1
|
Charoenpipatsin N, Yothachai P, Nuntawisuttiwong N, Wongpraparut O, Choosri P, Silpa-archa N. Dosimetry Assessment of Potential Hazard from Visible Light, Especially Blue Light, Emitted by Screen of Devices in Daily Use. Clin Cosmet Investig Dermatol 2025; 18:169-176. [PMID: 39867977 PMCID: PMC11761154 DOI: 10.2147/ccid.s490977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025]
Abstract
Visible light has been considered to have minimal impact on the skin. However, the increasing use of electronic devices has led to a significant increase in exposure to visible light, especially blue light. We measured the irradiance (mW/cm2) and estimated dose (J/cm2) of visible light and blue light emitted from various electronic devices including smartphones, tablets and computers. The measurement was done in normal screen mode and night shift mode at different brightness levels and distances across six screens. The irradiance and dose of visible light and blue light corresponded to the brightness, distance, and screen size of the devices. This study has shown that the irradiance and dose of visible light and blue light emitted from electronic devices in daily use are small and unlikely to be harmful to human skin.
Collapse
Affiliation(s)
- Norramon Charoenpipatsin
- Photodermatology Unit, Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Punyanut Yothachai
- Photodermatology Unit, Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nuttaporn Nuntawisuttiwong
- Photodermatology Unit, Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Pranomkorn Choosri
- Department of Biomedical Engineering, College of Health Sciences, Christian University of Thailand, Nakhon Pathom, Thailand
| | - Narumol Silpa-archa
- Photodermatology Unit, Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
2
|
Liu H, Schaeffel F, Feldkaemper MP. Effects of computer-generated patterns with different temporal and spatial frequencies on choroidal thickness, retinal dopamine and candidate genes in chickens wearing lenses. Front Med (Lausanne) 2024; 11:1469275. [PMID: 39720655 PMCID: PMC11666368 DOI: 10.3389/fmed.2024.1469275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/23/2024] [Indexed: 12/26/2024] Open
Abstract
Purpose Changes in choroidal thickness (ChT) are proposed to predict myopia development but evidence is mixed. We investigated time courses of choroidal responses, following different types of dynamic artificial stimulation in chicks with and without spectacle lenses, as well as changes in retinal dopamine metabolism and expression of candidate genes. Methods Chicks were kept in an arena surrounded by computer monitors presenting dynamic checkerboard fields of small, medium and large size. Fields were displayed with different cycle frequencies, as ON (rapid rise, slow decay) or OFF (slow rise, rapid decay) temporal luminance profile. Refractive errors, ocular biometry and ChT were assessed. Dopamine metabolism and candidate gene expression levels were also measured. Stimuli were applied for (1) 3 h with no lens, (2) 3 h and monocular treatment with -7D or +7D lenses, (3) 3 or 7 days. Results (1) The smallest fields caused the largest decrease in ChT. (2) Negative lens treatment induced on average 11.7 μm thinner choroids. ChT thinning was enhanced by 10 Hz-ON medium field size flicker which also reduced choroidal thickening with positive lenses. (3) With prolonged treatment, the choroid recovered from initial thinning in all groups although to varying degrees which were dependent on stimulus parameters. Relative ChT changes were positively correlated with the vitreal level of dopamine metabolites. Retinal EGR-1 mRNA level was positively correlated with choroidal thickness. Retinal melanopsin mRNA was increased by 10 Hz-ON stimulation and choroidal BMPR1A mRNA increased with 10 Hz-OFF stimulation. On average, early choroidal thinning did not predict the amount of negative lens-induced eye growth changes after 7 days, whereas later ChT changes showed a weak association. Conclusion Negative lenses caused long-lasting choroidal thinning, with some recovery during lens wear, especially after stimulation with 10 Hz. The dynamic stimuli modulated choroidal thinning but effects were small. There was little difference between ON and OFF stimulation, perhaps because the checkerboard patterns were too coarse. 10 Hz cycle frequency increased dopamine release. Less dopamine was correlated with thinner choroids. Result do not exclude a predictive value of choroidal thickening for future refractive development since we almost exclusively tested choroidal thinning effects.
Collapse
Affiliation(s)
- Hong Liu
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
- Aier Institute of Optometry and Vision Science, Aier Eye Hospital Group, Changsha, China
| | - Frank Schaeffel
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
| | - Marita Pauline Feldkaemper
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Du Y, Pang M, Chen H, Zhou X, Geng R, Zhang Y, Yang L, Li J, Han Y, Liu J, Zhang R, Bi H, Guo D. Inhibitory effect of Zhujing Pill on myopia progression: Mechanistic insights based on metabonomics and network pharmacology. PLoS One 2024; 19:e0312379. [PMID: 39625993 PMCID: PMC11614212 DOI: 10.1371/journal.pone.0312379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/02/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES This study endeavored to uncover the mechanisms by which Zhujing pill (ZJP) slows myopia progression. METHODS We employed biometric analyses to track diopter and axial length changes in guinea pigs with negative lens-induced myopia (LIM). Through integrating metabonomics and network pharmacology, we aimed to predict the anti-myopic targets and active ingredients of ZJP. Subsequent analysis, including real-time fluorescent quantitative PCR (qPCR) and Western blotting (WB), assessed the expression levels of CHRNA7, LPCAT1, and NOS2 in retinal tissues. KEY FINDINGS Our findings demonstrate that ZJP significantly mitigates diopter increase and axial elongation in LIM guinea pigs. Metabonomic analysis revealed significant changes in 13 serum metabolites, with ZJP reversing the expression of 5 key metabolites. By integrating metabonomics with network pharmacology, we identified core targets of ZJP against myopia and constructed a compound-gene-disease-metabolite network. The expressions of LPCAT1 and CHRNA7 were found to decrease in the LIM group but increase with ZJP treatment, whereas NOS2 expression showed the opposite pattern. CONCLUSIONS This investigation provides the first evidence of ZJP's multifaceted effectiveness in managing myopia, highlighting its impact on multiple components, targets, and pathways, including the novel involvement of LPCAT1 and CHRNA7 in myopia pathogenesis.
Collapse
Affiliation(s)
- Yongle Du
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengran Pang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haoyu Chen
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangkun Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruyue Geng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linqi Yang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawen Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yufeng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinpeng Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Amorim-de-Sousa A, Chakraborty R, Collins MJ, Fernandes P, González-Méijome J, Hannibal J, Hoseini-Yazdi H, Read SA, Ellrich J, Schilling T. Blue light stimulation of the blind spot in human: from melanopsin to clinically relevant biomarkers of myopia. Bioelectron Med 2024; 10:26. [PMID: 39491000 PMCID: PMC11533427 DOI: 10.1186/s42234-024-00159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024] Open
Abstract
The protective effects of time spent outdoors emphasize the major role of daylight in myopia. Based on the pathophysiology of myopia, the impact of blue light stimulation on the signaling cascade, from melanopsin at the blind spot to clinically relevant biomarkers for myopia, was investigated. Parameters and site of light stimulation are mainly defined by the photopigment melanopsin, that is sensitive to blue light with a peak wavelength of 480 nm and localized on the intrinsically photosensitive retinal ganglion cells (ipRGC) whose axons converge to the optic disc, corresponding to the physiological blind spot. Blue light at the blind spot (BluSpot) stimulation provides the opportunity to activate the vast majority of ipRGC and avoids additional involvement of rods and cones which may exert incalculable effects on the signaling cascade.Experimental studies have applied anatomical, histochemical, electrophysiological, imaging, and psychophysical methods to unravel the mode of action of BluSpot stimulation. Results indicate activation of melanopsin, improvement of contrast sensitivity, gain in electrical retinal activity, and increase of choroidal thickness following BluSpot stimulation. Short-term changes of clinically relevant biomarkers lead to the hypothesis that BluSpot stimulation may exert antimyopic effects with long-term application.
Collapse
Affiliation(s)
- Ana Amorim-de-Sousa
- Clinical & Experimental Optometry Research Lab, Physics Center of Minho and Porto Universities, University of Minho, Braga, Portugal
| | - Ranjay Chakraborty
- Myopia and Visual Development Lab, Flinders University College of Nursing and Health Sciences, Bedford Park, South Australia, Australia
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia
| | - Paulo Fernandes
- Clinical & Experimental Optometry Research Lab, Physics Center of Minho and Porto Universities, University of Minho, Braga, Portugal
| | - José González-Méijome
- Clinical & Experimental Optometry Research Lab, Physics Center of Minho and Porto Universities, University of Minho, Braga, Portugal
| | - Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg and Frederiksberg Hospital, Institute of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hosein Hoseini-Yazdi
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia
| | - Scott A Read
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, Australia
| | - Jens Ellrich
- Dopavision GmbH, Pfuelstrasse 5, 10997, Berlin, Germany
- Medical Faculty, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Tim Schilling
- Dopavision GmbH, Pfuelstrasse 5, 10997, Berlin, Germany.
| |
Collapse
|
5
|
Ling Y, Wang Y, Ye J, Luan C, Bi A, Gu Y, Shi X. Changes in Intrinsically Photosensitive Retinal Ganglion Cells, Dopaminergic Amacrine Cells, and Their Connectivity in the Retinas of Lid Suture Myopia. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39230992 PMCID: PMC11379095 DOI: 10.1167/iovs.65.11.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2024] Open
Abstract
Purpose This study investigates alterations in intrinsically photosensitive retinal ganglion cells (ipRGCs) and dopaminergic amacrine cells (DACs) in lid suture myopia (LSM) rats. Methods LSM was induced in rats by suturing the right eyes for 4 weeks. Double immunofluorescence staining of ipRGCs and DACs in whole-mount retinas was performed to analyze changes in the density and morphology of control, LSM, and fellow eyes. Real-time quantitative PCR and Western blotting were used to detect related genes and protein expression levels. Results Significant myopia was induced in the lid-sutured eye, but the fellow eye was not different to control. Decreased ipRGC density with paradoxically increased overall melanopsin expression and enlarged dendritic beads was observed in both the LSM and fellow eyes of the LSM rat retinas. In contrast, DAC changes occurred only in the LSM eyes, with reduced DAC density and tyrosine hydroxylase (TH) expression, sparser dendritic processes, and fewer varicosities. Interestingly, contacts between ipRGCs and DACs in the inner plexiform layer (IPL) and the expression of pituitary adenylate cyclase-activating polypeptide (PACAP) and vesicular monoamine transporter protein 2 (VMAT2) mRNA were decreased in the LSM eyes. Conclusions The ipRGCs and DACs in LSM rat retinas undergo multiple alterations in density, morphology, and related molecule expressions. However, the ipRGC changes alone appear not to be required for the development of myopia, given that myopia is only induced in the lid-sutured eye, and they are unlikely alone to drive the DAC changes. Reduced contacts between ipRGCs and DACs in the LSM eyes may be the structural foundation for the impaired signaling between them. PACAP and VMAT2, strongly associated with ipRGCs and DACs, may play important roles in LSM through complex mechanisms.
Collapse
Affiliation(s)
- Ying Ling
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yao Wang
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Jingjing Ye
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changlin Luan
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ailing Bi
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Gu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Xuefeng Shi
- Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Tianjin, China
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
- Institute of Ophthalmology, Nankai University, Tianjin, China
| |
Collapse
|
6
|
Zheleznyak L, Liu C, Winter S. Chromatic cues for the sign of defocus in the peripheral retina. BIOMEDICAL OPTICS EXPRESS 2024; 15:5098-5114. [PMID: 39296412 PMCID: PMC11407258 DOI: 10.1364/boe.537268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 09/21/2024]
Abstract
Detecting optical defocus at the retina is crucial for accurate accommodation and emmetropization. However, the optical characteristics of ocular defocus are not fully understood. To bridge this knowledge gap, we simulated polychromatic retinal image quality by considering both the monochromatic wavefront aberrations and chromatic aberrations of the eye, both in the fovea and the periphery (nasal visual field). Our study revealed two main findings: (1) chromatic and monochromatic aberrations interact to provide a signal to the retina (chromatic optical anisotropy) to discern positive from negative defocus and (2) that chromatic optical anisotropy exhibited notable differences among refractive error groups (myopes, emmetropes and hyperopes). These findings could enhance our understanding of the underlying mechanisms of defocus detection and their subsequent implications for myopia control therapies. Further research is needed to explore the retinal architecture's ability to utilize the optical signals identified in this study.
Collapse
Affiliation(s)
- Len Zheleznyak
- Clerio Vision, Inc., Rochester NY, USA
- Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Chang Liu
- The Institute of Optics, University of Rochester, Rochester, New York, USA
| | - Simon Winter
- Department of Biomedical Engineering, Wroclaw University of Science and Technology, Wroclaw, Poland
| |
Collapse
|
7
|
Barboni MTS, Széll N, Sohajda Z, Fehér T. Pupillary Light Reflex Reveals Melanopsin System Alteration in the Background of Myopia-26, the Female Limited Form of Early-Onset High Myopia. Invest Ophthalmol Vis Sci 2024; 65:6. [PMID: 38958970 PMCID: PMC11223624 DOI: 10.1167/iovs.65.8.6] [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: 09/19/2023] [Accepted: 05/17/2024] [Indexed: 07/04/2024] Open
Abstract
Purpose The purpose of this study was to evaluate pupillary light reflex (PLR) to chromatic flashes in patients with early-onset high-myopia (eoHM) without (myopic controls = M-CTRL) and with (female-limited myopia-26 = MYP-26) genetic mutations in the ARR3 gene encoding the cone arrestin. Methods Participants were 26 female subjects divided into 3 groups: emmetropic controls (E-CTRL, N = 12, mean age = 28.6 ± 7.8 years) and 2 myopic (M-CTRL, N = 7, mean age = 25.7 ± 11.5 years and MYP-26, N = 7, mean age = 28.3 ± 15.4 years) groups. In addition, one hemizygous carrier and one control male subject were examined. Direct PLRs were recorded after 10-minute dark adaptation. Stimuli were 1-second red (peak wavelength = 621 nm) and blue (peak wavelength = 470 nm) flashes at photopic luminance of 250 cd/m². A 2-minute interval between the flashes was introduced. Baseline pupil diameter (BPD), peak pupil constriction (PPC), and postillumination pupillary response (PIPR) were extracted from the PLR. Group comparisons were performed with ANOVAs. Results Dark-adapted BPD was comparable among the groups, whereas PPC to the red light was slightly reduced in patients with myopia (P = 0.02). PIPR at 6 seconds elicited by the blue flash was significantly weaker (P < 0.01) in female patients with MYP-26, whereas it was normal in the M-CTRL group and the asymptomatic male carrier. Conclusions L/M-cone abnormalities due to ARR3 gene mutation is currently claimed to underlie the pathological eye growth in MYP-26. Our results suggest that malfunction of the melanopsin system of intrinsically photosensitive retinal ganglion cells (ipRGCs) is specific to patients with symptomatic MYP-26, and may therefore play an additional role in the pathological eye growth of MYP-26.
Collapse
Affiliation(s)
| | - Noémi Széll
- Department of Ophthalmology, University of Debrecen, Debrecen, Hungary
| | - Zoltán Sohajda
- Kenézy Campus Department of Ophthalmology, University of Debrecen, Debrecen, Hungary
| | - Tamás Fehér
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| |
Collapse
|
8
|
Hoseini-Yazdi H, Read SA, Collins MJ, Bahmani H, Ellrich J, Schilling T. Increase in choroidal thickness after blue light stimulation of the blind spot in young adults. Bioelectron Med 2024; 10:13. [PMID: 38825695 PMCID: PMC11145801 DOI: 10.1186/s42234-024-00146-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024] Open
Abstract
BACKGROUND Blue light activates melanopsin, a photopigment that is expressed in intrinsically photosensitive retinal ganglion cells (ipRGCs). The axons of ipRGCs converge on the optic disc, which corresponds to the physiological blind spot in the visual field. Thus, a blue light stimulus aligned with the blind spot captures the ipRGCs axons at the optic disc. This study examined the potential changes in choroidal thickness and axial length associated with blue light stimulation of melanopsin-expressing ipRGCs at the blind spot. It was hypothesized that blue light stimulation at the blind spot in adults increases choroidal thickness. METHODS The blind spots of both eyes of 10 emmetropes and 10 myopes, with a mean age of 28 ± 6 years (SD), were stimulated locally for 1-minute with blue flickering light with a 460 nm peak wavelength. Measurements of choroidal thickness and axial length were collected from the left eye before stimulation and over a 60-minute poststimulation period. At a similar time of day, choroidal thickness and axial length were measured under sham control condition in all participants, while a subset of 3 emmetropes and 3 myopes were measured after 1-minute of red flickering light stimulation of the blind spot with a peak wavelength of 620 nm. Linear mixed model analyses were performed to examine the light-induced changes in choroidal thickness and axial length over time and between refractive groups. RESULTS Compared with sham control (2 ± 1 μm, n = 20) and red light (-1 ± 2 μm, n = 6) stimulation, subfoveal choroidal thickness increased within 60 min after blue light stimulation of the blind spot (7 ± 1 μm, n = 20; main effect of light, p < 0.001). Significant choroidal thickening after blue light stimulation occurred in emmetropes (10 ± 2 μm, p < 0.001) but not in myopes (4 ± 2 μm, p > 0.05). Choroidal thickening after blue light stimulation was greater in the fovea, diminishing in the parafoveal and perifoveal regions. There was no significant main effect of light, or light by refractive error interaction on the axial length after blind spot stimulation. CONCLUSIONS These findings demonstrate that stimulating melanopsin-expressing axons of ipRGCs at the blind spot with blue light increases choroidal thickness in young adults. This has potential implications for regulating eye growth.
Collapse
Affiliation(s)
- Hosein Hoseini-Yazdi
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, 4059, Australia
| | - Scott A Read
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, 4059, Australia
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, Centre for Vision and Eye Research, Optometry and Vision Science, Queensland University of Technology, Brisbane, 4059, Australia
| | - Hamed Bahmani
- Dopavision GmbH, Krausenstr. 9-10, 10117, Berlin, Germany
| | - Jens Ellrich
- Dopavision GmbH, Krausenstr. 9-10, 10117, Berlin, Germany
- Medical Faculty, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Tim Schilling
- Dopavision GmbH, Krausenstr. 9-10, 10117, Berlin, Germany.
| |
Collapse
|
9
|
Adhikari P, Uprety S, Feigl B, Zele AJ. Melanopsin-mediated amplification of cone signals in the human visual cortex. Proc Biol Sci 2024; 291:20232708. [PMID: 38808443 PMCID: PMC11285915 DOI: 10.1098/rspb.2023.2708] [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: 11/30/2023] [Accepted: 05/02/2024] [Indexed: 05/30/2024] Open
Abstract
The ambient daylight variation is coded by melanopsin photoreceptors and their luxotonic activity increases towards midday when colour temperatures are cooler, and irradiances are higher. Although melanopsin and cone photoresponses can be mediated via separate pathways, the connectivity of melanopsin cells across all levels of the retina enables them to modify cone signals. The downstream effects of melanopsin-cone interactions on human vision are however, incompletely understood. Here, we determined how the change in daytime melanopsin activation affects the human cone pathway signals in the visual cortex. A 5-primary silent-substitution method was developed to evaluate the dependence of cone-mediated signals on melanopsin activation by spectrally tuning the lights and stabilizing the rhodopsin activation under a constant cone photometric luminance. The retinal (white noise electroretinogram) and cortical responses (visual evoked potential) were simultaneously recorded with the photoreceptor-directed lights in 10 observers. By increasing the melanopsin activation, a reverse response pattern was observed with cone signals being supressed in the retina by 27% (p = 0.03) and subsequently amplified by 16% (p = 0.01) as they reach the cortex. We infer that melanopsin activity can amplify cone signals at sites distal to retinal bipolar cells to cause a decrease in the psychophysical Weber fraction for cone vision.
Collapse
Affiliation(s)
- Prakash Adhikari
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| | - Samir Uprety
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| | - Beatrix Feigl
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
- Queensland Eye Institute, Brisbane, Queensland 4101, Australia
| | - Andrew J Zele
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Queensland 4059, Australia
| |
Collapse
|
10
|
Matrone GM, van Doremaele ERW, Surendran A, Laswick Z, Griggs S, Ye G, McCulloch I, Santoro F, Rivnay J, van de Burgt Y. A modular organic neuromorphic spiking circuit for retina-inspired sensory coding and neurotransmitter-mediated neural pathways. Nat Commun 2024; 15:2868. [PMID: 38570478 PMCID: PMC10991258 DOI: 10.1038/s41467-024-47226-3] [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: 11/26/2023] [Accepted: 03/25/2024] [Indexed: 04/05/2024] Open
Abstract
Signal communication mechanisms within the human body rely on the transmission and modulation of action potentials. Replicating the interdependent functions of receptors, neurons and synapses with organic artificial neurons and biohybrid synapses is an essential first step towards merging neuromorphic circuits and biological systems, crucial for computing at the biological interface. However, most organic neuromorphic systems are based on simple circuits which exhibit limited adaptability to both external and internal biological cues, and are restricted to emulate only specific the functions of an individual neuron/synapse. Here, we present a modular neuromorphic system which combines organic spiking neurons and biohybrid synapses to replicate a neural pathway. The spiking neuron mimics the sensory coding function of afferent neurons from light stimuli, while the neuromodulatory activity of interneurons is emulated by neurotransmitters-mediated biohybrid synapses. Combining these functions, we create a modular connection between multiple neurons to establish a pre-processing retinal pathway primitive.
Collapse
Affiliation(s)
- Giovanni Maria Matrone
- Microsystems, Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AJ, Eindhoven, The Netherlands.
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
| | - Eveline R W van Doremaele
- Microsystems, Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AJ, Eindhoven, The Netherlands
| | - Abhijith Surendran
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Zachary Laswick
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Sophie Griggs
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, OX1 3TA, UK
| | - Gang Ye
- Center for Biomedical Optics and Photonics (CBOP) & College of Physics and Optoelectronic Engineering, Key Laboratory of Optoelectronic Devices and Systems, Shenzhen University, Shenzhen, 518060, PR China
| | - Iain McCulloch
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, OX1 3TA, UK
- King Abdullah University of Science and Technology (KAUST), KAUST Solar Center (KSC), Thuwal, 23955-6900, Saudi Arabia
| | - Francesca Santoro
- Tissue Electronics, Istituto Italiano di Tecnologia, Naples, 80125, Italy
- Institute of Biological Information Processing IBI-3 Bioelectronics, Forschungszentrum Juelich, 52428, Juelich, Germany
- Neuroelectronic Interfaces, Faculty of Electrical Engineering and IT, RWTH Aachen, 52074, Aachen, Germany
| | - Jonathan Rivnay
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yoeri van de Burgt
- Microsystems, Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AJ, Eindhoven, The Netherlands.
| |
Collapse
|
11
|
Chen YY, Tsai TH, Liu YL, Lin HJ, Wang IJ. The impact of light properties on ocular growth and myopia development. Taiwan J Ophthalmol 2024; 14:143-150. [PMID: 39027063 PMCID: PMC11253990 DOI: 10.4103/tjo.tjo-d-24-00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/03/2024] [Indexed: 07/20/2024] Open
Abstract
The objective of this article is to comprehensively review the effect of environmental lighting on ocular growth and refractive status in both animal and clinical studies, with an emphasis on the underlying mechanisms. This review was performed by searching research articles and reviews utilizing the terms "myopia," "light therapy," "axial length," "refractive error," and "emmetropization" in PubMed datasets. The review was finalized in December 2023. In the animal studies, high lighting brightness, illumination periods aligning with circadian rhythm, and color contrast signals including multiple wavelengths all help regulate ocular growth against myopia. Long wavelengths have been found to induce myopia in chicks, mice, fish, and guinea pigs, whereas shorter wavelengths lead to hyperopia. In contrast, red light has been observed to have a protective effect against myopia in tree shrews and rhesus monkeys. Apart from wavelength, flicker status also showed inconsistent effects on ocular growth, which could be attributed to differences in ocular refractive status, evolutionary disparities in retinal cone cells across species, and the selection of myopia induction models in experiments. In the clinical studies, current evidence suggests a control effect with red light therapy. Although the lighting conditions diverge from those in animal experiments, further reports are needed to assess the long-term effects. In conclusion, this review encompasses research related to the impact of light exposure on myopia and further explores the retinoscleral signaling pathway in refractive development. The aim is to establish a theoretical foundation for optimizing environmental factors in lighting design to address the epidemic of childhood myopia.
Collapse
Affiliation(s)
- Ying-Yi Chen
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Tzu-Hsun Tsai
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yao-Lin Liu
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Hui-Ju Lin
- Department of Ophthalmology, China Medical University Hospital, Taichung, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
12
|
Barrionuevo PA, Sandoval Salinas ML, Fanchini JM. Are ipRGCs involved in human color vision? Hints from physiology, psychophysics, and natural image statistics. Vision Res 2024; 217:108378. [PMID: 38458004 DOI: 10.1016/j.visres.2024.108378] [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: 11/30/2023] [Revised: 02/09/2024] [Accepted: 02/25/2024] [Indexed: 03/10/2024]
Abstract
Human photoreceptors consist of cones, rods, and melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). First studied in circadian regulation and pupillary control, ipRGCs project to a variety of brain centers suggesting a broader involvement beyond non-visual functions. IpRGC responses are stable, long-lasting, and with a particular codification of photoreceptor signals. In comparison with the transient and adaptive nature of cone and rod signals, ipRGCs' signaling might provide an ecological advantage to different attributes of color vision. Previous studies have indicated melanopsin's influence on visual responses yet its contribution to color perception in humans remains debated. We summarized evidence and hypotheses (from physiology, psychophysics, and natural image statistics) about direct and indirect involvement of ipRGCs in human color vision, by first briefly assessing the current knowledge about the role of melanopsin and ipRGCs in vision and codification of spectral signals. We then approached the question about melanopsin activation eliciting a color percept, discussing studies using the silent substitution method. Finally, we explore various avenues through which ipRGCs might impact color perception indirectly, such as through involvement in peripheral color matching, post-receptoral pathways, color constancy, long-term chromatic adaptation, and chromatic induction. While there is consensus about the role of ipRGCs in brightness perception, confirming its direct contribution to human color perception requires further investigation. We proposed potential approaches for future research, emphasizing the need for empirical validation and methodological thoroughness to elucidate the exact role of ipRGCs in human color vision.
Collapse
Affiliation(s)
- Pablo A Barrionuevo
- Allgemeine Psychologie, Justus-Liebig-Universität Gießen, Germany; Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina.
| | - María L Sandoval Salinas
- Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina; Instituto de Investigaciones de Biodiversidad Argentina (PIDBA), Facultad de Ciencias Naturales e Instituto Miguel Lillo, Universidad Nacional de Tucumán, Argentina
| | - José M Fanchini
- Instituto de Investigación en Luz, Ambiente y Visión (ILAV), CONICET - Universidad Nacional de Tucumán, Argentina; Departamento de Luminotecnia, Luz y Visión, Facultad de Ciencias Exactas y Tecnología, Universidad Nacional de Tucumán, Argentina
| |
Collapse
|
13
|
Reidy MG, Hartwick ATE, Mutti DO. The association between pupillary responses and axial length in children differs as a function of season. Sci Rep 2024; 14:598. [PMID: 38182869 PMCID: PMC10770316 DOI: 10.1038/s41598-024-51199-0] [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: 11/15/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024] Open
Abstract
The association between pupillary responses to repeated stimuli and adult refractive error has been previously demonstrated. This study evaluated whether this association exists in children and if it varies by season. Fifty children aged 8-17 years (average: 11.55 ± 2.75 years, 31 females) with refractive error between + 1.51 and - 5.69 diopters (non-cycloplegic) participated (n = 27 in summer, and n = 23 in winter). The RAPDx pupilometer measured pupil sizes while stimuli oscillated between colored light and dark at 0.1 Hz in three sequences: (1) alternating red and blue, (2) red-only, and (3) blue-only. The primary outcome was the difference in pupillary responses between the blue-only and red-only sequences. Pupillary constriction was greater in response to blue light than to red for those with shorter eyes in summer (β = - 9.42, P = 0.034) but not in winter (β = 3.42, P = 0.54). Greater constriction comprised faster pupillary escape following red light onset and slower redilation following stimulus offset of both colors (P = 0.017, 0.036, 0.035 respectively). The association between axial length and children's pupillary responses in summer, but not winter may be explained by greater light-associated release of retinal dopamine in summer. Shorter eyes' more robust responses are consistent with greater light exposure inhibiting axial elongation and reducing myopia risk.
Collapse
Affiliation(s)
| | | | - Donald O Mutti
- The Ohio State University College of Optometry, Columbus, USA
| |
Collapse
|
14
|
McMahon DG, Dowling JE. Neuromodulation: Actions of Dopamine, Retinoic Acid, Nitric Oxide, and Other Substances on Retinal Horizontal Cells. Eye Brain 2023; 15:125-137. [PMID: 37928979 PMCID: PMC10625386 DOI: 10.2147/eb.s420050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 11/07/2023] Open
Abstract
Whereas excitation and inhibition of neurons are well understood, it is clear that neuromodulatory influences on neurons and their synapses play a major role in shaping neural activity in the brain. Memory and learning, emotional and other complex behaviors, as well as cognitive disorders have all been related to neuromodulatory mechanisms. A number of neuroactive substances including monoamines such as dopamine and neuropeptides have been shown to act as neuromodulators, but other substances thought to play very different roles in the body and brain act as neuromodulators, such as retinoic acid. We still understand little about how neuromodulatory substances exert their effects, and the present review focuses on how two such substances, dopamine and retinoic acid, exert their effects. The emphasis is on the underlying neuromodulatory mechanisms down to the molecular level that allow the second order bipolar cells and the output neurons of the retina, the ganglion cells, to respond to different environmental (ie lighting) conditions. The modulation described affects a simple circuit in the outer retina, involves several neuroactive substances and is surprisingly complex and not fully understood.
Collapse
Affiliation(s)
- Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235, USA
| | - John E Dowling
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
15
|
Carpena-Torres C, Schilling T, Huete-Toral F, Bahmani H, Carracedo G. Increased ocular dopamine levels in rabbits after blue light stimulation of the optic nerve head. Exp Eye Res 2023; 234:109604. [PMID: 37499737 DOI: 10.1016/j.exer.2023.109604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023]
Abstract
The purpose was to quantify ocular dopamine in rabbits after stimulation of the optic nerve head with short-wavelength (blue) light to activate melanopsin expressed in the axons of intrinsically photosensitive retinal ganglion cells (ipRGCs). Dopamine levels in tears, aqueous humor, vitreous body, and retina (including choroid) were quantified after blue light stimulation of the optic nerve head of 15 rabbits with an optical fiber for 1 min, 10 min, or no stimulation (n = 5, each group). The left eye of all rabbits was operated on to introduce the optical fiber and stimulate the optic nerve, while the contralateral eye served as internal control. One minute of blue light stimulation significantly increased dopamine concentration in the vitreous body of the treated eyes compared to the contralateral ones (P = 0.015). Stimulation for 10 min significantly increased dopamine concentration in the vitreous body, as well as the aqueous humor (P < 0.05). Therefore, using an optical fiber approach to stimulate the optic nerve head with blue light significantly increased dopamine concentration in the aqueous humor and the vitreous body. This likely reflects an upregulation of retinal dopamine synthesis that could be attributed to ipRGC activation. However, the data provided in this study fell short of establishing a definitive link between dopamine release and ipRGC activation, mainly due to the lack of evidence supporting the expression of the melanopsin photopigment in the optic nerve.
Collapse
Affiliation(s)
- Carlos Carpena-Torres
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | | | - Fernando Huete-Toral
- Ocupharm Research Group, Department of Biochemistry and Molecular Biology, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain
| | | | - Gonzalo Carracedo
- Ocupharm Research Group, Department of Optometry and Vision, Faculty of Optics and Optometry, Complutense University of Madrid, Madrid, Spain.
| |
Collapse
|
16
|
Kinane C, Calligaro H, Jandot A, Coutanson C, Haddjeri N, Bennis M, Dkhissi-Benyahya O. Dopamine modulates the retinal clock through melanopsin-dependent regulation of cholinergic waves during development. BMC Biol 2023; 21:146. [PMID: 37365544 DOI: 10.1186/s12915-023-01647-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
BACKGROUND The mammalian retina contains an autonomous circadian clock that controls various aspects of retinal physiology and function, including dopamine (DA) release by amacrine cells. This neurotransmitter plays a critical role in retina development, visual signalling, and phase resetting of the retinal clock in adulthood. Interestingly, bidirectional regulation between dopaminergic cells and melanopsin-expressing retinal ganglion cells has been demonstrated in the adult and during development. Additionally, the adult melanopsin knockout mouse (Opn4 -/-) exhibits a shortening of the endogenous period of the retinal clock. However, whether DA and / or melanopsin influence the retinal clock mechanism during its maturation is still unknown. RESULTS Using wild-type Per2 Luc and melanopsin knockout (Opn4 -/-::Per2 Luc) mice at different postnatal stages, we found that the retina generates self-sustained circadian rhythms from postnatal day 5 in both genotypes and that the ability to express these rhythms emerges in the absence of external time cues. Intriguingly, only in wild-type explants, DA supplementation lengthened the endogenous period of the clock during the first week of postnatal development through both D1- and D2-like dopaminergic receptors. Furthermore, the blockade of spontaneous cholinergic retinal waves, which drive DA release in the early developmental stages, shortened the period and reduced the light-induced phase shift of the retinal clock only in wild-type retinas. CONCLUSIONS These data suggest that DA modulates the molecular core of the clock through melanopsin-dependent regulation of acetylcholine retinal waves, thus offering an unprecedented role of DA and melanopsin in the endogenous functioning and the light response of the retinal clock during development.
Collapse
Affiliation(s)
- Chaimaa Kinane
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, University Cadi Ayyad, Marrakech, Morocco
| | - Hugo Calligaro
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
- Salk Institute for Biological Studies, La Lolla, CA, USA
| | - Antonin Jandot
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Christine Coutanson
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Nasser Haddjeri
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France
| | - Mohamed Bennis
- Laboratory of Pharmacology, Neurobiology, Anthropobiology and Environment, University Cadi Ayyad, Marrakech, Morocco
| | - Ouria Dkhissi-Benyahya
- Inserm, Stem Cell and Brain Research Institute U1208, Univ Lyon, Université Claude Bernard Lyon 1, 18 Avenue du Doyen Lépine, 69500, Bron, France.
| |
Collapse
|
17
|
Jain V, Liang PJM, Raja S, Mikhael M, Cameron MA. Light activation of the dopaminergic system occurs after eye-opening in the mouse retina. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1184627. [PMID: 38983102 PMCID: PMC11182289 DOI: 10.3389/fopht.2023.1184627] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/13/2023] [Indexed: 07/11/2024]
Abstract
The neuromodulator dopamine plays a significant role in light adaptation, eye growth, and modulation of neuronal circuitry in the retina. Dopaminergic amacrine cells in the adult retina release dopamine in response to light stimulation, however, the light-induced activity of these cells in during postnatal development is not known. We assessed the activity of dopaminergic amacrine cells in the retina response to a light pulse in C57BL/6 wild-type animals across various postnatal ages. Expression of tyrosine hydroxylase (TH) in dopaminergic amacrine cells was apparent from postnatal day 3 (P3) and restricted to the dorso-temporal region; by P8 TH+ cells were uniformly distributed across the retina. TH cell density increased until P8 and then markedly decreased by P10 to then remain at this density into adulthood. Light-induced c-fos expression was observed in all light-pulsed retinae, however, no c-fos was ever found to be co-localised with TH prior to P12. At P14, one day after eye opening, 100% of TH cells co-localised with c-fos and this was maintained for all older ages analysed. Dopamine and its primary metabolite DOPAC were measured in the vitreous of animals P8-P30. Both analytes were found in the vitreous at all ages, however, a significant difference in dopamine concentration between dark and light-pulsed animals was only observed at P30. DOPAC concentration was found to be significantly light-induced from P16, and the amplitude of this difference increased over time. Our data suggests that dopaminergic cell activation and light-induced dopamine release in the retina is primarily driven by classical photoreceptors after eye-opening.
Collapse
Affiliation(s)
- Vrinda Jain
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | | | - Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Meena Mikhael
- Mass Spectrometry Facility, Western Sydney University, Sydney, NSW, Australia
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| |
Collapse
|
18
|
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: 44] [Impact Index Per Article: 22.0] [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
|
19
|
Bhoi JD, Goel M, Ribelayga CP, Mangel SC. Circadian clock organization in the retina: From clock components to rod and cone pathways and visual function. Prog Retin Eye Res 2023; 94:101119. [PMID: 36503722 PMCID: PMC10164718 DOI: 10.1016/j.preteyeres.2022.101119] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/13/2022]
Abstract
Circadian (24-h) clocks are cell-autonomous biological oscillators that orchestrate many aspects of our physiology on a daily basis. Numerous circadian rhythms in mammalian and non-mammalian retinas have been observed and the presence of an endogenous circadian clock has been demonstrated. However, how the clock and associated rhythms assemble into pathways that support and control retina function remains largely unknown. Our goal here is to review the current status of our knowledge and evaluate recent advances. We describe many previously-observed retinal rhythms, including circadian rhythms of morphology, biochemistry, physiology, and gene expression. We evaluate evidence concerning the location and molecular machinery of the retinal circadian clock, as well as consider findings that suggest the presence of multiple clocks. Our primary focus though is to describe in depth circadian rhythms in the light responses of retinal neurons with an emphasis on clock control of rod and cone pathways. We examine evidence that specific biochemical mechanisms produce these daily light response changes. We also discuss evidence for the presence of multiple circadian retinal pathways involving rhythms in neurotransmitter activity, transmitter receptors, metabolism, and pH. We focus on distinct actions of two dopamine receptor systems in the outer retina, a dopamine D4 receptor system that mediates circadian control of rod/cone gap junction coupling and a dopamine D1 receptor system that mediates non-circadian, light/dark adaptive regulation of gap junction coupling between horizontal cells. Finally, we evaluate the role of circadian rhythmicity in retinal degeneration and suggest future directions for the field of retinal circadian biology.
Collapse
Affiliation(s)
- Jacob D Bhoi
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA
| | - Manvi Goel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Christophe P Ribelayga
- Ruiz Department of Ophthalmology and Visual Science, McGovern Medical School, UTHEALTH-The University of Texas Health Science Center at Houston, Houston, TX, USA; Neuroscience Honors Research Program, William Marsh Rice University, Houston, TX, USA.
| | - Stuart C Mangel
- Department of Neuroscience, Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
20
|
Zhang C, Zhu Z, Zhao J, Li Y, Zhang Z, Zheng Y. Ubiquitous light-emitting diodes: Potential threats to retinal circadian rhythms and refractive development. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160809. [PMID: 36502986 DOI: 10.1016/j.scitotenv.2022.160809] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/08/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
The use of light-emitting diodes (LEDs) has increased considerably in the 21st century with humans living in a modern photoperiod with brighter nights and dimmer days. Prolonged exposure to LEDs, especially at night, is considered a new source of pollution because it may affect the synthesis and secretion of retinal melatonin and dopamine, resulting in negative impacts on retinal circadian clocks and potentially disrupting retinal circadian rhythms. The control of ocular refraction is believed to be related to retinal circadian rhythms. Moreover, the global prevalence of myopia has increased at an alarming rate in recent decades. The widespread use of LEDs and the rapid increase in the prevalence of myopia overlap, which is unlikely to be a coincidence. The connection among LEDs, retinal circadian rhythms, and refractive development is both fascinating and confusing. In this review, we aim to develop a systematic framework that includes LEDs, retinal circadian rhythms and refractive development. This paper summarizes the possible mechanisms by which LEDs may disrupt retinal circadian rhythms. We propose that prolonged exposure to LEDs may induce myopia by disrupting retinal circadian rhythms. Finally, we suggest several possible countermeasures to prevent LED interference on retinal circadian rhythms, with the hope of reducing the onset and progression of myopia.
Collapse
Affiliation(s)
- Chenchen Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Zhe Zhu
- Department of Ophthalmology, Eye Hospital of Shandong First Medical University, Shandong Eye Institute, Jinan 250000, China
| | - Jing Zhao
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Yanxia Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Zhaoying Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China
| | - Yajuan Zheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
21
|
Raja S, Milosavljevic N, Allen AE, Cameron MA. Burning the candle at both ends: Intraretinal signaling of intrinsically photosensitive retinal ganglion cells. Front Cell Neurosci 2023; 16:1095787. [PMID: 36687522 PMCID: PMC9853061 DOI: 10.3389/fncel.2022.1095787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 12/13/2022] [Indexed: 01/09/2023] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) are photoreceptors located in the ganglion cell layer. They project to brain regions involved in predominately non-image-forming functions including entrainment of circadian rhythms, control of the pupil light reflex, and modulation of mood and behavior. In addition to possessing intrinsic photosensitivity via the photopigment melanopsin, these cells receive inputs originating in rods and cones. While most research in the last two decades has focused on the downstream influence of ipRGC signaling, recent studies have shown that ipRGCs also act retrogradely within the retina itself as intraretinal signaling neurons. In this article, we review studies examining intraretinal and, in addition, intraocular signaling pathways of ipRGCs. Through these pathways, ipRGCs regulate inner and outer retinal circuitry through both chemical and electrical synapses, modulate the outputs of ganglion cells (both ipRGCs and non-ipRGCs), and influence arrangement of the correct retinal circuitry and vasculature during development. These data suggest that ipRGC function plays a significant role in the processing of image-forming vision at its earliest stage, positioning these photoreceptors to exert a vital role in perceptual vision. This research will have important implications for lighting design to optimize the best chromatic lighting environments for humans, both in adults and potentially even during fetal and postnatal development. Further studies into these unique ipRGC signaling pathways could also lead to a better understanding of the development of ocular dysfunctions such as myopia.
Collapse
Affiliation(s)
- Sushmitha Raja
- School of Medicine, Western Sydney University, Sydney, NSW, Australia
| | - Nina Milosavljevic
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Annette E. Allen
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Morven A. Cameron
- School of Medicine, Western Sydney University, Sydney, NSW, Australia,*Correspondence: Morven A. Cameron,
| |
Collapse
|
22
|
Chakraborty R, Collins MJ, Kricancic H, Davis B, Alonso-Caneiro D, Yi F, Baskaran K. The effect of intrinsically photosensitive retinal ganglion cell (ipRGC) stimulation on axial length changes to imposed optical defocus in young adults. JOURNAL OF OPTOMETRY 2023; 16:53-63. [PMID: 35589503 PMCID: PMC9811374 DOI: 10.1016/j.optom.2022.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/08/2022] [Accepted: 04/21/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE The intrinsically photosensitive retinal ganglion cells (ipRGCs) regulate pupil size and circadian rhythms. Stimulation of the ipRGCs using short-wavelength blue light causes a sustained pupil constriction known as the post-illumination pupil response (PIPR). Here we examined the effects of ipRGC stimulation on axial length changes to imposed optical defocus in young adults. MATERIALS AND METHODS Nearly emmetropic young participants were given either myopic (+3 D, n = 16) or hyperopic (-3 D, n = 17) defocus in their right eye for 2 h. Before and after defocus, a series of axial length measurements for up to 180 s were performed in the right eye using the IOL Master following exposure to 5 s red (625 nm, 3.74 × 1014 photons/cm2/s) and blue (470 nm, 3.29 × 1014 photons/cm2/s) stimuli. The pupil measurements were collected from the left eye to track the ipRGC activity. The 6 s and 30 s PIPR, early and late area under the curve (AUC), and time to return to baseline were calculated. RESULTS The PIPR with blue light was significantly stronger after 2 h of hyperopic defocus as indicated by a lower 6 and 30 s PIPR and a larger early and late AUC (all p<0.05). Short-wavelength ipRGC stimulation also significantly exaggerated the ocular response to hyperopic defocus, causing a significantly greater increase in axial length than that resulting from the hyperopic defocus alone (p = 0.017). Neither wavelength had any effect on axial length with myopic defocus. CONCLUSIONS These findings suggest an interaction between myopiagenic hyperopic defocus and ipRGC signaling.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- Caring Futures Institute, Flinders University, Bedford Park, SA 5042, Australia; College of Nursing and Health Sciences, Optometry and Vision Science, Sturt North, Flinders University, Bedford Park, SA 5042, Australia.
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Henry Kricancic
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Brett Davis
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - David Alonso-Caneiro
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Fan Yi
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | | |
Collapse
|
23
|
Hu S, Wang Y, Han X, Dai M, Zhang Y, Ma Y, Weng S, Xiao L. Activation of oxytocin receptors in mouse GABAergic amacrine cells modulates retinal dopaminergic signaling. BMC Biol 2022; 20:205. [PMID: 36127701 PMCID: PMC9490981 DOI: 10.1186/s12915-022-01405-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022] Open
Abstract
Background Oxytocin, secreted by oxytocin neurons in the hypothalamus, is an endogenous neuropeptide involved in modulating multiple sensory information processing pathways, and its roles in the brain have been associated with prosocial, maternal, and feeding-related behaviors. Visual information is necessary for initiating these behaviors, with the retina consisting of the first stage in the visual system mediating external stimulus perception. Oxytocin has been detected in the mammalian retina; however, the expression and possible function of oxytocin receptors (OxtR) in the retina remain unknown. Here, we explore the role of oxytocin in regulating visual information processing in the retina. Results We observed that OxtR mRNA and protein are expressed in the mouse retina. With Oxtr-Cre transgenic mice, immunostaining, and fluorescence in situ hybridization, we found that OxtRs are mainly expressed in GABAergic amacrine cells (ACs) in both the inner nuclear layer (INL) and ganglion cell layer (GCL). Further immunoreactivity studies showed that GABAergic OxtR+ neurons are mainly cholinergic and dopaminergic neurons in the INL and are cholinergic and corticotrophin-releasing hormone neurons in the GCL. Surprisingly, a high level of Oxtr mRNAs was detected in retinal dopaminergic neurons, and exogenous oxytocin application activated dopaminergic neurons to elevate the retinal dopamine level. Relying on in vivo electroretinographic recording, we found that activating retinal OxtRs reduced the activity of bipolar cells via OxtRs and dopamine receptors. Conclusions These data indicate the functional expression of OxtRs in retinal GABAergic ACs, especially dopaminergic ACs, and expand the interactions between oxytocinergic and dopaminergic systems. This study suggests that visual perception, from the first stage of information processing in the retina, is modulated by hypothalamic oxytocin signaling. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-022-01405-0.
Collapse
Affiliation(s)
- Songhui Hu
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yurong Wang
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Xu Han
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Min Dai
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yongxing Zhang
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Shijun Weng
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Lei Xiao
- The State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, and the Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
24
|
Wong NA, Bahmani H. A review of the current state of research on artificial blue light safety as it applies to digital devices. Heliyon 2022; 8:e10282. [PMID: 36042717 PMCID: PMC9420367 DOI: 10.1016/j.heliyon.2022.e10282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/13/2022] [Accepted: 08/09/2022] [Indexed: 12/24/2022] Open
Abstract
Light is necessary for human health and well-being. As we spend more time indoors, we are being increasingly exposed to artificial light. The development of artificial lighting has allowed us to control the brightness, colour, and timing of our light exposure. Yet, the widespread use of artificial light has raised concerns about the impact of altering our light environment on our health. The widespread adoption of personal digital devices over the past decade has exposed us to yet another source of artificial light. We spend a significant amount of time using digital devices with light-emitting screens, including smartphones and tablets, at close range. The light emitted from these devices, while appearing white, has an emission spectrum with a peak in the blue range. Blue light is often characterised as hazardous as its photon energy is higher than that of other wavelengths of visible light. Under certain conditions, visible blue light can cause harm to the retina and other ocular structures. Blue light can also influence the circadian rhythm and processes mediated by melanopsin-expressing intrinsically photosensitive retinal ganglion cells. While the blue component of sunlight is necessary for various physiological processes, whether the low-illuminance artificial blue light emitted from digital devices presents a risk to our health remains an ongoing area of debate. As technological advancements continue, it is relevant to understand how new devices may influence our well-being. This review examines the existing research on artificial blue light safety and the eye, visual performance, and circadian functions.
Collapse
Affiliation(s)
| | - Hamed Bahmani
- Dopavision GmbH, Berlin, Germany.,Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tuebingen, Germany
| |
Collapse
|
25
|
Rodriguez TC, Zhong L, Simpson H, Gleason E. Reduced Expression of TMEM16A Impairs Nitric Oxide-Dependent Cl− Transport in Retinal Amacrine Cells. Front Cell Neurosci 2022; 16:937060. [PMID: 35966201 PMCID: PMC9363626 DOI: 10.3389/fncel.2022.937060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/21/2022] [Indexed: 11/13/2022] Open
Abstract
Postsynaptic cytosolic Cl− concentration determines whether GABAergic and glycinergic synapses are inhibitory or excitatory. We have shown that nitric oxide (NO) initiates the release of Cl− from acidic internal stores into the cytosol of retinal amacrine cells (ACs) thereby elevating cytosolic Cl−. In addition, we found that cystic fibrosis transmembrane conductance regulator (CFTR) expression and Ca2+ elevations are necessary for the transient effects of NO on cytosolic Cl− levels, but the mechanism remains to be elucidated. Here, we investigated the involvement of TMEM16A as a possible link between Ca2+ elevations and cytosolic Cl− release. TMEM16A is a Ca2+-activated Cl− channel that is functionally coupled with CFTR in epithelia. Both proteins are also expressed in neurons. Based on this and its Ca2+ dependence, we test the hypothesis that TMEM16A participates in the NO-dependent elevation in cytosolic Cl− in ACs. Chick retina ACs express TMEM16A as shown by Western blot analysis, single-cell PCR, and immunocytochemistry. Electrophysiology experiments demonstrate that TMEM16A functions in amacrine cells. Pharmacological inhibition of TMEM16A with T16inh-AO1 reduces the NO-dependent Cl− release as indicated by the diminished shift in the reversal potential of GABAA receptor-mediated currents. We confirmed the involvement of TMEM16A in the NO-dependent Cl− release using CRISPR/Cas9 knockdown of TMEM16A. Two different modalities targeting the gene for TMEM16A (ANO1) were tested in retinal amacrine cells: an all-in-one plasmid vector and crRNA/tracrRNA/Cas9 ribonucleoprotein. The all-in-one CRISPR/Cas9 modality did not change the expression of TMEM16A protein and produced no change in the response to NO. However, TMEM16A-specific crRNA/tracrRNA/Cas9 ribonucleoprotein effectively reduces both TMEM16A protein levels and the NO-dependent shift in the reversal potential of GABA-gated currents. These results show that TMEM16A plays a role in the NO-dependent Cl− release from retinal ACs.
Collapse
|
26
|
Uprety S, Adhikari P, Feigl B, Zele AJ. Melanopsin photoreception differentially modulates rod-mediated and cone-mediated human temporal vision. iScience 2022; 25:104529. [PMID: 35754721 PMCID: PMC9218364 DOI: 10.1016/j.isci.2022.104529] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/04/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
To evaluate the nature of interactions between visual pathways transmitting the slower melanopsin and faster rod and cone signals, we implement a temporal phase summation paradigm in human observers using photoreceptor-directed stimuli. We show that melanopsin stimulation interacts with and alters both rod-mediated and cone-mediated vision regardless of whether it is perceptually visible or not. Melanopsin-rod interactions result in either inhibitory or facilitatory summation depending on the temporal frequency and photoreceptor pathway contrast sensitivity. Moreover, by isolating rod vision, we reveal a bipartite intensity response property of the rod pathway in photopic lighting that extends its operational range at lower frequencies to beyond its classic saturation limits but at the expense of attenuating sensitivity at higher frequencies. In comparison, melanopsin-cone interactions always lead to facilitation. These interactions can be described by linear or probability summations and potentially involve multiple intraretinal and visual cortical pathways to set human visual contrast sensitivity. Melanopsin ipRGCs support vision independent of the rod and cone signals Rod pathways mediate robust visual responses in daylight Temporal contrast sensitivity is contingent on the melanopsin excitation level Visual performance is collectively regulated by melanopsin, rod and cone pathways
Collapse
Affiliation(s)
- Samir Uprety
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.,School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Prakash Adhikari
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.,School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Beatrix Feigl
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.,Queensland Eye Institute, Brisbane, QLD 4101, Australia
| | - Andrew J Zele
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.,School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| |
Collapse
|
27
|
Liu AL, Liu YF, Wang G, Shao YQ, Yu CX, Yang Z, Zhou ZR, Han X, Gong X, Qian KW, Wang LQ, Ma YY, Zhong YM, Weng SJ, Yang XL. The role of ipRGCs in ocular growth and myopia development. SCIENCE ADVANCES 2022; 8:eabm9027. [PMID: 35675393 PMCID: PMC9176740 DOI: 10.1126/sciadv.abm9027] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The increasing global prevalence of myopia calls for elaboration of the pathogenesis of this disease. Here, we show that selective ablation and activation of intrinsically photosensitive retinal ganglion cells (ipRGCs) in developing mice induced myopic and hyperopic refractive shifts by modulating the corneal radius of curvature (CRC) and axial length (AL) in an opposite way. Melanopsin- and rod/cone-driven signals of ipRGCs were found to influence refractive development by affecting the AL and CRC, respectively. The role of ipRGCs in myopia progression is evidenced by attenuated form-deprivation myopia magnitudes in ipRGC-ablated and melanopsin-deficient animals and by enhanced melanopsin expression/photoresponses in form-deprived eyes. Cell subtype-specific ablation showed that M1 subtype cells, and probably M2/M3 subtype cells, are involved in ocular development. Thus, ipRGCs contribute substantially to mouse eye growth and myopia development, which may inspire novel strategies for myopia intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Shi-Jun Weng
- Corresponding author. (X.-L.Y.); (S.-J.W.); (Y.-M.Z.)
| | - Xiong-Li Yang
- Corresponding author. (X.-L.Y.); (S.-J.W.); (Y.-M.Z.)
| |
Collapse
|
28
|
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
|
29
|
Barrionuevo PA, Paz Filgueira C, Cao D. Is melanopsin activation affecting large field color-matching functions? JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2022; 39:1104-1110. [PMID: 36215541 DOI: 10.1364/josaa.457223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/11/2022] [Indexed: 06/16/2023]
Abstract
Color theory is based on the exclusive activation of cones. However, since the discovery of melanopsin expressing cells in the human retina, evidence of its intrusion in brightness and color vision is increasing. We aimed to assess if differences between peripheral or large field and foveal color matches can be accounted for by melanopsin activation or rod intrusion. Photopic color matches by young observers showed that differences between extrafoveal and foveal results cannot be explained by rod intrusion. Furthermore, statistical analyses on existing color-matching functions suggest a role of melanopsin activation, particularly, in large field S fundamentals.
Collapse
|
30
|
Chakraborty R, Collins MJ, Kricancic H, Moderiano D, Davis B, Alonso-Caneiro D, Yi F, Baskaran K. The intrinsically photosensitive retinal ganglion cell (ipRGC) mediated pupil response in young adult humans with refractive errors. JOURNAL OF OPTOMETRY 2022; 15:112-121. [PMID: 33402286 PMCID: PMC9068560 DOI: 10.1016/j.optom.2020.12.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/21/2020] [Accepted: 12/01/2020] [Indexed: 05/04/2023]
Abstract
PURPOSE The intrinsically photosensitive retinal ganglion cells (ipRGCs) signal environmental light, with axons projected to the midbrain that control pupil size and circadian rhythms. Post-illumination pupil response (PIPR), a sustained pupil constriction after short-wavelength light stimulation, is an indirect measure of ipRGC activity. Here, we measured the PIPR in young adults with various refractive errors using a custom-made optical system. METHODS PIPR was measured on myopic (-3.50 ± 1.82 D, n = 20) and non-myopic (+0.28 ± 0.23 D, n = 19) participants (mean age, 23.36 ± 3.06 years). The right eye was dilated and presented with long-wavelength (red, 625 nm, 3.68 × 1014 photons/cm2/s) and short-wavelength (blue, 470 nm, 3.24 × 1014 photons/cm2/s) 1 s and 5 s pulses of light, and the consensual response was measured in the left eye for 60 s following light offset. The 6 s and 30 s PIPR and early and late area under the curve (AUC) for 1 and 5 s stimuli were calculated. RESULTS For most subjects, the 6 s and 30 s PIPR were significantly lower (p < 0.001), and the early and late AUC were significantly larger for 1 s blue light compared to red light (p < 0.001), suggesting a strong ipRGC response. The 5 s blue stimulation induced a slightly stronger melanopsin response, compared to 1 s stimulation with the same wavelength. However, none of the PIPR metrics were different between myopes and non-myopes for either stimulus duration (p > 0.05). CONCLUSIONS We confirm previous research that there is no effect of refractive error on the PIPR.
Collapse
Affiliation(s)
- Ranjay Chakraborty
- College of Nursing and Health Sciences, Optometry and Vision Science, Sturt North, Flinders University, Sturt Rd, Bedford Park, SA 5042, Australia; Caring Futures Institute, Flinders University, Sturt Rd, Bedford Park, SA 5042, Australia.
| | - Michael J Collins
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Henry Kricancic
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Daniel Moderiano
- College of Nursing and Health Sciences, Optometry and Vision Science, Sturt North, Flinders University, Sturt Rd, Bedford Park, SA 5042, Australia
| | - Brett Davis
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - David Alonso-Caneiro
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | - Fan Yi
- Contact Lens and Visual Optics Laboratory, School of Optometry and Vision Science, Queensland University of Technology, Victoria Park Road, Kelvin Grove 4059, Brisbane, QLD, Australia
| | | |
Collapse
|
31
|
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: 8] [Impact Index Per Article: 2.7] [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
|
32
|
Endogenous functioning and light response of the retinal clock in vertebrates. PROGRESS IN BRAIN RESEARCH 2022; 273:49-69. [DOI: 10.1016/bs.pbr.2022.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
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
|
34
|
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only anterogradely to drive behavioral responses, but also retrogradely to some amacrine interneurons to modulate retinal physiology. We previously found that all displaced amacrine cells with spiking, tonic excitatory photoresponses receive gap-junction input from ipRGCs, but the connectivity patterns and functional roles of ipRGC-amacrine coupling remained largely unknown. Here, we injected PoPro1 fluorescent tracer into all six types of mouse ipRGCs to identify coupled amacrine cells, and analyzed the latter's morphological and electrophysiological properties. We also examined how genetically disrupting ipRGC-amacrine coupling affected ipRGC photoresponses. Results showed that ipRGCs couple with not just ON- and ON/OFF-stratified amacrine cells in the ganglion-cell layer as previously reported, but also OFF-stratified amacrine cells in both ganglion-cell and inner nuclear layers. M1- and M3-type ipRGCs couple mainly with ON/OFF-stratified amacrine cells, whereas the other ipRGC types couple almost exclusively with ON-stratified ones. ipRGCs transmit melanopsin-based light responses to at least 93% of the coupled amacrine cells. Some of the ON-stratifying ipRGC-coupled amacrine cells exhibit transient hyperpolarizing light responses. We detected bidirectional electrical transmission between an ipRGC and a coupled amacrine cell, although transmission was asymmetric for this particular cell pair, favoring the ipRGC-to-amacrine direction. We also observed electrical transmission between two amacrine cells coupled to the same ipRGC. In both scenarios of coupling, the coupled cells often spiked synchronously. While ipRGC-amacrine coupling somewhat reduces the peak firing rates of ipRGCs' intrinsic melanopsin-based photoresponses, it renders these responses more sustained and longer-lasting. In summary, ipRGCs' gap junctional network involves more amacrine cell types and plays more roles than previously appreciated.
Collapse
|
35
|
Roecklein KA, Franzen PL, Wescott DL, Hasler BP, Miller MA, Donofry SD, DuPont CM, Gratzmiller SM, Drexler SP, Wood-Vasey WM, Gamlin PD. Melanopsin-driven pupil response in summer and winter in unipolar seasonal affective disorder. J Affect Disord 2021; 291:93-101. [PMID: 34029883 PMCID: PMC8693789 DOI: 10.1016/j.jad.2021.04.084] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/19/2021] [Accepted: 04/25/2021] [Indexed: 11/30/2022]
Abstract
A retinal subsensitivity to environmental light may trigger Seasonal Affective Disorder (SAD) under low wintertime light conditions. The main aim of this study was to assess the responses of melanopsin-containing retinal ganglion cells in participants (N= 65) diagnosed with unipolar SAD compared to controls with no history of depression. Participants attended a summer visit, a winter visit, or both. Retinal responses to light were measured using the post-illumination pupil response (PIPR) to assess melanopsin-driven responses in the non-visual light input pathway. Linear mixed-effects modeling was used to test a group*season interaction on the Net PIPR (red minus blue light response, percent baseline). We observed a significant group*season interaction such that the PIPR decreased from summer to winter significantly in the SAD group while not in the control group. The SAD group PIPR was significantly lower in winter compared to controls but did not differ between groups in summer. Only 60% of the participants underwent an eye health exam, although all participants reported no history of retinal pathology, and eye exam status was neither associated with outcome nor different between groups. This seasonal variation in melanopsin driven non-visual responses to light may be a risk factor for SAD, and further highlights individual differences in responses to light for direct or indirect effects of light on mood.
Collapse
Affiliation(s)
- Kathryn A. Roecklein
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,The Center for the Neural Basis of Cognition, Pittsburgh, Pennsylvania, USA.,Corresponding Author:Kathryn A. Roecklein, Ph.D. Associate Professor Department of Psychology, University of Pittsburgh 210 S. Bouquet Street Pittsburgh, PA 15206 (412) 624-4553
| | - Peter L. Franzen
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Delainey L. Wescott
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brant P. Hasler
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Megan A. Miller
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shannon D. Donofry
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Caitlin M. DuPont
- Department of Psychology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sarah M. Gratzmiller
- Department of Psychiatry, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Scott P. Drexler
- Department of Ophthalmology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - W. Michael Wood-Vasey
- Department of Physics and Astronomy, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul D. Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
36
|
Pant M, Zele AJ, Feigl B, Adhikari P. Light adaptation characteristics of melanopsin. Vision Res 2021; 188:126-138. [PMID: 34315092 DOI: 10.1016/j.visres.2021.07.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 06/14/2021] [Accepted: 07/07/2021] [Indexed: 11/20/2022]
Abstract
Following photopigment bleaching, the rhodopsin and cone-opsins show a characteristic exponential regeneration in the dark with a photocycle dependent on the retinal pigment epithelium. Melanopsin pigment regeneration in animal models requires different pathways to rods and cones. To quantify melanopsin-mediated light adaptation in humans, we first estimated its photopigment regeneration kinetics through the photo-bleach recovery of the intrinsic melanopsin pupil light response (PLR). An intense broadband light (~120,000 Td) bleached 43% of melanopsin compared to 86% of the cone-opsins. Recovery from a 43% bleach was 3.4X slower for the melanopsin than cone-opsin. Post-bleach melanopsin regeneration followed an exponential growth with a 2.5 min time-constant (τ) that required 11.2 min for complete recovery; the half-bleaching level (Ip) was ~ 4.47 log melanopic Td (16.10 log melanopsin effective photons.cm-2.s-1; 8.25 log photoisomerisations.photoreceptor-1.s-1). The effect on the cone-directed PLR of the level of the melanopsin excitation during continuous light adaptation was then determined. We observed that cone-directed pupil constriction amplitudes increased by ~ 10% when adapting lights had a higher melanopic excitation but the same mean photometric luminance. Our findings suggest that melanopsin light adaptation enhances cone signalling along the non-visual retina-brain axis. Parameters τ and Ip will allow estimation of the level of melanopsin bleaching in any light units; the data have implications for quantifying the relative contributions of putative melanopsin pathways to regulate the post-bleach photopigment regeneration and adaptation.
Collapse
Affiliation(s)
- Mukund Pant
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Andrew J Zele
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Beatrix Feigl
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Queensland Eye Institute, Brisbane, Australia
| | - Prakash Adhikari
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
| |
Collapse
|
37
|
Vlasiuk A, Asari H. Feedback from retinal ganglion cells to the inner retina. PLoS One 2021; 16:e0254611. [PMID: 34292988 PMCID: PMC8297895 DOI: 10.1371/journal.pone.0254611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/29/2021] [Indexed: 11/19/2022] Open
Abstract
Retinal ganglion cells (RGCs) are thought to be strictly postsynaptic within the retina. They carry visual signals from the eye to the brain, but do not make chemical synapses onto other retinal neurons. Nevertheless, they form gap junctions with other RGCs and amacrine cells, providing possibilities for RGC signals to feed back into the inner retina. Here we identified such feedback circuitry in the salamander and mouse retinas. First, using biologically inspired circuit models, we found mutual inhibition among RGCs of the same type. We then experimentally determined that this effect is mediated by gap junctions with amacrine cells. Finally, we found that this negative feedback lowers RGC visual response gain without affecting feature selectivity. The principal neurons of the retina therefore participate in a recurrent circuit much as those in other brain areas, not being a mere collector of retinal signals, but are actively involved in visual computations.
Collapse
Affiliation(s)
- Anastasiia Vlasiuk
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Rome, Italy
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Hiroki Asari
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Monterotondo, Rome, Italy
- * E-mail:
| |
Collapse
|
38
|
Uprety S, Zele AJ, Feigl B, Cao D, Adhikari P. Optimizing methods to isolate melanopsin-directed responses. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2021; 38:1051-1064. [PMID: 34263761 DOI: 10.1364/josaa.423343] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/01/2021] [Indexed: 06/13/2023]
Abstract
The intrinsic melanopsin photoresponse may initiate visual signals that differ in spatiotemporal characteristics from the cone-opsin- and rhodopsin-mediated signals. Applying the CIE standard observer functions in silent-substitution methods can require individual differences in photoreceptor spectral sensitivities and pre-receptoral filtering to be corrected; failure to do so can lead to the intrusion of more sensitive cone processes with putative melanopsin-directed stimuli. Here we evaluate heterochromatic flicker photometry (HFP) and photoreceptor-directed temporal white noise as techniques to limit the effect of these individual differences. Individualized luminous efficiency functions (V(λ)) were compared to the CIE standard observer functions. We show that adapting chromaticities used in silent-substitution methods can deviate by up to 54% in luminance when estimated with the individual and standard observer functions. These deviations lead to inadvertent cone intrusions in the visual functions measured with melanopsin-directed stimuli. To eliminate the intrusions, individual HFP corrections are sufficient at low frequencies (∼1Hz) but temporal white noise is also required at higher frequencies to desensitize penumbral cones. We therefore recommend the selective application of individualized observer calibration and/or temporal white noise in silent-substitution paradigms when studying melanopsin-directed photoresponses.
Collapse
|
39
|
Goel M, Mangel SC. Dopamine-Mediated Circadian and Light/Dark-Adaptive Modulation of Chemical and Electrical Synapses in the Outer Retina. Front Cell Neurosci 2021; 15:647541. [PMID: 34025356 PMCID: PMC8131545 DOI: 10.3389/fncel.2021.647541] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/06/2021] [Indexed: 12/17/2022] Open
Abstract
The vertebrate retina, like most other brain regions, undergoes relatively slow alterations in neural signaling in response to gradual changes in physiological conditions (e.g., activity changes to rest), or in response to gradual changes in environmental conditions (e.g., day changes into night). As occurs elsewhere in the brain, the modulatory processes that mediate slow adaptation in the retina are driven by extrinsic signals (e.g., changes in ambient light level) and/or by intrinsic signals such as those of the circadian (24-h) clock in the retina. This review article describes and discusses the extrinsic and intrinsic modulatory processes that enable neural circuits in the retina to optimize their visual performance throughout day and night as the ambient light level changes by ~10 billion-fold. In the first synaptic layer of the retina, cone photoreceptor cells form gap junctions with rods and signal cone-bipolar and horizontal cells (HCs). Distinct extrinsic and intrinsic modulatory processes in this synaptic layer are mediated by long-range feedback of the neuromodulator dopamine. Dopamine is released by dopaminergic cells, interneurons whose cell bodies are located in the second synaptic layer of the retina. Distinct actions of dopamine modulate chemical and electrical synapses in day and night. The retinal circadian clock increases dopamine release in the day compared to night, activating high-affinity dopamine D4 receptors on cones. This clock effect controls electrical synapses between rods and cones so that rod-cone electrical coupling is minimal in the day and robust at night. The increase in rod-cone coupling at night improves the signal-to-noise ratio and the reliability of very dim multi-photon light responses, thereby enhancing detection of large dim objects on moonless nights.Conversely, maintained (30 min) bright illumination in the day compared to maintained darkness releases sufficient dopamine to activate low-affinity dopamine D1 receptors on cone-bipolar cell dendrites. This non-circadian light/dark adaptive process regulates the function of GABAA receptors on ON-cone-bipolar cell dendrites so that the receptive field (RF) surround of the cells is strong following maintained bright illumination but minimal following maintained darkness. The increase in surround strength in the day following maintained bright illumination enhances the detection of edges and fine spatial details.
Collapse
Affiliation(s)
- Manvi Goel
- Department of Neuroscience, Ohio State University College of Medicine, Columbus, OH, United States
| | - Stuart C Mangel
- Department of Neuroscience, Ohio State University College of Medicine, Columbus, OH, United States
| |
Collapse
|
40
|
Amorim-de-Sousa A, Schilling T, Fernandes P, Seshadri Y, Bahmani H, González-Méijome JM. Blue light blind-spot stimulation upregulates b-wave and pattern ERG activity in myopes. Sci Rep 2021; 11:9273. [PMID: 33927248 PMCID: PMC8085027 DOI: 10.1038/s41598-021-88459-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 04/05/2021] [Indexed: 01/03/2023] Open
Abstract
Upregulation of retinal dopaminergic activity may be a target treatment for myopia progression. This study aimed to explore the viability of inducing changes in retinal electrical activity with short-wavelength light targeting melanopsin-expressing retinal ganglion cells (ipRGCs) passing through the optic nerve head. Fifteen healthy non-myopic or myopic young adults were recruited and underwent stimulation with blue light using a virtual reality headset device. Amplitudes and implicit times from photopic 3.0 b-wave and pattern electroretinogram (PERG) were measured at baseline and 10 and 20 min after stimulation. Relative changes were compared between non-myopes and myopes. The ERG b-wave amplitude was significantly larger 20 min after blind-spot stimulation compared to baseline (p < 0.001) and 10 min (p < 0.001) post-stimulation. PERG amplitude P50-N95 also showed a significant main effect for ‘Time after stimulation’ (p < 0.050). Implicit times showed no differences following blind-spot stimulation. PERG and b-wave changes after blind-spot stimulation were stronger in myopes than non-myopes. It is possible to induce significant changes in retinal electrical activity by stimulating ipRGCs axons at the optic nerve head with blue light. The results suggest that the changes in retinal electrical activity are located at the inner plexiform layer and are likely to involve the dopaminergic system.
Collapse
Affiliation(s)
- Ana Amorim-de-Sousa
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal
| | | | - Paulo Fernandes
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal
| | | | - Hamed Bahmani
- Dopavision GmbH, Berlin, Germany.,Department of Physiology of Cognitive Processes, Max Planck Institute for Biological Cybernetics, Tübingen, Germany.,Bernstein Center for Computational Neuroscience, Tübingen, Germany
| | - José Manuel González-Méijome
- Clinical & Experimental Optometry Research Lab (CEORLab), Center of Physics (Optometry), School of Sciences, University of Minho, Gualtar, 4710-057, Braga, Portugal.
| |
Collapse
|
41
|
Markwell EL, Feigl B, Zele AJ. Intrinsically photosensitive melanopsin retinal ganglion cell contributions to the pupillary light reflex and circadian rhythm. Clin Exp Optom 2021; 93:137-49. [DOI: 10.1111/j.1444-0938.2010.00479.x] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Emma L Markwell
- Visual Science and Medical Retina Laboratory, Institute of Health and Biomedical Innovation and School of Optometry, Queensland University of Technology, Brisbane, Queensland, Australia
E‐mail:
| | - Beatrix Feigl
- Visual Science and Medical Retina Laboratory, Institute of Health and Biomedical Innovation and School of Optometry, Queensland University of Technology, Brisbane, Queensland, Australia
E‐mail:
| | - Andrew J Zele
- Visual Science and Medical Retina Laboratory, Institute of Health and Biomedical Innovation and School of Optometry, Queensland University of Technology, Brisbane, Queensland, Australia
E‐mail:
| |
Collapse
|
42
|
Najjar RP, Chao De La Barca JM, Barathi VA, Ho CEH, Lock JZ, Muralidharan AR, Tan RKY, Dhand C, Lakshminarayanan R, Reynier P, Milea D. Ocular growth and metabolomics are dependent upon the spectral content of ambient white light. Sci Rep 2021; 11:7586. [PMID: 33828194 PMCID: PMC8026599 DOI: 10.1038/s41598-021-87201-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Myopia results from an excessive axial growth of the eye, causing abnormal projection of remote images in front of the retina. Without adequate interventions, myopia is forecasted to affect 50% of the world population by 2050. Exposure to outdoor light plays a critical role in preventing myopia in children, possibly through the brightness and blue-shifted spectral composition of sunlight, which lacks in artificial indoor lighting. Here, we evaluated the impact of moderate levels of ambient standard white (SW: 233.1 lux, 3900 K) and blue-enriched white (BEW: 223.8 lux, 9700 K) lights on ocular growth and metabolomics in a chicken-model of form-deprivation myopia. Compared to SW light, BEW light decreased aberrant ocular axial elongation and accelerated recovery from form-deprivation. Furthermore, the metabolomic profiles in the vitreous and retinas of recovering form-deprived eyes were distinct from control eyes and were dependent on the spectral content of ambient light. For instance, exposure to BEW light was associated with deep lipid remodeling and metabolic changes related to energy production, cell proliferation, collagen turnover and nitric oxide metabolism. This study provides new insight on light-dependent modulations in ocular growth and metabolomics. If replicable in humans, our findings open new potential avenues for spectrally-tailored light-therapy strategies for myopia.
Collapse
Affiliation(s)
- Raymond P Najjar
- Singapore Eye Research Institute, Singapore, Singapore.
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore.
| | - Juan Manuel Chao De La Barca
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire d'Angers, Angers, France
- Unité Mixte de Recherche MITOVASC, CNRS 6015, INSERM U1083, Université d'Angers, Angers, France
| | - Veluchamy A Barathi
- Singapore Eye Research Institute, Singapore, Singapore
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | | - Royston K Y Tan
- Department of Ocular Bio-Engineering, National University of Singapore, Singapore, Singapore
| | - Chetna Dhand
- Singapore Eye Research Institute, Singapore, Singapore
- CSIR-Advanced Materials and Processes Research Institute, Hoshangabad Road, Bhopal, 462026, India
| | | | - Pascal Reynier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire d'Angers, Angers, France
- Unité Mixte de Recherche MITOVASC, CNRS 6015, INSERM U1083, Université d'Angers, Angers, France
| | - Dan Milea
- Singapore Eye Research Institute, Singapore, Singapore.
- The Ophthalmology and Visual Sciences ACP, Duke-NUS Medical School, Singapore, Singapore.
- Singapore National Eye Center, Singapore, Singapore.
| |
Collapse
|
43
|
Abstract
SIGNIFICANCE Pupillometry protocols evaluating rod/cone- and melanopsin-driven responses often use mydriatics to ensure maximal stimulus exposure; however, retinal effects of mydriatics are not fully understood. We demonstrate that dilation with either atropine or phenylephrine results in similar enhancements of rod/cone- and melanopsin-driven pupil responses. PURPOSE The purposes of this study were to compare the effects of atropine, a muscarinic antagonist, and phenylephrine, an adrenergic agonist, on consensual pupil responses and to assess the repeatability of pupil metrics without mydriasis. METHODS Right eye pupil responses of 20 adults aged 21 to 42 years were recorded before and 45 minutes after instillation of 0.5% atropine or 2.5% phenylephrine in the left eye. Stimuli were presented to the left eye and included six alternating 1-second 651-nm "red" and 456-nm "blue" flashes. Metrics included baseline pupil diameter, maximal constriction, 6- and 30-second post-illumination pupil responses, and early (0 to 10 seconds) and late (10 to 30 seconds) areas under the curve. RESULTS Dilation of the stimulated eye with either mydriatic significantly increased the 6-second post-illumination pupil response and early and late areas under the curve for blue stimuli, and early area under the curve for red stimuli (P < .05 for all). Melanopsin-driven post-illumination pupil responses, achieved with either phenylephrine or atropine, did not significantly differ from each other (P > .05 for all). Without mydriasis, intersession intraclass correlation coefficients for pupil metrics were 0.63 and 0.50 (6- and 30-second post-illumination pupil responses, respectively) and 0.78 and 0.44 (early and late areas under the curve, respectively) for blue stimuli, with no significant difference between sessions (P > .05 for all). CONCLUSIONS Dilation with phenylephrine or atropine resulted in similar enhancements of the rod/cone- and melanopsin-driven pupil responses, despite differing mechanisms. Early pupil metrics without mydriasis demonstrated moderate to good intersession repeatability.
Collapse
|
44
|
Pottackal J, Walsh HL, Rahmani P, Zhang K, Justice NJ, Demb JB. Photoreceptive Ganglion Cells Drive Circuits for Local Inhibition in the Mouse Retina. J Neurosci 2021; 41:1489-1504. [PMID: 33397711 PMCID: PMC7896016 DOI: 10.1523/jneurosci.0674-20.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 11/11/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs) exhibit melanopsin-dependent light responses that persist in the absence of rod and cone photoreceptor-mediated input. In addition to signaling anterogradely to the brain, ipRGCs signal retrogradely to intraretinal circuitry via gap junction-mediated electrical synapses with amacrine cells (ACs). However, the targets and functions of these intraretinal signals remain largely unknown. Here, in mice of both sexes, we identify circuitry that enables M5 ipRGCs to locally inhibit retinal neurons via electrical synapses with a nonspiking GABAergic AC. During pharmacological blockade of rod- and cone-mediated input, whole-cell recordings of corticotropin-releasing hormone-expressing (CRH+) ACs reveal persistent visual responses that require both melanopsin expression and gap junctions. In the developing retina, ipRGC-mediated input to CRH+ ACs is weak or absent before eye opening, indicating a primary role for this input in the mature retina (i.e., in parallel with rod- and cone-mediated input). Among several ipRGC types, only M5 ipRGCs exhibit consistent anatomical and physiological coupling to CRH+ ACs. Optogenetic stimulation of local CRH+ ACs directly drives IPSCs in M4 and M5, but not M1-M3, ipRGCs. CRH+ ACs also inhibit M2 ipRGC-coupled spiking ACs, demonstrating direct interaction between discrete networks of ipRGC-coupled interneurons. Together, these results demonstrate a functional role for electrical synapses in translating ipRGC activity into feedforward and feedback inhibition of local retinal circuits.SIGNIFICANCE STATEMENT Melanopsin directly generates light responses in intrinsically photosensitive retinal ganglion cells (ipRGCs). Through gap junction-mediated electrical synapses with retinal interneurons, these uniquely photoreceptive RGCs may also influence the activity and output of neuronal circuits within the retina. Here, we identified and studied an electrical synaptic circuit that, in principle, could couple ipRGC activity to the chemical output of an identified retinal interneuron. Specifically, we found that M5 ipRGCs form electrical synapses with corticotropin-releasing hormone-expressing amacrine cells, which locally release GABA to inhibit specific RGC types. Thus, ipRGCs are poised to influence the output of diverse retinal circuits via electrical synapses with interneurons.
Collapse
Affiliation(s)
| | | | | | | | - Nicholas J Justice
- Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas 77030
| | - Jonathan B Demb
- Interdepartmental Neuroscience Program
- Department of Ophthalmology and Visual Science
- Department of Cellular and Molecular Physiology
- Department of Neuroscience, Yale University, New Haven, Connecticut 06511
| |
Collapse
|
45
|
Wong KY, Fernandez FX. Circadian Responses to Light-Flash Exposure: Conceptualization and New Data Guiding Future Directions. Front Neurol 2021; 12:627550. [PMID: 33643205 PMCID: PMC7905211 DOI: 10.3389/fneur.2021.627550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/21/2021] [Indexed: 01/03/2023] Open
Abstract
A growing number of studies document circadian phase-shifting after exposure to millisecond light flashes. When strung together by intervening periods of darkness, these stimuli evoke pacemaker responses rivaling or outmatching those created by steady luminance, suggesting that the circadian system's relationship to light can be contextualized outside the principle of simple dose-dependence. In the current review, we present a brief chronology of this work. We then develop a conceptual model around it that attempts to relate the circadian effects of flashes to a natural integrative process the pacemaker uses to intermittently sample the photic information available at dawn and dusk. Presumably, these snapshots are employed as building blocks in the construction of a coherent representation of twilight the pacemaker consults to orient the next day's physiology (in that way, flash-resetting of pacemaker rhythms might be less an example of a circadian visual illusion and more an example of the kinds of gestalt inferences that the image-forming system routinely makes when identifying objects within the visual field; i.e., closure). We conclude our review with a discussion on the role of cones in the pacemaker's twilight predictions, providing new electrophysiological data suggesting that classical photoreceptors—but not melanopsin—are necessary for millisecond, intermediate-intensity flash responses in ipRGCs (intrinsically photosensitive retinal ganglion cells). Future investigations are necessary to confirm this “Cone Sentinel Model” of circadian flash-integration and twilight-prediction, and to further define the contribution of cones vs. rods in transducing pacemaker flash signals.
Collapse
Affiliation(s)
- Kwoon Y Wong
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, MI, United States.,Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Fabian-Xosé Fernandez
- Department of Psychology, BIO5 Research Institute, University of Arizona, Tucson, AZ, United States.,Department of Neurology, McKnight Brain Research Institute, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
46
|
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: 43] [Impact Index Per Article: 10.8] [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
|
47
|
Harrison KR, Chervenak AP, Resnick SM, Reifler AN, Wong KY. Amacrine Cells Forming Gap Junctions With Intrinsically Photosensitive Retinal Ganglion Cells: ipRGC Types, Neuromodulator Contents, and Connexin Isoform. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 33410914 PMCID: PMC7804497 DOI: 10.1167/iovs.62.1.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Intrinsically photosensitive retinal ganglion cells (ipRGCs) signal not only centrally to non-image-forming visual centers of the brain but also intraretinally to amacrine interneurons through gap junction electrical coupling, potentially modulating image-forming retinal processing. We aimed to determine (1) which ipRGC types couple with amacrine cells, (2) the neuromodulator contents of ipRGC-coupled amacrine cells, and (3) whether connexin36 (Cx36) contributes to ipRGC-amacrine coupling. Methods Gap junction-permeable Neurobiotin tracer was injected into green fluorescent protein (GFP)-labeled ipRGCs in Opn4Cre/+; Z/EG mice to stain coupled amacrine cells, and immunohistochemistry was performed to reveal the neuromodulator contents of the Neurobiotin-stained amacrine cells. We also created Opn4Cre/+; Cx36flox/flox; Z/EG mice to knock out Cx36 in GFP-labeled ipRGCs and looked for changes in the number of ipRGC-coupled amacrine cells. Results Seventy-three percent of ipRGCs, including all six types (M1-M6), were tracer-coupled with amacrine somas 5.7 to 16.5 µm in diameter but not with ganglion cells. Ninety-two percent of the ipRGC-coupled somas were in the ganglion cell layer and the rest in the inner nuclear layer. Some ipRGC-coupled amacrine cells were found to accumulate serotonin or to contain nitric oxide synthase or neuropeptide Y. Knocking out Cx36 in M2 and M4 dramatically reduced the number of coupled somas. Conclusions Heterologous gap junction coupling with amacrine cells is widespread across mouse ipRGC types. ipRGC-coupled amacrine cells probably comprise multiple morphologic types and use multiple neuromodulators, suggesting that gap junctional ipRGC-to-amacrine signaling likely exerts diverse modulatory effects on retinal physiology. ipRGC-amacrine coupling is mediated partly, but not solely, by Cx36.
Collapse
Affiliation(s)
- Krystal R. Harrison
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
| | - Andrew P. Chervenak
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Sarah M. Resnick
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Aaron N. Reifler
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| | - Kwoon Y. Wong
- Department of Molecular, Cellular, & Developmental Biology, University of Michigan, Ann Arbor, Michigan, United States
- Department of Ophthalmology & Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
48
|
Abstract
Retinal ganglion cells (RGCs) serve as a crucial communication channel from the retina to the brain. In the adult, these cells receive input from defined sets of presynaptic partners and communicate with postsynaptic brain regions to convey features of the visual scene. However, in the developing visual system, RGC interactions extend beyond their synaptic partners such that they guide development before the onset of vision. In this Review, we summarize our current understanding of how interactions between RGCs and their environment influence cellular targeting, migration and circuit maturation during visual system development. We describe the roles of RGC subclasses in shaping unique developmental responses within the retina and at central targets. Finally, we highlight the utility of RNA sequencing and genetic tools in uncovering RGC type-specific roles during the development of the visual system.
Collapse
Affiliation(s)
- Shane D'Souza
- The Visual Systems Group, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| | - Richard A Lang
- The Visual Systems Group, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Center for Chronobiology, Abrahamson Pediatric Eye Institute, Division of Pediatric Ophthalmology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
- Department of Ophthalmology, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
49
|
Ortuño-Lizarán I, Sánchez-Sáez X, Lax P, Serrano GE, Beach TG, Adler CH, Cuenca N. Dopaminergic Retinal Cell Loss and Visual Dysfunction in Parkinson Disease. Ann Neurol 2020; 88:893-906. [PMID: 32881029 PMCID: PMC10005860 DOI: 10.1002/ana.25897] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Considering the demonstrated implication of the retina in Parkinson disease (PD) pathology and the importance of dopaminergic cells in this tissue, we aimed to analyze the state of the dopaminergic amacrine cells and some of their main postsynaptic neurons in the retina of PD. METHODS Using immunohistochemistry and confocal microscopy, we evaluated morphology, number, and synaptic connections of dopaminergic cells and their postsynaptic cells, AII amacrine and melanopsin-containing retinal ganglion cells, in control and PD eyes from human donors. RESULTS In PD, dopaminergic amacrine cell number was reduced between 58% and 26% in different retinal regions, involving a decline in the number of synaptic contacts with AII amacrine cells (by 60%) and melanopsin cells (by 35%). Despite losing their main synaptic input, AII cells were not reduced in number, but they showed cellular alterations compromising their adequate function: (1) a loss of mitochondria inside their lobular appendages, which may indicate an energetic failure; and (2) a loss of connexin 36, suggesting alterations in the AII coupling and in visual signal transmission from the rod pathway. INTERPRETATION The dopaminergic system impairment and the affection of the rod pathway through the AII cells may explain and be partially responsible for the reduced contrast sensitivity or electroretinographic response described in PD. Also, dopamine reduction and the loss of synaptic contacts with melanopsin cells may contribute to the melanopsin retinal ganglion cell loss previously described and to the disturbances in circadian rhythm and sleep reported in PD patients. These data support the idea that the retina reproduces brain neurodegeneration and is highly involved in PD pathology. ANN NEUROL 2020;88:893-906.
Collapse
Affiliation(s)
- Isabel Ortuño-Lizarán
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | | | | | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
| |
Collapse
|
50
|
Feigl B, Dumpala S, Kerr GK, Zele AJ. Melanopsin Cell Dysfunction is Involved in Sleep Disruption in Parkinson’s Disease. JOURNAL OF PARKINSONS DISEASE 2020; 10:1467-1476. [DOI: 10.3233/jpd-202178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Melanopsin-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs) signal the environmental light to mediate circadian photoentrainment and sleep-wake cycles. There is high prevalence of circadian and sleep disruption in people with Parkinson’s disease, however the underlying mechanisms of these symptoms are not clear. Objective: Based on recent evidence of anatomical and functional loss of melanopsin ganglion cells in Parkinson’s disease, we evaluate the link between melanopsin function, circadian, and sleep behavior. Methods: The pupil light reflex and melanopsin-mediated post-illumination pupil response were measured using chromatic pupillometry in 30 optimally medicated people with Parkinson’s disease and 29 age-matched healthy controls. Circadian health was determined using dim light melatonin onset, sleep questionnaires, and actigraphy. Ophthalmic examination quantified eye health and optical coherence tomography measured retinal thickness. Results: The melanopsin-mediated post-illumination pupil response amplitudes were significantly reduced in Parkinson’s disease (p < 0.0001) and correlated with poor sleep quality (r2 = 33; p < 0.001) and nerve fiber layer thinning (r2 = 0.40; p < 0.001). People with Parkinson’s disease had significantly poorer sleep quality with higher subjective sleep scores (p < 0.05) and earlier melatonin onset (p = 0.01). Pupil light (outer retinal) response metrics, daily light exposure and outer retinal thickness were similar between the groups (p > 0.05). Conclusion: Our evidence-based data identify a mechanism through which inner retinal ipRGC dysfunction contributes to sleep disruption in Parkinson’s disease in the presence of normal outer retinal (rod-cone photoreceptor) function. Our findings provide a rationale for designing new treatment approaches in Parkinson’s disease through melanopsin photoreceptor-targeted light therapies for improving sleep-wake cycles.
Collapse
Affiliation(s)
- Beatrix Feigl
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Australia
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Australia
- Queensland Eye Institute (QEI), Brisbane, Australia
| | - Sunila Dumpala
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Australia
- School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, Australia
| | - Graham K. Kerr
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Movement Neuroscience Program, Queensland University of Technology (QUT), Brisbane, Australia
| | - Andrew J. Zele
- Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), Brisbane, Australia
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, Australia
- School of Optometry and Vision Science, Queensland University of Technology (QUT), Brisbane, Australia
| |
Collapse
|