1
|
Capper EN, Linton EF, Anders JJ, Kardon RH, Gramlich OW. MOG 35 - 55-induced EAE model of optic nerve inflammation compared to MS, MOGAD and NMOSD related subtypes of human optic neuritis. J Neuroinflammation 2025; 22:102. [PMID: 40197321 PMCID: PMC11977933 DOI: 10.1186/s12974-025-03424-4] [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: 12/18/2024] [Accepted: 03/19/2025] [Indexed: 04/10/2025] Open
Abstract
Optic neuritis (ON), or inflammation of the optic nerve, is a common presenting symptom of demyelinating neuroinflammatory conditions that result in significant, subacute vision loss. Given its association with visual impairment and varying extent of visual recovery, ON has been recognized as a significant health burden with a need for new therapeutic strategies to improve long-term visual outcomes. Among the resources utilized to study ON, animal models have emerged as powerful tools to examine the underlying pathophysiology and the effectiveness of proposed therapies. In the current review, we discuss the functional and structural phenotypes related to ON in currently used mouse models, and summarize how the pathophysiology and visual phenotype of the myelin oligodendrocyte glycoprotein 35-55 (MOG35 - 55) experimental autoimmune encephalomyelitis (EAE) mouse model recapitulates clinical features of multiple sclerosis (MS), myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD), and neuromyelitis optica spectrum disorder (NMOSD). The location of ON and the amount of visual recovery in the EAE model most closely resembles MS and NMOSD. However, we propose that the MOG35 - 55-induced EAE model of ON is primarily a MOGAD model given its similarity in pathophysiology, spinal cord demyelination pattern, and the degree of vision loss, retinal nerve fiber layer (RNFL) swelling, and disc edema. Overall, the MOG35 - 55-induced EAE animal model demonstrates overlapping features of autoimmune demyelinating conditions and serves as a comprehensive tool to further our understanding of visual impairment in all three conditions.
Collapse
Affiliation(s)
- Erin N Capper
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, 52246, USA
| | - Edward F Linton
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, 52246, USA
| | - Jeffrey J Anders
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, 52246, USA
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA
| | - Randy H Kardon
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, 52246, USA
| | - Oliver W Gramlich
- Department of Ophthalmology and Visual Sciences, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
- Center for the Prevention and Treatment of Visual Loss, Iowa City VA Health Care System, Iowa City, IA, 52246, USA.
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Chang K, Chen J, Rajagopalan A, Chen DF, Cho KS. Testing Visual Function by Assessment of the Optomotor Reflex in Glaucoma. Methods Mol Biol 2025; 2858:219-227. [PMID: 39433679 DOI: 10.1007/978-1-0716-4140-8_18] [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
Optomotor response/reflex (OMR) is a fast and efficient first-in-line visual screening method, especially for rodents. It has the potential to evaluate both the scotopic and photopic visions of nonrestrained animals through tracking head movement, providing a quantitative estimate of visual functions. In restrained animals, optokinetic response (OKR), compensatory eye movements for visual shifts in the surroundings, is utilized. Both OMR and OKR capitalize on an individual's innate reflex to stabilize images for the purpose of capturing clear vision. The two reflexes have similar reliability when evaluating stimulus luminance, contrast, spatial frequency, and velocity. They have emerged as powerful tools to evaluate the efficacy of pharmacological treatments and phenotypes of subjects undergoing study. With OMR and OKR accurately assessing visual acuity (VA) as well as contrast sensitivity (CS), the gold standards for measuring clinical vision, they provide reliable and easily accessible results that further eye and brain research. These methods of sight evaluation have been used in multiple animal models, particularly mice and zebrafish. Through OMR assays, these animal models have been utilized to investigate retinal degenerative diseases, helping researchers differentiate between worsening stages. Alongside tests such as optical coherence tomography (OCT), OMR provides confirmation of visual status, where increased OMR function often correlates with improved visual status. OMR has continued to be used outside of glaucoma in various retinal diseases, such as retinitis pigmentosa (RP), diabetic retinopathy, and age-related macular degeneration.In this chapter, we will introduce the concept and application of visual stimulus-induced head or eye reflex movement in different animal species and experimental models of eye diseases, such as glaucoma and other neurodegenerative disorders, and in patients with glaucoma.
Collapse
Affiliation(s)
- Karen Chang
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Julie Chen
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Aishwarya Rajagopalan
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Dong Feng Chen
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Kin-Sang Cho
- Department of Ophthalmology, Harvard Medical School, Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA.
| |
Collapse
|
3
|
Corydon TJ, Bek T. Multiple gene therapy as a tool for regulating the expression of molecules involved in neovascular age-related macular degeneration. Prog Retin Eye Res 2025; 104:101323. [PMID: 39672501 DOI: 10.1016/j.preteyeres.2024.101323] [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: 08/27/2024] [Revised: 11/08/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
Anti-vascular endothelial growth factor (VEGF) therapies have revolutionized the treatment of neovascular age-related macular degeneration (nAMD) and other retinal diseases. However, the necessity for repeated intravitreal injections and the observation of variable treatment responses calls for new treatment modalities where fewer and more effective interventions can result in a clinical effect. Gene therapy might be such an alternative, and therefore the development and clinical application of gene therapy aimed at modifying gene expression has received considerable attention. The article reviews current knowledge of the background, pathophysiological mechanisms, technologies, limitations, and future directions for gene therapy aimed at modifying the synthesis of compounds involved in acquired and senescent retinal disease. The authors have contributed to the field by developing gene therapy to reduce the expression of vascular endothelial growth factor (VEGF), as well as multiple gene therapy for simultaneous downregulation of the synthesis of VEGF and upregulation of pigment epithelium-derived factor (PEDF) using adeno-associated virus (AAV) vectors. It is suggested that such multi-target gene therapy might be included in future treatments of retinal diseases where the underlying mechanisms are complex and cannot be attributed to one specific mediator. Such diseases might include dry AMD (dAMD) with geographic atrophy, but also diabetic macular edema (DME) and retinal vein occlusion (RVO). Gene therapy can be expected to be most beneficial for the patients in need of multiple intra-vitreal injections and in whom the therapeutic response is insufficient. It is concluded, that in parallel with basic research, there is a need for clinical studies aimed at identifying factors that can be used to identify patients who will benefit from gene therapy already at the time of diagnosis of the retinal disease.
Collapse
Affiliation(s)
- Thomas J Corydon
- Department of Biomedicine, Hoegh Guldbergs Gade 10, Aarhus University, 8000, Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
| | - Toke Bek
- Department of Ophthalmology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
4
|
Wang Y, Liu N, Hu L, Yang J, Han M, Zhou T, Xing L, Jiang H. Nanoengineered mitochondria enable ocular mitochondrial disease therapy via the replacement of dysfunctional mitochondria. Acta Pharm Sin B 2024; 14:5435-5450. [PMID: 39807326 PMCID: PMC11725173 DOI: 10.1016/j.apsb.2024.08.007] [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: 04/30/2024] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 01/16/2025] Open
Abstract
Leber's hereditary optic neuropathy (LHON) is an ocular mitochondrial disease that involves the impairment of mitochondrial complex I, which is an important contributor to blindness among young adults across the globe. However, the disorder has no available cures, since the approved drug idebenone for LHON in Europe relies on bypassing complex I defects rather than fixing them. Herein, PARKIN mRNA-loaded nanoparticle (mNP)-engineered mitochondria (mNP-Mito) were designed to replace dysfunctional mitochondria with the delivery of exogenous mitochondria, normalizing the function of complex I for treating LHON. The mNP-Mito facilitated the supplementation of healthy mitochondria containing functional complex I via mitochondrial transfer, along with the elimination of dysfunctional mitochondria with impaired complex I via an enhanced PARKIN-mediated mitophagy process. In a mouse model induced with a complex I inhibitor (rotenone, Rot), mNP-Mito enhanced the presence of healthy mitochondria and exhibited a sharp increase in complex I activity (76.5%) compared to the group exposed to Rot damage (29.5%), which greatly promoted the restoration of ATP generation and mitigation of ocular mitochondrial disease-related phenotypes. This study highlights the significance of nanoengineered mitochondria as a promising and feasible tool for the replacement of dysfunctional mitochondria and the repair of mitochondrial function in mitochondrial disease therapies.
Collapse
Affiliation(s)
- Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Nahui Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lifan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jingsong Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Mengmeng Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Tianjiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hulin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- College of Pharmacy, Yanbian University, Yanji 133002, China
| |
Collapse
|
5
|
Hösel K, Chasan B, Tode J, Rose-John S, Roider JB, Ehlken C. Effect of intravitreal injection of anti-interleukin (IL)-6 antibody in experimental autoimmune uveitis in mice. J Ophthalmic Inflamm Infect 2024; 14:57. [PMID: 39497001 PMCID: PMC11535092 DOI: 10.1186/s12348-024-00441-x] [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: 03/22/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
PURPOSE The aim of this study was to assess the functional and clinical impact of intravitreal administration of a neutralizing anti-IL-6 antibody in the treatment of experimental autoimmune uveitis (EAU) in mice. METHODS EAU was induced in 17 female B10.RIII mice by administering Inter-Photoreceptor-Binding-Protein (IRBP) in complete Freund's adjuvant, followed by a boost with Pertussis toxin. Intravitreal injections of anti-Interleukin (IL)-6 antibody were administered on days 10, 13, and 16 after EAU induction (day 0) into the randomized treatment eye, with an isotype antibody similarly injected into the fellow control eye. Visual acuity was assessed using the optomotor reflex via OptoDrum, and clinical scoring was performed via fundus imaging (utilizing 6 EAU grades) in a single-blinded manner on days 0, 10, 13, 16, and 18. RESULTS Uveitis developed in all 17 mice. Significantly higher visual acuity was observed in treated eyes compared to control eyes on days 13, 16, and 18. The most pronounced effect was noted on days 16 and 18 (p < 0.001). On days 13, 16, and 18 the number of eyes with lower EAU-score was significantly higher in the treatment group, with the most notable effect observed on day 18 (p < 0.003). CONCLUSION Intravitreal administration of anti-IL-6 treatment notably mitigates experimental autoimmune uveitis in mice, both functionally and clinically. Further investigations are warranted to assess the potential of intravitreal anti-IL-6 therapy as a treatment option for non-infectious uveitis in humans.
Collapse
Affiliation(s)
- Kristin Hösel
- Department of Ophthalmology, UKSH Kiel, Kiel, Germany.
| | - Büsra Chasan
- Department of Ophthalmology, UKSH Kiel, Kiel, Germany
| | - Jan Tode
- Department of Ophthalmology, MHH Hannover, Hannover, Germany
| | - Stefan Rose-John
- Department of Biochemistry, Klinik für Augenheilkunde, CAU Kiel, Haus B2, Arnold-Heller-Str. 3, 24105, Kiel, Germany
| | | | | |
Collapse
|
6
|
Enayati S, Chang K, Lennikov A, Yang M, Lee C, Ashok A, Elzaridi F, Yen C, Gunes K, Xie J, Cho KS, Utheim TP, Chen DF. Optimal transcorneal electrical stimulation parameters for preserving photoreceptors in a mouse model of retinitis pigmentosa. Neural Regen Res 2024; 19:2543-2552. [PMID: 38526290 PMCID: PMC11090438 DOI: 10.4103/1673-5374.392888] [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: 12/31/2022] [Revised: 11/21/2023] [Accepted: 12/29/2023] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00034/figure1/v/2024-03-08T184507Z/r/image-tiff Retinitis pigmentosa is a hereditary retinal disease that affects rod and cone photoreceptors, leading to progressive photoreceptor loss. Previous research supports the beneficial effect of electrical stimulation on photoreceptor survival. This study aims to identify the most effective electrical stimulation parameters and functional advantages of transcorneal electrical stimulation (tcES) in mice affected by inherited retinal degeneration. Additionally, the study seeked to analyze the electric field that reaches the retina in both eyes in mice and post-mortem humans. In this study, we recorded waveforms and voltages directed to the retina during transcorneal electrical stimulation in C57BL/6J mice using an intraocular needle probe with rectangular, sine, and ramp waveforms. To investigate the functional effects of electrical stimulation on photoreceptors, we used human retinal explant cultures and rhodopsin knockout (Rho-/-) mice, demonstrating progressive photoreceptor degeneration with age. Human retinal explants isolated from the donors' eyes were then subjected to electrical stimulation and cultured for 48 hours to simulate the neurodegenerative environment in vitro. Photoreceptor density was evaluated by rhodopsin immunolabeling. In vivo Rho-/- mice were subjected to two 5-day series of daily transcorneal electrical stimulation using rectangular and ramp waveforms. Retinal function and visual perception of mice were evaluated by electroretinography and optomotor response (OMR), respectively. Immunolabeling was used to assess the morphological and biochemical changes of the photoreceptor and bipolar cells in mouse retinas. Oscilloscope recordings indicated effective delivery of rectangular, sine, and ramp waveforms to the retina by transcorneal electrical stimulation, of which the ramp waveform required the lowest voltage. Evaluation of the total conductive resistance of the post-mortem human compared to the mouse eyes indicated higher cornea-to-retina resistance in human eyes. The temperature recordings during and after electrical stimulation indicated no significant temperature change in vivo and only a subtle temperature increase in vitro (~0.5-1.5°C). Electrical stimulation increased photoreceptor survival in human retinal explant cultures, particularly at the ramp waveform. Transcorneal electrical stimulation (rectangular + ramp) waveforms significantly improved the survival and function of S and M-cones and enhanced visual acuity based on the optomotor response results. Histology and immunolabeling demonstrated increased photoreceptor survival, improved outer nuclear layer thickness, and increased bipolar cell sprouting in Rho-/- mice. These results indicate that transcorneal electrical stimulation effectively delivers the electrical field to the retina, improves photoreceptor survival in both human and mouse retinas, and increases visual function in Rho-/- mice. Combined rectangular and ramp waveform stimulation can promote photoreceptor survival in a minimally invasive fashion.
Collapse
Affiliation(s)
- Sam Enayati
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Institute of clinical medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Karen Chang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Department of Medical Biochemistry, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Anton Lennikov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Department of Medical Biochemistry, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Menglu Yang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Cherin Lee
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ajay Ashok
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Department of Medical Biochemistry, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Farris Elzaridi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Christina Yen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kasim Gunes
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Department of Histology and Embryology, School of Medicine, Marmara University, Istanbul, Turkiye
| | - Jia Xie
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Tor Paaske Utheim
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Institute of clinical medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, University of Oslo, Oslo, Norway
- Department of Ophthalmology, Drammen Hospital, Vestre Viken Hospital Trust, Drammen, Norway
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Wang Q, So C, Qiu C, Zhang T, Yang K, Pan F. Diminished light sensitivities of ON alpha retinal ganglion cells observed in a mouse model of hyperglycemia. Exp Eye Res 2024; 248:110113. [PMID: 39349092 DOI: 10.1016/j.exer.2024.110113] [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: 06/27/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024]
Abstract
This study aimed to investigate potential functional changes in retinal ganglion cells (RGCs) in a mouse model of hyperglycemia and explore possible therapeutic approaches. Hyperglycemia resembling type 1 diabetes mellitus (DM) was induced in C57BL/6 mice through intraperitoneal injection of streptozotocin (STZ). Blood glucose levels were confirmed to be elevated after 1 week and 4 weeks of injection. Mice with blood glucose levels above 350 mg/mL after 4 weeks of one-dose STZ injection were considered hyperglycemic. The light sensitivity of ON alpha (α) retinal ganglion cells (RGCs), not OFF αRGCs, was reduced in the hyperglycemic mouse model. The number of apoptotic cells, RGCs, and amacrine cells (ACs) remained unaffected at this stage. Similarly, the eletroretinogram (ERG) and optokinetic test results showed no significant differences. The application of picrotoxin (PTX) to block GABA receptors could increase the light sensitivity of ON αRGCs by 1 log unit in hyperglycemic mice. The results show that ON αRGCs may be more susceptible to microenvironmental changes caused by hyperglycemia than OFF αRGCs. This decline in light sensitivity may occur before cell apoptosis during the early stages of the hyperglycemic mouse model but has the potential to be reversed.
Collapse
Affiliation(s)
- Qin Wang
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Chunghim So
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chunting Qiu
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Ting Zhang
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Kangyi Yang
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong
| | - Feng Pan
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong; Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong; Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
8
|
Tian Z, Liu Q, Lin HY, Zhu YR, Ling L, Sung TC, Wang T, Li W, Gao M, Cheng S, Renuka RR, Subbiah SK, Fan G, Wu GJ, Higuchi A. Effects of ECM protein-coated surfaces on the generation of retinal pigment epithelium cells differentiated from human pluripotent stem cells. Regen Biomater 2024; 11:rbae091. [PMID: 39233867 PMCID: PMC11374035 DOI: 10.1093/rb/rbae091] [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: 04/01/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 09/06/2024] Open
Abstract
Retinal degeneration diseases, such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP), initially manifest as dysfunction or death of the retinal pigment epithelium (RPE). Subretinal transplantation of human pluripotent stem cell (hPSC)-derived RPE cells has emerged as a potential therapy for retinal degeneration. However, RPE cells differentiated from hPSCs using current protocols are xeno-containing and are rarely applied in clinical trials. The development of hPSC-derived RPE cell differentiation protocols using xeno-free biomaterials is urgently needed for clinical applications. In this study, two protocols (the activin A and NIC84 protocols) were selected for modification and use in the differentiation of hiPSCs into RPE cells; the chetomin concentration was gradually increased to achieve high differentiation efficiency of RPE cells. The xeno-free extracellular matrix (ECM) proteins, laminin-511, laminin-521 and recombinant vitronectin, were selected as plate-coating substrates, and a Matrigel (xeno-containing ECM)-coated surface was used as a positive control. Healthy, mature hPSC-derived RPE cells were transplanted into 21-day-old Royal College of Surgeons (RCS) rats, a model of retinal degeneration disease. The visual function of RCS rats was evaluated by optomotor response (qOMR) and electroretinography after transplantation of hPSC-derived RPE cells. Our study demonstrated that hPSCs can be efficiently differentiated into RPE cells on LN521-coated dishes using the NIC84 protocol, and that subretinal transplantation of the cell suspensions can delay the progression of vision loss in RCS rats.
Collapse
Affiliation(s)
- Zeyu Tian
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Hui-Yu Lin
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan, China
| | - Yu-Ru Zhu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan, China
| | - Ling Ling
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wanqi Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Min Gao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Sitian Cheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Remya Rajan Renuka
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Suresh Kumar Subbiah
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences and Department of Obstetrics & Gynecology, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan, China
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan, China
- R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan, China
| |
Collapse
|
9
|
Gunes K, Chang K, Lennikov A, Tai WL, Chen J, ElZaridi F, Cho KS, Utheim TP, Dong Feng C. Preservation of vision by transpalpebral electrical stimulation in mice with inherited retinal degeneration. Front Cell Dev Biol 2024; 12:1412909. [PMID: 39206091 PMCID: PMC11349514 DOI: 10.3389/fcell.2024.1412909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction The potential neuroprotective and regenerative properties of electrical stimulation (ES) were studied in rhodopsin knockout mice (Rho -/- ), a murine model of inherited retinal degeneration. The study focused on assessing the impact of varying ES frequencies on visual functions and photoreceptor cell survival in Rho -/- mice. Methods To elucidate the impact of electrical stimulation on cone survival, Rho -/- mice received either sham or transpalpebral ES using biphasic ramp or rectangular waveforms at 100 µA amplitude, starting at six weeks of age. The treatment duration spanned from one to three weeks. The optimal treatment frequency of ES sessions was determined by applying ES every one, two, or three days in three separate groups of Rho -/- mice. The sham group received daily treatments without the application of ES. Results Our study revealed significant improvement of visual function in Rho -/- mice following daily or every-other-day noninvasive transpalpebral ES, as evidenced by electroretinogram and optomotor response-based visual behavior assays. Concurrently, assessment of outer nuclear thickness and immunohistochemistry for the cone photoreceptor cell marker PNA demonstrated pronounced increases in the survival of rods and cones and improvement in the morphology of the inner and outer segments. Discussion This study underscores the protective effect of non-invasive ES in rhodopsin knockout-induced retinal degenerative disorders, providing a foundation for developing targeted therapeutic interventions for retinitis pigmentosa.
Collapse
Affiliation(s)
- Kasim Gunes
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
- Department of Histology and Embryology, School of Medicine, Marmara University, Istanbul, Türkiye
| | - Karen Chang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Wai Lydia Tai
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Julie Chen
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Farris ElZaridi
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Kin-Sang Cho
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Tor Paaske Utheim
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | - Chen Dong Feng
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Yuan M, Jin S, Tan G, Song S, Liu Y, Wang H, Shen Y. A Non-canonical Excitatory PV RGC-PV SC Visual Pathway for Mediating the Looming-evoked Innate Defensive Response. Neurosci Bull 2024; 40:310-324. [PMID: 37302108 PMCID: PMC10912393 DOI: 10.1007/s12264-023-01076-z] [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: 02/23/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Parvalbumin-positive retinal ganglion cells (PV+ RGCs) are an essential subset of RGCs found in various species. However, their role in transmitting visual information remains unclear. Here, we characterized PV+ RGCs in the retina and explored the functions of the PV+ RGC-mediated visual pathway. By applying multiple viral tracing strategies, we investigated the downstream of PV+ RGCs across the whole brain. Interestingly, we found that the PV+ RGCs provided direct monosynaptic input to PV+ excitatory neurons in the superficial layers of the superior colliculus (SC). Ablation or suppression of SC-projecting PV+ RGCs abolished or severely impaired the flight response to looming visual stimuli in mice without affecting visual acuity. Furthermore, using transcriptome expression profiling of individual cells and immunofluorescence colocalization for RGCs, we found that PV+ RGCs are predominant glutamatergic neurons. Thus, our findings indicate the critical role of PV+ RGCs in an innate defensive response and suggest a non-canonical subcortical visual pathway from excitatory PV+ RGCs to PV+ SC neurons that regulates looming visual stimuli. These results provide a potential target for intervening and treating diseases related to this circuit, such as schizophrenia and autism.
Collapse
Affiliation(s)
- Man Yuan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Sen Jin
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Gao Tan
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Siyuan Song
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, 77030, USA
| | - Yizong Liu
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China
| | - Huadong Wang
- The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, National Medical Products Administration Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen, 518055, China
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, CAS, Wuhan, 430071, China
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430060, China.
- Frontier Science Center of Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
11
|
Hetzer SM, O'Connell C, Lallo V, Robson M, Evanson NK. Model matters: Differential outcomes in traumatic optic neuropathy pathophysiology between blunt and blast-wave mediated head injuries. Exp Neurol 2024; 372:114613. [PMID: 37995952 PMCID: PMC10870099 DOI: 10.1016/j.expneurol.2023.114613] [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: 05/25/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Over 3 million people in the United States live with long-term disability because of a traumatic brain injury (TBI). The purpose of this study was to characterize and compare two different animal models of TBI (blunt head trauma and blast TBI) to determine common and divergent characteristics of these models. With recent literature reviews noting the prevalence of visual system injury in animal models of TBI, coupled with clinical estimates of 50-75% of all TBI cases, we decided to assess commonalities, if they existed, through visual system injury. A unilateral (left directed) blast and repeat blast model injury with coup-contra-coup injury patterns were compared to a midline blunt injury. Injuries were induced in adult male mice to observe and quantify visual deficits. Retinal ganglion cell loss and axonal degeneration in the optic tract, superior colliculus, and lateral geniculate nuclei were examined to trace injury outcomes throughout major vision-associated areas. Optokinetic response, immunohistochemistry, and western blots were analyzed. Where a single blunt injury produces significant visual deficits a single blast injury appears to have less severe visual consequences. Visual deficits after repeat blasts are similar to a single blast. Single blast injury induces contralateral damage to the right optic chiasm and tract whereas bilateral injury follows a single blunt TBI. Repeat blast injuries are required to see degeneration patterns in downstream regions similar to the damage seen in a single blunt injury. This finding is further supported by amyloid precursor protein (APP) staining in injured cohorts. Blunt injured groups present with staining 1.2 mm ahead of the optic nerve, indicating axonal breakage closer to the optic chiasm. In blast groups, APP was identifiable in a bilateral pattern only in the geniculate nucleus. Evidence for unilateral neuronal degeneration in brain tissue with bilateral axonal ruptures are pivotal discoveries in this model differentiation. Analysis of the two injury models suggests that there is a significant difference in the histological outcomes dependent on injury type, though visual system injury is likely present in more cases than are currently diagnosed clinically.
Collapse
Affiliation(s)
- S M Hetzer
- Neuroscience Graduate Program, University of Cincinnati College of Medicine, United States of America.
| | - C O'Connell
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, United States of America
| | - V Lallo
- College of Arts and Sciences, University of Cincinnati, United States of America
| | - M Robson
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, United States of America
| | - N K Evanson
- Department of Pediatrics, University of Cincinnati College of Medicine, United States of America; Division of Pediatric Rehabilitation Medicine, Cincinnati Children's Hospital Medical Center, United States of America
| |
Collapse
|
12
|
Mokady D, Charish J, Barretto-Burns P, Grisé KN, Coles BLK, Raab S, Ortin-Martinez A, Müller A, Fasching B, Jain P, Drukker M, van der Kooy D, Steger M. Small-Molecule-Directed Endogenous Regeneration of Visual Function in a Mammalian Retinal Degeneration Model. Int J Mol Sci 2024; 25:1521. [PMID: 38338800 PMCID: PMC10855388 DOI: 10.3390/ijms25031521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Degenerative retinal diseases associated with photoreceptor loss are a leading cause of visual impairment worldwide, with limited treatment options. Phenotypic profiling coupled with medicinal chemistry were used to develop a small molecule with proliferative effects on retinal stem/progenitor cells, as assessed in vitro in a neurosphere assay and in vivo by measuring Msx1-positive ciliary body cell proliferation. The compound was identified as having kinase inhibitory activity and was subjected to cellular pathway analysis in non-retinal human primary cell systems. When tested in a disease-relevant murine model of adult retinal degeneration (MNU-induced retinal degeneration), we observed that four repeat intravitreal injections of the compound improved the thickness of the outer nuclear layer along with the regeneration of the visual function, as measured with ERG, visual acuity, and contrast sensitivity tests. This serves as a proof of concept for the use of a small molecule to promote endogenous regeneration in the eye.
Collapse
Affiliation(s)
- Daphna Mokady
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Jason Charish
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | | | - Kenneth N. Grisé
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Brenda L. K. Coles
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Susanne Raab
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Arturo Ortin-Martinez
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Alex Müller
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Bernhard Fasching
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Payal Jain
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Micha Drukker
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Matthias Steger
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| |
Collapse
|
13
|
Chou TH, Hao Z, Alba D, Lazo A, Gallo Afflitto G, Eastwood JD, Porciatti V, Guy J, Yu H. Mitochondrially Targeted Gene Therapy Rescues Visual Loss in a Mouse Model of Leber's Hereditary Optic Neuropathy. Int J Mol Sci 2023; 24:17068. [PMID: 38069388 PMCID: PMC10707051 DOI: 10.3390/ijms242317068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/26/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Leber's hereditary optic neuropathy (LHON) is a common mitochondrial genetic disease, causing irreversible blindness in young individuals. Current treatments are inadequate, and there is no definitive cure. This study evaluates the effectiveness of delivering wildtype human NADH ubiquinone oxidoreductase subunit 4 (hND4) gene using mito-targeted AAV(MTSAAV) to rescue LHOH mice. We observed a declining pattern in electroretinograms amplitudes as mice aged across all groups (p < 0.001), with significant differences among groups (p = 0.023; Control vs. LHON, p = 0.008; Control vs. Rescue, p = 0.228). Inner retinal thickness and intraocular pressure did not change significantly with age or groups. Compared to LHON mice, those rescued with wildtype hND4 exhibited improved retinal visual acuity (0.29 ± 0.1 cy/deg vs. 0.15 ± 0.1 cy/deg) and increased functional hyperemia response (effect of flicker, p < 0.001, effect of Group, p = 0.004; Interaction Flicker × Group, p < 0.001). Postmortem analysis shows a marked reduction in retinal ganglion cell density in the LHON group compared to the other groups (Effect of Group, p < 0.001, Control vs. LHON, p < 0.001, Control vs. Rescue, p = 0.106). These results suggest that MTSAAV-delivered wildtype hND4 gene rescues, at least in part, visual impairment in an LHON mouse model and has the therapeutic potential to treat this disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (T.-H.C.); (Z.H.); (D.A.); (A.L.); (G.G.A.); (J.D.E.); (J.G.)
| | | | - Hong Yu
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (T.-H.C.); (Z.H.); (D.A.); (A.L.); (G.G.A.); (J.D.E.); (J.G.)
| |
Collapse
|
14
|
Tai WL, Cho KS, Kriukov E, Ashok A, Wang X, Monavarfeshani A, Yan W, Li Y, Guan T, Sanes JR, Baranov P, Chen DF. Suppressing DNMT3a Alleviates the Intrinsic Epigenetic Barrier for Optic Nerve Regeneration and Restores Vision in Adult Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567614. [PMID: 38014168 PMCID: PMC10680854 DOI: 10.1101/2023.11.17.567614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The limited regenerative potential of the optic nerve in adult mammals presents a major challenge for restoring vision after optic nerve trauma or disease. The mechanisms of this regenerative failure are not fully understood1,2. Here, through small-molecule and genetic screening for epigenetic modulators3, we identify DNA methyltransferase 3a (DNMT3a) as a potent inhibitor of axon regeneration in mouse and human retinal explants. Selective suppression of DNMT3a in retinal ganglion cells (RGCs) by gene targeting or delivery of shRNA leads to robust, full-length regeneration of RGC axons through the optic nerve and restoration of vision in adult mice after nerve crush injury. Genome-wide bisulfite and transcriptome profiling in combination with single nucleus RNA-sequencing of RGCs revealed selective DNA demethylation and reactivation of genetic programs supporting neuronal survival and axonal growth/regeneration by DNMT3a deficiency. This was accompanied by the suppression of gene networks associated with apoptosis and inflammation. Our results identify DNMT3a as the central orchestrator of an RGC-intrinsic mechanism that limits optic nerve regeneration. Suppressing DNMT3a expression in RGCs unlocks the epigenetic switch for optic nerve regeneration and presents a promising therapeutic avenue for effectively reversing vision loss resulted from optic nerve trauma or diseases.
Collapse
Affiliation(s)
- Wai Lydia Tai
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Emil Kriukov
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Ajay Ashok
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Xuejian Wang
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China
| | - Aboozar Monavarfeshani
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA, USA
| | - Wenjun Yan
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA, USA
| | - Yingqian Li
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Timothy Guan
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Joshua R Sanes
- Department of Cellular and Molecular Biology, Center for Brain Science, Harvard University, MA, USA
| | - Petr Baranov
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dong Feng Chen
- Schepens Eye Research Institute of Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Hassan S, Hsu Y, Mayer SK, Thomas J, Kothapalli A, Helms M, Baker SA, Laird JG, Bhattarai S, Drack AV. A visually guided swim assay for mouse models of human retinal disease recapitulates the multi-luminance mobility test in humans. Saudi J Ophthalmol 2023; 37:313-320. [PMID: 38155679 PMCID: PMC10752274 DOI: 10.4103/sjopt.sjopt_155_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 12/30/2023] Open
Abstract
PURPOSE The purpose of this study was to develop a visually guided swim assay (VGSA) for measuring vision in mouse retinal disease models comparable to the multi-luminance mobility test (MLMT) utilized in human clinical trials. METHODS Three mouse retinal disease models were studied: Bardet-Biedl syndrome type 1 (Bbs1M390R/M390R), n = 5; Bardet-Biedl syndrome type 10 (Bbs10-/-), n = 11; and X linked retinoschisis (retinoschisin knockout; Rs1-KO), n = 5. Controls were normally-sighted mice, n = 10. Eyeless Pax6Sey-Dey mice, n = 4, were used to determine the performance of animals without vision in VGSA. RESULTS Eyeless Pax6Sey-Dey mice had a VGSA time-to-platform (TTP) 7X longer than normally-sighted controls (P < 0.0001). Controls demonstrated no difference in their TTP in both lighting conditions; the same was true for Pax6Sey-Dey. At 4-6 M, Rs1-KO and Bbs10-/- had longer TTP in the dark than controls (P = 0.0156 and P = 1.23 × 10-8, respectively). At 9-11 M, both BBS models had longer TTP than controls in light and dark with times similar to Pax6Sey-Dey (P < 0.0001), demonstrating progressive vision loss in BBS models, but not in controls nor in Rs1-KO. At 1 M, Bbs10-/- ERG light-adapted (cone) amplitudes were nonrecordable, resulting in a floor effect. VGSA did not reach a floor until 9-11 M. ERG combined rod/cone b-wave amplitudes were nonrecordable in all three mutant groups at 9-11 M, but VGSA still showed differences in visual function. ERG values correlate non-linearly with VGSA, and VGSA measured the continual decline of vision. CONCLUSION ERG is no longer a useful endpoint once the nonrecordable level is reached. VGSA differentiates between different levels of vision, different ages, and different disease models even after ERG is nonrecordable, similar to the MLMT in humans.
Collapse
Affiliation(s)
- Salma Hassan
- Department of Anatomy and Cell Biology, Biomedical Science- Cell and Developmental Biology Graduate Program, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
| | - Ying Hsu
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
| | - Sara K. Mayer
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
- Interdisciplinary Genetics Program, University of Iowa, Iowa City, IA, USA
| | - Jacintha Thomas
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
| | | | - Megan Helms
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
| | - Sheila A. Baker
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Joseph G. Laird
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Sajag Bhattarai
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
| | - Arlene V. Drack
- Department of Anatomy and Cell Biology, Biomedical Science- Cell and Developmental Biology Graduate Program, Iowa City, IA, USA
- Department of Ophthalmology and Visual Sciences, IVR, Iowa City, IA, USA
- Interdisciplinary Genetics Program, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
16
|
Pan L, Cho KS, Wei X, Xu F, Lennikov A, Hu G, Tang J, Guo S, Chen J, Kriukov E, Kyle R, Elzaridi F, Jiang S, Dromel PA, Young M, Baranov P, Do CW, Williams RW, Chen J, Lu L, Chen DF. IGFBPL1 is a master driver of microglia homeostasis and resolution of neuroinflammation in glaucoma and brain tauopathy. Cell Rep 2023; 42:112889. [PMID: 37527036 PMCID: PMC10528709 DOI: 10.1016/j.celrep.2023.112889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/08/2023] [Accepted: 07/12/2023] [Indexed: 08/03/2023] Open
Abstract
Microglia shift toward an inflammatory phenotype during aging that is thought to exacerbate age-related neurodegeneration. The molecular and cellular signals that resolve neuroinflammation post-injury are largely undefined. Here, we exploit systems genetics methods based on the extended BXD murine reference family and identify IGFBPL1 as an upstream cis-regulator of microglia-specific genes to switch off inflammation. IGFBPL1 is expressed by mouse and human microglia, and higher levels of its expression resolve lipopolysaccharide-induced neuroinflammation by resetting the transcriptome signature back to a homeostatic state via IGF1R signaling. Conversely, IGFBPL1 deficiency or selective deletion of IGF1R in microglia shifts these cells to an inflammatory landscape and induces early manifestation of brain tauopathy and retinal neurodegeneration. Therapeutic administration of IGFBPL1 drives pro-homeostatic microglia and prevents glaucomatous neurodegeneration and vision loss in mice. These results identify IGFBPL1 as a master driver of the counter-inflammatory microglial modulator that presents an endogenous resolution of neuroinflammation to prevent neurodegeneration in eye and brain.
Collapse
Affiliation(s)
- Li Pan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; School of Optometry, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Xin Wei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Fuyi Xu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, School of Pharmacy, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Anton Lennikov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Guangan Hu
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jing Tang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuai Guo
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Julie Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Emil Kriukov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Robert Kyle
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Farris Elzaridi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Shuhong Jiang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Pierre A Dromel
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Michael Young
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Petr Baranov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Chi-Wai Do
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jianzhu Chen
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Ahn SY, Jung EH, Ahn H, Lee JS, Bak JH, Kim ED, Song JH, Shin HS, Jamiyansharav M, Seo KY. Automatic measurement of mouse visual acuity based on optomotor response: SKY optomotry. Lab Anim 2023; 57:412-423. [PMID: 36708198 DOI: 10.1177/00236772221148576] [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: 01/29/2023]
Abstract
In the field of visual science study using rodents, several assessment methods have been developed for measuring visual function. However, methods such as electroretinograms tests, visual evoked potentials tests and maze tests have limitations in that they measure function of only a specific type of cells, are difficult to quantify or require sufficient training time. The method which uses an optokinetic reflex and optomotor response, a compensatory eye and head movement in response to changes in the visual scene, became the most widely used method. However, this method requires highly trained experimenters and is time consuming. We showed that measured visual acuity values are significantly different between beginner and expert. Here we suggest an automated optometry program, 'SKY optomotry', which automatically tracks rodents' optomotor response to overcome subjectivity and the lengthy scoring procedure of the existing method. To evaluate the performance of SKY optomotry using 8-12-week-old C57BL/6 mice we compared the binomial decision of SKY optomotry with a skilled expert, and the area under the curve of SKY optomotry was 0.845. Comparing the final visual acuity, the intraclass correlation coefficient value between SKY optomotry and an expert was 0.860 (95% confidence interval (CI) 0.709-0.928), whereas that between an expert and a beginner was 0.642 (95% CI 0.292-0.811). SKY optomotry showed an excellent level of performance with good inter-rater agreements based on the visual acuity measured by an expert. With the use of our application, researchers will be able to test an experimental animal's eyesight more accurately while saving time on specialized training.
Collapse
Affiliation(s)
- So Yeon Ahn
- Department of Medicine, Yonsei University College of Medicine, Republic of Korea
| | - Eun Hye Jung
- Department of Medicine, Yonsei University College of Medicine, Republic of Korea
| | - Hyunmin Ahn
- Department of Medicine, Yonsei University College of Medicine, Republic of Korea
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Republic of Korea
| | - Jihei Sara Lee
- Department of Medicine, Yonsei University College of Medicine, Republic of Korea
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Republic of Korea
| | - Jeong Hyeon Bak
- Department of Mechanical Engineering, Hanyang University, Republic of Korea
| | - Eun-do Kim
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, USA
| | - Ja-Hyun Song
- Korea Mouse Sensory Phenotyping Center (KMSPC), Yonsei University College of Medicine, Republic of Korea
| | - Hae-Sol Shin
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Republic of Korea
- Korea Mouse Sensory Phenotyping Center (KMSPC), Yonsei University College of Medicine, Republic of Korea
| | | | - Kyoung Yul Seo
- Department of Medicine, Yonsei University College of Medicine, Republic of Korea
- Department of Ophthalmology, The Institute of Vision Research, Yonsei University College of Medicine, Republic of Korea
| |
Collapse
|
18
|
Wang H, Peng Z, Li Y, Sahn JJ, Hodges TR, Chou TH, Liu Q, Zhou X, Jiao S, Porciatti V, Liebl DJ, Martin SF, Wen R. σ 2R/TMEM97 in retinal ganglion cell degeneration. Sci Rep 2022; 12:20753. [PMID: 36456686 PMCID: PMC9715665 DOI: 10.1038/s41598-022-24537-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
The sigma 2 receptor (σ2R) was recently identified as an endoplasmic reticulum (ER) membrane protein known as transmembrane protein 97 (TMEM97). Studies have shown that σ2R/TMEM97 binding compounds are neuroprotective, suggesting a role of σ2R/TMEM97 in neurodegenerative processes. To understand the function of σ2R/TMEM97 in neurodegeneration pathways, we characterized ischemia-induced retinal ganglion cell (RGC) degeneration in TMEM97-/- mice and found that RGCs in TMEM97-/- mice are resistant to degeneration. In addition, intravitreal injection of a selective σ2R/TMEM97 ligand DKR-1677 significantly protects RGCs from ischemia-induced degeneration in wildtype mice. Our results provide conclusive evidence that σ2R/TMEM97 plays a role to facilitate RGC death following ischemic injury and that inhibiting the function of σ2R/TMEM97 is neuroprotective. This work is a breakthrough toward elucidating the biology and function of σ2R/TMEM97 in RGCs and likely in other σ2R/TMEM97 expressing neurons. Moreover, these findings support future studies to develop new neuroprotective approaches for RGC degenerative diseases by inhibiting σ2R/TMEM97.
Collapse
Affiliation(s)
- Hua Wang
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Zhiyou Peng
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Yiwen Li
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - James J Sahn
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Timothy R Hodges
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA
| | - Tsung-Han Chou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Qiong Liu
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Xuezhi Zhou
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Shuliang Jiao
- Department of Biomedical Engineering, Florida International University, Miami, FL, 33174, USA
| | - Vittorio Porciatti
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Daniel J Liebl
- Department of Neurosurgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Stephen F Martin
- Department of Chemistry and Biochemistry, University of Texas at Austin, Austin, TX, 78712, USA.
| | - Rong Wen
- Bascom Palmer Eye Institute, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
19
|
Mansouri V. X-Linked Retinitis Pigmentosa Gene Therapy: Preclinical Aspects. Ophthalmol Ther 2022; 12:7-34. [PMID: 36346573 PMCID: PMC9641696 DOI: 10.1007/s40123-022-00602-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
The most common inherited eye disease is retinitis pigmentosa (RP). X-linked RP (XLRP) is one of the most severe types of RP, with a considerable disease burden. Patients with XLRP experience a decrease in their vision and become blind in their 4th decade of life, causing much morbidity after starting a rather normal life. Treatment of XLRP remains challenging, and current treatments are not effective enough in restoring vision. Gene therapy of XLRP, capable of restoring the functional RPGR gene, showed promising results in preclinical studies and clinical trials; however, to date, no approved product has entered the market. The development of a gene therapy product needs through preliminary assessment of the drug in animal models before administration to humans. In this article, we reviewed the genetic pathology of XLRP, along with the preclinical aspects of the XLRP gene therapy, animal models, associated assessments, and future challenges and directions.
Collapse
Affiliation(s)
- Vahid Mansouri
- Gene Therapy Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Chen BY, Liou JC, Wu JL, Chen CH, Yang SL. Photoreceptor and vision protective effects of astragaloside IV in mice model with light-evoked retinal damage. Biomed Pharmacother 2022; 153:113404. [DOI: 10.1016/j.biopha.2022.113404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/01/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022] Open
|
21
|
Hou M, Shao Z, Zhang S, Liu X, Fan P, Jiang M, Zhao Y, Xiao R, Yuan H. Age-related visual impairments and retinal ganglion cells axonal degeneration in a mouse model harboring OPTN (E50K) mutation. Cell Death Dis 2022; 13:362. [PMID: 35436991 PMCID: PMC9016082 DOI: 10.1038/s41419-022-04836-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 11/09/2022]
Abstract
Retinal ganglion cells (RGCs) axons are the signal carriers of visual information between retina and brain. Therefore, they play one of the important roles affected in many optic neurodegenerative diseases like glaucoma. Among the genetic risks associated with glaucoma, the E50K mutation in the Optineurin (OPTN) gene are known to result in glaucoma in the absence of increased intraocular pressure (IOP), whereas the relevant pathological mechanism and neurological issues remain to be further investigated. In this study, the OPTN (E50K) mutant mouse model was established through CRISPR/Cas9-mediated genome editing, and aging-related RGCs loss and the visual dysfunction were identified. In E50K mice 16 months old, the axonal transport decreased comparing to wild-type (WT) mice at the same age. Furthermore, results of electron microscopy demonstrated significant morphological anomaly of mitochondria in RGCs axons of young E50K mice 3 months old, and these changes were aggravated with age. These indicated that the damaged mitochondria-associated dysfunction of RGCs axon should play an etiological role in glaucoma as an age-related outcome of OPTN (E50K) mutation. The findings of this study have potential implications for the targeted prevention and treatment of NTG.
Collapse
|
22
|
Niu F, Han P, Zhang J, She Y, Yang L, Yu J, Zhuang M, Tang K, Shi Y, Yang B, Liu C, Peng B, Ji SJ. The m 6A reader YTHDF2 is a negative regulator for dendrite development and maintenance of retinal ganglion cells. eLife 2022; 11:75827. [PMID: 35179492 PMCID: PMC8906807 DOI: 10.7554/elife.75827] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/16/2022] [Indexed: 11/29/2022] Open
Abstract
The precise control of growth and maintenance of the retinal ganglion cell (RGC) dendrite arborization is critical for normal visual functions in mammals. However, the underlying mechanisms remain elusive. Here, we find that the N6-methyladenosine (m6A) reader YTHDF2 is highly expressed in the mouse RGCs. Conditional knockout (cKO) of Ythdf2 in the retina leads to increased RGC dendrite branching, resulting in more synapses in the inner plexiform layer. Interestingly, the Ythdf2 cKO mice show improved visual acuity compared with control mice. We further demonstrate that Ythdf2 cKO in the retina protects RGCs from dendrite degeneration caused by the experimental acute glaucoma model. We identify the m6A-modified YTHDF2 target transcripts which mediate these effects. This study reveals mechanisms by which YTHDF2 restricts RGC dendrite development and maintenance. YTHDF2 and its target mRNAs might be valuable in developing new treatment approaches for glaucomatous eyes.
Collapse
Affiliation(s)
- Fugui Niu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Peng Han
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jian Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Yuanchu She
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Lixin Yang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Jun Yu
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Mengru Zhuang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Kezhen Tang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuwei Shi
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Baisheng Yang
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Chunqiao Liu
- Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bo Peng
- Department of Neurosurgery, Fudan University, Shanghai, China
| | - Sheng-Jian Ji
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
23
|
Markov DA, Petrucco L, Kist AM, Portugues R. A cerebellar internal model calibrates a feedback controller involved in sensorimotor control. Nat Commun 2021; 12:6694. [PMID: 34795244 PMCID: PMC8602262 DOI: 10.1038/s41467-021-26988-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/28/2021] [Indexed: 11/18/2022] Open
Abstract
Animals must adapt their behavior to survive in a changing environment. Behavioral adaptations can be evoked by two mechanisms: feedback control and internal-model-based control. Feedback controllers can maintain the sensory state of the animal at a desired level under different environmental conditions. In contrast, internal models learn the relationship between the motor output and its sensory consequences and can be used to recalibrate behaviors. Here, we present multiple unpredictable perturbations in visual feedback to larval zebrafish performing the optomotor response and show that they react to these perturbations through a feedback control mechanism. In contrast, if a perturbation is long-lasting, fish adapt their behavior by updating a cerebellum-dependent internal model. We use modelling and functional imaging to show that the neuronal requirements for these mechanisms are met in the larval zebrafish brain. Our results illustrate the role of the cerebellum in encoding internal models and how these can calibrate neuronal circuits involved in reactive behaviors depending on the interactions between animal and environment.
Collapse
Affiliation(s)
- Daniil A Markov
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
| | - Luigi Petrucco
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
- Institute of Neuroscience, Technical University of Munich, 80802, Munich, Germany
| | - Andreas M Kist
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany
- Division of Phoniatrics and Pediatric Audiology, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Ruben Portugues
- Sensorimotor Control Research Group, Max Planck Institute of Neurobiology, 82152, Martinsried, Germany.
- Institute of Neuroscience, Technical University of Munich, 80802, Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
24
|
Retinal and cortical visual acuity in a common inbred albino mouse. PLoS One 2021; 16:e0242394. [PMID: 34048428 PMCID: PMC8162811 DOI: 10.1371/journal.pone.0242394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 05/12/2021] [Indexed: 11/19/2022] Open
Abstract
While albino mice are widely used in research which includes the use of visually guided behavioral tests, information on their visual capability is scarce. We compared the spatial resolution (acuity) of albino mice (BALB/c) with that of pigmented mice (C57BL/6J). We used a high-throughput pattern electroretinogram (PERG) and pattern visual evoked potential (PVEP) method for objective assessment of retinal and cortical acuity, as well as optomotor head-tracking response/ reflex (OMR). We found that PERG, PVEP, and OMR acuities of C57BL/6J mice were all in the range of 0.5-0.6 cycles/degree (cyc/deg). BALB/c mice had PERG and PVEP acuities in the range of 0.1-0.2 cyc/deg but were unresponsive to OMR stimulus. Results indicate that retinal and cortical acuity can be reliably determined with electrophysiological methods in BALB/c mice, although PERG/PVEP acuities are lower than those of C57BL/6J mice. The reduced acuity of BALB/c mice appears to be primarily determined at retinal level.
Collapse
|
25
|
Abstract
Gamma-aminobutyric acid (GABA) is regarded as the most important inhibitory neurotransmitter in the central nervous system, including the retina. However, the roles of GABA-immunolabeled retinal ganglion cells (RGCs) have not been explored. Here, we report the expression of GABAergic RGCs that project to many brain areas in mice, including the superior colliculus. Selective ablation of the superior colliculus-projecting GABAergic RGCs, leaving other GABAergic RGCs intact, reduces the looming stimulus-induced defensive response without affecting image-forming functions; it also significantly enhances glucose metabolism in the superior colliculus, as determined by [18F]-fluorodeoxyglucose PET (FDG PET). Our findings demonstrate that superior colliculus-projecting GABAergic RGCs control the visually active defensive response by regulating superior colliculus neurons.
Collapse
Affiliation(s)
- Danrui Cai
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei
| | - Xue Luo
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei
- Division of Life Science, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Kejiong Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei
| | - Yin Shen
- Eye Center, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei
- Medical Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Ainsbury EA, Dalke C, Hamada N, Benadjaoud MA, Chumak V, Ginjaume M, Kok JL, Mancuso M, Sabatier L, Struelens L, Thariat J, Jourdain JR. Radiation-induced lens opacities: Epidemiological, clinical and experimental evidence, methodological issues, research gaps and strategy. ENVIRONMENT INTERNATIONAL 2021; 146:106213. [PMID: 33276315 DOI: 10.1016/j.envint.2020.106213] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/11/2020] [Accepted: 08/25/2020] [Indexed: 06/12/2023]
Abstract
In 2011, the International Commission on Radiological Protection (ICRP) recommended reducing the occupational equivalent dose limit for the lens of the eye from 150 mSv/year to 20 mSv/year, averaged over five years, with no single year exceeding 50 mSv. With this recommendation, several important assumptions were made, such as lack of dose rate effect, classification of cataracts as a tissue reaction with a dose threshold at 0.5 Gy, and progression of minor opacities into vision-impairing cataracts. However, although new dose thresholds and occupational dose limits have been set for radiation-induced cataract, ICRP clearly states that the recommendations are chiefly based on epidemiological evidence because there are a very small number of studies that provide explicit biological and mechanistic evidence at doses under 2 Gy. Since the release of the 2011 ICRP statement, the Multidisciplinary European Low Dose Initiative (MELODI) supported in April 2019 a scientific workshop that aimed to review epidemiological, clinical and biological evidence for radiation-induced cataracts. The purpose of this article is to present and discuss recent related epidemiological and clinical studies, ophthalmic examination techniques, biological and mechanistic knowledge, and to identify research gaps, towards the implementation of a research strategy for future studies on radiation-induced lens opacities. The authors recommend particularly to study the effect of ionizing radiation on the lens in the context of the wider, systemic effects, including in the retina, brain and other organs, and as such cataract is recommended to be studied as part of larger scale programs focused on multiple radiation health effects.
Collapse
Affiliation(s)
- Elizabeth A Ainsbury
- Public Health England (PHE) Centre for Radiation, Chemical and Environmental Hazards, Oxon, United Kingdom.
| | - Claudia Dalke
- Helmholtz Zentrum München GmbH, German Research Center for Environmental Health, Germany.
| | - Nobuyuki Hamada
- Radiation Safety Research Center, Nuclear Technology Research Laboratory, Central Research Institute of Electric Power Industry (CRIEPI), Tokyo, Japan.
| | - Mohamed Amine Benadjaoud
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), BP 17, 31 avenue de la division Leclerc, Fontenay-aux-Roses, France.
| | - Vadim Chumak
- National Research Centre for Radiation Medicine, Ukraine.
| | | | - Judith L Kok
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Italian National Agency for New Technologies, Energy and Sustainable Economic Development, (ENEA), Rome, Italy.
| | - Laure Sabatier
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Saclay, France.
| | | | - Juliette Thariat
- Laboratoire de physique corpusculaire IN2P3/ENSICAEN -UMR6534 - Unicaen - Normandie University, France
| | - Jean-René Jourdain
- Institut de Radioprotection et de Sûreté Nucléaire (IRSN), BP 17, 31 avenue de la division Leclerc, Fontenay-aux-Roses, France.
| |
Collapse
|
27
|
Rabiee B, Anwar KN, Shen X, Putra I, Liu M, Jung R, Afsharkhamseh N, Rosenblatt MI, Fishman GA, Liu X, Ghassemi M, Djalilian AR. Gene dosage manipulation alleviates manifestations of hereditary PAX6 haploinsufficiency in mice. Sci Transl Med 2020; 12:eaaz4894. [PMID: 33298563 DOI: 10.1126/scitranslmed.aaz4894] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 04/16/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
In autosomal dominant conditions with haploinsufficiency, a single functional allele cannot maintain sufficient dosage for normal function. We hypothesized that pharmacologic induction of the wild-type allele could lead to gene dosage compensation and mitigation of the disease manifestations. The paired box 6 (PAX6) gene is crucial in tissue development and maintenance particularly in eye, brain, and pancreas. Aniridia is a panocular condition with impaired eye development and limited vision due to PAX6 haploinsufficiency. To test our hypothesis, we performed a chemical screen and found mitogen-activated protein kinase kinase (MEK) inhibitors to induce PAX6 expression in normal and mutant corneal cells. Treatment of newborn Pax6-deficient mice (Pax6Sey-Neu/+ ) with topical or systemic MEK inhibitor PD0325901 led to increased corneal PAX6 expression, improved corneal morphology, reduced corneal opacity, and enhanced ocular function. These results suggest that induction of the wild-type allele by drug repurposing is a potential therapeutic strategy for haploinsufficiencies, which is not limited to specific mutations.
Collapse
Affiliation(s)
- Behnam Rabiee
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Khandaker N Anwar
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Xiang Shen
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ilham Putra
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mingna Liu
- Departments of Biology and Psychology, University of Virginia, Charlottesville, VA 22903, USA
| | - Rebecca Jung
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Neda Afsharkhamseh
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Gerald A Fishman
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
- Pangere Center for Inherited Retinal Diseases, The Chicago Lighthouse, Chicago, IL 60608, USA
| | - Xiaorong Liu
- Departments of Biology and Psychology, University of Virginia, Charlottesville, VA 22903, USA
| | - Mahmood Ghassemi
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
28
|
Tong M, Yu X, Shao J, Shao Z, Li W, Lin W. Automated measuring method based on Machine learning for optomotor response in mice. Neurocomputing 2020. [DOI: 10.1016/j.neucom.2020.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
29
|
Tang Y, Xiao Z, Pan L, Zhuang D, Cho KS, Robert K, Chen X, Shu L, Tang G, Wu J, Sun X, Chen DF. Therapeutic Targeting of Retinal Immune Microenvironment With CSF-1 Receptor Antibody Promotes Visual Function Recovery After Ischemic Optic Neuropathy. Front Immunol 2020; 11:585918. [PMID: 33281816 PMCID: PMC7691249 DOI: 10.3389/fimmu.2020.585918] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/13/2020] [Indexed: 01/02/2023] Open
Abstract
Retinal ischemia/reperfusion injury (RI) is a common cause of irreversible visual impairment and blindness in elderly and critical unmet medical need. While no effective treatment is available for RI, microglial activation and local immune responses in the retina are thought to play important roles in the pathophysiology of neurodegeneration. While survival and activation of microglia depend critically on colony-stimulating factor receptor (CSF-1R) signaling, it remains unclear if targeting the retinal immune microenvironments by CSF-1RAb after RI is sufficient to rescue vision and present a potentially effective therapy. Here we used rodent models of RI and showed that retinal ischemia induced by acute elevation of intraocular pressure triggered an early activation of microglia and macrophages in the retina within 12 h. This was followed by lymphocyte infiltration and increased production of pro-inflammatory cytokines. Intravitreal injection of CSF-1R neutralizing antibody (CSF-1RAb) after RI significantly blocked microglial activation and the subsequent T cell recruitment. This also led to improved retinal ganglion cell survival and function measured by cell quantification and electroretinogram positive scotopic threshold responses, as well as increased visual acuity and contrast sensitivity as assessed by optomotor reflex-based assays, when compared to the isotype-treated control group. Moreover, the administration of CSF-1RAb efficiently attenuated inflammatory responses and activation of human microglia in culture, suggesting a therapeutic target with human relevance. These results, together with the existing clinical safety profiles, support that CSF-1RAb may present a promising therapeutic avenue for RI, a currently untreatable condition, by targeting microglia and the immune microenvironment in the retina to facilitate neural survival and visual function recovery.
Collapse
Affiliation(s)
- Yizhen Tang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Zebin Xiao
- Department of Radiology, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Li Pan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
- School of Optometry, The Hong Kong Polytechnic University, Hong Kong, Hong Kong
| | - Dongli Zhuang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Kyle Robert
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Xiaoxiao Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Lian Shu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Guangxian Tang
- Department of Ophthalmology, 1st Hospital of Shijiazhuang, Shijiazhuang, China
| | - Jihong Wu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Dong F. Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
30
|
Gudapati K, Singh A, Clarkson-Townsend D, Feola AJ, Allen RS. Behavioral Assessment of Visual Function via Optomotor Response and Cognitive Function via Y-Maze in Diabetic Rats. J Vis Exp 2020. [PMID: 33165321 DOI: 10.3791/61806] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The optomotor response and the Y-maze are behavioral tests useful for assessing visual and cognitive function, respectively. The optomotor response is a valuable tool to track changes in spatial frequency (SF) and contrast sensitivity (CS) thresholds over time in a number of retinal disease models, including diabetic retinopathy. Similarly, the Y-maze can be used to monitor spatial cognition (as measured by spontaneous alternation) and exploratory behavior (as measured by a number of entries) in a number of disease models that affect the central nervous system. Advantages of the optomotor response and the Y-maze include sensitivity, speed of testing, the use of innate responses (training is not needed), and the ability to be performed on awake (non-anesthetized) animals. Here, protocols are described for both the optomotor response and the Y-maze and examples of their use shown in models of Type I and Type II diabetes. Methods include preparation of rodents and equipment, performance of the optomotor response and the Y-maze, and post-test data analysis.
Collapse
Affiliation(s)
- Kaavya Gudapati
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology
| | - Anayesha Singh
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Neuroscience, Emory University
| | - Danielle Clarkson-Townsend
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Gangarosa Department of Environmental Health, Emory University
| | - Andrew J Feola
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology;
| |
Collapse
|
31
|
Xiao J, Adil MY, Chang K, Yu Z, Yang L, Utheim TP, Chen DF, Cho KS. Visual Contrast Sensitivity Correlates to the Retinal Degeneration in Rhodopsin Knockout Mice. Invest Ophthalmol Vis Sci 2020; 60:4196-4204. [PMID: 31618423 PMCID: PMC6795341 DOI: 10.1167/iovs.19-26966] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Clinical manifestations of photoreceptor degeneration include gradual thinning of the outer nuclear layer (ONL) and progressive reduction of electroretinogram (ERG) amplitudes and vision loss. Although preclinical evaluations of treatment strategies greatly depend on rodent models, the courses of these changes in mice remain unclear. We thus sought to investigate the temporal correlations in changes of spatial vision, ERG response, and ONL thickness in mice with progressive photoreceptor degeneration. Methods Adult wild-type (WT) mice and mice carrying rhodopsin deficiency (Rho−/−), a frequently used mouse model of human retinitis pigmentosa, were selected for investigation. Mouse spatial vision, including visual acuity (VA) and contrast sensitivity (CS), was determined using optomotor response (OMR) assays; ONL thickness was quantified by spectral-domain optical coherence tomography (SD-OCT), and ERG was performed to evaluate retinal functions. The mice were killed when they were 14 weeks old, and the cone photoreceptors in retinal sections were counted. Results Spatial vision, ONL thickness, and ERG amplitudes remained stable in WT mice at all examined time points. While 6-week-old Rho−/− mice had VA, CS, as well as ERG responses similar to those of WT mice, progressive reductions in the spatial vision and retinal functions were recorded thereafter. Most tested 12-week-old Rho−/− mice had no visual-evoked OMR and ERG responses. Moreover, CS, but not VA, displayed a linear decline that was closely associated with ONL thinning, reduction of ERG amplitudes, and loss of cones. Conclusions We presented a comprehensive study of the relation between the changes of spatial vision, retinal function, and ONL thickness in postnatal week (PW)6 to PW12 Rho−/− mice. CS is a more sensitive indicator of spatial vision compared to VA, although both are required as separate parameters for monitoring the visual changes in retina undergoing photoreceptor degeneration.
Collapse
Affiliation(s)
- Jiaxin Xiao
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Muhammed Yasin Adil
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karen Chang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,National Taiwan University, Taiwan
| | - Zicheng Yu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Lanbo Yang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Tor P Utheim
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Norway
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Kin-Sang Cho
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States.,Geriatric Research Education and Clinical Center, Office of Research and Development, Edith Nourse Rogers Memorial Veterans Hospital, Bedford, Massachusetts, United States
| |
Collapse
|
32
|
Optomotor Swimming in Larval Zebrafish Is Driven by Global Whole-Field Visual Motion and Local Light-Dark Transitions. Cell Rep 2019; 29:659-670.e3. [DOI: 10.1016/j.celrep.2019.09.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/22/2019] [Accepted: 09/08/2019] [Indexed: 01/28/2023] Open
|
33
|
Berry MH, Holt A, Salari A, Veit J, Visel M, Levitz J, Aghi K, Gaub BM, Sivyer B, Flannery JG, Isacoff EY. Restoration of high-sensitivity and adapting vision with a cone opsin. Nat Commun 2019; 10:1221. [PMID: 30874546 PMCID: PMC6420663 DOI: 10.1038/s41467-019-09124-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 02/20/2019] [Indexed: 01/27/2023] Open
Abstract
Inherited and age-related retinal degenerative diseases cause progressive loss of rod and cone photoreceptors, leading to blindness, but spare downstream retinal neurons, which can be targeted for optogenetic therapy. However, optogenetic approaches have been limited by either low light sensitivity or slow kinetics, and lack adaptation to changes in ambient light, and not been shown to restore object vision. We find that the vertebrate medium wavelength cone opsin (MW-opsin) overcomes these limitations and supports vision in dim light. MW-opsin enables an otherwise blind retinitis pigmenotosa mouse to discriminate temporal and spatial light patterns displayed on a standard LCD computer tablet, displays adaption to changes in ambient light, and restores open-field novel object exploration under incidental room light. By contrast, rhodopsin, which is similar in sensitivity but slower in light response and has greater rundown, fails these tests. Thus, MW-opsin provides the speed, sensitivity and adaptation needed to restore patterned vision.
Collapse
Affiliation(s)
- Michael H Berry
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Department of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR, 97239, USA
| | - Amy Holt
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Autoosa Salari
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Julia Veit
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
| | - Meike Visel
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Joshua Levitz
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Department of Biochemistry, Weill Cornell Medicine, New York, NY, 10024, USA
| | - Krisha Aghi
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
| | - Benjamin M Gaub
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
- Department of Biosystems Science Engineering, ETH Zürich, Mattenstrasse 26, Basel, 8092, Switzerland
| | - Benjamin Sivyer
- Department of Physiology and Pharmacology, Oregon Health and Sciences University, Portland, OR, 97239, USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - John G Flannery
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
- School of Optometry, University of California, Berkeley, CA, 94720, USA
| | - Ehud Y Isacoff
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA.
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA.
- Bioscience Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
34
|
Gasparini SJ, Llonch S, Borsch O, Ader M. Transplantation of photoreceptors into the degenerative retina: Current state and future perspectives. Prog Retin Eye Res 2018; 69:1-37. [PMID: 30445193 DOI: 10.1016/j.preteyeres.2018.11.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022]
Abstract
The mammalian retina displays no intrinsic regenerative capacities, therefore retinal degenerative diseases such as age-related macular degeneration (AMD) or retinitis pigmentosa (RP) result in a permanent loss of the light-sensing photoreceptor cells. The degeneration of photoreceptors leads to vision impairment and, in later stages, complete blindness. Several therapeutic strategies have been developed to slow down or prevent further retinal degeneration, however a definitive cure i.e. replacement of the lost photoreceptors, has not yet been established. Cell-based treatment approaches, by means of photoreceptor transplantation, have been studied in pre-clinical animal models over the last three decades. The introduction of pluripotent stem cell-derived retinal organoids represents, in principle, an unlimited source for the generation of transplantable human photoreceptors. However, safety, immunological and reproducibility-related issues regarding the use of such cells still need to be solved. Moreover, the recent finding of cytoplasmic material transfer between donor and host photoreceptors demands reinterpretation of several former transplantation studies. At the same time, material transfer between healthy donor and dysfunctional patient photoreceptors also offers a potential alternative strategy for therapeutic intervention. In this review we discuss the history and current state of photoreceptor transplantation, the techniques used to assess rescue of visual function, the prerequisites for effective transplantation as well as the main roadblocks, including safety and immune response to the graft, that need to be overcome for successful clinical translation of photoreceptor transplantation approaches.
Collapse
Affiliation(s)
- Sylvia J Gasparini
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Sílvia Llonch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Oliver Borsch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany.
| |
Collapse
|