathway, linking near work and myopia.
Collapse
Affiliation(s)
- Hosein Hoseini-Yazdi
- 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
| | - 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
| | - Michael J Collins
- 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
| |
Collapse
|
26
|
Karouta C, Kucharski R, Hardy K, Thomson K, Maleszka R, Morgan I, Ashby R. Transcriptome-based insights into gene networks controlling myopia prevention. FASEB J 2021; 35:e21846. [PMID: 34405458 DOI: 10.1096/fj.202100350rr] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 11/11/2022]
Abstract
Myopia (short-sightedness), usually caused by excessive elongation of the eye during development, has reached epidemic proportions worldwide. In animal systems including the chicken model, several treatments have been shown to inhibit ocular elongation and experimental myopia. Although diverse in their apparent mechanism of action, each one leads to a reduction in the rate of ocular growth. We hypothesize that a defined set of retinal molecular changes may underlie growth inhibition, irrespective of the treatment agent used. Accordingly, across five well-established but diverse methods of inhibiting myopia, significant overlap is seen in the retinal transcriptome profile (transcript levels and alternative splicing events) in chicks when analyzed by RNA-seq. Within the two major pathway networks enriched during growth inhibition, that of cell signaling and circadian entrainment, transcription factors form the largest functional grouping. Importantly, a large percentage of those genes forming the defined retinal response are downstream targets of the transcription factor EGR1 which itself shows a universal response to all five growth-inhibitory treatments. This supports EGR1's previously implicated role in ocular growth regulation. Finally, by contrasting our data with human linkage and GWAS studies on refractive error, we confirm the applicability of our study to the human condition. Together, these findings suggest that a universal set of transcriptome changes, which sit within a well-defined retinal network that cannot be bypassed, is fundamental to growth regulation, thus paving a way for designing novel targets for myopia therapies.
Collapse
Affiliation(s)
- Cindy Karouta
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Robert Kucharski
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Kristine Hardy
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Kate Thomson
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
| | - Ryszard Maleszka
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Ian Morgan
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Regan Ashby
- Centre for Research in Therapeutic Solutions, Biomedical Sciences, Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia.,Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
27
|
Summers JA, Schaeffel F, Marcos S, Wu H, Tkatchenko AV. Functional integration of eye tissues and refractive eye development: Mechanisms and pathways. Exp Eye Res 2021; 209:108693. [PMID: 34228967 PMCID: PMC11697408 DOI: 10.1016/j.exer.2021.108693] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/16/2022]
Abstract
Refractive eye development is a tightly coordinated developmental process. The general layout of the eye and its various components are established during embryonic development, which involves a complex cross-tissue signaling. The eye then undergoes a refinement process during the postnatal emmetropization process, which relies heavily on the integration of environmental and genetic factors and is controlled by an elaborate genetic network. This genetic network encodes a multilayered signaling cascade, which converts visual stimuli into molecular signals that guide the postnatal growth of the eye. The signaling cascade underlying refractive eye development spans across all ocular tissues and comprises multiple signaling pathways. Notably, tissue-tissue interaction plays a key role in both embryonic eye development and postnatal eye emmetropization. Recent advances in eye biometry, physiological optics and systems genetics of refractive error have significantly advanced our understanding of the biological processes involved in refractive eye development and provided a framework for the development of new treatment options for myopia. In this review, we summarize the recent data on the mechanisms and signaling pathways underlying refractive eye development and discuss new evidence suggesting a wide-spread signal integration across different tissues and ocular components involved in visually guided eye growth.
Collapse
Affiliation(s)
- Jody A Summers
- Department of Cell Biology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Tuebingen, Germany; Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Basel, Switzerland
| | - Susana Marcos
- Instituto de Óptica "Daza de Valdés", Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Hao Wu
- Department of Ophthalmology, Columbia University, New York, USA
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, USA; Department of Pathology and Cell Biology, Columbia University, New York, USA.
| |
Collapse
|
28
|
Small Leucine-Rich Proteoglycans (SLRPs) in the Retina. Int J Mol Sci 2021; 22:ijms22147293. [PMID: 34298915 PMCID: PMC8305803 DOI: 10.3390/ijms22147293] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 01/17/2023] Open
Abstract
Retinal diseases such as age-related macular degeneration (AMD), retinopathy of prematurity (ROP), and diabetic retinopathy (DR) are the leading causes of visual impairment worldwide. There is a critical need to understand the structural and cellular components that play a vital role in the pathophysiology of retinal diseases. One potential component is the family of structural proteins called small leucine-rich proteoglycans (SLRPs). SLRPs are crucial in many fundamental biological processes involved in the maintenance of retinal homeostasis. They are present within the extracellular matrix (ECM) of connective and vascular tissues and contribute to tissue organization and modulation of cell growth. They play a vital role in cell–matrix interactions in many upstream signaling pathways involved in fibrillogenesis and angiogenesis. In this comprehensive review, we describe the expression patterns and function of SLRPs in the retina, including Biglycan and Decorin from class I; Fibromodulin, Lumican, and a Proline/arginine-rich end leucine-rich repeat protein (PRELP) from class II; Opticin and Osteoglycin/Mimecan from class III; and Chondroadherin (CHAD), Tsukushi and Nyctalopin from class IV.
Collapse
|
29
|
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: 1.3] [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
|
30
|
Ji S, Mao X, Zhang Y, Ye L, Dai J. Contribution of M-opsin-based color vision to refractive development in mice. Exp Eye Res 2021; 209:108669. [PMID: 34126082 DOI: 10.1016/j.exer.2021.108669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 11/26/2022]
Abstract
M-opsin, encoded by opn1mw gene, is involved in green-light perception of mice. The role of M-opsin in emmetropization of mice remains uncertain. To answer the above question, 4-week-old wild-type (WT) mice were exposed to white light or green light (460-600 nm, a peak at 510 nm) for 12 weeks. Refractive development was estimated biweekly. After treatment, retinal function was assessed using electroretinogram (ERG). Dopamine (DA) in the retina was evaluated by high-performance liquid chromatography, M-opsin and S-opsin protein levels by Western blot and ELISA, and mRNA expressions of opn1mw and opn1sw by RT-PCR. Effects of M-opsin were further verified in Opn1mw-/- and WT mice raised in white light for 4 weeks. Refractive development was examined at 4, 6, and 8 weeks after birth. The retinal structure was estimated through hematoxylin and eosin staining (H&E) and transmission electron microscopy (TEM). Retinal wholemounts from WT and Opn1mw-/- mice were co-immunolabeled with M-opsin and S-opsin, their distribution and quantity were then assayed by immunofluorescence staining (IF). Expression of S-opsin protein and opn1sw mRNA were determined by Western blot, ELISA, or RT-PCR. Retinal function and DA content were analyzed by ERG and liquid chromatography tandem-mass spectrometry (LC-MS/MS), respectively. Lastly, visual cliff test was used to evaluate the depth perception of the Opn1mw-/- mice. We found that green light-treated WT mice were more myopic with increased M-opsin expression and decreased DA content than white light-treated WT mice after 12-week illumination. No electrophysiologic abnormalities were recorded in mice exposed to green light compared to those exposed to white light. A more hyperopic shift was further observed in 8-week-old Opn1mw-/- mice in white light with lower DA level and weakened cone function than the WT mice under white light. Neither obvious structural disruption of the retina nor abnormal depth perception was found in Opn1mw-/- mice. Together, these results suggested that the M-opsin-based color vision participated in the refractive development of mice. Overexposure to green light caused myopia, but less perception of the middle-wavelength components in white light promoted hyperopia in mice. Furthermore, possible dopaminergic signaling pathway was suggested in myopia induced by green light.
Collapse
Affiliation(s)
- Shunmei Ji
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Xiuyu Mao
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Yifan Zhang
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Lin Ye
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences (Fudan University), Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Eye & ENT Hospital Affiliated to Fudan University, Shanghai, China; Department of Ophthalmology, Zhongshan Hospital Affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
31
|
Tkatchenko TV, Tkatchenko AV. Genome-wide analysis of retinal transcriptome reveals common genetic network underlying perception of contrast and optical defocus detection. BMC Med Genomics 2021; 14:153. [PMID: 34107987 PMCID: PMC8190860 DOI: 10.1186/s12920-021-01005-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Refractive eye development is regulated by optical defocus in a process of emmetropization. Excessive exposure to negative optical defocus often leads to the development of myopia. However, it is still largely unknown how optical defocus is detected by the retina. METHODS Here, we used genome-wide RNA-sequencing to conduct analysis of the retinal gene expression network underlying contrast perception and refractive eye development. RESULTS We report that the genetic network subserving contrast perception plays an important role in optical defocus detection and emmetropization. Our results demonstrate an interaction between contrast perception, the retinal circadian clock pathway and the signaling pathway underlying optical defocus detection. We also observe that the relative majority of genes causing human myopia are involved in the processing of optical defocus. CONCLUSIONS Together, our results support the hypothesis that optical defocus is perceived by the retina using contrast as a proxy and provide new insights into molecular signaling underlying refractive eye development.
Collapse
Affiliation(s)
| | - Andrei V. Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY USA
- Edward S. Harkness Eye Institute, Research Annex Room 415, 635 W. 165th Street, New York, NY 10032 USA
| |
Collapse
|
32
|
Quint WH, Tadema KCD, de Vrieze E, Lukowicz RM, Broekman S, Winkelman BHJ, Hoevenaars M, de Gruiter HM, van Wijk E, Schaeffel F, Meester-Smoor M, Miller AC, Willemsen R, Klaver CCW, Iglesias AI. Loss of Gap Junction Delta-2 (GJD2) gene orthologs leads to refractive error in zebrafish. Commun Biol 2021; 4:676. [PMID: 34083742 PMCID: PMC8175550 DOI: 10.1038/s42003-021-02185-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 05/04/2021] [Indexed: 12/20/2022] Open
Abstract
Myopia is the most common developmental disorder of juvenile eyes, and it has become an increasing cause of severe visual impairment. The GJD2 locus has been consistently associated with myopia in multiple independent genome-wide association studies. However, despite the strong genetic evidence, little is known about the functional role of GJD2 in refractive error development. Here, we find that depletion of gjd2a (Cx35.5) or gjd2b (Cx35.1) orthologs in zebrafish, cause changes in the biometry and refractive status of the eye. Our immunohistological and scRNA sequencing studies show that Cx35.5 (gjd2a) is a retinal connexin and its depletion leads to hyperopia and electrophysiological changes in the retina. These findings support a role for Cx35.5 (gjd2a) in the regulation of ocular biometry. Cx35.1 (gjd2b) has previously been identified in the retina, however, we found an additional lenticular role. Lack of Cx35.1 (gjd2b) led to a nuclear cataract that triggered axial elongation. Our results provide functional evidence of a link between gjd2 and refractive error.
Collapse
Affiliation(s)
- Wim H Quint
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Kirke C D Tadema
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Erik de Vrieze
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Rachel M Lukowicz
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Sanne Broekman
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Beerend H J Winkelman
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Cerebellar Coordination and Cognition, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Melanie Hoevenaars
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Erwin van Wijk
- Department of Otorhinolaryngology, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank Schaeffel
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Magda Meester-Smoor
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Adam C Miller
- Institute of Neuroscience, University of Oregon, Eugene, United States
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Caroline C W Klaver
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Ophthalmology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Adriana I Iglesias
- Department of Ophthalmology, Erasmus Medical Center, Rotterdam, The Netherlands.
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
33
|
Angée C, Nedelec B, Erjavec E, Rozet JM, Fares Taie L. Congenital Microcoria: Clinical Features and Molecular Genetics. Genes (Basel) 2021; 12:genes12050624. [PMID: 33922078 PMCID: PMC8143514 DOI: 10.3390/genes12050624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 01/05/2023] Open
Abstract
Iris integrity is required to regulate both the amount of light reaching the retina and intraocular pressure (IOP), with elevated IOP being a major risk factor for glaucoma. Congenital microcoria (MCOR) is an extremely rare, autosomal dominant disease affecting iris development and hindering both of these functions. It is characterized by absent or underdeveloped dilator muscle fibers and immaturity of the iridocorneal angle—where the aqueous humor is drained—which play a central role in IOP regulation. The dilator muscle anomaly is manifested in pinhole pupils (<2 mm) and thin transilluminable irises, causing both hemeralopia and photoaversion. Axial myopia and juvenile open-angle glaucoma are very frequent (80% and 30% of all cases, respectively). It has been suggested that the immaturity of the chamber angle contributes to glaucoma, and myopia has been ascribed to photoaversion and elevated IOP. Though possible, these mechanisms are insufficient. The disease has been tied to chromosome 13q32.1 structural variations. In addition to compromising iris development, modification of the 13q32.1 architecture could alter signaling pathways for axial ocular length and IOP regulation. Here, we summarize the clinical, histological, and molecular features of this disease, and we discuss the possible etiology of associated anomalies.
Collapse
|
34
|
Sze YH, Zhao Q, Cheung JKW, Li KK, Tse DYY, To CH, Lam TC. High-pH reversed-phase fractionated neural retina proteome of normal growing C57BL/6 mouse. Sci Data 2021; 8:27. [PMID: 33500412 PMCID: PMC7838270 DOI: 10.1038/s41597-021-00813-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The retina is a key sensory tissue composed of multiple layers of cell populations that work coherently to process and decode visual information. Mass spectrometry-based proteomics approach has allowed high-throughput, untargeted protein identification, demonstrating the presence of these proteins in the retina and their involvement in biological signalling cascades. The comprehensive wild-type mouse retina proteome was prepared using a novel sample preparation approach, the suspension trapping (S-Trap) filter, and further fractionated with high-pH reversed phase chromatography involving a total of 28 injections. This data-dependent acquisition (DDA) approach using a Sciex TripleTOF 6600 mass spectrometer identified a total of 7,122 unique proteins (1% FDR), and generated a spectral library of 5,950 proteins in the normal C57BL/6 mouse retina. Data-independent acquisition (DIA) approach relies on a large and high-quality spectral library to analyse chromatograms, this spectral library would enable access to SWATH-MS acquisition to provide unbiased, multiplexed, and quantification of proteins in the mouse retina, acting as the most extensive reference library to investigate retinal diseases using the C57BL/6 mouse model.
Collapse
Affiliation(s)
- Ying Hon Sze
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
| | - Qian Zhao
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research, Hong Kong, China
| | - Jimmy Ka Wai Cheung
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
| | - King Kit Li
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
| | - Dennis Yan Yin Tse
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research, Hong Kong, China
| | - Chi Ho To
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China
- Centre for Eye and Vision Research, Hong Kong, China
| | - Thomas Chuen Lam
- Laboratory of Experimental Optometry, Centre for Myopia Research, School of Optometry, Hong Kong Polytechnic University, Hong Kong, China.
- Centre for Eye and Vision Research, Hong Kong, China.
| |
Collapse
|
35
|
Landis EG, Park HN, Chrenek M, He L, Sidhu C, Chakraborty R, Strickland R, Iuvone PM, Pardue MT. Ambient Light Regulates Retinal Dopamine Signaling and Myopia Susceptibility. Invest Ophthalmol Vis Sci 2021; 62:28. [PMID: 33502461 PMCID: PMC7846952 DOI: 10.1167/iovs.62.1.28] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Purpose Exposure to high-intensity or outdoor lighting has been shown to decrease the severity of myopia in both human epidemiological studies and animal models. Currently, it is not fully understood how light interacts with visual signaling to impact myopia. Previous work performed in the mouse retina has demonstrated that functional rod photoreceptors are needed to develop experimentally-induced myopia, alluding to an essential role for rod signaling in refractive development. Methods To determine whether dim rod-dominated illuminance levels influence myopia susceptibility, we housed male C57BL/6J mice under 12:12 light/dark cycles with scotopic (1.6 × 10−3 candela/m2), mesopic (1.6 × 101 cd/m2), or photopic (4.7 × 103 cd/m2) lighting from post-natal day 23 (P23) to P38. Half the mice received monocular exposure to −10 diopter (D) lens defocus from P28–38. Molecular assays to measure expression and content of DA-related genes and protein were conducted to determine how illuminance and lens defocus alter dopamine (DA) synthesis, storage, uptake, and degradation and affect myopia susceptibility in mice. Results We found that mice exposed to either scotopic or photopic lighting developed significantly less severe myopic refractive shifts (lens treated eye minus contralateral eye; –1.62 ± 0.37D and −1.74 ± 0.44D, respectively) than mice exposed to mesopic lighting (–3.61 ± 0.50D; P < 0.005). The 3,4-dihydroxyphenylacetic acid /DA ratio, indicating DA activity, was highest under photopic light regardless of lens defocus treatment (controls: 0.09 ± 0.011 pg/mg, lens defocus: 0.08 ± 0.008 pg/mg). Conclusions Lens defocus interacted with ambient conditions to differentially alter myopia susceptibility and DA-related genes and proteins. Collectively, these results show that scotopic and photopic lighting protect against lens-induced myopia, potentially indicating that a broad range of light levels are important in refractive development.
Collapse
Affiliation(s)
- Erica G Landis
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States
| | - Han Na Park
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Li He
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Curran Sidhu
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Ranjay Chakraborty
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States
| | - Ryan Strickland
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States
| | - P Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States.,Department of Pharmacology, Emory University, Atlanta, Georgia, United States
| | - Machelle T Pardue
- Department of Neuroscience, Emory University, Atlanta, Georgia, United States.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Atlanta, Georgia, United States.,Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, Georgia, United States
| |
Collapse
|
36
|
Tian L, Guo YT, Ying M, Liu YC, Li X, Wang Y. Co-existence of myopia and amblyopia in a guinea pig model with monocular form deprivation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:110. [PMID: 33569412 PMCID: PMC7867913 DOI: 10.21037/atm-20-5433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Form deprivation myopia is a type of ametropia, with identifiable causes in humans, that has been induced in many animals. The age of onset of myopia induced by monocular form deprivation coincides with the period of visual development in guinea pigs. However, visual acuity of form-deprived eyes in guinea pigs is not understood yet. In this study, we investigated whether monocular form deprivation would affect visual acuity in infant guinea pigs by evaluating the development of myopia and amblyopia after monocular form deprivation, and whether form deprivation myopia and amblyopia occurred simultaneously or successively. Methods Twenty pigmented guinea pigs (2 weeks old) were randomly assigned to two groups: monocularly form-deprived (n=10), in which facemasks modified from latex balloons covered the right eye, and normal controls (n=10). Refraction, axial length, and visual acuity were measured at 4 intervals (after 0, 1, 4, and 8 weeks of form deprivation), using cycloplegic streak retinoscopy, A-scan ultrasonography (with an oscillation frequency of 10 MHz), and sweep visual evoked potentials (sweep VEPs), respectively. Sweep VEPs were performed with correction of the induced myopic refractive error. Results Longer deprivation periods resulted in significant refractive errors in form-deprived eyes compared with those in contralateral and normal control eyes; the axial lengths of form-deprived eyes increased significantly after 4 and 8 weeks of form deprivation. These results revealed that myopia was established at 4 weeks. The acuity of form-deprived eyes was unchanged compared to that at the pretreatment time point, while that of contralateral eyes and eyes in normal control guinea pigs improved; there were significant differences between the deprived eyes and the other two open eyes from 1 to 8 weeks of form deprivation, showing that amblyopia was possibly established during 1 week of form deprivation. Conclusions This study demonstrated the feasibility of using sweep VEPs to estimate the visual acuity of guinea pigs. Further, our results revealed that amblyopia likely occurred earlier than myopia; amblyopia and myopia coexisted after a long duration of monocular form deprivation in guinea pigs. Understanding this relationship may help provide insights into failures of treatment of amblyopia associated with myopic anisometropia.
Collapse
Affiliation(s)
- Lu Tian
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| | - Ya-Tu Guo
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| | - Ming Ying
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| | - Yang-Chen Liu
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| | - Xuan Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| | - Yan Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China.,Tianjin Eye Institute, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Hospital, Tianjin, China
| |
Collapse
|
37
|
Muralidharan AR, Lança C, Biswas S, Barathi VA, Wan Yu Shermaine L, Seang-Mei S, Milea D, Najjar RP. Light and myopia: from epidemiological studies to neurobiological mechanisms. Ther Adv Ophthalmol 2021; 13:25158414211059246. [PMID: 34988370 PMCID: PMC8721425 DOI: 10.1177/25158414211059246] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/25/2021] [Indexed: 12/22/2022] Open
Abstract
Myopia is far beyond its inconvenience and represents a true, highly prevalent, sight-threatening ocular condition, especially in Asia. Without adequate interventions, the current epidemic of myopia is projected to affect 50% of the world population by 2050, becoming the leading cause of irreversible blindness. Although blurred vision, the predominant symptom of myopia, can be improved by contact lenses, glasses or refractive surgery, corrected myopia, particularly high myopia, still carries the risk of secondary blinding complications such as glaucoma, myopic maculopathy and retinal detachment, prompting the need for prevention. Epidemiological studies have reported an association between outdoor time and myopia prevention in children. The protective effect of time spent outdoors could be due to the unique characteristics (intensity, spectral distribution, temporal pattern, etc.) of sunlight that are lacking in artificial lighting. Concomitantly, studies in animal models have highlighted the efficacy of light and its components in delaying or even stopping the development of myopia and endeavoured to elucidate possible mechanisms involved in this process. In this narrative review, we (1) summarize the current knowledge concerning light modulation of ocular growth and refractive error development based on studies in human and animal models, (2) summarize potential neurobiological mechanisms involved in the effects of light on ocular growth and emmetropization and (3) highlight a potential pathway for the translational development of noninvasive light-therapy strategies for myopia prevention in children.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Dan Milea
- Singapore Eye Research Institute, Singapore
| | - Raymond P Najjar
- Visual Neurosciences Group, Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower Level 6, Singapore 169856
| |
Collapse
|
38
|
Zhao F, Li Q, Chen W, Zhu H, Zhou D, Reinach PS, Yang Z, He M, Xue A, Wu D, Liu T, Fu Q, Zeng C, Qu J, Zhou X. Dysfunction of VIPR2 leads to myopia in humans and mice. J Med Genet 2020; 59:88-100. [DOI: 10.1136/jmedgenet-2020-107220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/27/2020] [Accepted: 11/01/2020] [Indexed: 01/01/2023]
Abstract
BackgroundMyopia is the leading cause of refractive errors. As its pathogenesis is poorly understood, we determined if the retinal VIP-VIPR2 signalling pathway axis has a role in controlling signalling output that affects myopia development in mice.MethodsAssociation analysis meta-study, single-cell transcriptome, bulk RNA sequencing, pharmacological manipulation and VIPR2 gene knockout studies were used to clarify if changes in the VIP-VIPR2 signalling pathway affect refractive development in mice.ResultsThe SNP rs6979985 of the VIPR2 gene was associated with high myopia in a Chinese Han cohort (randomceffect model: p=0.013). After either 1 or 2 days’ form deprivation (FD) retinal VIP mRNA expression was downregulated. Retinal single-cell transcriptome sequencing showed that VIPR2 was expressed mainly by bipolar cells. Furthermore, the cAMP signalling pathway axis was inhibited in some VIPR2+ clusters after 2 days of FD. The selective VIPR2 antagonist PG99-465 induced relative myopia, whereas the selective VIPR2 agonist Ro25-1553 inhibited this response. In Vipr2 knockout (Vipr2-KO) mice, refraction was significantly shifted towards myopia (p<0.05). The amplitudes of the bipolar cell derived b-waves in 7-week-old Vipr2-KO mice were significantly larger than those in their WT littermates (p<0.05).ConclusionsLoss of VIPR2 function likely compromises bipolar cell function based on presumed changes in signal transduction due to altered signature electrical wave activity output in these mice. As these effects correspond with increases in form deprivation myopia (FDM), the VIP-VIPR2 signalling pathway axis is a viable novel target to control the development of this condition.
Collapse
|
39
|
Strickland R, Landis EG, Pardue MT. Short-Wavelength (Violet) Light Protects Mice From Myopia Through Cone Signaling. Invest Ophthalmol Vis Sci 2020; 61:13. [PMID: 32049342 PMCID: PMC7326482 DOI: 10.1167/iovs.61.2.13] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Exposure to short-wavelength light influences refractive development and inhibits myopic development in many animal models. Retinal mechanisms underlying this response remain unknown. This study used a mouse model of lens-induced myopia to evaluate the effect of different wavelength light on refractive development and dopamine levels in the retina. A possible retinal pathway is tested using a mutant mouse with dysfunctional cones. Methods Wild-type C57BL/6J (WT) and ALS/LtJ/Gnat2cpfl3 (Gnat2−/−) mice were exposed to one of three different light conditions beginning at postnatal day 28: broad-spectrum “white” (420-680 nm), medium wavelength “green” (525 ± 40 nm), and short wavelength “violet” (400 ± 20 nm). One-half of the mice received hyperopic lens defocus. All mice were exposed to the light for 4 weeks; animals were measured weekly for refractive error and axial parameters. Retinal dopamine and the dopamine metabolite 3,4-dihydroxyphenylacetic acid were measured by HPLC. Results In WT mice, short-wavelength violet light induced hyperopia and violet light inhibited lens-induced myopia when compared with mice exposed to white light. Hyperopia could be attributed to shallower vitreous chambers in WT animals. There were no changes in the levels of dopamine or its metabolite. In Gnat2−/− mice, violet light did not induce hyperopia or inhibit lens-induced myopia. Conclusions These findings show that short-wavelength light slows refractive eye growth, producing hyperopic responses in mice and inhibiting lens-induced myopia. The lack of inhibition in mice with dysfunctional cones suggests that cone signaling plays a role in the hyperopic response to short-wavelength (violet) light.
Collapse
|
40
|
Increased endogenous dopamine prevents myopia in mice. Exp Eye Res 2020; 193:107956. [PMID: 32032629 DOI: 10.1016/j.exer.2020.107956] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/31/2020] [Accepted: 02/01/2020] [Indexed: 02/01/2023]
Abstract
Experimental evidence suggests that dopamine (DA) modulates refractive eye growth. We evaluated whether increasing endogenous DA activity using pharmacological or genetic approaches decreased myopia susceptibility in mice. First, we assessed the effects of systemic L-3,4-dihydroxyphenylalanine (L-DOPA) injections on form deprivation myopia (FDM) in C57BL/6 J (WTC57) mice. WTC57 mice received daily systemic injections of L-DOPA (n = 11), L-DOPA + ascorbic acid (AA, n = 22), AA (n = 20), or Saline (n = 16). Second, we tested transgenic mice with increased or decreased expression of vesicular monoamine transporter 2 (VMAT2HI, n = 22; WTHI, n = 18; VMAT2LO, n = 18; or WTLO, n = 9) under normal and form deprivation conditions. VMAT2 packages DA into vesicles, affecting DA release. At post-natal day 28 (P28), monocular FD was induced in each genotype. Weekly measurements of refractive error, corneal curvature, and ocular biometry were performed until P42 or P49. WTC57 mice exposed to FD developed a significant myopic shift (treated-contralateral eye) in AA (-3.27 ± 0.73D) or saline (-3.71 ± 0.80D) treated groups that was significantly attenuated by L-DOPA (-0.73 ± 0.90D, p = 0.0002) or L-DOPA + AA (-0.11 ± 0.46D, p = 0.0103). The VMAT2LO mice, with under-expression of VMAT2, were most susceptible to FDM. VMAT2LO mice developed significant myopic shifts to FD after one week compared to VMAT2HI and WT mice (VMAT2LO: -5.48 ± 0.54D; VMAT2HI: -0.52 ± 0.92D, p < 0.05; WT: -2.13 ± 0.78D, p < 0.05; ungoggled control: -0.22 ± 0.24D, p < 0.001). These results indicate that endogenously increasing DA synthesis and release by genetic and pharmacological methods prevents FDM in mice.
Collapse
|
41
|
Wang M, Aleman AC, Schaeffel F. Probing the Potency of Artificial Dynamic ON or OFF Stimuli to Inhibit Myopia Development. Invest Ophthalmol Vis Sci 2019; 60:2599-2611. [PMID: 31219534 DOI: 10.1167/iovs.18-26471] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To determine whether equiluminant artificial dynamic ON or OFF stimuli on a computer screen can induce bidirectional changes in choroidal thickness (ChTh) in both humans and chickens, and whether such changes are associated with bidirectional changes in retinal dopamine release in chickens. Methods Experiment 1: Before and after ON or OFF stimulation for 1 hour, ChTh was measured with optical coherence tomography (OCT). Experiment 2: chicks (n = 14) were raised under ON or OFF stimulation for 3 hours. ChTh was determined by OCT. Experiment 3: chicks were raised for 7 days either under room light (500 lux, n = 11), dynamic ON stimulus (700 lux, n = 15), or dynamic OFF stimulus (700 lux, n = 7). In addition, negative lenses were attached to their right eyes. After experiments 2 and 3, retinal and vitreal dopamine (DA), and its metabolites, were measured by HPLC-electrochemical detection. Results Experiment 1: Dynamic ON stimuli caused thicker choroids (+5.3 ± 2.0 μm), whereas OFF stimuli caused choroidal thinning (-4.7 ± 0.5 μm) (right eye data only, P < 0.001). Experiment 2: After 3 hours, chickens developed thicker choroids with ON stimuli (+37.4 ± 12.4 μm) and thinner choroids with OFF stimuli (-11.3 ± 3.6 μm, difference P < 0.01). Vitreal DA, 3-methoxytyramine, and homovanillic acid levels were elevated after ON stimulation, compared with the OFF (P < 0.05). Experiment 3: After 7 days, chickens with lenses developed more myopia both with ON and OFF stimulation, compared with room light. ON stimulation increased vitreal DA compared with OFF. Conclusions Artificial dynamic ON or OFF stimuli had similar effects on ChTh in humans and chickens, but more work will be necessary to determine whether such stimuli can be used as novel interventions of myopia.
Collapse
Affiliation(s)
- Min Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Hunan Province, China.,Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| | - Andrea C Aleman
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, Germany
| |
Collapse
|
42
|
Tkatchenko TV, Tkatchenko AV. Pharmacogenomic Approach to Antimyopia Drug Development: Pathways Lead the Way. Trends Pharmacol Sci 2019; 40:833-852. [PMID: 31676152 DOI: 10.1016/j.tips.2019.09.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 09/04/2019] [Accepted: 09/16/2019] [Indexed: 12/29/2022]
Abstract
Myopia is the most common eye disorder in the world which is caused by a mismatch between the optical power of the eye and its excessively long axial length. Recent studies revealed that the regulation of the axial length of the eye occurs via a complex signaling cascade, which originates in the retina and propagates across all ocular tissues to the sclera. The complexity of this regulatory cascade has made it particularly difficult to develop effective antimyopia drugs. The current pharmacological treatment options for myopia are limited to atropine and 7-methylxanthine, which have either significant adverse effects or low efficacy. In this review, we focus on the recent advances in genome-wide studies of the signaling pathways underlying myopia development and discuss the potential of systems genetics and pharmacogenomic approaches for the development of antimyopia drugs.
Collapse
Affiliation(s)
| | - Andrei V Tkatchenko
- Department of Ophthalmology, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
43
|
Gong X, Wu XH, Liu AL, Qian KW, Li YY, Ma YY, Huang F, Wang Q, Wu H, Zhou X, Qu J, Yuan F, Zhong YM, Yang XL, Weng SJ. Optic nerve crush modulates refractive development of the C57BL/6 mouse by changing multiple ocular dimensions. Brain Res 2019; 1726:146537. [PMID: 31672473 DOI: 10.1016/j.brainres.2019.146537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 10/17/2019] [Accepted: 10/26/2019] [Indexed: 01/16/2023]
Abstract
Higher visual centers could modulate visually-guided ocular growth, in addition to local mechanisms intrinsic to the eye. There is evidence that such central modulations could be species (even subspecies)-dependent. While the mouse has recently become an important experimental animal in myopia studies, it remains unclear whether and how visual centers modulate refractive development in mice, an issue that was examined in the present study. We found that optic nerve crush (ONC), performed at P18, could modify normal refractive development in the C57BL/6 mouse raised in normal visual environment. Unexpectedly, sham surgery caused a steeper cornea, leading to a modest myopic refractive shift, but did not induce significant changes in ocular axis length. ONC caused corneal flattening and re-calibrated the refractive set-point in a bidirectional manner, causing significant myopic (<-3 D, 54.5%) or hyperopic (>+3 D, 18.2%) shifts in refractive error in most (totally 72.7%) animals, both due to changes in ocular axial length. ONC did not change the density of dopaminergic amacrine cells, but increased retinal levels of dopamine and DOPAC. We conclude that higher visual centers are likely to play a role in fine-tuning of ocular growth, thus modifying refractive development in the C57BL/6 mouse. The changes in refractive error induced by ONC are accounted for by alternations in multiple ocular dimensions, including corneal curvature and axial length.
Collapse
Affiliation(s)
- Xue Gong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Hua Wu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China; Discipline of Neuroscience and Department of Anatomy and Physiology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ai-Lin Liu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kang-Wei Qian
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun-Yun Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuan-Yuan Ma
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Furong Huang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiongsi Wang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hao Wu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangtian Zhou
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fei Yuan
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong-Mei Zhong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiong-Li Yang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shi-Jun Weng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology and Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
44
|
Xu C, Wu J, Wang J. Associations of rs524952 and rs634990 gene polymorphisms in 15q14 with high myopia: A meta-analysis. Mol Vis 2019; 25:603-609. [PMID: 31673225 PMCID: PMC6798703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 10/12/2019] [Indexed: 11/09/2022] Open
Abstract
Purpose Many studies have been conducted to investigate the association between the rs524952 and rs634990 polymorphisms and high myopia (HM). However, the results were conflicting. Thus, a meta-analysis was needed to reveal the real association between the two single nucleotide polymorphisms (SNPs) and HM. Methods All eligible studies published in Pubmed, Embase, China Biologic Medicine (CBM), the China National Knowledge Infrastructure (CNKI), the Cochrane Library, and the Web of Science from 2010 to March 2019 were examined. Results Six comparison groups in four studies with 5,293 subjects for the rs524952 polymorphism and five studies with 6,750 subjects for the rs634990 polymorphism were included. No statistically significant associations were observed between the rs524952 and rs634990 polymorphisms and HM under the allelic model, recessive genetic model, and dominant genetic model in this meta-analysis. Subgroup analysis was conducted by dividing the studies into two groups according to the case sample size, which showed that the association between the rs524952 polymorphism and HM was found only in a subgroup of fewer than 300 cases under the dominant genetic model (OR=0.64; 95% confidence interval [CI]:0.43-0.96). Sensitivity analysis for the rs524952 polymorphism suggested the results of this study were stable under all the genetic models. However, the association between the rs634990 polymorphism and HM turned out to be statistically significant in the allelic, recessive, and dominant genetic models after the omission of Qiang et al.'s study. No publication bias was found. Conclusions The results of this meta-analysis suggested the rs524952 and rs634990 polymorphisms may have nothing to do with the development of HM. The present results must be confirmed with larger-scale studies in the future.
Collapse
|
45
|
Altered ocular parameters from circadian clock gene disruptions. PLoS One 2019; 14:e0217111. [PMID: 31211778 PMCID: PMC6581257 DOI: 10.1371/journal.pone.0217111] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
The pathophysiology of refractive errors is poorly understood. Myopia (nearsightedness) in particular both blurs vision and predisposes the eye to many blinding diseases during adulthood. Based on past findings of diurnal variations in the dimensions of the eyes of humans and other vertebrates, altered diurnal rhythms of these ocular dimensions with experimentally induced myopia, and evolving evidence that ambient light exposures influence refractive development, we assessed whether disturbances in circadian signals might alter the refractive development of the eye. In mice, retinal-specific knockout of the clock gene Bmal1 induces myopia and elongates the vitreous chamber, the optical compartment separating the lens and the retina. These alterations simulate common ocular findings in clinical myopia. In Drosophila melanogaster, knockouts of the clock genes cycle or period lengthen the pseudocone, the optical component of the ommatidium that separates the facet lens from the photoreceptors. Disrupting circadian signaling thus alters optical development of the eye in widely separated species. We propose that mechanisms of myopia include circadian dysregulation, a frequent occurrence in modern societies where myopia also is both highly prevalent and increasing at alarming rates. Addressing circadian dysregulation may improve understanding of the pathogenesis of refractive errors and introduce novel therapeutic approaches to ameliorate myopia development in children.
Collapse
|
46
|
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: 250] [Impact Index Per Article: 41.7] [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
|
47
|
Szczerkowska KI, Petrezselyova S, Lindovsky J, Palkova M, Dvorak J, Makovicky P, Fang M, Jiang C, Chen L, Shi M, Liu X, Zhang J, Kubik-Zahorodna A, Schuster B, Beck IM, Novosadova V, Prochazka J, Sedlacek R. Myopia disease mouse models: a missense point mutation (S673G) and a protein-truncating mutation of the Zfp644 mimic human disease phenotype. Cell Biosci 2019; 9:21. [PMID: 30834109 PMCID: PMC6385473 DOI: 10.1186/s13578-019-0280-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/01/2019] [Indexed: 12/24/2022] Open
Abstract
Zinc finger 644 (Zfp644 in mouse, ZNF644 in human) gene is a transcription factor whose mutation S672G is considered a potential genetic factor of inherited high myopia. ZNF644 interacts with G9a/GLP complex, which functions as a H3K9 methyltransferase to silence transcription. In this study, we generated mouse models to unravel the mechanisms leading to symptoms associated with high myopia. Employing TALEN technology, two mice mutants were generated, either with the disease-carrying mutation (Zfp644S673G) or with a truncated form of Zfp644 (Zfp644Δ8). Eye morphology and visual functions were analysed in both mutants, revealing a significant difference in a vitreous chamber depth and lens diameter, however the physiological function of retina was preserved as found under the high-myopia conditions. Our findings prove that ZNF644/Zfp644 is involved in the development of high-myopia, indicating that mutations such as, Zfp644S673G and Zfp644Δ8 are causative for changes connected with the disease. The developed models represent a valuable tool to investigate the molecular basis of myopia pathogenesis and its potential treatment.
Collapse
Affiliation(s)
- Katarzyna I Szczerkowska
- 1Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics CAS, Prumyslova 595, Vestec, 252 50 Prague, Czech Republic
| | - Silvia Petrezselyova
- 1Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics CAS, Prumyslova 595, Vestec, 252 50 Prague, Czech Republic.,2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Jiri Lindovsky
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Marcela Palkova
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Jan Dvorak
- 1Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics CAS, Prumyslova 595, Vestec, 252 50 Prague, Czech Republic
| | - Peter Makovicky
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Mingyan Fang
- 3Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden.,4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Chongyi Jiang
- 4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Lingyan Chen
- 4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Mingming Shi
- 4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Xiao Liu
- 4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | - Jianguo Zhang
- 4BGI-Shenzhen, Shenzhen, 518083 China.,5China National GeneBank, BGI-Shenzhen, Shenzhen, 518120 China
| | | | - Bjoern Schuster
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Inken M Beck
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic.,6Animal Research Center, Ulm University, Ulm, Germany
| | - Vendula Novosadova
- 2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Jan Prochazka
- 1Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics CAS, Prumyslova 595, Vestec, 252 50 Prague, Czech Republic.,2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| | - Radislav Sedlacek
- 1Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics CAS, Prumyslova 595, Vestec, 252 50 Prague, Czech Republic.,2Czech Centre for Phenogenomics, Institute of Molecular Genetics CAS, Prague, Czech Republic
| |
Collapse
|
48
|
Murphy MJ, Riddell N, Crewther DP, Simpson D, Crewther SG. Temporal whole field sawtooth flicker without a spatial component elicits a myopic shift following optical defocus irrespective of waveform direction in chicks. PeerJ 2019; 7:e6277. [PMID: 30697484 PMCID: PMC6347968 DOI: 10.7717/peerj.6277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 12/11/2018] [Indexed: 01/17/2023] Open
Abstract
Purpose Myopia (short-sightedness) is the commonest visual disorder and greatest risk factor for sight threatening secondary pathologies. Myopia and hyperopia can be induced in animal models by rearing with optical lens defocus of opposite sign. The degree of refractive compensation to lens-induced defocus in chicks has been shown to be modified by directionally drifting sawtooth spatio-temporal luminance diamond plaids, with Fast-ON sawtooth spatio-temporal luminance profiles inhibiting the myopic shift in response to negative lenses, and Fast-OFF profiles inhibiting the hyperopic shift in response to positive lenses. What is unknown is whether similar sign-of-defocus dependent results produced by spatio-temporal modulation of sawtooth patterns could be achieved by rearing chicks under whole field low temporal frequency sawtooth luminance profiles at 1 or 4 Hz without a spatial component, or whether such stimuli would indiscriminately elicit a myopic shift such as that previously shown with symmetrical (or near-symmetrical) low frequency flicker across a range of species. Methods Hatchling chicks (n = 166) were reared from days five to nine under one of three defocus conditions (No Lens, +10D lens, or -10D lens) and five light conditions (No Flicker, 1 Hz Fast-ON/Slow-OFF sawtooth flicker, 4 Hz Fast-ON/Slow-OFF sawtooth flicker, 1 Hz Fast-OFF/Slow-ON sawtooth flicker, or 4Hz Fast-OFF/Slow-ON sawtooth flicker). The sawtooth flicker was produced by light emitting diodes (white LEDs, 1.2 -183 Lux), and had no measurable dark phase. Biometrics (refraction and ocular axial dimensions) were measured on day nine. Results Both 1 Hz and 4 Hz Fast-ON and Fast-OFF sawtooth flicker induced an increase in vitreous chamber depth that was greater in the presence of negative compared to positive lens defocus. Both sawtooth profiles at both temporal frequencies inhibited the hyperopic shift in response to +10D lenses, whilst full myopic compensation (or over-compensation) in response to -10D lenses was observed. Conclusions Whole field low temporal frequency Fast-ON and Fast-OFF sawtooth flicker induces a generalized myopic shift, similar to that previously shown for symmetrical sine-wave and square-wave flicker. Our findings highlight that temporal modulation of retinal ON/OFF pathways per se (without a spatial component) is insufficient to produce strong sign-of-defocus dependent effect.
Collapse
Affiliation(s)
- Melanie J Murphy
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - Nina Riddell
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| | - David P Crewther
- Centre for Human Psychopharmacology, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - David Simpson
- Brain Sciences Institute, Swinburne University of Technology, Melbourne, Victoria, Australia
| | - Sheila G Crewther
- School of Psychology & Public Health, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
49
|
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: 2.6] [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
|
50
|
Riddell N, Faou P, Crewther SG. Short term optical defocus perturbs normal developmental shifts in retina/RPE protein abundance. BMC DEVELOPMENTAL BIOLOGY 2018; 18:18. [PMID: 30157773 PMCID: PMC6116556 DOI: 10.1186/s12861-018-0177-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 08/16/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Myopia (short-sightedness) affects approximately 1.4 billion people worldwide, and prevalence is increasing. Animal models induced by defocusing lenses show striking similarity with human myopia in terms of morphology and the implicated genetic pathways. Less is known about proteome changes in animals. Thus, the present study aimed to improve understanding of protein pathway responses to lens defocus, with an emphasis on relating expression changes to no lens control development and identifying bidirectional and/or distinct pathways across myopia and hyperopia (long-sightedness) models. RESULTS Quantitative label-free proteomics and gene set enrichment analysis (GSEA) were used to examine protein pathway expression in the retina/RPE of chicks following 6 h and 48 h of myopia induction with - 10 dioptre (D) lenses, hyperopia induction with +10D lenses, or normal no lens rearing. Seventy-one pathways linked to cell development and neuronal maturation were differentially enriched between 6 and 48 h in no lens chicks. The majority of these normal developmental changes were disrupted by lens-wear (47 of 71 pathways), however, only 11 pathways displayed distinct expression profiles across the lens conditions. Most notably, negative lens-wear induced up-regulation of proteins involved in ATP-driven ion transport, calcium homeostasis, and GABA signalling between 6 and 48 h, while the same proteins were down-regulated over time in normally developing chicks. Glutamate and bicarbonate/chloride transporters were also down-regulated over time in normally developing chicks, and positive lens-wear inhibited this down-regulation. CONCLUSIONS The chick retina/RPE proteome undergoes extensive pathway expression shifts during normal development. Most of these pathways are further disrupted by lens-wear. The identified expression patterns suggest close interactions between neurotransmission (as exemplified by increased GABA receptor and synaptic protein expression), cellular ion homeostasis, and associated energy resources during myopia induction. We have also provided novel evidence for changes to SLC-mediated transmembrane transport during hyperopia induction, with potential implications for signalling at the photoreceptor-bipolar synapse. These findings reflect a key role for perturbed neurotransmission and ionic homeostasis in optically-induced refractive errors, and are predicted by our Retinal Ion Driven Efflux (RIDE) model.
Collapse
Affiliation(s)
- Nina Riddell
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Plenty Rd., Bundoora, Melbourne, VIC, 3083, Australia.
| | - Pierre Faou
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, VIC, Australia
| | - Sheila G Crewther
- Department of Psychology and Counselling, School of Psychology and Public Health, La Trobe University, Plenty Rd., Bundoora, Melbourne, VIC, 3083, Australia
| |
Collapse
|