1
|
Barta CL, Thoreson WB. Retinal inputs that drive optomotor responses of mice under mesopic conditions. IBRO Neurosci Rep 2024; 17:138-144. [PMID: 39170059 PMCID: PMC11338136 DOI: 10.1016/j.ibneur.2024.07.003] [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: 03/05/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/23/2024] Open
Abstract
Optomotor responses are a popular way to assess sub-cortical visual responses in mice. We studied photoreceptor inputs into optomotor circuits using genetically-modified mice lacking the exocytotic calcium sensors synaptotagmin 1 (Syt1) and 7 (Syt7) in rods or cones. We also tested mice that in which cone transducin, GNAT2, had been eliminated. We studied spatial frequency sensitivity under mesopic conditions by varying the spatial frequency of a grating rotating at 12 deg/s and contrast sensitivity by varying luminance contrast of 0.2c/deg gratings. We found that eliminating Syt1 from rods reduced responses to a low spatial frequency grating (0.05c/deg) consistent with low resolution in this pathway. Conversely, eliminating the ability of cones to respond to light (by eliminating GNAT2) or transmit light responses (by selectively eliminating Syt1) showed weaker responses to a high spatial frequency grating (3c/deg). Eliminating Syt7 from the entire optomotor pathway in a global knockout had no significant effect on optomotor responses. We isolated the secondary rod pathway involving transmission of rod responses to cones via gap junctions by simultaneously eliminating Syt1 from rods and GNAT2 from cones. We found that the secondary rod pathway is sufficient to drive robust optomotor responses under mesopic conditions. Finally, eliminating Syt1 from both rods and cones almost completely abolished optomotor responses, but we detected weak responses to large, bright rotating gratings that are likely driven by input from intrinsically photosensitive retinal ganglion cells.
Collapse
Affiliation(s)
- CL Barta
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
| | - WB Thoreson
- Truhlsen Eye Institute and Department of Ophthalmology and Visual Sciences, USA
- Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
2
|
Wilmet B, Michiels C, Zhang J, Callebert J, Sahel JA, Picaud S, Audo I, Zeitz C. Loss of ON-Pathway Function in Mice Lacking Lrit3 Decreases Recovery From Lens-Induced Myopia. Invest Ophthalmol Vis Sci 2024; 65:18. [PMID: 39250117 PMCID: PMC11385651 DOI: 10.1167/iovs.65.11.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Abstract
Purpose To determine whether the Lrit3-/- mouse model of complete congenital stationary night blindness with an ON-pathway defect harbors myopic features and whether the genetic defect influences the recovery from lens-induced myopia. Methods Retinal levels of dopamine (DA) and 3,4 dihydroxyphenylacetic acid (DOPAC) from adult isolated Lrit3-/- retinas were quantified using ultra performance liquid chromatography after light adaptation. Natural refractive development of Lrit3-/- mice was measured from three weeks to nine weeks of age using an infrared photorefractometer. Susceptibility to myopia induction was assessed using a lens-induced myopia protocol with -25 D lenses placed in front of the right eye of the animals for three weeks; the mean interocular shift was measured with an infrared photorefractometer after two and three weeks of goggling and after one and two weeks after removal of goggles. Results Compared to wild-type littermates (Lrit3+/+), both DA and DOPAC were drastically reduced in Lrit3-/- retinas. Natural refractive development was normal but Lrit3-/- mice showed a higher myopic shift and a lower ability to recover from induced myopia. Conclusions Our data consolidate the link between ON pathway defect altered dopaminergic signaling and myopia. We document for the first time the role of ON pathway on the recovery from myopia induction.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Jingyi Zhang
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburg, PA, United States
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, Centre de Référence Maladies Rares REFERET and INSERM-DGOS CIC 1423, Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
3
|
Schaeffel F, Swiatczak B. Mechanisms of emmetropization and what might go wrong in myopia. Vision Res 2024; 220:108402. [PMID: 38705024 DOI: 10.1016/j.visres.2024.108402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024]
Abstract
Studies in animal models and humans have shown that refractive state is optimized during postnatal development by a closed-loop negative feedback system that uses retinal image defocus as an error signal, a mechanism called emmetropization. The sensor to detect defocus and its sign resides in the retina itself. The retina and/or the retinal pigment epithelium (RPE) presumably releases biochemical messengers to change choroidal thickness and modulate the growth rates of the underlying sclera. A central question arises: if emmetropization operates as a closed-loop system, why does it not stop myopia development? Recent experiments in young human subjects have shown that (1) the emmetropic retina can perfectly distinguish between real positive defocus and simulated defocus, and trigger transient axial eye shortening or elongation, respectively. (2) Strikingly, the myopic retina has reduced ability to inhibit eye growth when positive defocus is imposed. (3) The bi-directional response of the emmetropic retina is elicited with low spatial frequency information below 8 cyc/deg, which makes it unlikely that optical higher-order aberrations play a role. (4) The retinal mechanism for the detection of the sign of defocus involves a comparison of defocus blur in the blue (S-cone) and red end of the spectrum (L + M-cones) but, again, the myopic retina is not responsive, at least not in short-term experiments. This suggests that it cannot fully trigger the inhibitory arm of the emmetropization feedback loop. As a result, with an open feedback loop, myopia development becomes "open-loop".
Collapse
Affiliation(s)
- Frank Schaeffel
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland; Section Neurobiology of the Eye, Institute of Ophthalmic Research, University of Tübingen, Germany; Zeiss Vision Lab, Institute of Ophthalmic Research, University of Tübingen, Germany.
| | - Barbara Swiatczak
- Myopia Research Group, Institute of Molecular and Clinical Ophthalmology Basel (IOB), Switzerland
| |
Collapse
|
4
|
Bales KL, Karesh AM, Hogan K, Chacko AS, Douglas GL, Feola AJ, Nickerson JM, Pybus A, Wood L, Boatright JH, Pardue MT. Voluntary exercise preserves visual function and reduces inflammatory response in an adult mouse model of autosomal dominant retinitis pigmentosa. Sci Rep 2024; 14:6940. [PMID: 38521799 PMCID: PMC10960803 DOI: 10.1038/s41598-024-57027-9] [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: 06/08/2023] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Whole-body physical exercise has been shown to promote retinal structure and function preservation in animal models of retinal degeneration. It is currently unknown how exercise modulates retinal inflammatory responses. In this study, we investigated cytokine alterations associated with retinal neuroprotection induced by voluntary running wheel exercise in a retinal degeneration mouse model of class B1 autosomal dominant retinitis pigmentosa, I307N Rho. I307N Rho mice undergo rod photoreceptor degeneration when exposed to bright light (induced). Our data show, active induced mice exhibited significant preservation of retinal and visual function compared to inactive induced mice after 4 weeks of exercise. Retinal cytokine expression revealed significant reductions of proinflammatory chemokines, keratinocyte-derived chemokine (KC) and interferon gamma inducible protein-10 (IP-10) expression in active groups compared to inactive groups. Through immunofluorescence, we found KC and IP-10 labeling localized to retinal vasculature marker, collagen IV. These data show that whole-body exercise lowers specific retinal cytokine expression associated with retinal vasculature. Future studies should determine whether suppression of inflammatory responses is requisite for exercise-induced retinal protection.
Collapse
Affiliation(s)
- Katie L Bales
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | - Austin M Karesh
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Kelleigh Hogan
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | - Alicia S Chacko
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - GianMarco L Douglas
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
| | - Andrew J Feola
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | | | - Alyssa Pybus
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Levi Wood
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jeffrey H Boatright
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Atlanta VA Medical Center for Visual and Neurocognitive Rehabilitation, Decatur, GA, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.
- Department of Ophthalmology, Emory University, 1365B Clifton Road NE, Rm. 2600, Atlanta, GA, 30332, USA.
| |
Collapse
|
5
|
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: 28] [Impact Index Per Article: 28.0] [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
|
6
|
Wilmet B, Callebert J, Duvoisin R, Goulet R, Tourain C, Michiels C, Frederiksen H, Schaeffel F, Marre O, Sahel JA, Audo I, Picaud S, Zeitz C. Mice Lacking Gpr179 with Complete Congenital Stationary Night Blindness Are a Good Model for Myopia. Int J Mol Sci 2022; 24:ijms24010219. [PMID: 36613663 PMCID: PMC9820543 DOI: 10.3390/ijms24010219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/25/2022] Open
Abstract
Mutations in GPR179 are one of the most common causes of autosomal recessive complete congenital stationary night blindness (cCSNB). This retinal disease is characterized in patients by impaired dim and night vision, associated with other ocular symptoms, including high myopia. cCSNB is caused by a complete loss of signal transmission from photoreceptors to ON-bipolar cells. In this study, we hypothesized that the lack of Gpr179 and the subsequent impaired ON-pathway could lead to myopic features in a mouse model of cCSNB. Using ultra performance liquid chromatography, we show that adult Gpr179-/- mice have a significant decrease in both retinal dopamine and 3,4-dihydroxyphenylacetic acid, compared to Gpr179+/+ mice. This alteration of the dopaminergic system is thought to be correlated with an increased susceptibility to lens-induced myopia but does not affect the natural refractive development. Altogether, our data added a novel myopia model, which could be used to identify therapeutic interventions.
Collapse
Affiliation(s)
- Baptiste Wilmet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| | - Jacques Callebert
- Service of Biochemistry and Molecular Biology, INSERM U942, Hospital Lariboisière, AP-HP, 75010 Paris, France
| | - Robert Duvoisin
- Department of Chemical Physiology & Biochemistry, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruben Goulet
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christophe Tourain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Wavefront-Engineering Microscopy Group, Neurophotonics Laboratory, CNRS UMR8250, Paris Descartes University, 75270 Paris, France
| | - Christelle Michiels
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Helen Frederiksen
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Frank Schaeffel
- Institute of Molecular and Clinical Ophthalmology Basel (IOB), 4056 Basel, Switzerland
- Section of Neurobiology of the Eye, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
- Zeiss Vision Lab, Ophthalmic Research Institute, University of Tuebingen, 72076 Tuebingen, Germany
| | - Olivier Marre
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - José Alain Sahel
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
- Fondation Ophtalmologique Adolphe de Rothschild, 75019 Paris, France
- Académie des Sciences, Institut de France, 75006 Paris, France
| | - Isabelle Audo
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Centre Hospitalier National d’Ophtalmologie des Quinze-Vingts, INSERM-DGOS CIC 1423, 75012 Paris, France
| | - Serge Picaud
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
| | - Christina Zeitz
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 75012 Paris, France
- Correspondence: (B.W.); (C.Z.); Tel.: +33-1-53-46-25-26 (B.W.); +33-1-53-46-25-40 (C.Z.)
| |
Collapse
|
7
|
Ma W, Li X, Yang L, Pan J, Chen Y, Lu Y, Dong X, Li D, Gan W. High VSX1 expression promotes the aggressiveness of clear cell renal cell carcinoma by transcriptionally regulating FKBP10. J Transl Med 2022; 20:554. [PMID: 36463181 PMCID: PMC9719260 DOI: 10.1186/s12967-022-03772-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/12/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC), the most common urological malignancy, has an unfavorable prognosis and an unknown mechanism of progression. Through survival analyses screening of The Cancer Genome Atlas (TCGA) dataset, we identified Visual system homeobox1 (VSX1) as a novel potential prognostic biomarker in ccRCC and subsequently investigated the oncogenic role of VSX1 in ccRCC. METHODS The differential expression of VSX1 in human tumors and the clinical prognoses were analyzed in the TCGA dataset and Gene Expression Omnibus. Spearman's correlation coefficient was determined for the correlation analysis of VSX1 expression and other genes of interest. The roles of VSX1 in cell proliferation, invasion, and migration of ccRCC cells were evaluated via the CCK-8 assay, colony formation assay, and Transwell assay, respectively. Further results were demonstrated by western blotting, immunohistochemistry, qRT-PCR, tumor sphere formation, flow cytometry, and the dual‑luciferase reporter assay. RESULTS VSX1 mRNA upregulation was generally observed in multiple human malignancies from the TCGA database and was confirmed in ccRCC clinical specimens from our department. High VSX1 expression usually indicated that overall and disease-free survival were unfavorable for patients with ccRCC. In terms of mechanism, knockdown or overexpression of VSX1 affected ccRCC aggressiveness in vitro. The dual-luciferase reporter gene assay implied that VSX1 overexpression significantly increased the luciferase activity of TMEM44, FKBP10, and TRIB3, which indicated that VSX1 promoted ccRCC invasiveness via transcriptional regulation of these genes. The significantly enhanced growth in vitro that was induced by stable VSX1 overexpression was almost restored to normal by the knockdown of FKBP10. CONCLUSIONS This study demonstrated that VSX1 was a novel prognostic biomarker in ccRCC and that high VSX1 expression promoted cell proliferation, invasion, and migration in ccRCC via transcriptional activation of downstream target genes.
Collapse
Affiliation(s)
- Wenliang Ma
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Xin Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Lei Yang
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Jun Pan
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Yi Chen
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China
| | - Yanwen Lu
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Xiang Dong
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China
| | - Dongmei Li
- Immunology and Reproduction Biology Laboratory & State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, People's Republic of China.
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, 210093, Jiangsu, China.
| | - Weidong Gan
- Department of Urology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Ichinose T, Habib S. ON and OFF Signaling Pathways in the Retina and the Visual System. FRONTIERS IN OPHTHALMOLOGY 2022; 2:989002. [PMID: 36926308 PMCID: PMC10016624 DOI: 10.3389/fopht.2022.989002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Visual processing starts at the retina of the eye, and signals are then transferred primarily to the visual cortex and the tectum. In the retina, multiple neural networks encode different aspects of visual input, such as color and motion. Subsequently, multiple neural streams in parallel convey unique aspects of visual information to cortical and subcortical regions. Bipolar cells, which are the second order neurons of the retina, separate visual signals evoked by light and dark contrasts and encode them to ON and OFF pathways, respectively. The interplay between ON and OFF neural signals is the foundation for visual processing for object contrast which underlies higher order stimulus processing. ON and OFF pathways have been classically thought to signal in a mirror-symmetric manner. However, while these two pathways contribute synergistically to visual perception in some instances, they have pronounced asymmetries suggesting independent operation in other cases. In this review, we summarize the role of the ON-OFF dichotomy in visual signaling, aiming to contribute to the understanding of visual recognition.
Collapse
Affiliation(s)
- Tomomi Ichinose
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
- Correspondence: Tomomi Ichinose, MD, PhD,
| | - Samar Habib
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Medical Parasitology, Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|