1
|
Morgan JE, Bevan RJ, Cimaglia G. Retinal Ganglion Cell Subtypes and Their Vulnerability in Glaucoma. Methods Mol Biol 2025; 2858:191-205. [PMID: 39433677 DOI: 10.1007/978-1-0716-4140-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The detection of selective retinal ganglion cell damage in glaucoma has been a long sought-after goal, not just for the development of clinical tests for the early detection of glaucoma but for the elucidation of potential mechanisms underlying retinal ganglion cell loss. Early reports of the selective vulnerability of larger retinal ganglion cells (RGCs) in human studies did not translate simply to the loss of a particular class of RGC but more likely reflected shrinkage and degeneration across all RGC classes. Subsequent studies of nonhuman primate (NHP) models of glaucoma indicated some selectivity with great damage to the magnocellular vs parvocellular pathways. More recently, rodent models of experimental glaucoma have highlighted a selective vulnerability of OFF-centered RGCs-particularly those with transient responses. Selectivity for OFF pathway damage is also seen as a trend in a rat model of glaucoma. These data support the concept that some RGCs are more vulnerable to the effects of glaucoma damage. This chapter covers some of the methods to elucidate RGC damage and the relevance of model selection to mimic human glaucoma rather than just RGC death.
Collapse
Affiliation(s)
- James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Ryan J Bevan
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| | - Gloria Cimaglia
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| |
Collapse
|
2
|
Lu W, Wang Y, Hu W, Lin X, Tong X, Tian Y, Chen Y, Wang Y, Xiao Y, Yang H, Feng Y, Sun X. A novel three-dimensional method for detailed analysis of RGC central projections under acute ocular hypertension. Exp Eye Res 2025; 250:110157. [PMID: 39571780 DOI: 10.1016/j.exer.2024.110157] [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: 08/12/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024]
Abstract
Normal perception of visual information relies not only on the quantity and quality of retinal ganglion cells (RGCs), but also on the integrity of the visual pathway, within which RGC central projection predominates. However, the exact changes of RGC central projection under particular pathological conditions remain to be elucidated. Here, we report a whole-brain clearing method modified from iDISCO for 3D visualization of RGC central projection. The CTB-labeled RGC central projection was visualized three-dimensionally with minimized both fluorescence quenching and the time taken. For observation of RGC axonal degeneration pattern under pathological conditions, we took acute ocular hypertension (AOH) as an example. Mice were intracamerally irrigated, and fluorescent signal in brain subregions where RGC axons projected to were quantified. The novel methodology is well-applied for rapid clearing and observation of RGC central projection in C57BL/6J, showing damaged RGC central projection on the AOH side and the most statistically significant degeneration in the superior colliculi (SC). Detailed analysis also revealed a distinct injury pattern among lateral geniculate nuclei (LGN) subregions, with the parvocellular part of the pregeniculate nuclei (PrGPC) being more vulnerable compared with the magnocellular part (PrGMC). The intracranial retrograde labeling of RGC subgroups based on brain damage variation showed PrGPC-projecting RGCs (Plgn RGC) being smaller than PrGMC-projecting RGCs (Mlgn RGC) in size and less in number, yet more vulnerable in terms of degeneration under AOH. Our data revealed the methodology for visualizing selective neuronal vulnerability under AOH, and in the meantime provided novel approach for future mechanisms exploration regarding RGC degeneration.
Collapse
Affiliation(s)
- Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200032, China; Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China
| | - Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China
| | - Xinyi Lin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China
| | - Yi Tian
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200032, China
| | - Yuning Chen
- Reproductive Medicine Center, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China
| | - Yan Xiao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China
| | - Hongfang Yang
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200032, China.
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, Brain Science Collaborative Innovation Center, State Key Laboratory of Medical Neurobiology, Institute of Acupuncture and Moxibustion, Fudan Institutes of Integrative Medicine, Fudan University, Shanghai, 200032, China; Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai, 200032, China.
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200032, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, 200032, China.
| |
Collapse
|
3
|
Roy S. Emerging strategies targeting genes and cells in glaucoma. Vision Res 2024; 227:108533. [PMID: 39644708 DOI: 10.1016/j.visres.2024.108533] [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: 09/05/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Glaucoma comprises a heterogeneous set of eye conditions that cause progressive vision loss. Glaucoma has a complex etiology, with different genetic and non-genetic risk factors that differ across populations. Although difficult to diagnose in early stages, compromised cellular signaling, dysregulation of genes, and homeostatic imbalance are common precursors to injury and subsequent death of retinal ganglion cells (RGCs). Lowering intraocular pressure (IOP) remains the primary approach for managing glaucoma but IOP alone does not explain all glaucoma risks. Orthogonal approaches such as large-scale genetic screening, combined with studies of animal models have been instrumental in identifying genes and molecular pathways involved in glaucoma pathogenesis. Cell type dependent vulnerability among RGCs can reveal genetic basis for specific visual deficits. A growing body of knowledge and availability of modern tools to perform targeted assessments of cellular health in different animal models facilitate development of effective and timely interventions for vision rescue. This review highlights recent findings on genes, molecules, and cell types in the context of glaucoma pathophysiology and treatment.
Collapse
Affiliation(s)
- Suva Roy
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
4
|
Phu J, Khuu SK, Nivison-Smith L, Kalloniatis M. Standard automated perimetry for glaucoma and diseases of the retina and visual pathways: current and future perspectives. Prog Retin Eye Res 2024:101307. [PMID: 39413870 DOI: 10.1016/j.preteyeres.2024.101307] [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: 06/14/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Static automated perimetry (SAP) remains a mainstay of functional assessment of the visual field in diseases of the visual pathway, such as glaucoma and age-related macular degeneration. The fundamental psychophysical task of responding to stimuli of different levels of contrast has remained minimally changed since its inception in the 1980s, and this is potentially the root of several unresolved issues involving the technique. Enduring issues include the optimisation of SAP parameters for maximising defect detection, the influence of subjective behaviour on the response, structure-function discordance, and ageing- and disease-related changes of the visual pathway. Addressing these issues has been a focus of our research program and is the subject of this manuscript. We will review some of the basic psychophysical principles and methods that have contributed to the development of SAP and their contributions to its output measurements. Parameters that are interrogated include stimulus size and background luminance and their modification to improve defect defection in glaucoma and age-related macular degeneration. We propose frameworks for optimising testing parameters and leveraging the results for changing clinical care. In our pursuit of optimising the structure-function relationship in the eye, several areas of research have been developed and explored, including: the reconciliation of subjective responses in perimetry; by minimising sources of biases, such as Method of Limits we have been able to equate static and kinetic perimetry outputs in relation to underlying structural loci. This also formed the basis for our clustering framework, which groups together statistically similar structural and functional test locations to maximise structure-function concordance. Throughout the manuscript, we review the scientific underpinnings of clinical measurements, framing application into real-world patients to improve clinical practice.
Collapse
Affiliation(s)
- Jack Phu
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW, Australia; Centre for Eye Health, University of New South Wales, Kensington, NSW, Australia; Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Concord Clinical School, Concord Repatriation General Hospital, Concord, NSW, Australia; School of Medicine (Optometry), Deakin University, Waurn Ponds, VIC, Australia.
| | - Sieu K Khuu
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW, Australia.
| | - Lisa Nivison-Smith
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW, Australia; Centre for Eye Health, University of New South Wales, Kensington, NSW, Australia.
| | - Michael Kalloniatis
- School of Optometry and Vision Science, University of New South Wales, Kensington, NSW, Australia; School of Medicine (Optometry), Deakin University, Waurn Ponds, VIC, Australia; College of Optometry, University of Houston, Houston, TX, USA.
| |
Collapse
|
5
|
Huang G, Chen L, Lin Y, Tang F, Huang H, Chen Q, Cui L, Xu F, Shen C. Comparative proteomic analysis of retinal hypoxia-ischemia in an acute ocular hypertension model using tandem mass tag-based quantitative proteomics. Exp Eye Res 2024; 247:110063. [PMID: 39216638 DOI: 10.1016/j.exer.2024.110063] [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: 02/02/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
The main symptom of acute glaucoma is acute ocular hypertension (AOH), which leads to the death of retinal ganglion cells (RGCs) and permanent loss of vision. However, effective treatments for these conditions are lacking. This study aimed to identify major regulators and overall protein changes involved in AOH-induced RGC death. Proteomic patterns of the retinal protein extracts from the AOH and sham groups were analyzed using mass spectrometry (MS), followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. Proteomic analysis revealed 92 proteins in the AOH group compared to the control group; 58 proteins were upregulated and 34 were downregulated. Alterations in fatty acid-binding protein 7 (FABP7) and caveolin-1 (Cav-1), which are related to fatty acid metabolism and ocular inflammatory signaling, were detected using western blotting and biochemical assays. Variations in the expression of galectin-1 (Gal-1), S100 calcium-binding protein A6 (S100a6), and visinin-like protein-1 (VILIP) have been associated with neuronal ischemia. Our investigation demonstrates that neuroinflammation and fatty acid metabolism are involved in retinal impairment following AOH, suggesting a possible treatment approach for acute glaucoma.
Collapse
Affiliation(s)
- Guangyi Huang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Lifei Chen
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Yunru Lin
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China; Youjiang Medical University for Nationalities, Baise, 533000, China
| | - Fen Tang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Hui Huang
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Qi Chen
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Ling Cui
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Fan Xu
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China
| | - Chaolan Shen
- Department of Ophthalmology, the People's Hospital of Guangxi Zhuang Autonomous Region & Guangxi Key Laboratory of Eye Health & Guangxi Health Commission Key Laboratory of Ophthalmology and Related Systemic Diseases Artificial Intelligence Screening Technology &Institute of Ophthalmic Diseases, Guangxi Academy of Medical Sciences, Nanning, 530021, China.
| |
Collapse
|
6
|
Garcia-Sanchez J, Lin D, Liu WW. Mechanosensitive ion channels in glaucoma pathophysiology. Vision Res 2024; 223:108473. [PMID: 39180975 PMCID: PMC11398070 DOI: 10.1016/j.visres.2024.108473] [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: 06/17/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
Force sensing is a fundamental ability that allows cells and organisms to interact with their physical environment. The eye is constantly subjected to mechanical forces such as blinking and eye movements. Furthermore, elevated intraocular pressure (IOP) can cause mechanical strain at the optic nerve head, resulting in retinal ganglion cell death (RGC) in glaucoma. How mechanical stimuli are sensed and affect cellular physiology in the eye is unclear. Recent studies have shown that mechanosensitive ion channels are expressed in many ocular tissues relevant to glaucoma and may influence IOP regulation and RGC survival. Furthermore, variants in mechanosensitive ion channel genes may be associated with risk for primary open angle glaucoma. These findings suggest that mechanosensitive channels may be important mechanosensors mediating cellular responses to pressure signals in the eye. In this review, we focus on mechanosensitive ion channels from three major channel families-PIEZO, two-pore potassium and transient receptor potential channels. We review the key properties of these channels, their effects on cell function and physiology, and discuss their possible roles in glaucoma pathophysiology.
Collapse
Affiliation(s)
- Julian Garcia-Sanchez
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Danting Lin
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wendy W Liu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
7
|
Akiba R, Lind Boniec S, Knecht S, Uyama H, Tu HY, Baba T, Takahashi M, Mandai M, Wong RO. Cellular and circuit remodeling of the primate foveal midget pathway after acute photoreceptor loss. Proc Natl Acad Sci U S A 2024; 121:e2413104121. [PMID: 39231211 PMCID: PMC11406236 DOI: 10.1073/pnas.2413104121] [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: 06/30/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024] Open
Abstract
The retinal fovea in human and nonhuman primates is essential for high acuity and color vision. Within the fovea lies specialized circuitry in which signals from a single cone photoreceptor are largely conveyed to one ON and one OFF type midget bipolar cell (MBC), which in turn connect to a single ON or OFF midget ganglion cell (MGC), respectively. Restoring foveal vision requires not only photoreceptor replacement but also appropriate reconnection with surviving ON and OFF MBCs and MGCs. However, our current understanding of the effects of cone loss on the remaining foveal midget pathway is limited. We thus used serial block-face electron microscopy to determine the degree of plasticity and potential remodeling of this pathway in adult Macaca fascicularis several months after acute photoreceptor loss upon photocoagulation. We reconstructed MBC structure and connectivity within and adjacent to the region of cone loss. We found that MBC dendrites within the scotoma retracted and failed to reach surviving cones to form new connections. However, both surviving cones and ON and OFF MBC dendrites at the scotoma border exhibited remodeling, suggesting that these neurons can demonstrate plasticity and rewiring at maturity. At six months postlesion, disconnected OFF MBCs clearly lost output ribbon synapses with their postsynaptic partners, whereas the majority of ON MBCs maintained their axonal ribbon numbers, suggesting differential timing or extent in ON and OFF midget circuit remodeling after cone loss. Our findings raise rewiring considerations for cell replacement approaches in the restoration of foveal vision.
Collapse
Affiliation(s)
- Ryutaro Akiba
- Department of Biological Structure, University of Washington, Seattle, WA 98195
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Department of Ophthalmology and Visual Sciences, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Shane Lind Boniec
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - Sharm Knecht
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| | - Hirofumi Uyama
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
| | - Hung-Ya Tu
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka 565-0871, Japan
| | - Takayuki Baba
- Department of Ophthalmology and Visual Sciences, Chiba University Graduate School of Medicine, Chiba 260-8677, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Research Center, Kobe City Eye Hospital Research Center, Kobe, Hyogo 650-0047, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo 650-0047, Japan
- Research Center, Kobe City Eye Hospital Research Center, Kobe, Hyogo 650-0047, Japan
| | - Rachel O Wong
- Department of Biological Structure, University of Washington, Seattle, WA 98195
| |
Collapse
|
8
|
Sotozono A, Namekata K, Guo X, Shinozaki Y, Harada C, Noro T, Nakano T, Harada T. Membrane-anchored intracellular insulin receptor or insulin-like growth factor-1 receptor elicits ligand-independent downstream signaling. Biochem Biophys Rep 2024; 39:101799. [PMID: 39161576 PMCID: PMC11332076 DOI: 10.1016/j.bbrep.2024.101799] [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: 04/23/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Neurodegenerative diseases including glaucoma affect insulin signaling, and insulin treatment has been shown to reverse the neurodegenerative loss of dendritic complexity in retinal ganglion cells. Therefore, strategies for enhancing or maintaining insulin signaling are worth pursuing to establish new therapies for these diseases. In the present study, we generated constitutively active insulin receptor (F-iIR) and insulin-like growth factor-1 receptor (F-iIGF1R) using a system that forces membrane localization of the intracellular domains of these receptors by farnesylation. Immunohistochemistry and Western blot analysis revealed that F-iIR and F-iIGF1R caused the activation of ERK and AKT in the absence of ligands in vitro. Our results suggest that in vivo effects of F-iIR and F-iIGF1R on the progression of neurodegenerative diseases should be investigated in the future.
Collapse
Affiliation(s)
- Akiko Sotozono
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Youichi Shinozaki
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Takahiko Noro
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Tadashi Nakano
- Department of Ophthalmology, The Jikei University School of Medicine, Tokyo, Japan
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
9
|
Scott MTW, Xu H, Yakovleva A, Tibshirani R, Goldberg JL, Norcia AM. Glaucoma classification through SSVEP derived ON- and OFF-pathway features. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.22.24312443. [PMID: 39228700 PMCID: PMC11370506 DOI: 10.1101/2024.08.22.24312443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Recent evidence from small animal models and human electrophysiology suggests that the OFF-pathway is more vulnerable to glaucomatous insult than the ON-pathway. Thus, OFF-pathway based measurements of visual function may be useful in the diagnosis of Glaucoma. The steady-state visually evoked potential (SSVEP) can be used to non-invasively make such functional measurements. Here, we examine whether OFF- and ON-pathway biasing SSVEP measurements differently predict glaucoma diagnosis using a large cohort of 98 glaucoma patients and 71 controls. Using both a logistic regression with k-fold cross-validation and a random forest classifier, we show that OFF-pathway biasing features produce a small improvement in predictive accuracy over ON-pathway biasing features. However, despite our inclusion of many more response features and the retention of both participants' eyes, our classifier did not perform as well as previous reports that used the isolated-check VEP. This is likely a result of the relatively small amount of data we collected for each participant, but may also be explained by the absence of any train-test splitting in preexisting work. Nevertheless, our results support further exploration of the diagnostic potential of OFF-pathway biasing functional biomarkers for glaucoma.
Collapse
Affiliation(s)
| | - Hui Xu
- Department of Statistics, Stanford University
| | - Alexandra Yakovleva
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University
| | | | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University
| | - Anthony M Norcia
- Wu-Tsai Neurosciences Institute, Department of Psychology, Stanford University
| |
Collapse
|
10
|
El Hajji S, Shiga Y, Belforte N, Solorio YC, Tastet O, D’Onofrio P, Dotigny F, Prat A, Arbour N, Fortune B, Di Polo A. Insulin restores retinal ganglion cell functional connectivity and promotes visual recovery in glaucoma. SCIENCE ADVANCES 2024; 10:eadl5722. [PMID: 39110798 PMCID: PMC11305393 DOI: 10.1126/sciadv.adl5722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
Dendrite pathology and synaptic loss result in neural circuit dysfunction, a common feature of neurodegenerative diseases. There is a lack of strategies that target dendritic and synaptic regeneration to promote neurorecovery. We show that daily human recombinant insulin eye drops stimulate retinal ganglion cell (RGC) dendrite and synapse regeneration during ocular hypertension, a risk factor to develop glaucoma. We demonstrate that the ribosomal protein p70S6 kinase (S6K) is essential for insulin-dependent dendritic regrowth. Furthermore, S6K phosphorylation of the stress-activated protein kinase-interacting protein 1 (SIN1), a link between the mammalian target of rapamycin complexes 1 and 2 (mTORC1/2), is required for insulin-induced dendritic regeneration. Using two-photon microscopy live retinal imaging, we show that insulin rescues single-RGC light-evoked calcium (Ca2+) dynamics. We further demonstrate that insulin enhances neuronal survival and retina-brain connectivity leading to improved optomotor reflex-elicited behaviors. Our data support that insulin is a compelling pro-regenerative strategy with potential clinical implications for the treatment and management of glaucoma.
Collapse
Affiliation(s)
- Sana El Hajji
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Nicolas Belforte
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Yves Carpentier Solorio
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Olivier Tastet
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Philippe D’Onofrio
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Florence Dotigny
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Nathalie Arbour
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO box 6128, Station centre-ville, Montreal, Quebec, Canada
- Neuroscience Division, Centre de recherche du Centre Hospitalier de l’Université de Montréal (CR-CHUM), 900 Saint Denis Street, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Yu A, Tan LX, Lakkaraju A, Santina LD, Ou Y. Microglia target synaptic sites early during excitatory circuit disassembly in neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.13.598914. [PMID: 38915631 PMCID: PMC11195198 DOI: 10.1101/2024.06.13.598914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
During development, microglia prune excess synapses to refine neuronal circuits. In neurodegeneration, the role of microglia-mediated synaptic pruning in circuit remodeling and dysfunction is important for developing therapies aimed at modulating microglial function. Here we analyzed the role of microglia in the synapse disassembly of degenerating postsynaptic neurons in the inner retina. After inducing transient intraocular pressure elevation to injure retinal ganglion cells, microglia increase in number, shift to ameboid morphology, and exhibit greater process movement. Furthermore, due to the greater number of microglia, there is increased colocalization of microglia with synaptic components throughout the inner plexiform layer and with excitatory synaptic sites along individual ganglion cell dendrites. Microglia depletion partially restores ganglion cell function, suggesting that microglia activation may be neurotoxic in early neurodegeneration. Our results demonstrate the important role of microglia in synapse disassembly in degenerating circuits, highlighting their recruitment to synaptic sites early after neuronal injury.
Collapse
Affiliation(s)
- Alfred Yu
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Li Xuan Tan
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Aparna Lakkaraju
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| | - Luca Della Santina
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
- College of Optometry, University of Houston, Houston, TX, USA
| | - Yvonne Ou
- Department of Ophthalmology, UCSF School of Medicine, San Francisco, CA, USA
| |
Collapse
|
12
|
Du R, Wang P, Tian N. CD3ζ-Mediated Signaling Protects Retinal Ganglion Cells in Glutamate Excitotoxicity of the Retina. Cells 2024; 13:1006. [PMID: 38920637 PMCID: PMC11201742 DOI: 10.3390/cells13121006] [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: 03/30/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Excessive levels of glutamate activity could potentially damage and kill neurons. Glutamate excitotoxicity is thought to play a critical role in many CNS and retinal diseases. Accordingly, glutamate excitotoxicity has been used as a model to study neuronal diseases. Immune proteins, such as major histocompatibility complex (MHC) class I molecules and their receptors, play important roles in many neuronal diseases, while T-cell receptors (TCR) are the primary receptors of MHCI. We previously showed that a critical component of TCR, CD3ζ, is expressed by mouse retinal ganglion cells (RGCs). The mutation of CD3ζ or MHCI molecules compromises the development of RGC structure and function. In this study, we investigated whether CD3ζ-mediated molecular signaling regulates RGC death in glutamate excitotoxicity. We show that mutation of CD3ζ significantly increased RGC survival in NMDA-induced excitotoxicity. In addition, we found that several downstream molecules of TCR, including Src (proto-oncogene tyrosine-protein kinase) family kinases (SFKs) and spleen tyrosine kinase (Syk), are expressed by RGCs. Selective inhibition of an SFK member, Hck, or Syk members, Syk or Zap70, significantly increased RGC survival in NMDA-induced excitotoxicity. These results provide direct evidence to reveal the underlying molecular mechanisms that control RGC death under disease conditions.
Collapse
Affiliation(s)
- Rui Du
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
| | - Ping Wang
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
| | - Ning Tian
- Department of Ophthalmology and Visual Science, University of Utah School of Medicine, Salt Lake City, UT 84132, USA; (R.D.); (P.W.)
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84132, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84132, USA
- Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA
| |
Collapse
|
13
|
Tsai YC, Lee HP, Tsung TH, Chen YH, Lu DW. Unveiling Novel Structural Biomarkers for the Diagnosis of Glaucoma. Biomedicines 2024; 12:1211. [PMID: 38927418 PMCID: PMC11200849 DOI: 10.3390/biomedicines12061211] [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: 04/29/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Glaucoma, a leading cause of irreversible blindness, poses a significant global health burden. Early detection is crucial for effective management and prevention of vision loss. This study presents a collection of novel structural biomarkers in glaucoma diagnosis. By employing advanced imaging techniques and data analysis algorithms, we now can recognize indicators of glaucomatous progression. Many research studies have revealed a correlation between the structural changes in the eye or brain, particularly in the optic nerve head and retinal nerve fiber layer, and the progression of glaucoma. These biomarkers demonstrate value in distinguishing glaucomatous eyes from healthy ones, even in the early stages of the disease. By facilitating timely detection and monitoring, they hold the potential to mitigate vision impairment and improve patient outcomes. This study marks an advancement in the field of glaucoma, offering a promising avenue for enhancing the diagnosis and possible management.
Collapse
Affiliation(s)
- Yu-Chien Tsai
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
- Department of Ophthalmology, Taoyuan Armed Forces General Hospital, Taoyuan 325, Taiwan
| | - Hsin-Pei Lee
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Ta-Hsin Tsung
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Yi-Hao Chen
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Da-Wen Lu
- Department of Ophthalmology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| |
Collapse
|
14
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
15
|
Cilingir V, Seven E. Retinal clues for selective neuronal loss in multiple sclerosis. Neurol Sci 2024; 45:1163-1171. [PMID: 37837508 DOI: 10.1007/s10072-023-07110-2] [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: 05/03/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023]
Abstract
OBJECTIVE The relationship between the cell body layer and the dendritic network layer of the retina and cognitive performance (CP) in MS patients has not been examined separately. The objective of this study is to predict cognitive impairment (CI) in RRMS patients and to examine the relationship between CP and ganglion cell layer (GCL), inner plexiform layer (IPL), and GCL divided by IPL (GCL/IPL). METHODS Ophthalmological evaluation, retinal segmentation, and Symbol Digit Modalities Test (SDMT) were performed on 102 RRMS patients and 54 healthy subjects. The relationships of GCL, IPL, and GCL/IPL with CP in eyes without a history of optic neuritis were investigated using Spearman's correlation. Models were created by accepting 1 standard deviation less of the SDMT mean of the control group as the limit for CI. The cutoff value of the GCL/IPL variable that could predict CI was calculated by ROC analysis, and the ability to accurately predict CI was tested with binary logistic regression. RESULTS No correlation was found between OCT parameters and CP in healthy subjects. Correlation was found between GCL thickness and GCL/IPL variable and CP in RRMS patients (r=0.235, r=0.667 respectively). A GCL/IPL value of 1.255 was able to identify CI with 81.8% sensitivity and 75.9% specificity (AUC=0.844, LR=3.38) and predicted CI with 74.5% accuracy (Nagelkerke R2=0.439). CONCLUSION In RRMS patients, the IPL thickness is unrelated to CP. Therewithal, the GCL/IPL-CP relationship is stronger than the GCL-CP relationship and GCL/IPL can predict CI.
Collapse
Affiliation(s)
- Vedat Cilingir
- Department of Neurology, Van Yuzuncu Yil University, Bardakcı, Tusba, 65300, Van, Turkey.
- Department of Neuroscience Research, Van Yuzuncu Yil University, Tusba, Van, Turkey.
| | - Erbil Seven
- Department of Ophthalmology, Van Yuzuncu Yil University, Tusba, Van, Turkey
| |
Collapse
|
16
|
van Koeverden AK, Afiat BC, Nguyen CT, Bui BV, Lee PY. Understanding how ageing impacts ganglion cell susceptibility to injury in glaucoma. Clin Exp Optom 2024; 107:147-155. [PMID: 37980904 DOI: 10.1080/08164622.2023.2279734] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023] Open
Abstract
Glaucoma is a leading cause of blindness worldwide, with a marked increase in prevalence with advancing age. Due to the multifactorial nature of glaucoma pathogenesis, dissecting how ageing impacts upon glaucoma risk requires analysis and synthesis of evidence from a vast literature. While there is a wealth of human clinical studies examining glaucoma pathogenesis and why older patients have increased risk, many aspects of the disease such as adaptations of retinal ganglion cells to stress, autophagy and the role of glial cells in glaucoma, require the use of animal models to study the complex cellular processes and interactions. Additionally, the accelerated nature of ageing in rodents facilitates the longitudinal study of changes that would not be feasible in human clinical studies. This review article examines evidence derived predominantly from rodent models on how the ageing process impacts upon various aspects of glaucoma pathology from the retinal ganglion cells themselves, to supporting cells and tissues such as glial cells, connective tissue and vasculature, in addition to oxidative stress and autophagy. An improved understanding of how ageing modifies these factors may lead to the development of different therapeutic strategies that target specific risk factors or processes involved in glaucoma.
Collapse
Affiliation(s)
- Anna K van Koeverden
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Brianna C Afiat
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Christine To Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Pei Ying Lee
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Zueva MV, Neroeva NV, Zhuravleva AN, Bogolepova AN, Kotelin VV, Fadeev DV, Tsapenko IV. Fractal Phototherapy in Maximizing Retina and Brain Plasticity. ADVANCES IN NEUROBIOLOGY 2024; 36:585-637. [PMID: 38468055 DOI: 10.1007/978-3-031-47606-8_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The neuroplasticity potential is reduced with aging and impairs during neurodegenerative diseases and brain and visual system injuries. This limits the brain's capacity to repair the structure and dynamics of its activity after lesions. Maximization of neuroplasticity is necessary to provide the maximal CNS response to therapeutic intervention and adaptive reorganization of neuronal networks in patients with degenerative pathology and traumatic injury to restore the functional activity of the brain and retina.Considering the fractal geometry and dynamics of the healthy brain and the loss of fractality in neurodegenerative pathology, we suggest that the application of self-similar visual signals with a fractal temporal structure in the stimulation therapy can reactivate the adaptive neuroplasticity and enhance the effectiveness of neurorehabilitation. This proposition was tested in the recent studies. Patients with glaucoma had a statistically significant positive effect of fractal photic therapy on light sensitivity and the perimetric MD index, which shows that methods of fractal stimulation can be a novel nonpharmacological approach to neuroprotective therapy and neurorehabilitation. In healthy rabbits, it was demonstrated that a long-term course of photostimulation with fractal signals does not harm the electroretinogram (ERG) and retina structure. Rabbits with modeled retinal atrophy showed better dynamics of the ERG restoration during daily stimulation therapy for a week in comparison with the controls. Positive changes in the retinal function can indirectly suggest the activation of its adaptive plasticity and the high potential of stimulation therapy with fractal visual stimuli in a nonpharmacological neurorehabilitation, which requires further study.
Collapse
Affiliation(s)
- Marina V Zueva
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Natalia V Neroeva
- Department of Pathology of the Retina and Optic Nerve, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Anastasia N Zhuravleva
- Department of Glaucoma, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Anna N Bogolepova
- Department of neurology, neurosurgery and medical genetics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladislav V Kotelin
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Denis V Fadeev
- Scientific Experimental Center Department, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Irina V Tsapenko
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
18
|
McCracken S, Fitzpatrick MJ, Hall AL, Wang Z, Kerschensteiner D, Morgan JL, Williams PR. Diversity in homeostatic calcium set points predicts retinal ganglion cell survival following optic nerve injury in vivo. Cell Rep 2023; 42:113165. [PMID: 37751356 PMCID: PMC10947246 DOI: 10.1016/j.celrep.2023.113165] [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: 10/17/2022] [Revised: 06/29/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Retinal ganglion cell (RGC) degeneration drives vision loss in blinding conditions. RGC death is often triggered by axon degeneration in the optic nerve. Here, we study the contributions of dynamic and homeostatic Ca2+ levels to RGC death from axon injury. We find that axonal Ca2+ elevations from optic nerve injury do not propagate over distance or reach RGC somas, and acute and chronic Ca2+ dynamics do not affect RGC survival. Instead, we discover that baseline Ca2+ levels vary widely between RGCs and predict their survival after axon injury, and that lowering these levels reduces RGC survival. Further, we find that well-surviving RGC types have higher baseline Ca2+ levels than poorly surviving types. Finally, we observe considerable variation in the baseline Ca2+ levels of different RGCs of the same type, which are predictive of within-type differences in survival.
Collapse
Affiliation(s)
- Sean McCracken
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael J Fitzpatrick
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Allison L Hall
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Postbaccalaureate Program in Developmental Biology & Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zelun Wang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Graduate Program in Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Daniel Kerschensteiner
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Josh L Morgan
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Philip R Williams
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
19
|
Prigge CL, Dembla M, Sharma A, El-Quessny M, Kozlowski C, Paisley CE, Miltner AM, Johnson TM, Della Santina L, Feller MB, Kay JN. Rejection of inappropriate synaptic partners in mouse retina mediated by transcellular FLRT2-UNC5 signaling. Dev Cell 2023; 58:2080-2096.e7. [PMID: 37557174 PMCID: PMC10615732 DOI: 10.1016/j.devcel.2023.07.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 05/26/2023] [Accepted: 07/18/2023] [Indexed: 08/11/2023]
Abstract
During nervous system development, neurons choose synaptic partners with remarkable specificity; however, the cell-cell recognition mechanisms governing rejection of inappropriate partners remain enigmatic. Here, we show that mouse retinal neurons avoid inappropriate partners by using the FLRT2-uncoordinated-5 (UNC5) receptor-ligand system. Within the inner plexiform layer (IPL), FLRT2 is expressed by direction-selective (DS) circuit neurons, whereas UNC5C/D are expressed by non-DS neurons projecting to adjacent IPL sublayers. In vivo gain- and loss-of-function experiments demonstrate that FLRT2-UNC5 binding eliminates growing DS dendrites that have strayed from the DS circuit IPL sublayers. Abrogation of FLRT2-UNC5 binding allows mistargeted arbors to persist, elaborate, and acquire synapses from inappropriate partners. Conversely, UNC5C misexpression within DS circuit sublayers inhibits dendrite growth and drives arbors into adjacent sublayers. Mechanistically, UNC5s promote dendrite elimination by interfering with FLRT2-mediated adhesion. Based on their broad expression, FLRT-UNC5 recognition is poised to exert widespread effects upon synaptic partner choices across the nervous system.
Collapse
Affiliation(s)
- Cameron L Prigge
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Mayur Dembla
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Arsha Sharma
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Malak El-Quessny
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christopher Kozlowski
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Caitlin E Paisley
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Adam M Miltner
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Tyler M Johnson
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA
| | - Luca Della Santina
- Department of Vision Sciences, University of Houston College of Optometry, Houston, TX 77204, USA
| | - Marla B Feller
- Helen Wills Neuroscience Institute and Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeremy N Kay
- Departments of Neurobiology, Ophthalmology, and Cell Biology, Duke University School of Medicine, Box 3802, Durham, NC 27710, USA.
| |
Collapse
|
20
|
Martucci A, Di Giuliano F, Minosse S, Pocobelli G, Nucci C, Garaci F. MRI and Clinical Biomarkers Overlap between Glaucoma and Alzheimer's Disease. Int J Mol Sci 2023; 24:14932. [PMID: 37834380 PMCID: PMC10573932 DOI: 10.3390/ijms241914932] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Glaucoma is the leading cause of blindness worldwide. It is classically associated with structural and functional changes in the optic nerve head and retinal nerve fiber layer, but the damage is not limited to the eye. The involvement of the central visual pathways and disruption of brain network organization have been reported using advanced neuroimaging techniques. The brain structural changes at the level of the areas implied in processing visual information could justify the discrepancy between signs and symptoms and underlie the analogy of this disease with neurodegenerative dementias, such as Alzheimer's disease, and with the complex group of pathologies commonly referred to as "disconnection syndromes." This review aims to summarize the current state of the art on the use of advanced neuroimaging techniques in glaucoma and Alzheimer's disease, highlighting the emerging biomarkers shared by both diseases.
Collapse
Affiliation(s)
- Alessio Martucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Francesca Di Giuliano
- Neuroradiology Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy;
| | - Silvia Minosse
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (S.M.); (F.G.)
| | - Giulio Pocobelli
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Carlo Nucci
- Ophthalmology Unit, Department of Experimental Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (A.M.); (G.P.)
| | - Francesco Garaci
- Diagnostic Imaging Unit, Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (S.M.); (F.G.)
- San Raffaele Cassino, 03043 Frosinone, Italy
| |
Collapse
|
21
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
22
|
Yao K, Mou Q, Lou X, Ye M, Zhao B, Hu Y, Luo J, Zhang H, Li X, Zhao Y. Microglial SIRT1 activation attenuates synapse loss in retinal inner plexiform layer via mTORC1 inhibition. J Neuroinflammation 2023; 20:202. [PMID: 37670386 PMCID: PMC10481494 DOI: 10.1186/s12974-023-02886-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/30/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Optic nerve injury (ONI) is a key cause of irreversible blindness and triggers retinal ganglion cells (RGCs) change and synapse loss. Microglia is the resistant immune cell in brain and retina and has been demonstrated to be highly related with neuron and synapse injury. However, the function of Sirtuin 1 (SIRT1), a neuroprotective molecule, in mediating microglial activation, retinal synapse loss and subsequent retinal ganglion cells death in optic nerve injury model as well as the regulatory mechanism remain unclear. METHOD To this end, optic nerve crush (ONC) model was conducted to mimic optic nerve injury. Resveratrol and EX527, highly specific activator and inhibitor of SIRT1, respectively, were used to explore the function of SIRT1 in vivo and vitro. Cx3Cr1-CreERT2/RaptorF/F mice were used to delete Raptor for inhibiting mammalian target of rapamycin complex 1 (mTORC1) activity in microglia. HEK293 and BV2 cells were transfected with plasmids to explore the regulatory mechanism of SIRT1. RESULTS We discovered that microglial activation and synapse loss in retinal inner plexiform layer (IPL) occurred after optic nerve crush, with later-development retinal ganglion cells death. SIRT1 activation induced by resveratrol inhibited microglial activation and attenuated synapse loss and retinal ganglion cells injury. After injury, microglial phagocytosed synapse and SIRT1 inhibited this process to protect synapse and retinal ganglion cells. Moreover, SIRT1 exhibited neuron protective effects via activating tuberous sclerosis complex 2 (TSC2) through deacetylation, and enhancing the inhibition effect of tuberous sclerosis complex 2 on mammalian target of rapamycin complex 1 activity. CONCLUSION Our research provides novel insights into microglial SIRT1 in optic nerve injury and suggests a potential strategy for neuroprotective treatment of optic nerve injury disease.
Collapse
Affiliation(s)
- Ke Yao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qianxue Mou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaotong Lou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meng Ye
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bowen Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuanyuan Hu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
23
|
Soliño M, Yu A, Della Santina L, Ou Y. Large-scale survey of excitatory synapses reveals sublamina-specific and asymmetric synapse disassembly in a neurodegenerative circuit. iScience 2023; 26:107262. [PMID: 37609630 PMCID: PMC10440711 DOI: 10.1016/j.isci.2023.107262] [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: 03/10/2023] [Revised: 05/09/2023] [Accepted: 06/27/2023] [Indexed: 08/24/2023] Open
Abstract
In the nervous system, parallel circuits are organized in part by the lamina-specific compartmentalization of synaptic connections. In sensory systems such as mammalian retina, degenerating third-order neurons remodel their local presynaptic connectivity with second-order neurons. To determine whether there are sublamina-specific perturbations after injury of adult retinal ganglion cells, we comprehensively analyzed excitatory synapses across the inner plexiform layer (IPL) where bipolar cells connect to ganglion cells. Here, we show that pre- and postsynaptic component loss occurs throughout the IPL in a sublamina-dependent fashion after transient intraocular pressure elevation. Partnered synaptic components are lost as neurodegeneration progresses, while unpartnered synaptic components remain stable. Furthermore, presynaptic components are either lost first or simultaneously with the postsynaptic component. Our results demonstrate that this degenerating neural circuit exhibits differential vulnerability of excitatory synapses depending on IPL depth, highlighting the ordered disassembly of synapses that is specific to laminar compartments of the retina.
Collapse
Affiliation(s)
- Manuel Soliño
- Department of Ophthalmology, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Alfred Yu
- Department of Ophthalmology, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| | - Luca Della Santina
- Department of Ophthalmology, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
- College of Optometry, University of Houston, Houston, TX 77204, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California San Francisco School of Medicine, San Francisco, CA 94143, USA
| |
Collapse
|
24
|
Tribble JR, Hui F, Quintero H, El Hajji S, Bell K, Di Polo A, Williams PA. Neuroprotection in glaucoma: Mechanisms beyond intraocular pressure lowering. Mol Aspects Med 2023; 92:101193. [PMID: 37331129 DOI: 10.1016/j.mam.2023.101193] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flora Hui
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Department of Optometry & Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Katharina Bell
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Eye ACP Duke-NUS, Singapore
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
25
|
Van Dyck A, Masin L, Bergmans S, Schevenels G, Beckers A, Vanhollebeke B, Moons L. A new microfluidic model to study dendritic remodeling and mitochondrial dynamics during axonal regeneration of adult zebrafish retinal neurons. Front Mol Neurosci 2023; 16:1196504. [PMID: 37396787 PMCID: PMC10307971 DOI: 10.3389/fnmol.2023.1196504] [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: 03/29/2023] [Accepted: 05/10/2023] [Indexed: 07/04/2023] Open
Abstract
Unlike mammals, adult zebrafish are able to fully regenerate axons and functionally recover from neuronal damage in the mature central nervous system (CNS). Decades of research have tried to identify the mechanisms behind their spontaneous regenerative capacity, but the exact underlying pathways and molecular drivers remain to be fully elucidated. By studying optic nerve injury-induced axonal regrowth of adult zebrafish retinal ganglion cells (RGCs), we previously reported transient dendritic shrinkage and changes in the distribution and morphology of mitochondria in the different neuronal compartments throughout the regenerative process. These data suggest that dendrite remodeling and temporary changes in mitochondrial dynamics contribute to effective axonal and dendritic repair upon optic nerve injury. To further elucidate these interactions, we here present a novel adult zebrafish microfluidic model in which we can demonstrate compartment-specific alterations in resource allocation in real-time at single neuron level. First, we developed a pioneering method that enables to isolate and culture adult zebrafish retinal neurons in a microfluidic setup. Notably, with this protocol, we report on a long-term adult primary neuronal culture with a high number of surviving and spontaneously outgrowing mature neurons, which was thus far only very limitedly described in literature. By performing time-lapse live cell imaging and kymographic analyses in this setup, we can explore changes in dendritic remodeling and mitochondrial motility during spontaneous axonal regeneration. This innovative model system will enable to discover how redirecting intraneuronal energy resources supports successful regeneration in the adult zebrafish CNS, and might facilitate the discovery of new therapeutic targets to promote neuronal repair in humans.
Collapse
Affiliation(s)
- Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Lagali PS, Shanmugalingam U, Baker AN, Mezey N, Smith PD, Coupland SG, Tsilfidis C. Assessment of the uniform field electroretinogram for mouse retinal ganglion cell functional analysis. Doc Ophthalmol 2023:10.1007/s10633-023-09933-y. [PMID: 37106219 DOI: 10.1007/s10633-023-09933-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 03/26/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE The uniform field electroretinogram (UF-ERG) has been suggested as an alternative to the pattern electroretinogram (PERG) for non-invasive assessment of retinal ganglion cell (RGC) function in primates. We evaluated the validity of the UF-ERG to assess mouse RGC activity in vivo. METHODS Unilateral optic nerve crush (ONC) was performed on adult C57BL/6J mice. Contralateral eyes with uncrushed optic nerves and eyes from surgically naive mice served as experimental controls. Electrophysiological visual assessment was performed at 12 weeks post-ONC. Flash-mediated visual-evoked cortical potentials (VEPs) were measured to confirm the robustness of the ONC procedure. Full-field flash ERGs were used to interrogate photoreceptor and retinal bipolar cell function. RGC function was assessed with pattern ERGs. Summed onset and offset UF-ERG responses to alternating dark and light uniform field flash stimuli of different intensities and wavelengths were recorded from ONC and control eyes, and relative differences were compared to the PERG results. Following electrophysiological analysis, RGC loss was monitored by immunohistochemical staining of the RGC marker protein, RBPMS, in post-mortem retinal tissues. RESULTS ONC dramatically impacts RGC integrity and optic nerve function, demonstrated by reduced RGC counts and near complete elimination of VEPs. ONC did not affect scotopic ERG a-wave and b-wave amplitudes, while PERG amplitudes of eyes subjected to ONC were reduced by approximately 50% compared to controls. Summation of ON and OFF UF-ERG responses did not reveal statistically significant differences between ONC and control eyes, regardless of visual stimulus. CONCLUSIONS PERG responses are markedly impaired upon ONC, while UF-ERG responses are not significantly affected by surgical trauma to RGC axons in mice. The more closely related pattern and uniform field ERGs recorded in primates suggests species-specific differences in RGC features or subpopulations corresponding to PERG and UF-ERG response generators, limiting the utility of the UF-ERG for mouse RGC functional analysis.
Collapse
Affiliation(s)
- Pamela S Lagali
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | | | - Adam N Baker
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
| | - Natalie Mezey
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Patrice D Smith
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Stuart G Coupland
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada
- Department of Ophthalmology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Catherine Tsilfidis
- Neuroscience Program, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- University of Ottawa Eye Institute, The Ottawa Hospital, Ottawa, ON, K1H 8L6, Canada.
- Department of Ophthalmology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
27
|
Pang JJ, Gao F, Wu SM. Light responses and amacrine cell modulation of morphologically-identified retinal ganglion cells in the mouse retina. Vision Res 2023; 205:108187. [PMID: 36758452 PMCID: PMC11349081 DOI: 10.1016/j.visres.2023.108187] [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: 11/21/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/10/2023]
Abstract
By analyzing light-evoked spike responses, cation currents (ΔIC) and chloride currents (ΔICl) of over 100 morphologically-identified retinal ganglion cells (GCs) in dark-adapted mouse retina, we found there are at least 14 functionally- and morphologically-distinct types of RGCs. These cells can be divided into 5 groups based on their patterns of spike response to whole field light steps (SRWFLS), a GC identification scheme commonly used in studies with extracellular recording techniques. We also found that all GCs in the mouse retina express strychnine-sensitive glycine receptors, and receive light-elicited chloride current (ΔICl) accompanied by a conductance increase from narrow-field, glycinergic amacrine cells. As the dark membrane potential of RGC are near the chloride-equilibrium potential, mouse GCs' spike responses are mediated primarily by bipolar cells inputs, and modulated by "shunting inhibition" from narrow-field amacrine cells. Analysis of strychnine actions on light-evoked cation current ΔIC (bipolar cell inputs) in GCs suggests that narrow-field amacrine cells modulate GCs by sending ON-OFF crossover feedback signals to presynaptic bipolar cell axon terminals via sign-inverting glycinergic synapses, and the feedback signals are synergistic to the bipolar cell light responses. Therefore narrow-field amacrine cells enhance light-evoked bipolar cell inputs to GCs by presynaptic "synergistic addition", besides the abovementioned postsynaptic "shunting inhibition" in GCs.
Collapse
Affiliation(s)
- Ji-Jie Pang
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - Fan Gao
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States
| | - Samuel M Wu
- Cullen Eye Institute, Baylor College of Medicine, Houston, TX 77030, United States.
| |
Collapse
|
28
|
Smith BJ, McHugh CF, Hirano AA, Brecha NC, Barnes S. Transient and Sustained Ganglion Cell Light Responses Are Differentially Modulated by Intrinsically Produced Reactive Oxygen Species Acting upon Specific Voltage-Gated Na + Channel Isoforms. J Neurosci 2023; 43:2291-2304. [PMID: 36828637 PMCID: PMC10072295 DOI: 10.1523/jneurosci.1723-22.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/26/2023] Open
Abstract
Increasing spike rates drive greater neuronal energy demand. In turn, mitochondrial ATP production leads to the generation of reactive oxygen species (ROS) that can modulate ion channel gating. Does ROS production autoregulate the excitability of a neuron? We investigated the links between retinal ganglion cell (RGC) excitability and spike activity-driven ROS production in male and female mice. Changes to the light-evoked and current-evoked spike patterns of functionally identified αRGC subtypes, along with their NaV channel-gating properties, were recorded during experimentally induced decreases and increases of intracellular ROS. During periods of highest spike rates (e.g., following light onset in ON sustained RGCs and light offset in OFF sustained RGCs), these αRGC subtypes responded to reductions of ROS (induced by catalase or glutathione monoethyl ester) with higher spike rates. Increases in ROS (induced by mercaptosuccinate, antimycin-A, or H2O2) lowered spike rates. In ON and OFF transient RGCs, there were no changes in spike rate during ROS decreases but increased ROS increased spiking. This suggests that endogenous ROS are intrinsic neuromodulators in RGCs having high metabolic demands but not in RGCs with lower energy needs. We identified ROS-induced shifts in the voltage-dependent gating of specific isoforms of NaV channels that account for the modulation of ON and OFF sustained RGC spike frequency by ROS-mediated feedback. ROS-induced changes to NaV channel gating, affecting activation and inactivation kinetics, are consistent with the differing spike pattern alterations observed in RGC subtypes. Cell-autonomous generation of ROS during spiking contributes to tuning the spike patterns of RGCs.SIGNIFICANCE STATEMENT Energy production within retinal ganglion cells (RGCs) is accompanied by metabolic by-products harmful to cellular function. How these by-products modulate the excitability of RGCs bears heavily on visual function and the etiology of optic neuropathies. A novel hypothesis of how RGC metabolism can produce automodulation of electrical signaling was tested by identifying the characteristics and biophysical origins of changes to the excitability of RGCs caused by oxidizing by-products in the retina. This impacts our understanding of the pathophysiology of RGC dysfunction, supporting an emerging model in which increases in oxidizing chemical species during energy production, but not necessarily bioenergetic failure, lead to preferential degeneration of specific subtypes of RGCs, yielding loss of different aspects of visual capacity.
Collapse
Affiliation(s)
- Benjamin J Smith
- Doheny Eye Institute, University of California, Los Angeles, California 91103
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Cyrus F McHugh
- Doheny Eye Institute, University of California, Los Angeles, California 91103
| | - Arlene A Hirano
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Nicholas C Brecha
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California 90073
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Stein Eye Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| | - Steven Barnes
- Doheny Eye Institute, University of California, Los Angeles, California 91103
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
- Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California 90095
| |
Collapse
|
29
|
Boal AM, McGrady NR, Holden JM, Risner ML, Calkins DJ. Retinal ganglion cells adapt to ionic stress in experimental glaucoma. Front Neurosci 2023; 17:1142668. [PMID: 37051140 PMCID: PMC10083336 DOI: 10.3389/fnins.2023.1142668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/10/2023] [Indexed: 03/28/2023] Open
Abstract
IntroductionIdentification of early adaptive and maladaptive neuronal stress responses is an important step in developing targeted neuroprotective therapies for degenerative disease. In glaucoma, retinal ganglion cells (RGCs) and their axons undergo progressive degeneration resulting from stress driven by sensitivity to intraocular pressure (IOP). Despite therapies that can effectively manage IOP many patients progress to vision loss, necessitating development of neuronal-based therapies. Evidence from experimental models of glaucoma indicates that early in the disease RGCs experience altered excitability and are challenged with dysregulated potassium (K+) homeostasis. Previously we demonstrated that certain RGC types have distinct excitability profiles and thresholds for depolarization block, which are associated with sensitivity to extracellular K+.MethodsHere, we used our inducible mouse model of glaucoma to investigate how RGC sensitivity to K+ changes with exposure to elevated IOP.ResultsIn controls, conditions of increased K+ enhanced membrane depolarization, reduced action potential generation, and widened action potentials. Consistent with our previous work, 4 weeks of IOP elevation diminished RGC light-and current-evoked responses. Compared to controls, we found that IOP elevation reduced the effects of increased K+ on depolarization block threshold, with IOP-exposed cells maintaining greater excitability. Finally, IOP elevation did not alter axon initial segment dimensions, suggesting that structural plasticity alone cannot explain decreased K+ sensitivity.DiscussionThus, in response to prolonged IOP elevation RGCs undergo an adaptive process that reduces sensitivity to changes in K+ while diminishing excitability. These experiments give insight into the RGC response to IOP stress and lay the groundwork for mechanistic investigation into targets for neuroprotective therapy.
Collapse
|
30
|
Harris SC, Dunn FA. Asymmetric retinal direction tuning predicts optokinetic eye movements across stimulus conditions. eLife 2023; 12:e81780. [PMID: 36930180 PMCID: PMC10023158 DOI: 10.7554/elife.81780] [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: 07/11/2022] [Accepted: 02/02/2023] [Indexed: 03/18/2023] Open
Abstract
Across species, the optokinetic reflex (OKR) stabilizes vision during self-motion. OKR occurs when ON direction-selective retinal ganglion cells (oDSGCs) detect slow, global image motion on the retina. How oDSGC activity is integrated centrally to generate behavior remains unknown. Here, we discover mechanisms that contribute to motion encoding in vertically tuned oDSGCs and leverage these findings to empirically define signal transformation between retinal output and vertical OKR behavior. We demonstrate that motion encoding in vertically tuned oDSGCs is contrast-sensitive and asymmetric for oDSGC types that prefer opposite directions. These phenomena arise from the interplay between spike threshold nonlinearities and differences in synaptic input weights, including shifts in the balance of excitation and inhibition. In behaving mice, these neurophysiological observations, along with a central subtraction of oDSGC outputs, accurately predict the trajectories of vertical OKR across stimulus conditions. Thus, asymmetric tuning across competing sensory channels can critically shape behavior.
Collapse
Affiliation(s)
- Scott C Harris
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Neuroscience Graduate Program, University of California, San FranciscoSan FranciscoUnited States
| | - Felice A Dunn
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
31
|
Dan QQ, Chen L, Shi LL, Zhou X, Wang TH, Liu H. Urine-derived mesenchymal stem cells-derived exosomes enhances survival and proliferation of aging retinal ganglion cells. BMC Mol Cell Biol 2023; 24:8. [PMID: 36879194 PMCID: PMC9990288 DOI: 10.1186/s12860-023-00467-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 02/06/2023] [Indexed: 03/08/2023] Open
Abstract
OBJECTIVES This study was designed to investigate to test the effect of exosomes from urine-derived mesenchymal stem cells (USCs) on the survival and viability of aging retinal ganglion cells (RGCs), and explored the preliminary related mechanisms. METHODS Primary USCs were cultured and identified by immunofluorescence staining. Aging RGCs models were established by D-galactose treatment and identified by β-Galactosidase staining. After treatment with USCs conditioned medium (with USCs removal), flow cytometry was performed to examine the apoptosis and cell cycle of RGCs. Cell viability of RGCs was detected by Cell-counting Kit 8 (CCK8) assay. Moreover, gene sequencing and bioinformatics analysis were applied to analyze the genetic variation after medium treatment in RGCs along with the biological functions of differentially expressed genes (DEGs). RESULTS The number of apoptotic aging RGCs was significantly reduced in USCs medium-treated RGCs. Besides, USCs-derived exosomes exert significant promotion on the cell viability and proliferation of aging RGCs. Further, sequencing data analyzed and identified DEGs expressed in aging RGCs and aging RGCs treated with USCs conditioned medium. The sequencing outcomes demonstrated 117 upregulated genes and 186 downregulated genes in normal RGCs group vs aging RGCs group, 137 upregulated ones and 517 downregulated ones in aging RGCs group vs aging RGCs + USCs medium group. These DEGs involves in numerous positive molecular activities to promote the recovery of RGCs function. CONCLUSIONS Collectively, the therapeutic potentials of USCs-derived exosomes include suppression on cell apoptosis, enhancement on cell viability and proliferation of aging RGCs. The underlying mechanism involves multiple genetic variation and changes of transduction signaling pathways.
Collapse
Affiliation(s)
- Qi-Qin Dan
- Institute of Neurological Disease, West China Hospital, Sichuan University, No. 88 Keyuan South Road, Chengdu, 610041, China
| | - Li Chen
- Institute of Neurological Disease, West China Hospital, Sichuan University, No. 88 Keyuan South Road, Chengdu, 610041, China
| | - Lan-Lan Shi
- Department of Human Anatomy and Tissue Embryology, Kunming Medical University, Kunming, 650500, China
| | - Xiu Zhou
- Laboratory Animal Department, Institute of Neuroscience, Kunming Medical University, Kunming, 650500, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, West China Hospital, Sichuan University, No. 88 Keyuan South Road, Chengdu, 610041, China.
| | - Hua Liu
- Department of Anatomy, Institute of Eyes, Jinzhou Medical University, No.40, Section 3, Songpo Road, Linghe District, JinzhouLiaoning, 121001, China.
| |
Collapse
|
32
|
Naguib S, DeJulius CR, Backstrom JR, Haider AA, Ang JM, Boal AM, Calkins DJ, Duvall CL, Rex TS. Intraocular Sustained Release of EPO-R76E Mitigates Glaucoma Pathogenesis by Activating the NRF2/ARE Pathway. Antioxidants (Basel) 2023; 12:556. [PMID: 36978804 PMCID: PMC10045745 DOI: 10.3390/antiox12030556] [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: 01/12/2023] [Revised: 02/03/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
Erythropoietin (EPO) is neuroprotective in multiple models of neurodegenerative diseases, including glaucoma. EPO-R76E retains the neuroprotective effects of EPO but diminishes the effects on hematocrit. Treatment with EPO-R76E in a glaucoma model increases expression of antioxidant proteins and is neuroprotective. A major pathway that controls the expression of antioxidant proteins is the NRF2/ARE pathway. This pathway is activated endogenously after elevation of intraocular pressure (IOP) and contributes to the slow onset of pathology in glaucoma. In this study, we explored if sustained release of EPO-R76E in the eye would activate the NRF2/ARE pathway and if this pathway was key to its neuroprotective activity. Treatment with PLGA.EPO-E76E prevented increases in retinal superoxide levels in vivo, and caused phosphorylation of NRF2 and upregulation of antioxidants. Further, EPO-R76E activates NRF2 via phosphorylation by the MAPK pathway rather than the PI3K/Akt pathway, used by the endogenous antioxidant response to elevated IOP.
Collapse
Affiliation(s)
- Sarah Naguib
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
| | - Carlisle R. DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Jon R. Backstrom
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Ameer A. Haider
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
| | - John M. Ang
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrew M. Boal
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
| | - David J. Calkins
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37232, USA
| | - Tonia S. Rex
- Neuroscience Program, Vanderbilt University, Nashville, TN 37232, USA
- Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
33
|
Luminance Contrast Shifts Dominance Balance between ON and OFF Pathways in Human Vision. J Neurosci 2023; 43:993-1007. [PMID: 36535768 PMCID: PMC9908321 DOI: 10.1523/jneurosci.1672-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/14/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
Human vision processes light and dark stimuli in visual scenes with separate ON and OFF neuronal pathways. In nature, stimuli lighter or darker than their local surround have different spatial properties and contrast distributions (Ratliff et al., 2010; Cooper and Norcia, 2015; Rahimi-Nasrabadi et al., 2021). Similarly, in human vision, we show that luminance contrast affects the perception of lights and darks differently. At high contrast, human subjects of both sexes locate dark stimuli faster and more accurately than light stimuli, which is consistent with a visual system dominated by the OFF pathway. However, at low contrast, they locate light stimuli faster and more accurately than dark stimuli, which is consistent with a visual system dominated by the ON pathway. Luminance contrast was strongly correlated with multiple ON/OFF dominance ratios estimated from light/dark ratios of performance errors, missed targets, or reaction times (RTs). All correlations could be demonstrated at multiple eccentricities of the central visual field with an ON-OFF perimetry test implemented in a head-mounted visual display. We conclude that high-contrast stimuli are processed faster and more accurately by OFF pathways than ON pathways. However, the OFF dominance shifts toward ON dominance when stimulus contrast decreases, as expected from the higher-contrast sensitivity of ON cortical pathways (Kremkow et al., 2014; Rahimi-Nasrabadi et al., 2021). The results highlight the importance of contrast polarity in visual field measurements and predict a loss of low-contrast vision in humans with ON pathway deficits, as demonstrated in animal models (Sarnaik et al., 2014).SIGNIFICANCE STATEMENT ON and OFF retino-thalamo-cortical pathways respond differently to luminance contrast. In both animal models and humans, low contrasts drive stronger responses from ON pathways, whereas high contrasts drive stronger responses from OFF pathways. We demonstrate that these ON-OFF pathway differences have a correlate in human vision. At low contrast, humans locate light targets faster and more accurately than dark targets but, as contrast increases, dark targets become more visible than light targets. We also demonstrate that contrast is strongly correlated with multiple light/dark ratios of visual performance in central vision. These results provide a link between neuronal physiology and human vision while emphasizing the importance of stimulus polarity in measurements of visual fields and contrast sensitivity.
Collapse
|
34
|
Analysis of Axonal Regrowth and Dendritic Remodeling After Optic Nerve Crush in Adult Zebrafish. Methods Mol Biol 2023; 2636:163-190. [PMID: 36881300 DOI: 10.1007/978-1-0716-3012-9_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Neurodegenerative diseases and central nervous system (CNS) injuries are frequently characterized by axonal damage, as well as dendritic pathology. In contrast to mammals, adult zebrafish show a robust regeneration capacity after CNS injury and form the ideal model organism to further unravel the underlying mechanisms for both axonal and dendritic regrowth upon CNS damage. Here, we first describe an optic nerve crush injury model in adult zebrafish, an injury paradigm that inflicts de- and regeneration of the axons of retinal ganglion cells (RGCs), but also triggers RGC dendrite disintegration and subsequent recovery in a stereotyped and timed process. Next, we outline protocols for quantifying axonal regeneration and synaptic recovery in the brain, using retro- and anterograde tracing experiments and an immunofluorescent staining for presynaptic compartments, respectively. Finally, methods to analyze RGC dendrite retraction and subsequent regrowth in the retina are delineated, using morphological measurements and immunofluorescent staining for dendritic and synaptic markers.
Collapse
|
35
|
Križaj D, Cordeiro S, Strauß O. Retinal TRP channels: Cell-type-specific regulators of retinal homeostasis and multimodal integration. Prog Retin Eye Res 2023; 92:101114. [PMID: 36163161 PMCID: PMC9897210 DOI: 10.1016/j.preteyeres.2022.101114] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 02/05/2023]
Abstract
Transient receptor potential (TRP) channels are a widely expressed family of 28 evolutionarily conserved cationic ion channels that operate as primary detectors of chemical and physical stimuli and secondary effectors of metabotropic and ionotropic receptors. In vertebrates, the channels are grouped into six related families: TRPC, TRPV, TRPM, TRPA, TRPML, and TRPP. As sensory transducers, TRP channels are ubiquitously expressed across the body and the CNS, mediating critical functions in mechanosensation, nociception, chemosensing, thermosensing, and phototransduction. This article surveys current knowledge about the expression and function of the TRP family in vertebrate retinas, which, while dedicated to transduction and transmission of visual information, are highly susceptible to non-visual stimuli. Every retinal cell expresses multiple TRP subunits, with recent evidence establishing their critical roles in paradigmatic aspects of vertebrate vision that include TRPM1-dependent transduction of ON bipolar signaling, TRPC6/7-mediated ganglion cell phototransduction, TRP/TRPL phototransduction in Drosophila and TRPV4-dependent osmoregulation, mechanotransduction, and regulation of inner and outer blood-retina barriers. TRP channels tune light-dependent and independent functions of retinal circuits by modulating the intracellular concentration of the 2nd messenger calcium, with emerging evidence implicating specific subunits in the pathogenesis of debilitating diseases such as glaucoma, ocular trauma, diabetic retinopathy, and ischemia. Elucidation of TRP channel involvement in retinal biology will yield rewards in terms of fundamental understanding of vertebrate vision and therapeutic targeting to treat diseases caused by channel dysfunction or over-activation.
Collapse
Affiliation(s)
- David Križaj
- Departments of Ophthalmology, Neurobiology, and Bioengineering, University of Utah, Salt Lake City, USA
| | - Soenke Cordeiro
- Institute of Physiology, Faculty of Medicine, Christian-Albrechts-University Kiel, Germany
| | - Olaf Strauß
- Experimental Ophthalmology, Department of Ophthalmology, Charité - Universitätsmedizin Berlin, a Corporate Member of Freie Universität, Humboldt-University, The Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
36
|
Loss of Retinogeniculate Synaptic Function in the DBA/2J Mouse Model of Glaucoma. eNeuro 2022; 9:ENEURO.0421-22.2022. [PMID: 36526366 PMCID: PMC9794376 DOI: 10.1523/eneuro.0421-22.2022] [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: 10/11/2022] [Revised: 11/22/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Retinal ganglion cell (RGC) axons comprise the optic nerve and carry information to the dorsolateral geniculate nucleus (dLGN), which is then relayed to the cortex for conscious vision. Glaucoma is a blinding neurodegenerative disease that commonly results from intraocular pressure (IOP)-associated injury leading to RGC axonal pathology, disruption of RGC outputs to the brain, and eventual apoptotic loss of RGC somata. The consequences of elevated IOP and glaucomatous pathology on RGC signaling to the dLGN are largely unknown yet are likely to contribute to vision loss. Here, we used anatomic and physiological approaches to study the structure and function of retinogeniculate (RG) synapses in male and female DBA/2J (D2) mice with inherited glaucoma before and after IOP elevation. D2 mice showed progressive loss of anterograde optic tract transport to the dLGN and vGlut2 labeling of RGC axon terminals while patch-clamp measurements of RG synaptic function showed that synaptic transmission was reduced in 9-month and 12-month D2 mice because of the loss of individual RGC axon inputs. TC neuron dendrites had reduced Sholl complexity at 12 months, suggestive of delayed reorganization following reduced synaptic input. There was no detectable change in RGC density in 11- to 12-month D2 retinas, quantified as the number of ganglion cell layer-residing somata immuno-positive for NeuN and immuno-negative for the amacrine marker choline acetyltransferase (ChAT). Thus, observed synaptic defects appear to precede RGC somatic loss. These findings identify glaucoma-associated and IOP-associated deficits in an important subcortical RGC projection target, shedding light on processes linking IOP to vision loss.
Collapse
|
37
|
Mohammadi M, Su E, Chew L, Mohammadzadeh V, Caprioli J, Weiss RE, Nouri-Mahdavi K. Comparison of Ganglion Cell Layer and Inner Plexiform Layer Rates of Change in Suspected and Established Glaucoma. Am J Ophthalmol 2022; 249:12-20. [PMID: 36516918 PMCID: PMC10106372 DOI: 10.1016/j.ajo.2022.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/28/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022]
Abstract
PURPOSE We compared ganglion cell layer (GCL) and inner plexiform layer (IPL) rates of change (RoC) in patients with glaucoma suspect (GS) and established glaucoma (EG) to test the hypothesis that IPL thickness changes would occur earlier than GCL changes in eyes with early damage. DESIGN Prospective, cohort study. METHODS A total of 64 GS eyes (46 patients) and 112 EG eyes (112 patients) with ≥2 years of follow-up and ≥3 macular optical coherence tomography scans were included. GCL and IPL superpixel thickness measurements were exported. A Bayesian hierarchical model with random intercepts/slopes and random residual variances was fitted to estimate RoC in individual superpixels. Normalized RoC and proportions of superpixels with significantly negative and positive GCL and IPL RoC were compared within the groups. RESULTS The average (SD) follow-up time and number of scans were 3.5 (0.7) years and 4.2 (1.0), respectively, in the GS group and 3.6 (0.4) years and 7.3 (1.1) in the EG group. Mean (SD) normalized RoC was faster for GCL than IPL (-0.69 [0.05] vs -0.33 [0.04]) in the GS group, whereas it was faster for IPL (-0.47 [0.03] vs -0.28 [0.02]) in EG eyes. GCL RoC were significantly negative in 24 of 36 superpixels compared with 8 of 36 for IPL (P < .001) in GS eyes. In the EG group, 23 of 36 superpixels had significant negative IPL RoC compared with 13 of 36 superpixels for GCL (P = .006). CONCLUSIONS GCL thickness is more likely to demonstrate change over time compared with IPL in glaucoma suspects. There is no evidence of preferential IPL thinning in eyes with suspected early glaucoma damage.
Collapse
Affiliation(s)
- Massood Mohammadi
- From the Glaucoma Division, Stein Eye Institute, David Geffen School of Medicine (M.M., L.C., V.M., J.C., K.N-M.), University of California Los Angeles, Los Angeles, California, USA
| | - Erica Su
- Department of Biostatistics, Fielding School of Public Health (E.S., R.E.W.), University of California Los Angeles, Los Angeles, California, USA
| | - Leila Chew
- From the Glaucoma Division, Stein Eye Institute, David Geffen School of Medicine (M.M., L.C., V.M., J.C., K.N-M.), University of California Los Angeles, Los Angeles, California, USA
| | - Vahid Mohammadzadeh
- From the Glaucoma Division, Stein Eye Institute, David Geffen School of Medicine (M.M., L.C., V.M., J.C., K.N-M.), University of California Los Angeles, Los Angeles, California, USA
| | - Joseph Caprioli
- From the Glaucoma Division, Stein Eye Institute, David Geffen School of Medicine (M.M., L.C., V.M., J.C., K.N-M.), University of California Los Angeles, Los Angeles, California, USA
| | - Robert E Weiss
- Department of Biostatistics, Fielding School of Public Health (E.S., R.E.W.), University of California Los Angeles, Los Angeles, California, USA
| | - Kouros Nouri-Mahdavi
- From the Glaucoma Division, Stein Eye Institute, David Geffen School of Medicine (M.M., L.C., V.M., J.C., K.N-M.), University of California Los Angeles, Los Angeles, California, USA.
| |
Collapse
|
38
|
Garner MA, Strickland RG, Girkin CA, Gross AK. Mechanisms of retinal ganglion cell injury following acute increases in intraocular pressure. FRONTIERS IN OPHTHALMOLOGY 2022; 2:1007103. [PMID: 38983517 PMCID: PMC11182138 DOI: 10.3389/fopht.2022.1007103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/26/2022] [Indexed: 07/11/2024]
Abstract
The maintenance of intraocular pressure (IOP) is critical to preserving the pristine optics required for vision. Disturbances in IOP can directly impact the optic nerve and retina, and inner retinal injury can occur following acute and chronic IOP elevation. There are a variety of animal models that have been developed to study the effects of acute and chronic elevation of IOP on the retina, retinal ganglion cell (RGC) morphology, intracellular signaling, gene expression changes, and survival. Acute IOP models induce injury that allows for the study of RGC response to well characterized injury and potential recovery. This review will focus on the initial impact of acute IOP elevation on RGC injury and recovery as these early responses may be the best targets for potential therapeutic interventions to promote RGC survival in glaucoma.
Collapse
Affiliation(s)
- Mary Anne Garner
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ryan G. Strickland
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Christopher A. Girkin
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Alecia K. Gross
- Department of Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Ophthalmology and Visual Sciences, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
39
|
Pitale PM, Shen G, Sigireddi RR, Polo-Prieto M, Park YH, Gibson SE, Westenskow PD, Channa R, Frankfort BJ. Selective vulnerability of the intermediate retinal capillary plexus precedes retinal ganglion cell loss in ocular hypertension. Front Cell Neurosci 2022; 16:1073786. [PMID: 36545655 PMCID: PMC9760765 DOI: 10.3389/fncel.2022.1073786] [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: 10/18/2022] [Accepted: 11/15/2022] [Indexed: 12/08/2022] Open
Abstract
Introduction: Glaucoma, a disease of retinal ganglion cell (RGC) injury and potentially devastating vision loss, is associated with both ocular hypertension (OHT) and reduced ocular blood flow. However, the relationship between OHT and retinal capillary architecture is not well understood. In this project, we studied microvasculature damage in mice exposed to mild levels of induced OHT. Methods: Mild OHT was induced with the microbead model for 2 weeks. At this time point, some retinas were immunostained with CD31 (endothelium), Collagen IV (basement membrane), and RBPMS (RGCs) for z-stack confocal microscopy. We processed these confocal images to distinguish the three retinal capillary plexi (superficial, intermediate, and deep). We manually counted RGC density, analyzed vascular complexity, and identified topographical and spatial vascular features of the retinal capillaries using a combination of novel manual and automated workflows. Other retinas were dissociated and immunopanned to isolate RGCs and amacrine cells (ACs) for hypoxia gene array analysis. Results: RGC counts were normal but there was decreased overall retinal capillary complexity. This reduced complexity could be explained by abnormalities in the intermediate retinal capillary plexus (IRCP) that spared the other plexi. Capillary junction density, vessel length, and vascular area were all significantly reduced, and the number of acellular capillaries was dramatically increased. ACs, which share a neurovascular unit (NVU) with the IRCP, displayed a marked increase in the relative expression of many hypoxia-related genes compared to RGCs from the same preparations. Discussion: We have discovered a rapidly occurring, IRCP-specific, OHT-induced vascular phenotype that precedes RGC loss. AC/IRCP NVU dysfunction may be a mechanistic link for early vascular remodeling in glaucoma.
Collapse
Affiliation(s)
- Priyamvada M. Pitale
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Guofu Shen
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Rohini R. Sigireddi
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Maria Polo-Prieto
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Yong H. Park
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Solomon E. Gibson
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Peter D. Westenskow
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Roomasa Channa
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Benjamin J. Frankfort
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States,Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States,*Correspondence: Benjamin J. Frankfort
| |
Collapse
|
40
|
Youale J, Bigot K, Kodati B, Jaworski T, Fan Y, Nsiah NY, Pappenhagen N, Inman DM, Behar-Cohen F, Bordet T, Picard E. Neuroprotective Effects of Transferrin in Experimental Glaucoma Models. Int J Mol Sci 2022; 23:ijms232112753. [PMID: 36361544 PMCID: PMC9659282 DOI: 10.3390/ijms232112753] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 01/25/2023] Open
Abstract
Iron is essential for retinal metabolism, but an excess of ferrous iron causes oxidative stress. In glaucomatous eyes, retinal ganglion cell (RGC) death has been associated with dysregulation of iron homeostasis. Transferrin (TF) is an endogenous iron transporter that controls ocular iron levels. Intraocular administration of TF is neuroprotective in various models of retinal degeneration, preventing iron overload and reducing iron-induced oxidative stress. Herein, we assessed the protective effects of TF on RGC survival, using ex vivo rat retinal explants exposed to iron, NMDA-induced excitotoxicity, or CoCl2-induced hypoxia, and an in vivo rat model of ocular hypertension (OHT). TF significantly preserved RGCs against FeSO4-induced toxicity, NMDA-induced excitotoxicity, and CoCl2-induced hypoxia. TF protected RGCs from apoptosis, ferroptosis, and necrosis. In OHT rats, TF reduced RGC loss by about 70% compared to vehicle-treated animals and preserved about 47% of the axons. Finally, increased iron staining was shown in the retina of a glaucoma patient's eye as compared to non-glaucomatous eyes. These results indicate that TF can interfere with different cell-death mechanisms involved in glaucoma pathogenesis and demonstrate the ability of TF to protect RGCs exposed to elevated IOP. Altogether, these results suggest that TF is a promising treatment against glaucoma neuropathy.
Collapse
Affiliation(s)
- Jenny Youale
- Eyevensys, 11 Rue Watt, 75013 Paris, France
- Centre de Recherche des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, From Physiopathology of Ocular Diseases to Clinical Development, 75006 Paris, France
| | | | - Bindu Kodati
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Thara Jaworski
- Centre de Recherche des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, From Physiopathology of Ocular Diseases to Clinical Development, 75006 Paris, France
| | - Yan Fan
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nana Yaa Nsiah
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Nathaniel Pappenhagen
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Denise M. Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Francine Behar-Cohen
- Eyevensys, 11 Rue Watt, 75013 Paris, France
- Centre de Recherche des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, From Physiopathology of Ocular Diseases to Clinical Development, 75006 Paris, France
- Cochin Hospital, AP-HP, Assistance Publique Hôpitaux de Paris, 24 rue du Faubourg Saint Jacques, 75014 Paris, France
| | - Thierry Bordet
- Eyevensys, 11 Rue Watt, 75013 Paris, France
- Correspondence: (T.B.); (E.P.)
| | - Emilie Picard
- Centre de Recherche des Cordeliers, INSERM, Université de Paris Cité, Sorbonne Université, From Physiopathology of Ocular Diseases to Clinical Development, 75006 Paris, France
- Correspondence: (T.B.); (E.P.)
| |
Collapse
|
41
|
Norcia AM, Yakovleva A, Jehangir N, Goldberg JL. Preferential Loss of Contrast Decrement Responses in Human Glaucoma. Invest Ophthalmol Vis Sci 2022; 63:16. [PMID: 36264656 PMCID: PMC9587510 DOI: 10.1167/iovs.63.11.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to determine whether glaucoma in human patients produces preferential damage to OFF visual pathways, as suggested by animal experimental models, patient electroretinogram (ERG), and retinal imaging data. Methods Steady-state visual evoked potentials (SSVEPs) were recorded monocularly from 50 patients with glaucoma and 28 age-similar controls in response to equal Weber contrast increments and decrements presented using 2.73 hertz (Hz) sawtooth temporal waveforms. Results The eyes of patients with glaucoma were separated into mild (better than -6 decibel [dB] mean deviation; n = 28) and moderate to severe (worse than -6 dB mean deviation, n = 22) groups based on their Humphrey 24-2 visual field measurements. Response amplitudes and phases from the two glaucoma-severity groups were compared to controls at the group level. SSVEP amplitudes were depressed in both glaucoma groups, more so in the moderate to severe glaucoma group. The differences between controls and the moderate-severe glaucoma groups were more statistically reliable for decrements than for increments. Mean responses to decremental sawtooth stimuli were larger than those to increments in controls and in the mild glaucoma but not in the moderate to severe glaucoma group at the first harmonic. OFF/decrement responses at the first harmonic were faster in controls, but not in patients. Conclusions The observed pattern of preferential loss of decremental responses in human glaucoma is consistent with prior reports of selective damage to OFF retinal ganglion cells in murine models and in data from human ERG and retinal imaging. These data motivate pursuit of SSVEP as a biomarker for glaucoma progression.
Collapse
Affiliation(s)
- Anthony M. Norcia
- Department of Psychology, Wu Tsai Neurosciences Institute, Stanford University, Stanford, California, United States
| | - Alexandra Yakovleva
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Stanford, California, United States
| | - Naz Jehangir
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Stanford, California, United States
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Stanford, California, United States
| |
Collapse
|
42
|
Sarossy M, Crowston J, Kumar D, Weymouth A, Wu Z. Time-Frequency Analysis of ERG With Discrete Wavelet Transform and Matching Pursuits for Glaucoma. Transl Vis Sci Technol 2022; 11:19. [PMID: 36227605 PMCID: PMC9583752 DOI: 10.1167/tvst.11.10.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/13/2022] [Indexed: 02/01/2023] Open
Abstract
Purpose To examine the performance of two time-frequency feature extraction techniques applied to electroretinograms (ERGs) for the prediction of glaucoma severity. Methods ERGs targeting the photopic negative response were obtained in 103 eyes of 55 patients with glaucoma. Features from the ERG recordings were extracted using two time-frequency extraction techniques based on the discrete wavelet transform (DWT) and the matching pursuit (MP) decomposition. Amplitude markers of the time-domain signal were also extracted. Linear and multivariate adaptive regression spline (MARS) models were fitted using combinations of these features to predict estimated retinal ganglion cell counts, a measure of glaucoma disease severity derived from standard automated perimetry and optical coherence tomography imaging. Results Predictive models using features from the time-frequency analyses-using both DWT and MP-combined with amplitude markers outperformed predictive models using the markers alone with linear (P = 0.001) and MARS (P ≤ 0.011) models. For example, the proportions of variance (R2) explained by the MARS model using the DWT and MP features with amplitude markers were 0.53 and 0.63, respectively, compared to 0.34 for the model using the markers alone (P = 0.011 and P = 0.001, respectively). Conclusions Novel time-frequency features extracted from the photopic ERG substantially added to the prediction of glaucoma severity compared to using the time-domain amplitude markers alone. Translational Relevance Substantial information about retinal ganglion cell dysfunction exists in the time-frequency domain of ERGs that could be useful in the management of glaucoma.
Collapse
Affiliation(s)
- Marc Sarossy
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | | | | | - Anne Weymouth
- Department of Optometry and Vision Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Zhichao Wu
- Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Victoria, Australia
| |
Collapse
|
43
|
McGrady NR, Holden JM, Ribeiro M, Boal AM, Risner ML, Calkins DJ. Axon hyperexcitability in the contralateral projection following unilateral optic nerve crush in mice. Brain Commun 2022; 4:fcac251. [PMID: 36267329 PMCID: PMC9576152 DOI: 10.1093/braincomms/fcac251] [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: 03/16/2022] [Revised: 08/02/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022] Open
Abstract
Optic neuropathies are characterized by degeneration of retinal ganglion cell axonal projections to the brain, including acute conditions like optic nerve trauma and progressive conditions such as glaucoma. Despite different aetiologies, retinal ganglion cell axon degeneration in traumatic optic neuropathy and glaucoma share common pathological signatures. We compared how early pathogenesis of optic nerve trauma and glaucoma influence axon function in the mouse optic projection. We assessed pathology by measuring anterograde axonal transport from retina to superior colliculus, current-evoked optic nerve compound action potential and retinal ganglion cell density 1 week following unilateral optic nerve crush or intraocular pressure elevation. Nerve crush reduced axon transport, compound axon potential and retinal ganglion cell density, which were unaffected by intraocular pressure elevation. Surprisingly, optic nerves contralateral to crush demonstrated 5-fold enhanced excitability in compound action potential compared with naïve nerves. Enhanced excitability in contralateral sham nerves is not due to increased accumulation of voltage-gated sodium channel 1.6, or ectopic voltage-gated sodium channel 1.2 expression within nodes of Ranvier. Our results indicate hyperexcitability is driven by intrinsic responses of αON-sustained retinal ganglion cells. We found αON-sustained retinal ganglion cells in contralateral, sham and eyes demonstrated increased responses to depolarizing currents compared with those from naïve eyes, while light-driven responses remained intact. Dendritic arbours of αON-sustained retinal ganglion cells of the sham eye were like naïve, but soma area and non-phosphorylated neurofilament H increased. Current- and light-evoked responses of sham αOFF-sustained retinal ganglion cells remained stable along with somato-dendritic morphologies. In retinas directly affected by crush, light responses of αON- and αOFF-sustained retinal ganglion cells diminished compared with naïve cells along with decreased dendritic field area or branch points. Like light responses, αOFF-sustained retinal ganglion cell current-evoked responses diminished, but surprisingly, αON-sustained retinal ganglion cell responses were similar to those from naïve retinas. Optic nerve crush reduced dendritic length and area in αON-sustained retinal ganglion cells in eyes ipsilateral to injury, while crush significantly reduced dendritic branching in αOFF-sustained retinal ganglion cells. Interestingly, 1 week of intraocular pressure elevation only affected αOFF-sustained retinal ganglion cell physiology, depolarizing resting membrane potential in cells of affected eyes and blunting current-evoked responses in cells of saline-injected eyes. Collectively, our results suggest that neither saline nor sham surgery provide a true control, chronic versus acute optic neuropathies differentially affect retinal ganglion cells composing the ON and OFF pathways, and acute stress can have near-term effects on the contralateral projection.
Collapse
Affiliation(s)
- Nolan R McGrady
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave. S., Nashville, TN 37232, USA
| | - Joseph M Holden
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave. S., Nashville, TN 37232, USA
| | - Marcio Ribeiro
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave. S., Nashville, TN 37232, USA
| | - Andrew M Boal
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave. S., Nashville, TN 37232, USA
| | - Michael L Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, AA7103 MCN/VUIIS, 1161 21st Ave. S., Nashville, TN 37232, USA
| | - David J Calkins
- Correspondence to: David J. Calkins, PhD AA7103 MCN/VUIIS 1161 21st Ave. S., Nashville, TN 37232, USA E-mail:
| |
Collapse
|
44
|
Van Hook MJ. Influences of Glaucoma on the Structure and Function of Synapses in the Visual System. Antioxid Redox Signal 2022; 37:842-861. [PMID: 35044228 PMCID: PMC9587776 DOI: 10.1089/ars.2021.0253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 12/31/2021] [Indexed: 11/12/2022]
Abstract
Significance: Glaucoma is an age-related neurodegenerative disorder of the visual system associated with sensitivity to intraocular pressure (IOP). It is the leading irreversible cause of vision loss worldwide, and vision loss results from damage and dysfunction of the retinal output neurons known as retinal ganglion cells (RGCs). Recent Advances: Elevated IOP and optic nerve injury triggers pruning of RGC dendrites, altered morphology of excitatory inputs from presynaptic bipolar cells, and disrupted RGC synaptic function. Less is known about RGC outputs, although evidence to date indicates that glaucoma is associated with altered mitochondrial and synaptic structure and function in RGC-projection targets in the brain. These early functional changes likely contribute to vision loss and might be a window into early diagnosis and treatment. Critical Issues: Glaucoma affects different RGC populations to varying extents and along distinct time courses. The influence of glaucoma on RGC synaptic function as well as the mechanisms underlying these effects remain to be determined. Since RGCs are an especially energetically demanding population of neurons, altered intracellular axon transport of mitochondria and mitochondrial function might contribute to RGC synaptic dysfunction in the retina and brain as well as RGC vulnerability in glaucoma. Future Directions: The mechanisms underlying differential RGC vulnerability remain to be determined. Moreover, the timing and mechanisms of RGCs synaptic dysfunction and degeneration will provide valuable insight into the disease process in glaucoma. Future work will be able to capitalize on these findings to better design diagnostic and therapeutic approaches to detect disease and prevent vision loss. Antioxid. Redox Signal. 37, 842-861.
Collapse
Affiliation(s)
- Matthew J. Van Hook
- Department of Ophthalmology & Visual Science and Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
- Department of Cellular & Integrative Physiology, Truhlsen Eye Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| |
Collapse
|
45
|
Gao J, Provencio I, Liu X. Intrinsically photosensitive retinal ganglion cells in glaucoma. Front Cell Neurosci 2022; 16:992747. [PMID: 36212698 PMCID: PMC9537624 DOI: 10.3389/fncel.2022.992747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/05/2022] [Indexed: 11/30/2022] Open
Abstract
Glaucoma is a group of eye diseases afflicting more than 70 million people worldwide. It is characterized by damage to retinal ganglion cells (RGCs) that ultimately leads to the death of the cells and vision loss. The diversity of RGC types has been appreciated for decades, and studies, including ours, have shown that RGCs degenerate and die in a type-specific manner in rodent models of glaucoma. The type-specific loss of RGCs results in differential damage to visual and non-visual functions. One type of RGC, the intrinsically photosensitive retinal ganglion cell (ipRGC), expressing the photopigment melanopsin, serves a broad array of non-visual responses to light. Since its discovery, six subtypes of ipRGC have been described, each contributing to various image-forming and non-image-forming functions such as circadian photoentrainment, the pupillary light reflex, the photic control of mood and sleep, and visual contrast sensitivity. We recently demonstrated a link between type-specific ipRGC survival and behavioral deficits in a mouse model of chronic ocular hypertension. This review focuses on the type-specific ipRGC degeneration and associated behavioral changes in animal models and glaucoma patients. A better understanding of how glaucomatous insult impacts the ipRGC-based circuits will have broad impacts on improving the treatment of glaucoma-associated non-visual disorders.
Collapse
Affiliation(s)
- Jingyi Gao
- Department of Biology, University of Virginia, Charlottesville, VA, United States
| | - Ignacio Provencio
- Department of Biology, University of Virginia, Charlottesville, VA, United States
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, United States
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, United States
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, VA, United States
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, United States
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, United States
- Department of Psychology, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
46
|
Fraenkl SA, Simon Q, Yucel Y, Gupta N, Wittwer VV, Frueh BE, Tschanz SA. Impact of cerebral hypoperfusion-reperfusion on optic nerve integrity and visual function in the DBA/2J mouse model of glaucoma. BMJ Open Ophthalmol 2022; 7:bmjophth-2022-001078. [PMID: 36161839 PMCID: PMC9476133 DOI: 10.1136/bmjophth-2022-001078] [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: 06/01/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
Objective One of the most important risk factors for developing a glaucomatous optic neuropathy is elevated intraocular pressure. Moreover, mechanisms such as altered perfusion have been postulated to injure the optical path. In a mouse model, we compare first negative effects of cerebral perfusion/reperfusion on the optic nerve structure versus alterations by elevated intraocular pressure. Second, we compare the alterations by isolated hypoperfusion-reperfusion and isolated intraocular pressure to the combination of both. Methods and analysis Mice were divided in four groups: (1) controls; (2) perfusion altered mice that underwent transient bi-common carotid artery occlusion (BCCAO) for 40 min; (3) glaucoma group (DBA/2J mice); (4) combined glaucoma and altered perfusion (DBA/2J mice with transient BCCAO). Optic nerve sections were stereologically examined 10–12 weeks after intervention. Results All experimental groups showed a decreased total axon number per optic nerve compared with controls. In DBA/2J and combined DBA/2J & BCCAO mice the significant decrease was roughly 50%, while BCCAO leaded to a 23% reduction of axon number, however reaching significance only in the direct t-test. The difference in axon number between BCCAO and both DBA/2J mice was almost 30%, lacking statistical significance due to a remarkably high variation in both DBA/2J groups. Conclusion Elevated intraocular pressure in the DBA/2J mouse model of glaucoma leads to a much more pronounced optic nerve atrophy compared with transient forebrain hypoperfusion and reperfusion by BCCAO. A supposed worsening effect of an altered perfusion added to the pressure-related damage could not be detected.
Collapse
Affiliation(s)
| | - Quentin Simon
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Yeni Yucel
- Keenan Research Centre, St Michael's Hospital Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Neeru Gupta
- Keenan Research Centre, St Michael's Hospital Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada.,Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Valéry V Wittwer
- Department of Ophthalmology, Inselspital, University of Bern, Bern, Switzerland.,Ophthalmologic Network Organization (ONO), Geneva, Switzerland
| | - Beatrice E Frueh
- Department of Ophthalmology, Inselspital, University of Bern, Bern, Switzerland
| | | |
Collapse
|
47
|
Meah A, Boodram V, Bucinca-Cupallari F, Lim H. Axonal architecture of the mouse inner retina revealed by second harmonic generation. PNAS NEXUS 2022; 1:pgac160. [PMID: 36106183 PMCID: PMC9463061 DOI: 10.1093/pnasnexus/pgac160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/11/2022] [Indexed: 01/29/2023]
Abstract
We describe a novel method for visualizing the network of axons in the unlabeled fresh wholemount retina. The intrinsic radiation of second harmonic generation (SHG) was utilized to visualize single axons of all major retinal neurons, i.e., photoreceptors, horizontal cells, bipolar cells, amacrine cells, and the retinal ganglion cells. The cell types of SHG+ axons were determined using transgenic GFP/YFP mice. New findings were obtained with retinal SHG imaging: Müller cells do not maintain uniformly polarized microtubules in the processes; SHG+ axons of bipolar cells terminate in the inner plexiform layer (IPL) in a subtype-specific manner; a subset of amacrine cells, presumably the axon-bearing types, emits SHG; and the axon-like neurites of amacrine cells provide a cytoskeletal scaffolding for the IPL stratification. To demonstrate the utility, retinal SHG imaging was applied to testing whether the inner retina is preserved in glaucoma, using DBA/2 mice as a model of glaucoma and DBA/2-Gpnmb+ as the nonglaucomatous control. It was found that the morphology of the inner retina was largely intact in glaucoma and the presynaptic compartments to the retinal ganglion cells were uncompromised. It proves retinal SHG imaging as a promising technology for studying the physiological and diseased retinas in 3D.
Collapse
Affiliation(s)
- Arafat Meah
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Vinessia Boodram
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA
| | - Festa Bucinca-Cupallari
- Department of Physics and Astronomy, Hunter College, New York, NY 10065, USA,The Graduate Centre of the City University of New York, New York, NY 10065, USA
| | | |
Collapse
|
48
|
Boal AM, McGrady NR, Risner ML, Calkins DJ. Sensitivity to extracellular potassium underlies type-intrinsic differences in retinal ganglion cell excitability. Front Cell Neurosci 2022; 16:966425. [PMID: 35990894 PMCID: PMC9390602 DOI: 10.3389/fncel.2022.966425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Neuronal type-specific physiologic heterogeneity can be driven by both extrinsic and intrinsic mechanisms. In retinal ganglion cells (RGCs), which carry visual information from the retina to central targets, evidence suggests intrinsic properties shaping action potential (AP) generation significantly impact the responses of RGCs to visual stimuli. Here, we explored how differences in intrinsic excitability further distinguish two RCG types with distinct presynaptic circuits, alpha ON-sustained (αON-S) cells and alpha OFF-sustained (αOFF-S) cells. We found that αOFF-S RGCs are more excitable to modest depolarizing currents than αON-S RGCs but excitability plateaued earlier as depolarization increased (i.e., depolarization block). In addition to differences in depolarization block sensitivity, the two cell types also produced distinct AP shapes with increasing stimulation. αOFF-S AP width and variability increased with depolarization magnitude, which correlated with the onset of depolarization block, while αON-S AP width and variability remained stable. We then tested if differences in depolarization block observed in αON-S and αOFF-S RGCs were due to sensitivity to extracellular potassium. We found αOFF-S RGCs more sensitive to increased extracellular potassium concentration, which shifted αON-S RGC excitability to that of αOFF-S cells under baseline potassium conditions. Finally, we investigated the influence of the axon initial segment (AIS) dimensions on RGC spiking. We found that the relationship between AIS length and evoked spike rate varied not only by cell type, but also by the strength of stimulation, suggesting AIS structure alone cannot fully explain the observed differences RGC excitability. Thus, sensitivity to extracellular potassium contributes to differences in intrinsic excitability, a key factor that shapes how RGCs encode visual information.
Collapse
|
49
|
Gao J, Griner EM, Liu M, Moy J, Provencio I, Liu X. Differential effects of experimental glaucoma on intrinsically photosensitive retinal ganglion cells in mice. J Comp Neurol 2022; 530:1494-1506. [PMID: 34958682 PMCID: PMC9010357 DOI: 10.1002/cne.25293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/08/2022]
Abstract
Glaucoma is a group of eye diseases characterized by retinal ganglion cell (RGC) loss and optic nerve damage. Studies, including this study, support that RGCs degenerate and die in a type-specific manner following the disease insult. Here we specifically examined one RGC type, the intrinsically photosensitive retinal ganglion cell (ipRGC), and its associated functional deficits in a mouse model of experimental glaucoma. We induced chronic ocular hypertension (OHT) by laser photocoagulation and then characterized the survival of ipRGC subtypes. We found that ipRGCs suffer significant loss, similar to the general RGC population, but ipRGC subtypes are differentially affected following chronic OHT. M4 ipRGCs, which are involved in pattern vision, are susceptible to chronic OHT. Correspondingly, mice with chronic OHT experience reduced contrast sensitivity and visual acuity. By comparison, M1 ipRGCs, which project to the suprachiasmatic nuclei to regulate circadian rhythmicity, exhibit almost no cell loss following chronic OHT. Accordingly, we observed that circadian re-entrainment and circadian rhythmicity are largely not disrupted in OHT mice. Our study demonstrates the link between subtype-specific ipRGC survival and behavioral deficits in glaucomatous mice. These findings provide insight into glaucoma-induced visual behavioral deficits and their underlying mechanisms.
Collapse
Affiliation(s)
- Jingyi Gao
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Erin M. Griner
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Mingna Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Joanna Moy
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Ignacio Provencio
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Xiaorong Liu
- Department of Biology, University of Virginia, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia, Charlottesville, VA, USA
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
- Program in Fundamental Neuroscience, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
50
|
Frame G, Schuller A, Smith MA, Crish SD, Dengler-Crish CM. Alterations in Retinal Signaling Across Age and Sex in 3xTg Alzheimer’s Disease Mice. J Alzheimers Dis 2022; 88:471-492. [PMID: 35599482 PMCID: PMC9398084 DOI: 10.3233/jad-220016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Background: Visual disturbances often precede cognitive dysfunction in patients with Alzheimer’s disease (AD) and may coincide with early accumulation of amyloid-β (Aβ) protein in the retina. These findings have inspired critical research on in vivo ophthalmic Aβ imaging for disease biomarker detection but have not fully answered mechanistic questions on how retinal pathology affects visual signaling between the eye and brain. Objective: The goal of this study was to provide a functional and structural assessment of eye-brain communication between retinal ganglion cells (RGCs) and their primary projection target, the superior colliculus, in female and male 3xTg-AD mice across disease stages. Methods: Retinal electrophysiology, axonal transport, and immunofluorescence were used to determine RGC projection integrity, and retinal and collicular Aβ levels were assessed with advanced protein quantitation techniques. Results: 3xTg mice exhibited nuanced deficits in RGC electrical signaling, axonal transport, and synaptic integrity that exceeded normal age-related decrements in RGC function in age- and sex-matched healthy control mice. These deficits presented in sex-specific patterns among 3xTg mice, differing in the timing and severity of changes. Conclusion: These data support the premise that retinal Aβ is not just a benign biomarker in the eye, but may contribute to subtle, nuanced visual processing deficits. Such disruptions might enhance the biomarker potential of ocular amyloid and differentiate patients with incipient AD from patients experiencing normal age-related decrements in visual function.
Collapse
Affiliation(s)
- Gabrielle Frame
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
- Biomedical Sciences Graduate Program, Kent State University, Kent, OH, USA
| | - Adam Schuller
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Matthew A. Smith
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, Akron, OH, USA
| | - Samuel D. Crish
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | | |
Collapse
|