1
|
Soucy JR, Malechka VV, Baranov P. Retinal Ganglion Cell Transplantation: Differentiation and Quantification Strategies. Methods Mol Biol 2025; 2858:173-190. [PMID: 39433676 DOI: 10.1007/978-1-0716-4140-8_15] [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
Glaucoma is a leading cause of irreversible blindness worldwide. Current treatments focus on reducing intraocular pressure but cannot restore lost visual function once it is lost due to retinal ganglion cell (RGC) degeneration and death. Recent advances suggest that transplantation of stem cell-derived RGCs could offer new therapeutic approaches for glaucoma and vision restoration. Here, we present a detailed protocol for differentiating human RGCs from embryonic stem cells using both three-dimensional retinal organoid and two-dimensional culture approaches. Following differentiation, we describe methods for isolating and purifying retinal ganglion cells from these cultures and their subsequent transplantation into the mouse retina, with careful monitoring and postoperative care to ensure successful integration. Finally, we describe a quantitative method for assessing transplantation outcomes involving confocal imaging of retinal flat-mounts and custom ImageJ and MATLAB scripts to map and analyze the spatial distribution of donor RGCs within the host retina. Altogether, this approach provides a robust framework for exploring RGC transplantation as a potential therapy for vision loss in glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Volha V Malechka
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA
| | - Petr Baranov
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA, USA.
| |
Collapse
|
2
|
Goldberg JL. Bringing Eye Transplant Into the Light. JAMA 2024; 332:1523-1524. [PMID: 39250112 DOI: 10.1001/jama.2024.14811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Affiliation(s)
- Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California
| |
Collapse
|
3
|
Wu KY, Dhaliwal JK, Sasitharan A, Kalevar A. Cell Therapy for Retinal Degenerative Diseases: Progress and Prospects. Pharmaceutics 2024; 16:1299. [PMID: 39458628 PMCID: PMC11510658 DOI: 10.3390/pharmaceutics16101299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) and retinitis pigmentosa (RP) are leading causes of vision loss, with AMD affecting older populations and RP being a rarer, genetically inherited condition. Both diseases result in progressive retinal degeneration, for which current treatments remain inadequate in advanced stages. This review aims to provide an overview of the retina's anatomy and physiology, elucidate the pathophysiology of AMD and RP, and evaluate emerging cell-based therapies for these conditions. Methods: A comprehensive review of the literature was conducted, focusing on cell therapy approaches, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and retinal progenitor cells. Preclinical and clinical studies were analyzed to assess therapeutic potential, with attention to mechanisms such as cell replacement, neuroprotection, and paracrine effects. Relevant challenges, including ethical concerns and clinical translation, were also explored. Results: Cell-based therapies demonstrate potential for restoring retinal function and slowing disease progression through mechanisms like neuroprotection and cell replacement. Preclinical trials show promising outcomes, but clinical studies face significant hurdles, including challenges in cell delivery and long-term efficacy. Combination therapies integrating gene editing and biomaterials offer potential future advancements. Conclusions: While cell-based therapies for AMD and RP have made significant progress, substantial barriers to clinical application remain. Further research is essential to overcome these obstacles, improve delivery methods, and ensure the safe and effective translation of these therapies into clinical practice.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Jaskarn K. Dhaliwal
- Faculty of Health Sciences, Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada
| | - Akash Sasitharan
- Faculty of Medicine and Health Sciences, Department of Medicine, McGill University, Montreal, QC H3A 0GA, Canada
| | - Ananda Kalevar
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| |
Collapse
|
4
|
Wang LH, Huang CH, Lin IC. Advances in Neuroprotection in Glaucoma: Pharmacological Strategies and Emerging Technologies. Pharmaceuticals (Basel) 2024; 17:1261. [PMID: 39458902 PMCID: PMC11510571 DOI: 10.3390/ph17101261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Glaucoma is a major global health concern and the leading cause of irreversible blindness worldwide, characterized by the progressive degeneration of retinal ganglion cells (RGCs) and their axons. This review focuses on the need for neuroprotective strategies in glaucoma management, addressing the limitations of current treatments that primarily target intraocular pressure (IOP) reduction. Despite effective IOP management, many patients continue to experience RGC degeneration, leading to irreversible blindness. This review provides an overview of both pharmacological interventions and emerging technologies aimed at directly protecting RGCs and the optic nerve, independent of IOP reduction. Pharmacological agents such as brimonidine, neurotrophic factors, memantine, Ginkgo biloba extract, citicoline, nicotinamide, insulin, and resveratrol show promise in preclinical and early clinical studies for their neuroprotective properties. Emerging technologies, including stem cell therapy, gene therapy, mitochondrial-targeted therapies, and nanotechnologies, offer innovative approaches for neuroprotection and regeneration of damaged RGCs. While these interventions hold significant potential, further research and clinical trials are necessary to confirm their efficacy and establish their role in clinical practice. This review highlights the multifaceted nature of neuroprotection in glaucoma, aiming to guide future research and clinical practice toward more effective management of glaucoma-induced neurodegeneration.
Collapse
Affiliation(s)
- Li-Hsin Wang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan;
| | - Chun-Hao Huang
- Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan;
| | - I-Chan Lin
- Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, Taipei 110301, Taiwan;
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
5
|
Chen Y, Yu J, Ge S, Jia R, Song X, Wang Y, Fan X. An Overview of Optic Pathway Glioma With Neurofibromatosis Type 1: Pathogenesis, Risk Factors, and Therapeutic Strategies. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 38837168 PMCID: PMC11160950 DOI: 10.1167/iovs.65.6.8] [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: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 06/06/2024] Open
Abstract
Optic pathway gliomas (OPGs) are most predominant pilocytic astrocytomas, which are typically diagnosed within the first decade of life. The majority of affected children with OPGs also present with neurofibromatosis type 1 (NF1), the most common tumor predisposition syndrome. OPGs in individuals with NF1 primarily affect the optic pathway and lead to visual disturbance. However, it is challenging to assess risk in asymptomatic patients without valid biomarkers. On the other hand, for symptomatic patients, there is still no effective treatment to prevent or recover vision loss. Therefore, this review summarizes current knowledge regarding the pathogenesis of NF1-associated OPGs (NF1-OPGs) from preclinical studies to seek potential prognostic markers and therapeutic targets. First, the loss of the NF1 gene activates 3 distinct Ras effector pathways, including the PI3K/AKT/mTOR pathway, the MEK/ERK pathway, and the cAMP pathway, which mediate glioma tumorigenesis. Meanwhile, non-neoplastic cells from the tumor microenvironment (microglia, T cells, neurons, etc.) also contribute to gliomagenesis via various soluble factors. Subsequently, we investigated potential genetic risk factors, molecularly targeted therapies, and neuroprotective strategies for tumor prevention and vision recovery. Last, potential directions and promising preclinical models of NF1-OPGs are presented for further research. On the whole, NF1-OPGs develop as a result of the interaction between glioma cells and the tumor microenvironment. Developing effective treatments require a better understanding of tumor molecular characteristics, as well as multistage interventions targeting both neoplastic cells and non-neoplastic cells.
Collapse
Affiliation(s)
- Ying Chen
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Jie Yu
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Shengfang Ge
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xin Song
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Yefei Wang
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| | - Xianqun Fan
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
6
|
Soucy JR, Kriukov E, Oswald J, Phay M, Masland J, Pernstich C, Baranov P. Sustained neurotrophic factor cotreatment enhances donor and host retinal ganglion cell survival in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583961. [PMID: 38558999 PMCID: PMC10979869 DOI: 10.1101/2024.03.07.583961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Retinal ganglion cells (RGCs) lack regenerative capacity in mammals, and their degeneration in glaucoma leads to irreversible blindness. The transplantation of stem cell-derived RGCs lacks clinically relevant effect due to insufficient survival and integration of donor cells. We hypothesize that the retinal microenvironment plays a critical role in this process, and we can engineer a more acceptable setting for transplantation. Since the adult mammalian retina does not have regenerative capacity, we turned to the developing human retina to reconstruct cell-cell interactions at a single-cell level. We established a human fetal retina atlas by integrating currently available single-cell RNA sequencing datasets of human fetal retinas into a unified resource. We align RGC transcriptomes in pseudotime to map RGC developmental fate trajectories against the broader timeline of retinal development. Through this analysis, we identified brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) as key factors in RGC survival, highly expressed during fetal development but significantly reduced in adulthood despite the persistence of their receptors. To demonstrate the practical application of these findings, we show that using a slow-release formulation of BDNF and GDNF enhances RGC differentiation, survival, and function in vitro and improves RGC transplantation outcomes in a mouse model. BNDF/GDNF co-treatment not only increased survival and coverage of donor RGCs within the retina but also showed neuroprotective effects on host RGCs, preserving retinal function in a model of optic neuropathy. Altogether, our findings suggest that manipulating the retinal microenvironment with slow-release neurotrophic factors holds promise in regenerative medicine for treating glaucoma and other optic neuropathies. This approach not only improves donor cell survival and integration but also provides a neuroprotective benefit to host cells, indicating a significant advancement for glaucoma therapies.
Collapse
|
7
|
Karthikeyan SK, McKendrick AM, Pai VH, Kuzhuppilly NIR, Ganeshrao SB. Contrast Discrimination and Global Form Perception in Primary Open-Angle Glaucoma and Primary Angle-Closure Glaucoma. Invest Ophthalmol Vis Sci 2024; 65:33. [PMID: 38771569 PMCID: PMC11114617 DOI: 10.1167/iovs.65.5.33] [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: 12/15/2023] [Accepted: 05/03/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose This study explored early (contrast discrimination) and intermediate (global form perception) visual processing in primary subtypes of glaucoma: primary open-angle glaucoma (POAG) and primary angle-closure glaucoma (PACG). We aimed to understand early and intermediate visual processing in POAG and PACG, matched for similar visual field defect severity. Methods Early visual processing was measured using a contrast discrimination task described by Porkorny and Smith (1997), and intermediate processing using a global form perception task using glass pattern coherence thresholds. Thresholds were determined centrally and at a single midperipheral location (12.5°) in a quadrant without visual field defects. Controls were tested in corresponding quadrants to individuals with glaucoma. Results Sixty participants (20 POAG, 20 PACG, and 20 age-matched controls), aged 50 to 77 years, were included. Visual field defects were matched between POAG and PACG, with mean deviation values of -6.53 ± 4.46 (range: -1.5 to -16.85) dB and -6.2 ± 4.24 (range: -1.37 to -16.42) dB, respectively. Two-Way ANOVA revealed significant differences in thresholds between the glaucoma groups and the control group for both contrast discrimination and global form perception tasks, with higher thresholds in the glaucoma groups. Post hoc analyses showed no significant contrast discrimination difference between POAG and PACG, but POAG had significantly higher thresholds than PACG for form perception. Conclusions In form perception, POAG showed slightly worse performance than PACG, suggesting that individuals with POAG may experience more severe functional damage than PACG of similar visual field severity.
Collapse
Affiliation(s)
- Siddharth K Karthikeyan
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Allison M McKendrick
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Australia
- Lions Eye Institute, Nedlands, WA, Australia
- Division of Optometry, School of Allied Health, University of Western Australia, Perth, Australia
| | - Vijaya H Pai
- Department of Ophthalmology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Neetha I R Kuzhuppilly
- Department of Ophthalmology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Shonraj B Ganeshrao
- Department of Optometry, Manipal College of Health Professions, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
8
|
Luo Z, Chang KC. Cell replacement with stem cell-derived retinal ganglion cells from different protocols. Neural Regen Res 2024; 19:807-810. [PMID: 37843215 PMCID: PMC10664109 DOI: 10.4103/1673-5374.381494] [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: 03/21/2023] [Revised: 05/20/2023] [Accepted: 06/13/2023] [Indexed: 10/17/2023] Open
Abstract
Glaucoma, characterized by a degenerative loss of retinal ganglion cells, is the second leading cause of blindness worldwide. There is currently no cure for vision loss in glaucoma because retinal ganglion cells do not regenerate and are not replaced after injury. Human stem cell-derived retinal ganglion cell transplant is a potential therapeutic strategy for retinal ganglion cell degenerative diseases. In this review, we first discuss a 2D protocol for retinal ganglion cell differentiation from human stem cell culture, including a rapid protocol that can generate retinal ganglion cells in less than two weeks and focus on their transplantation outcomes. Next, we discuss using 3D retinal organoids for retinal ganglion cell transplantation, comparing cell suspensions and clusters. This review provides insight into current knowledge on human stem cell-derived retinal ganglion cell differentiation and transplantation, with an impact on the field of regenerative medicine and especially retinal ganglion cell degenerative diseases such as glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
9
|
Iwanaga R, Yahagi N, Hakeda‐Suzuki S, Suzuki T. Cell adhesion and actin dynamics factors promote axonal extension and synapse formation in transplanted Drosophila photoreceptor cells. Dev Growth Differ 2024; 66:205-218. [PMID: 38403285 PMCID: PMC11457513 DOI: 10.1111/dgd.12916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/27/2024]
Abstract
Vision is formed by the transmission of light stimuli to the brain through axons extending from photoreceptor cells. Damage to these axons leads to loss of vision. Despite research on neural circuit regeneration through transplantation, achieving precise axon projection remains challenging. To achieve optic nerve regeneration by transplantation, we employed the Drosophila visual system. We previously established a transplantation method for Drosophila utilizing photoreceptor precursor cells extracted from the eye disc. However, little axonal elongation of transplanted cells into the brain, the lamina, was observed. We verified axonal elongation to the lamina by modifying the selection process for transplanted cells. Moreover, we focused on N-cadherin (Ncad), a cell adhesion factor, and Twinstar (Tsr), which has been shown to promote actin reorganization and induce axon elongation in damaged nerves. Overexpression of Ncad and tsr promoted axon elongation to the lamina, along with presynaptic structure formation in the elongating axons. Furthermore, overexpression of Neurexin-1 (Nrx-1), encoding a protein identified as a synaptic organizer, was found to not only promote presynapse formation but also enhance axon elongation. By introducing Ncad, tsr, and Nrx-1, we not only successfully achieved axonal projection of transplanted cells to the brain beyond the retina, but also confirmed the projection of transplanted cells into a deeper ganglion, the medulla. The present study offers valuable insights to realize regeneration through transplantation in a more complex nervous system.
Collapse
Affiliation(s)
- Riku Iwanaga
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Nagisa Yahagi
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| | - Satoko Hakeda‐Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
- Research Initiatives and Promotion OrganizationYokohama National UniversityYokohamaJapan
| | - Takashi Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokahamaJapan
| |
Collapse
|
10
|
Li G, Luo Y. Enriching new transplantable RGC-like cells from retinal organoids for RGC replacement therapy. Biochem Biophys Res Commun 2024; 700:149509. [PMID: 38306929 DOI: 10.1016/j.bbrc.2024.149509] [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: 12/05/2023] [Revised: 12/16/2023] [Accepted: 01/08/2024] [Indexed: 02/04/2024]
Abstract
Optic neuropathies, such as glaucoma, are due to progressive retinal ganglion cells (RGCs) degeneration, result in irreversible vision loss. The promising RGCs replacement therapy for restoring vision are impeded by insufficient RGC-like cells sources. The present work was enriched one new type RGC-like cells using two surface markers CD184 and CD171 from human induced pluripotent stem cells (hiPSCs) by FACS sorting firstly. These new kind cells have well proliferation ability and possessed passage tolerance in vitro 2D or 3D spheroids culture, which kept expressing Pax6, Brn3b and βIII-Tubulin and so on. The transplanted CD184+CD171+ RGC-like cells could survive and integrate into the normal and optic nerve crush (ONC) mice retina, especially they were more inclined to across the optic nerve head and extend to the damaged optic nerve. These data support the feasible application for cell replacement therapy in RGC degenerative diseases, as well as help to develop new commercial cells sorting reagents and establish good manufacturing practice (GMP) grade RGC-like donor cells for further clinical application.
Collapse
Affiliation(s)
- Guilan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Yuanting Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| |
Collapse
|
11
|
Xu Z, Guo Y, Xiang K, Xiao D, Xiang M. Rapid and efficient generation of a transplantable population of functional retinal ganglion cells from fibroblasts. Cell Prolif 2024; 57:e13550. [PMID: 37740641 PMCID: PMC10849786 DOI: 10.1111/cpr.13550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/24/2023] Open
Abstract
Glaucoma and other optic neuropathies lead to progressive and irreversible vision loss by damaging retinal ganglion cells (RGCs) and their axons. Cell replacement therapy is a potential promising treatment. However, current methods to obtain RGCs have inherent limitations, including time-consuming procedures, inefficient yields and complex protocols, which hinder their practical application. Here, we have developed a straightforward, rapid and efficient approach for directly inducing RGCs from mouse embryonic fibroblasts (MEFs) using a combination of triple transcription factors (TFs): ASCL1, BRN3B and PAX6 (ABP). We showed that on the 6th day following ABP induction, neurons with molecular characteristics of RGCs were observed, and more than 60% of induced neurons became iRGCs (induced retinal ganglion cells) in the end. Transplanted iRGCs had the ability to survive and appropriately integrate into the RGC layer of mouse retinal explants and N-methyl-D-aspartic acid (NMDA)-damaged retinas. Moreover, they exhibited electrophysiological properties typical of RGCs, and were able to regrow dendrites and axons and form synaptic connections with host retinal cells. Together, we have established a rapid and efficient approach to acquire functional RGCs for potential cell replacement therapy to treat glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Zihui Xu
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Yanan Guo
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Kangjian Xiang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Dongchang Xiao
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
| | - Mengqing Xiang
- State Key Laboratory of OphthalmologyZhongshan Ophthalmic Center, Sun Yat‐sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual ScienceGuangzhouChina
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
12
|
Esposito EP, Han IC, Johnson TV. Gene and cell-based therapies for retinal and optic nerve disease. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:243-262. [PMID: 39341657 DOI: 10.1016/b978-0-323-90120-8.00016-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Leading causes of blindness worldwide include neurodegenerative diseases of the retina, which cause irreversible loss of retinal pigment epithelium (RPE) and photoreceptors, and optic neuropathies, which result in retinal ganglion cell (RGC) death. Because photoreceptor and RGCs do not spontaneously regenerate in mammals, including humans, vision loss from these conditions is, at present, permanent. Recent advances in gene and cell-based therapies have provided new hope to patients affected by these conditions. This chapter reviews the current state and future of these approaches to treating ocular neurodegenerative disease. Gene therapies for retinal degeneration and optic neuropathies primarily focus on correcting known pathogenic mutations that cause inherited conditions to halt progression. There are multiple retinal and optic neuropathy gene therapies in clinical trials, and one retinal gene therapy is approved in the United States, Canada, Europe, and Australia. Cell-based therapies are mutation agnostic and have the potential to repopulate neurons regardless of the underlying etiology of degeneration. While photoreceptor cell replacement is nearing a human clinical trial, RPE transplantation is currently in phase I/II clinical trials. RGC replacement faces numerous logistical challenges, but preclinical research has laid the foundation for functional repair of optic neuropathies to be feasible.
Collapse
Affiliation(s)
- Edward P Esposito
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ian C Han
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Thomas V Johnson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
13
|
Zueva MV, Neroeva NV, Zhuravleva AN, Bogolepova AN, Kotelin VV, Fadeev DV, Tsapenko IV. Fractal Phototherapy in Maximizing Retina and Brain Plasticity. ADVANCES IN NEUROBIOLOGY 2024; 36:585-637. [PMID: 38468055 DOI: 10.1007/978-3-031-47606-8_31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
The neuroplasticity potential is reduced with aging and impairs during neurodegenerative diseases and brain and visual system injuries. This limits the brain's capacity to repair the structure and dynamics of its activity after lesions. Maximization of neuroplasticity is necessary to provide the maximal CNS response to therapeutic intervention and adaptive reorganization of neuronal networks in patients with degenerative pathology and traumatic injury to restore the functional activity of the brain and retina.Considering the fractal geometry and dynamics of the healthy brain and the loss of fractality in neurodegenerative pathology, we suggest that the application of self-similar visual signals with a fractal temporal structure in the stimulation therapy can reactivate the adaptive neuroplasticity and enhance the effectiveness of neurorehabilitation. This proposition was tested in the recent studies. Patients with glaucoma had a statistically significant positive effect of fractal photic therapy on light sensitivity and the perimetric MD index, which shows that methods of fractal stimulation can be a novel nonpharmacological approach to neuroprotective therapy and neurorehabilitation. In healthy rabbits, it was demonstrated that a long-term course of photostimulation with fractal signals does not harm the electroretinogram (ERG) and retina structure. Rabbits with modeled retinal atrophy showed better dynamics of the ERG restoration during daily stimulation therapy for a week in comparison with the controls. Positive changes in the retinal function can indirectly suggest the activation of its adaptive plasticity and the high potential of stimulation therapy with fractal visual stimuli in a nonpharmacological neurorehabilitation, which requires further study.
Collapse
Affiliation(s)
- Marina V Zueva
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Natalia V Neroeva
- Department of Pathology of the Retina and Optic Nerve, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Anastasia N Zhuravleva
- Department of Glaucoma, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Anna N Bogolepova
- Department of neurology, neurosurgery and medical genetics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vladislav V Kotelin
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Denis V Fadeev
- Scientific Experimental Center Department, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| | - Irina V Tsapenko
- Department of Clinical Physiology of Vision, Helmholtz National Medical Research Center of Eye Diseases, Moscow, Russia
| |
Collapse
|
14
|
Johnson TV, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Zack DJ. The Retinal Ganglion Cell Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration Consortium. OPHTHALMOLOGY SCIENCE 2023; 3:100390. [PMID: 38025164 PMCID: PMC10630665 DOI: 10.1016/j.xops.2023.100390] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/24/2023] [Accepted: 08/18/2023] [Indexed: 12/01/2023]
Abstract
Purpose The Retinal Ganglion Cell (RGC) Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) consortium was founded in 2021 to help address the numerous scientific and clinical obstacles that impede development of vision-restorative treatments for patients with optic neuropathies. The goals of the RReSTORe consortium are: (1) to define and prioritize the most critical challenges and questions related to RGC regeneration; (2) to brainstorm innovative tools and experimental approaches to meet these challenges; and (3) to foster opportunities for collaborative scientific research among diverse investigators. Design and Participants The RReSTORe consortium currently includes > 220 members spanning all career stages worldwide and is directed by an organizing committee comprised of 15 leading scientists and physician-scientists of diverse backgrounds. Methods Herein, we describe the structure and organization of the RReSTORe consortium, its activities to date, and the perceived impact that the consortium has had on the field based on a survey of participants. Results In addition to helping propel the field of regenerative medicine as applied to optic neuropathies, the RReSTORe consortium serves as a framework for developing large collaborative groups aimed at tackling audacious goals that may be expanded beyond ophthalmology and vision science. Conclusions The development of innovative interventions capable of restoring vision for patients suffering from optic neuropathy would be transformative for the ophthalmology field, and may set the stage for functional restoration in other central nervous system disorders. By coordinating large-scale, international collaborations among scientists with diverse and complementary expertise, we are confident that the RReSTORe consortium will help to accelerate the field toward clinical translation. Financial Disclosures Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Thomas V. Johnson
- Wilmer Eye Institute and Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Petr Baranov
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, Maryland
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada, Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, Oregon
| | | | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, California
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Alex L. Kolodkin
- Solomon H Snyder Department of Neuroscience and Department of Molecular Biology & Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carol A. Mason
- Zuckerman Mind Brain Behavior Institute, Department of Neuroscience, Department of Pathology & Cell Biology, and Department of Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, New York
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, California
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, Washington
| | - Ahmara G. Ross
- Scheie Eye Institute, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brian C. Samuels
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Donald J. Zack
- Departments of Ophthalmology (Wilmer Eye Institute), Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
15
|
Do JL, Pedroarena-Leal N, Louie M, Avila Garcia P, Alnihmy A, Patel A, Weinreb RN, Wahlin KJ, La Torre Vila A, Welsbie DS. Mechanical Disruption of the Inner Limiting Membrane In Vivo Enhances Targeting to the Inner Retina. Invest Ophthalmol Vis Sci 2023; 64:25. [PMID: 38117244 PMCID: PMC10741092 DOI: 10.1167/iovs.64.15.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/19/2023] [Indexed: 12/21/2023] Open
Abstract
Purpose To evaluate the effects of mechanical disruption of the inner limiting membrane (ILM) on the ability to target interventions to the inner neurosensory retina in a rodent model. Our study used an animal model to gain insight into the normal physiology of the ILM and advances our understanding of the effects of mechanical ILM removal on the viral transduction of retinal ganglion cells and retinal ganglion cell transplantation. Methods The ILM in the in vivo rat eye was disrupted using mechanical forces applied to the vitreoretinal interface. Immunohistology and electron microscopy were used to verify the removal of the ILM in retina flatmounts and sections. To assess the degree to which ILM disruption enhanced transvitreal access to the retina, in vivo studies involving intravitreal injections of adeno-associated virus (AAV) to transduce retinal ganglion cells (RGCs) and ex vivo studies involving co-culture of human stem cell-derived RGCs (hRGCs) on retinal explants were performed. RGC transduction efficiency and transplanted hRGC integration with retinal explants were evaluated by immunohistology of the retinas. Results Mechanical disruption of the ILM in the rodent eye was sufficient to remove the ILM from targeted retinal areas while preserving the underlying retinal nerve fiber layer and RGCs. Removal of the ILM enhanced the transduction efficiency of intravitreally delivered AAV threefold (1380.0 ± 290.1 vs. 442.0 ± 249.3 cells/mm2; N = 6; P = 0.034). Removal of the ILM was also sufficient to promote integration of transplanted RGCs within the inner retina. Conclusions The ILM is a barrier to transvitreally delivered agents including viral vectors and cells. Mechanical removal of the ILM is sufficient to enhance access to the inner retina, improve viral transduction efficiencies of RGCs, and enhance cellular integration of transplanted RGCs with the retina.
Collapse
Affiliation(s)
- Jiun L. Do
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Nicole Pedroarena-Leal
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Mikaela Louie
- Department of Cell Biology and Human Anatomy, University of California Davis, California, United States
| | - Paula Avila Garcia
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Adam Alnihmy
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Amit Patel
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Robert N. Weinreb
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Karl J. Wahlin
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Anna La Torre Vila
- Department of Cell Biology and Human Anatomy, University of California Davis, California, United States
| | - Derek S. Welsbie
- Gleiberman Center for Glaucoma Research, Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
16
|
Lo J, Mehta K, Dhillon A, Huang YK, Luo Z, Nam MH, Al Diri I, Chang KC. Therapeutic strategies for glaucoma and optic neuropathies. Mol Aspects Med 2023; 94:101219. [PMID: 37839232 PMCID: PMC10841486 DOI: 10.1016/j.mam.2023.101219] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/02/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Glaucoma is a neurodegenerative eye disease that causes permanent vision impairment. The main pathological characteristics of glaucoma are retinal ganglion cell (RGC) loss and optic nerve degeneration. Glaucoma can be caused by elevated intraocular pressure (IOP), although some cases are congenital or occur in patients with normal IOP. Current glaucoma treatments rely on medicine and surgery to lower IOP, which only delays disease progression. First-line glaucoma medicines are supported by pharmacotherapy advancements such as Rho kinase inhibitors and innovative drug delivery systems. Glaucoma surgery has shifted to safer minimally invasive (or microinvasive) glaucoma surgery, but further trials are needed to validate long-term efficacy. Further, growing evidence shows that adeno-associated virus gene transduction and stem cell-based RGC replacement therapy hold potential to treat optic nerve fiber degeneration and glaucoma. However, better understanding of the regulatory mechanisms of RGC development is needed to provide insight into RGC differentiation from stem cells and help choose target genes for viral therapy. In this review, we overview current progress in RGC development research, optic nerve fiber regeneration, and human stem cell-derived RGC differentiation and transplantation. We also provide an outlook on perspectives and challenges in the field.
Collapse
Affiliation(s)
- Jung Lo
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Kamakshi Mehta
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA
| | - Armaan Dhillon
- Sue Anschutz-Rodgers Eye Center and Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Yu-Kai Huang
- Department of Neurosurgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Mi-Hyun Nam
- Sue Anschutz-Rodgers Eye Center and Department of Ophthalmology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
| | - Issam Al Diri
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA.
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15219, USA; Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA; Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
17
|
Lv W, Wu X, Dou Y, Yan Y, Chen L, Fei Z, Fei F. Homer1 Protects against Retinal Ganglion Cell Pyroptosis by Inhibiting Endoplasmic Reticulum Stress-Associated TXNIP/NLRP3 Inflammasome Activation after Middle Cerebral Artery Occlusion-Induced Retinal Ischemia. Int J Mol Sci 2023; 24:16811. [PMID: 38069134 PMCID: PMC10706256 DOI: 10.3390/ijms242316811] [Citation(s) in RCA: 2] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
Retinal ischemia, after cerebral ischemia, is an easily overlooked pathophysiological problem in which inflammation is considered to play an important role. Pyroptosis is a kind of cell death pattern accompanied by inflammation. Homer scaffold protein 1 (Homer1) has anti-inflammation properties and protects against ischemic injury. However, little is known about pyroptosis following middle cerebral artery occlusion (MCAO)-induced retinal ischemia and the regulatory mechanisms involved by Homer1 for the development of pyroptosis. In the present study, retinal ischemic injury was induced in mice by permanent MCAO in vivo, and retinal ganglion cells (RGCs) were subjected to Oxygen and Glucose Deprivation (OGD) to establish an in vitro model. It was shown that TXNIP/NLRP3-mediated pyroptosis was located predominantly in RGCs, which gradually increased after retinal ischemia and peaked at 24 h after retinal ischemia. Interestingly, the RGCs pyroptosis occurred not only in the cell body but also in the axon. Notably, the occurrence of pyroptosis coincided with the change of Homer1 expression in the retina after retinal ischemia and Homer1 also co-localized with RGCs. It was demonstrated that overexpression of Homer1 not only alleviated RGCs pyroptosis and inhibited the release of pro-inflammatory factors but also led to the increase in phosphorylation of AMPK, inhibition of ER stress, and preservation of visual function after retinal ischemia. In conclusion, it was suggested that Homer1 may protect against MCAO-induced retinal ischemia and RGCs pyroptosis by inhibiting endoplasmic reticulum stress-associated TXNIP/NLRP3 inflammasome activation after MCAO-induced retinal ischemia.
Collapse
Affiliation(s)
- Weihao Lv
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China; (W.L.); (X.W.); (Y.D.); (Y.Y.)
| | - Xiuquan Wu
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China; (W.L.); (X.W.); (Y.D.); (Y.Y.)
| | - Yanan Dou
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China; (W.L.); (X.W.); (Y.D.); (Y.Y.)
| | - Yiwen Yan
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China; (W.L.); (X.W.); (Y.D.); (Y.Y.)
| | - Leiying Chen
- Department of Ophthalmology, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China
| | - Zhou Fei
- Department of Neurosurgery, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China; (W.L.); (X.W.); (Y.D.); (Y.Y.)
| | - Fei Fei
- Department of Ophthalmology, Xijing Hospital, Air Force Medical University, No. 127, Changle West Road, Xincheng District, Xi’an 710032, China
| |
Collapse
|
18
|
Zhang KY, Nagalingam A, Mary S, Aguzzi EA, Li W, Chetla N, Smith B, Paulaitis ME, Edwards MM, Quigley HA, Zack DJ, Johnson TV. Rare intercellular material transfer as a confound to interpreting inner retinal neuronal transplantation following internal limiting membrane disruption. Stem Cell Reports 2023; 18:2203-2221. [PMID: 37802075 PMCID: PMC10679651 DOI: 10.1016/j.stemcr.2023.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 10/08/2023] Open
Abstract
Intercellular cytoplasmic material transfer (MT) occurs between transplanted and developing photoreceptors and ambiguates cell origin identification in developmental, transdifferentiation, and transplantation experiments. Whether MT is a photoreceptor-specific phenomenon is unclear. Retinal ganglion cell (RGC) replacement, through transdifferentiation or transplantation, holds potential for restoring vision in optic neuropathies. During careful assessment for MT following human stem cell-derived RGC transplantation into mice, we identified RGC xenografts occasionally giving rise to labeling of donor-derived cytoplasmic, nuclear, and mitochondrial proteins within recipient Müller glia. Critically, nuclear organization is distinct between human and murine retinal neurons, which enables unequivocal discrimination of donor from host cells. MT was greatly facilitated by internal limiting membrane disruption, which also augments retinal engraftment following transplantation. Our findings demonstrate that retinal MT is not unique to photoreceptors and challenge the isolated use of species-specific immunofluorescent markers for xenotransplant identification. Assessment for MT is critical when analyzing neuronal replacement interventions.
Collapse
Affiliation(s)
- Kevin Y Zhang
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arumugam Nagalingam
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Stella Mary
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Erika A Aguzzi
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Weifeng Li
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nitin Chetla
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara Smith
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael E Paulaitis
- Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malia M Edwards
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Harry A Quigley
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Donald J Zack
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Departments of Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Glaucoma Center for Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
19
|
Soucy JR, Todd L, Kriukov E, Phay M, Malechka VV, Rivera JD, Reh TA, Baranov P. Controlling donor and newborn neuron migration and maturation in the eye through microenvironment engineering. Proc Natl Acad Sci U S A 2023; 120:e2302089120. [PMID: 37931105 PMCID: PMC10655587 DOI: 10.1073/pnas.2302089120] [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: 02/07/2023] [Accepted: 09/30/2023] [Indexed: 11/08/2023] Open
Abstract
Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.
Collapse
Affiliation(s)
- Jonathan R. Soucy
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Levi Todd
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Emil Kriukov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Monichan Phay
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Volha V. Malechka
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - John Dayron Rivera
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA98195
| | - Petr Baranov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA02114
- Department of Ophthalmology, Harvard Medical School, Boston, MA02114
| |
Collapse
|
20
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
21
|
Patel SH, Lamba DA. Factors Affecting Stem Cell-Based Regenerative Approaches in Retinal Degeneration. Annu Rev Vis Sci 2023; 9:155-175. [PMID: 37713278 DOI: 10.1146/annurev-vision-120222-012817] [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: 09/17/2023]
Abstract
Inherited and age-associated vision loss is often associated with degeneration of the cells of the retina, the light-sensitive layer at the back of the eye. The mammalian retina, being a postmitotic neural tissue, does not have the capacity to repair itself through endogenous regeneration. There has been considerable excitement for the development of cell replacement approaches since the isolation and development of culture methods for human pluripotent stem cells, as well as the generation of induced pluripotent stem cells. This has now been combined with novel three-dimensional organoid culture systems that closely mimic human retinal development in vitro. In this review, we cover the current state of the field, with emphasis on the cell delivery challenges, role of the recipient immunological microenvironment, and challenges related to connectivity between transplanted cells and host circuitry both locally and centrally to the different areas of the brain.
Collapse
Affiliation(s)
- Sachin H Patel
- Department of Ophthalmology, University of California, San Francisco, California, USA;
| | - Deepak A Lamba
- Department of Ophthalmology, University of California, San Francisco, California, USA;
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California, San Francisco, California, USA
| |
Collapse
|
22
|
Mcloughlin KJ, Aladdad AM, Payne AJ, Boda AI, Nieto-Gomez S, Kador KE. Purification of retinal ganglion cells using low-pressure flow cytometry. Front Mol Neurosci 2023; 16:1149024. [PMID: 37547921 PMCID: PMC10400357 DOI: 10.3389/fnmol.2023.1149024] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
Purified Retinal Ganglion Cells (RGCs) for in vitro study have been a valuable tool in the study of neural regeneration and in the development of therapies to treat glaucoma. Traditionally, RGCs have been isolated from early postnatal rats and mice, and more recently from human in vitro derived retinal organoids using a two-step immunopanning technique based upon the expression of Thy-1. This technique, however, limits the time periods from which RGCs can be isolated, missing the earliest born RGCs at which time the greatest stage of axon growth occurs, as well as being limited in its use with models of retinal degeneration as Thy-1 is downregulated following injury. While fluorescence associated cell sorting (FACS) in combination with new optogenetically labeled RGCs would be able to overcome this limitation, the use of traditional FACS sorters has been limited to genomic and proteomic studies, as RGCs have little to no survival post-sorting. Here we describe a new method for RGC isolation utilizing a combined immunopanning-fluorescence associated cell sorting (IP-FACS) protocol that initially depletes macrophages and photoreceptors, using immunopanning to enrich for RGCs before using low-pressure FACS to isolate these cells. We demonstrate that RGCs isolated via IP-FACS when compared to RGCs isolated via immunopanning at the same age have similar purity as measured by antibody staining and qRT-PCR; survival as measured by live dead staining; neurite outgrowth; and electrophysiological properties as measured by calcium release response to glutamate. Finally, we demonstrate the ability to isolate RGCs from early embryonic mice prior to the expression of Thy-1 using Brn3b-eGFP optogenetically labeled cells. This method provides a new approach for the isolation of RGCs for the study of early developed RGCs, the study of RGC subtypes and the isolation of RGCs for cell transplantation studies.
Collapse
Affiliation(s)
- Kiran J. Mcloughlin
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Afnan M. Aladdad
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Andrew J. Payne
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Anna I. Boda
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Sayra Nieto-Gomez
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Karl E. Kador
- Department of Ophthalmology, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, Kansas City, MO, United States
| |
Collapse
|
23
|
Subramani M, Van Hook MJ, Ahmad I. Reproducible generation of human retinal ganglion cells from banked retinal progenitor cells: analysis of target recognition and IGF-1-mediated axon regeneration. Front Cell Dev Biol 2023; 11:1214104. [PMID: 37519299 PMCID: PMC10373790 DOI: 10.3389/fcell.2023.1214104] [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: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
The selective degeneration of retinal ganglion cells (RGCs) is a common feature in glaucoma, a complex group of diseases, leading to irreversible vision loss. Stem cell-based glaucoma disease modeling, cell replacement, and axon regeneration are viable approaches to understand mechanisms underlying glaucomatous degeneration for neuroprotection, ex vivo stem cell therapy, and therapeutic regeneration. These approaches require direct and facile generation of human RGCs (hRGCs) from pluripotent stem cells. Here, we demonstrate a method for rapid generation of hRGCs from banked human pluripotent stem cell-derived retinal progenitor cells (hRPCs) by recapitulating the developmental mechanism. The resulting hRGCs are stable, functional, and transplantable and have the potential for target recognition, demonstrating their suitability for both ex vivo stem cell approaches to glaucomatous degeneration and disease modeling. Additionally, we demonstrate that hRGCs derived from banked hRPCs are capable of regenerating their axons through an evolutionarily conserved mechanism involving insulin-like growth factor 1 and the mTOR axis, demonstrating their potential to identify and characterize the underlying mechanism(s) that can be targeted for therapeutic regeneration.
Collapse
Affiliation(s)
| | | | - Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
24
|
Johnson TV, Calkins DJ, Fortune B, Goldberg JL, La Torre A, Lamba DA, Meyer JS, Reh TA, Wallace VA, Zack DJ, Baranov P. The importance of unambiguous cell origin determination in neuronal repopulation studies. iScience 2023; 26:106361. [PMID: 37009209 PMCID: PMC10060674 DOI: 10.1016/j.isci.2023.106361] [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] [Indexed: 03/12/2023] Open
Abstract
Neuronal repopulation achieved through transplantation or transdifferentiation from endogenous sources holds tremendous potential for restoring function in chronic neurodegenerative disease or acute injury. Key to the evaluation of neuronal engraftment is the definitive discrimination of new or donor neurons from preexisting cells within the host tissue. Recent work has identified mechanisms by which genetically encoded donor cell reporters can be transferred to host neurons through intercellular material transfer. In addition, labeling transplanted and endogenously transdifferentiated neurons through viral vector transduction can yield misexpression in host cells in some circumstances. These issues can confound the tracking and evaluation of repopulated neurons in regenerative experimental paradigms. Using the retina as an example, we discuss common reasons for artifactual labeling of endogenous host neurons with donor cell reporters and suggest strategies to prevent erroneous conclusions based on misidentification of cell origin.
Collapse
Affiliation(s)
- Thomas V. Johnson
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J. Calkins
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Anna La Torre
- Department of Cell Biology & Human Anatomy, University of California Davis, Davis, CA, USA
| | - Deepak A. Lamba
- Department of Ophthalmology and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Jason S. Meyer
- Departments of Medical & Molecular Genetics, Ophthalmology (Glick Eye Institute), Pharmacology & Toxicology, and Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Valerie A. Wallace
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Departments of Laboratory Medicine & Pathobiology, and Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Donald J. Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Petr Baranov
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Tao Y, Zhang Q, Meng M, Huang J. A bibliometric analysis of the application of stem cells in glaucoma research from 1999 to 2022. Front Cell Dev Biol 2023; 11:1081898. [PMID: 36743419 PMCID: PMC9889543 DOI: 10.3389/fcell.2023.1081898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Background: Glaucoma, a neurodegenerative disease of the retina, is the leading cause of irreversible blindness. Stem cells have therapeutic potential for glaucoma. However, few bibliometric studies have been published in this field. Concerning a visual map, this article aims to characterize the research context, cooperation relationship, hotspots, and trends concerning the application of stem cells in glaucoma research. Methods: Publications focusing on stem cell research and glaucoma were retrieved from the Web of Science Core Collection. VOSviewer, CiteSpace, Microsoft Excel, and Scimago Graphica were used to map the contributions of countries or regions, authors, organizations, and journals. Journal Impact Factor data were obtained from the Web of Science Core Collection. We analyzed the tendencies, hotspots, and knowledge networks using VOSviewer, and CiteSpace. Results: We analyzed 518 articles published from 1999 through 2022. In the first decade, the number of articles in this field increased slowly, and there was a marked acceleration in publication frequency after 2010. The United States, China, and England were the main contributors. Yiqin Du was the most prolific author, and among the top 10 prolific writers, Keith R. Martin's work was cited most frequently. Investigative Ophthalmology and Visual Science, Experimental Eye Research, and Cornea published the most articles in this domain. The three most commonly co-cited journals were Investigative Ophthalmology and Visual Science, Experimental Eye Research, and Proceedings of the National Academy of Sciences of the United States of America. The Central South University, the University of Pittsburgh, and the National Institutes of Health National Eye Institute were highly prolific institutions in this research area. Our keywords analysis with VOSviewer suggested directions of future research and yielded the following recent key themes, extracellular vesicles, exosomes, mitochondria, growth factors, oxidative stress, and ocular diseases. Four co-cited references had a citation burst duration until 2022. Conclusion: With improvements in overall quality of life and demographic transitions toward population aging, research and clinical focus on eye care has increased, with glaucoma as a key area of emphasis. This study added to our understanding of the global landscape and Frontier hotspots in this field.
Collapse
Affiliation(s)
- Yuanyuan Tao
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qian Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ming Meng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
26
|
Wu S, Mo X. Optic Nerve Regeneration in Diabetic Retinopathy: Potentials and Challenges Ahead. Int J Mol Sci 2023; 24:ijms24021447. [PMID: 36674963 PMCID: PMC9865663 DOI: 10.3390/ijms24021447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/31/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
Diabetic retinopathy (DR), the most common microvascular compilation of diabetes, is the leading cause of vision loss and blindness worldwide. Recent studies indicate that retinal neuron impairment occurs before any noticeable vascular changes in DR, and retinal ganglion cell (RGC) degeneration is one of the earliest signs. Axons of RGCs have little capacity to regenerate after injury, clinically leading the visual functional defects to become irreversible. In the past two decades, tremendous progress has been achieved to enable RGC axon regeneration in animal models of optic nerve injury, which holds promise for neural repair and visual restoration in DR. This review summarizes these advances and discusses the potential and challenges for developing optic nerve regeneration strategies treating DR.
Collapse
Affiliation(s)
| | - Xiaofen Mo
- Correspondence: ; Tel.: +86-021-64377134
| |
Collapse
|
27
|
Tawfik M, Chen F, Goldberg JL, Sabel BA. Nanomedicine and drug delivery to the retina: current status and implications for gene therapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2022; 395:1477-1507. [PMID: 36107200 PMCID: PMC9630211 DOI: 10.1007/s00210-022-02287-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
Blindness affects more than 60 million people worldwide. Retinal disorders, including age-related macular degeneration (AMD), diabetic retinopathy (DR), and glaucoma, are the leading causes of blindness. Finding means to optimize local and sustained delivery of drugs or genes to the eye and retina is one goal to advance the development of new therapeutics. Despite the ease of accessibility of delivering drugs via the ocular surface, the delivery of drugs to the retina is still challenging due to anatomic and physiologic barriers. Designing a suitable delivery platform to overcome these barriers should enhance drug bioavailability and provide a safe, controlled, and sustained release. Current inventions for posterior segment treatments include intravitreal implants and subretinal viral gene delivery that satisfy these criteria. Several other novel drug delivery technologies, including nanoparticles, micelles, dendrimers, microneedles, liposomes, and nanowires, are now being widely studied for posterior segment drug delivery, and extensive research on gene delivery using siRNA, mRNA, or aptamers is also on the rise. This review discusses the current state of retinal drug/gene delivery and highlights future therapeutic opportunities.
Collapse
Affiliation(s)
- Mohamed Tawfik
- Institute of Medical Psychology, Medical Faculty, Otto-Von-Guericke University, Magdeburg, Germany
| | - Fang Chen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Bernhard A Sabel
- Institute of Medical Psychology, Medical Faculty, Otto-Von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
28
|
Luo Z, Chang KC, Wu S, Sun C, Xia X, Nahmou M, Bian M, Wen RR, Zhu Y, Shah S, Tanasa B, Wernig M, Goldberg JL. Directly induced human retinal ganglion cells mimic fetal RGCs and are neuroprotective after transplantation in vivo. Stem Cell Reports 2022; 17:2690-2703. [PMID: 36368332 PMCID: PMC9768574 DOI: 10.1016/j.stemcr.2022.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Retinal ganglion cell (RGC) replacement therapy could restore vision in glaucoma and other optic neuropathies. We developed a rapid protocol for directly induced RGC (iRGC) differentiation from human stem cells, leveraging overexpression of NGN2. Neuronal morphology and neurite growth were observed within 1 week of induction; characteristic RGC-specific gene expression confirmed identity. Calcium imaging demonstrated γ-aminobutyric acid (GABA)-induced excitation characteristic of immature RGCs. Single-cell RNA sequencing showed more similarities between iRGCs and early-stage fetal human RGCs than retinal organoid-derived RGCs. Intravitreally transplanted iRGCs survived and migrated into host retinas independent of prior optic nerve trauma, but iRGCs protected host RGCs from neurodegeneration. These data demonstrate rapid iRGC generation in vitro into an immature cell with high similarity to human fetal RGCs and capacity for retinal integration after transplantation and neuroprotective function after optic nerve injury. The simplicity of this system may benefit translational studies on human RGCs.
Collapse
Affiliation(s)
- Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Kun-Che Chang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Department of Ophthalmology and Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Suqian Wu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Shanghai Key Laboratory of Visual Impairment and Restoration, Department of Ophthalmology and Vision Science, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai 200031, China
| | - Catalina Sun
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Xin Xia
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Michael Nahmou
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Minjuan Bian
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Rain R. Wen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Ying Zhu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sahil Shah
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Bogdan Tanasa
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Marius Wernig
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Corresponding author
| |
Collapse
|
29
|
Yang YP, Hsiao YJ, Chang KJ, Foustine S, Ko YL, Tsai YC, Tai HY, Ko YC, Chiou SH, Lin TC, Chen SJ, Chien Y, Hwang DK. Pluripotent Stem Cells in Clinical Cell Transplantation: Focusing on Induced Pluripotent Stem Cell-Derived RPE Cell Therapy in Age-Related Macular Degeneration. Int J Mol Sci 2022; 23:ijms232213794. [PMID: 36430270 PMCID: PMC9696562 DOI: 10.3390/ijms232213794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Human pluripotent stem cells (PSCs), including both embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), represent valuable cell sources to replace diseased or injured tissues in regenerative medicine. iPSCs exhibit the potential for indefinite self-renewal and differentiation into various cell types and can be reprogrammed from somatic tissue that can be easily obtained, paving the way for cell therapy, regenerative medicine, and personalized medicine. Cell therapies using various iPSC-derived cell types are now evolving rapidly for the treatment of clinical diseases, including Parkinson's disease, hematological diseases, cardiomyopathy, osteoarthritis, and retinal diseases. Since the first interventional clinical trial with autologous iPSC-derived retinal pigment epithelial cells (RPEs) for the treatment of age-related macular degeneration (AMD) was accomplished in Japan, several preclinical trials using iPSC suspensions or monolayers have been launched, or are ongoing or completed. The evolution and generation of human leukocyte antigen (HLA)-universal iPSCs may facilitate the clinical application of iPSC-based therapies. Thus, iPSCs hold great promise in the treatment of multiple retinal diseases. The efficacy and adverse effects of iPSC-based retinal therapies should be carefully assessed in ongoing and further clinical trials.
Collapse
Affiliation(s)
- Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yu-Jer Hsiao
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Kao-Jung Chang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shania Foustine
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yu-Ling Ko
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yi-Ching Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Hsiao-Yun Tai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yu-Chieh Ko
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Genomics Research Center, Academia Sinica, Taipei 115201, Taiwan
| | - Tai-Chi Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
| | - Yueh Chien
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Correspondence: (Y.C.); (D.-K.H.); Tel.: +886-2-2875-2121 (D.-K.H.)
| | - De-Kuang Hwang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 112201, Taiwan
- Correspondence: (Y.C.); (D.-K.H.); Tel.: +886-2-2875-2121 (D.-K.H.)
| |
Collapse
|
30
|
Transplanted human induced pluripotent stem cells- derived retinal ganglion cells embed within mouse retinas and are electrophysiologically functional. iScience 2022; 25:105308. [DOI: 10.1016/j.isci.2022.105308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/22/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
|
31
|
Croteau LP, Risner ML, Wareham LK, McGrady NR, Chamling X, Zack DJ, Calkins DJ. Ex Vivo Integration of Human Stem Retinal Ganglion Cells into the Mouse Retina. Cells 2022; 11:cells11203241. [PMID: 36291110 PMCID: PMC9600680 DOI: 10.3390/cells11203241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/04/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cell replacement therapies may be key in achieving functional recovery in neurodegenerative optic neuropathies diseases such as glaucoma. One strategy that holds promise in this regard is the use of human embryonic stem cell and induced pluripotent stem-derived retinal ganglion cells (hRGCs). Previous hRGC transplantation studies have shown modest success. This is in part due to the low survival and integration of the transplanted cells in the host retina. The field is further challenged by mixed assays and outcome measurements that probe and determine transplantation success. Thefore, we have devised a transplantation assay involving hRGCs and mouse retina explants that bypasses physical barriers imposed by retinal membranes. We show that hRGC neurites and somas are capable of invading mouse explants with a subset of hRGC neurites being guided by mouse RGC axons. Neonatal mouse retina explants, and to a lesser extent, adult explants, promote hRGC integrity and neurite outgrowth. Using this assay, we tested whether suppmenting cultures with brain derived neurotrophic factor (BDNF) and the adenylate cyclase activator, forskolin, enhances hRGC neurite integration, neurite outgrowth, and integrity. We show that supplementing cultures with a combination BDNF and forskolin strongly favors hRGC integrity, increasing neurite outgrowth and complexity as well as the invasion of mouse explants. The transplantation assay presented here is a practical tool for investigating strategies for testing and optimizing the integration of donor cells into host tissues.
Collapse
Affiliation(s)
- Louis-Philippe Croteau
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael L. Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren K. Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nolan R. McGrady
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David J. Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
32
|
Johnson TV, Polo AD, Sahel JA, Schuman JS. Neuroprotection, Neuroenhancement, and Neuroregeneration of the Retina and Optic Nerve. OPHTHALMOLOGY SCIENCE 2022; 2:100216. [PMID: 36245765 PMCID: PMC9559091 DOI: 10.1016/j.xops.2022.100216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
33
|
Levin LA, Patrick C, Choudry NB, Sharif NA, Goldberg JL. Neuroprotection in neurodegenerations of the brain and eye: Lessons from the past and directions for the future. Front Neurol 2022; 13:964197. [PMID: 36034312 PMCID: PMC9412944 DOI: 10.3389/fneur.2022.964197] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Background Neurological and ophthalmological neurodegenerative diseases in large part share underlying biology and pathophysiology. Despite extensive preclinical research on neuroprotection that in many cases bridges and unifies both fields, only a handful of neuroprotective therapies have succeeded clinically in either. Main body Understanding the commonalities among brain and neuroretinal neurodegenerations can help develop innovative ways to improve translational success in neuroprotection research and emerging therapies. To do this, analysis of why translational research in neuroprotection fails necessitates addressing roadblocks at basic research and clinical trial levels. These include optimizing translational approaches with respect to biomarkers, therapeutic targets, treatments, animal models, and regulatory pathways. Conclusion The common features of neurological and ophthalmological neurodegenerations are useful for outlining a path forward that should increase the likelihood of translational success in neuroprotective therapies.
Collapse
Affiliation(s)
- Leonard A. Levin
- Departments of Ophthalmology and Visual Sciences, Neurology & Neurosurgery, McGill University, Montreal, QC, Canada
| | | | - Nozhat B. Choudry
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| | - Najam A. Sharif
- Global Alliances and External Research, Ophthalmology Innovation Center, Santen Inc., Emeryville, CA, United States
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, CA, United States
| |
Collapse
|
34
|
Van Gelder RN, Chiang MF, Dyer MA, Greenwell TN, Levin LA, Wong RO, Svendsen CN. Regenerative and restorative medicine for eye disease. Nat Med 2022; 28:1149-1156. [PMID: 35715505 PMCID: PMC10718186 DOI: 10.1038/s41591-022-01862-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/06/2022] [Indexed: 12/11/2022]
Abstract
Causes of blindness differ across the globe; in higher-income countries, most blindness results from the degeneration of specific classes of cells in the retina, including retinal pigment epithelium (RPE), photoreceptors, and retinal ganglion cells. Advances over the past decade in retinal regenerative medicine have allowed each of these cell types to be produced ex vivo from progenitor stem cells. Here, we review progress in applying these technologies to cell replacement - with the goal of vision restoration in degenerative disease. We discuss the landscape of human clinical trials for RPE transplantation and advanced preclinical studies for other cell types. We also review progress toward in situ repair of retinal degeneration using endogenous progenitor cells. Finally, we provide a high-level overview of progress toward prosthetic ocular vision restoration, including advanced photovoltaic devices, opsin-based gene therapy, and small-molecule photoswitches. Progress in each of these domains is at or near the human clinical-trial stage, bringing the audacious goal of vision restoration within sight.
Collapse
Affiliation(s)
- Russell N Van Gelder
- Karalis-Johnson Retina Center, Department of Ophthalmology, University of Washington School of Medicine, Seattle, WA, USA.
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA, USA.
- Department of Pathology and Laboratory Medicine, University of Washington School of Medicine, Seattle, WA, USA.
- Roger and Angie Karalis Johnson Retina Center, University of Washington School of Medicine, Seattle, WA, USA.
| | - Michael F Chiang
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael A Dyer
- Department of Developmental Neurobiology, St. Jude's Research Hospital, Memphis, TN, USA
| | - Thomas N Greenwell
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - Leonard A Levin
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, Quebec, Canada
- Department of Neurology & Neurosurgery, McGill University, Montreal, Quebec, Canada
| | - Rachel O Wong
- Karalis-Johnson Retina Center, Department of Ophthalmology, University of Washington School of Medicine, Seattle, WA, USA
- Department of Biological Structure, University of Washington School of Medicine, Seattle, WA, USA
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Los Angeles, CA, USA
- Department of Biomedical Sciences, Cedars Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
35
|
Wareham LK, Liddelow SA, Temple S, Benowitz LI, Di Polo A, Wellington C, Goldberg JL, He Z, Duan X, Bu G, Davis AA, Shekhar K, Torre AL, Chan DC, Canto-Soler MV, Flanagan JG, Subramanian P, Rossi S, Brunner T, Bovenkamp DE, Calkins DJ. Solving neurodegeneration: common mechanisms and strategies for new treatments. Mol Neurodegener 2022; 17:23. [PMID: 35313950 PMCID: PMC8935795 DOI: 10.1186/s13024-022-00524-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Across neurodegenerative diseases, common mechanisms may reveal novel therapeutic targets based on neuronal protection, repair, or regeneration, independent of etiology or site of disease pathology. To address these mechanisms and discuss emerging treatments, in April, 2021, Glaucoma Research Foundation, BrightFocus Foundation, and the Melza M. and Frank Theodore Barr Foundation collaborated to bring together key opinion leaders and experts in the field of neurodegenerative disease for a virtual meeting titled "Solving Neurodegeneration". This "think-tank" style meeting focused on uncovering common mechanistic roots of neurodegenerative disease and promising targets for new treatments, catalyzed by the goal of finding new treatments for glaucoma, the world's leading cause of irreversible blindness and the common interest of the three hosting foundations. Glaucoma, which causes vision loss through degeneration of the optic nerve, likely shares early cellular and molecular events with other neurodegenerative diseases of the central nervous system. Here we discuss major areas of mechanistic overlap between neurodegenerative diseases of the central nervous system: neuroinflammation, bioenergetics and metabolism, genetic contributions, and neurovascular interactions. We summarize important discussion points with emphasis on the research areas that are most innovative and promising in the treatment of neurodegeneration yet require further development. The research that is highlighted provides unique opportunities for collaboration that will lead to efforts in preventing neurodegeneration and ultimately vision loss.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Sally Temple
- Neural Stem Cell Institute, NY, 12144, Rensselaer, USA
| | - Larry I Benowitz
- Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, CA, Palo Alto, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, MA, Boston, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Albert A Davis
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, CA, 91125, Pasadena, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Aurora, CO, USA
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | | | | | | | | | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
36
|
Gibbons A, Henderson AD. Non-Arteritic Anterior Ischemic Optic Neuropathy: Challenges for the Future. FRONTIERS IN OPHTHALMOLOGY 2022; 2:848710. [PMID: 38983540 PMCID: PMC11182325 DOI: 10.3389/fopht.2022.848710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/23/2022] [Indexed: 07/11/2024]
Affiliation(s)
- Alison Gibbons
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amanda D Henderson
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
37
|
Zhang Q, Li Y, Zhuo Y. Synaptic or Non-synaptic? Different Intercellular Interactions with Retinal Ganglion Cells in Optic Nerve Regeneration. Mol Neurobiol 2022; 59:3052-3072. [PMID: 35266115 PMCID: PMC9016027 DOI: 10.1007/s12035-022-02781-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 02/24/2022] [Indexed: 12/31/2022]
Abstract
Axons of adult neurons in the mammalian central nervous system generally fail to regenerate by themselves, and few if any therapeutic options exist to reverse this situation. Due to a weak intrinsic potential for axon growth and the presence of strong extrinsic inhibitors, retinal ganglion cells (RGCs) cannot regenerate their axons spontaneously after optic nerve injury and eventually undergo apoptosis, resulting in permanent visual dysfunction. Regarding the extracellular environment, research to date has generally focused on glial cells and inflammatory cells, while few studies have discussed the potentially significant role of interneurons that make direct connections with RGCs as part of the complex retinal circuitry. In this study, we provide a novel angle to summarize these extracellular influences following optic nerve injury as "intercellular interactions" with RGCs and classify these interactions as synaptic and non-synaptic. By discussing current knowledge of non-synaptic (glial cells and inflammatory cells) and synaptic (mostly amacrine cells and bipolar cells) interactions, we hope to accentuate the previously neglected but significant effects of pre-synaptic interneurons and bring unique insights into future pursuit of optic nerve regeneration and visual function recovery.
Collapse
Affiliation(s)
- Qi Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
38
|
Varadarajan SG, Hunyara JL, Hamilton NR, Kolodkin AL, Huberman AD. Central nervous system regeneration. Cell 2022; 185:77-94. [PMID: 34995518 PMCID: PMC10896592 DOI: 10.1016/j.cell.2021.10.029] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
Neurons of the mammalian central nervous system fail to regenerate. Substantial progress has been made toward identifying the cellular and molecular mechanisms that underlie regenerative failure and how altering those pathways can promote cell survival and/or axon regeneration. Here, we summarize those findings while comparing the regenerative process in the central versus the peripheral nervous system. We also highlight studies that advance our understanding of the mechanisms underlying neural degeneration in response to injury, as many of these mechanisms represent primary targets for restoring functional neural circuits.
Collapse
Affiliation(s)
| | - John L Hunyara
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Natalie R Hamilton
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alex L Kolodkin
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Andrew D Huberman
- Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Ophthalmology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
39
|
Dromel PC, Singh D, Andres E, Likes M, Kurisawa M, Alexander-Katz A, Spector M, Young M. A bioinspired gelatin-hyaluronic acid-based hybrid interpenetrating network for the enhancement of retinal ganglion cells replacement therapy. NPJ Regen Med 2021; 6:85. [PMID: 34930951 PMCID: PMC8688498 DOI: 10.1038/s41536-021-00195-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/20/2021] [Indexed: 11/25/2022] Open
Abstract
Biomaterial-based cell replacement approaches to regenerative medicine are emerging as promising treatments for a wide array of profound clinical problems. Here we report an interpenetrating polymer network (IPN) composed of gelatin-hydroxyphenyl propionic acid and hyaluronic acid tyramine that is able to enhance intravitreal retinal cell therapy. By tuning our bioinspired hydrogel to mimic the vitreous chemical composition and mechanical characteristics we were able to improve in vitro and in vivo viability of human retinal ganglion cells (hRGC) incorporated into the IPN. In vivo vitreal injections of cell-bearing IPN in rats showed extensive attachment to the inner limiting membrane of the retina, improving with hydrogels stiffness. Engrafted hRGC displayed signs of regenerating processes along the optic nerve. Of note was the decrease in the immune cell response to hRGC delivered in the gel. The findings compel further translation of the gelatin-hyaluronic acid IPN for intravitreal cell therapy.
Collapse
Affiliation(s)
- Pierre C Dromel
- Massachusetts Institute of Technology, Cambridge, MA, USA
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Deepti Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Eliot Andres
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | | | - Motoichi Kurisawa
- A*STAR Institute of Bioengineering and Nanotechnology, Singapore, Singapore
| | | | - Myron Spector
- VA Boston Healthcare System, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Young
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
40
|
Xiao D, Jin K, Qiu S, Lei Q, Huang W, Chen H, Su J, Xu Q, Xu Z, Gou B, Tie X, Liu F, Liu S, Liu Y, Xiang M. In vivo Regeneration of Ganglion Cells for Vision Restoration in Mammalian Retinas. Front Cell Dev Biol 2021; 9:755544. [PMID: 34671605 PMCID: PMC8520940 DOI: 10.3389/fcell.2021.755544] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
Glaucoma and other optic neuropathies affect millions of people worldwide, ultimately causing progressive and irreversible degeneration of retinal ganglion cells (RGCs) and blindness. Previous research into cell replacement therapy of these neurodegenerative diseases has been stalled due to the incapability for grafted RGCs to integrate into the retina and project properly along the long visual pathway. In vivo RGC regeneration would be a promising alternative approach but mammalian retinas lack regenerative capacity. It therefore has long been a great challenge to regenerate functional and properly projecting RGCs for vision restoration in mammals. Here we show that the transcription factors (TFs) Math5 and Brn3b together are able to reprogram mature mouse Müller glia (MG) into RGCs. The reprogrammed RGCs extend long axons that make appropriate intra-retinal and extra-retinal projections through the entire visual pathway to innervate both image-forming and non-image-forming brain targets. They exhibit typical neuronal electrophysiological properties and improve visual responses in RGC loss mouse models. Together, our data provide evidence that mammalian MG can be reprogrammed by defined TFs to achieve in vivo regeneration of functional RGCs as well as a promising new therapeutic approach to restore vision to patients with glaucoma and other optic neuropathies.
Collapse
Affiliation(s)
- Dongchang Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Kangxin Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Suo Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qiannan Lei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haiqiao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jing Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Qiang Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zihui Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Bin Gou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoxiu Tie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Feng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Sheng Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Mengqing Xiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
41
|
Luo Z, Xian B, Li K, Li K, Yang R, Chen M, Xu C, Tang M, Rong H, Hu D, Ye M, Yang S, Lu S, Zhang H, Ge J. Biodegradable scaffolds facilitate epiretinal transplantation of hiPSC-Derived retinal neurons in nonhuman primates. Acta Biomater 2021; 134:289-301. [PMID: 34314890 DOI: 10.1016/j.actbio.2021.07.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/30/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Transplantation of stem cell-derived retinal neurons is a promising regenerative therapy for optic neuropathy. However, significant anatomic differences compromise its efficacy in large animal models. The present study describes the procedure and outcomes of human-induced pluripotent stem cell (hiPSC)-derived retinal sheet transplantation in primate models using biodegradable materials. Stem cell-derived retinal organoids were seeded on polylactic-coglycolic acid (PLGA) scaffolds and directed toward a retinal ganglion cell (RGC) fate. The seeded tissues showed active proliferation, typical neuronal morphology, and electrical excitability. The cellular scaffolds were then epiretinally transplanted onto the inner surface of rhesus monkey retinas. With sufficient graft-host contact provided by the scaffold, the transplanted tissues survived for up to 1 year without tumorigenesis. Histological examinations indicated survival, further maturation, and migration. Moreover, green fluorescent protein-labeled axonal projections toward the host optic nerve were observed. Cryopreserved organoids were also able to survive and migrate after transplantation. Our results suggest the potential efficacy of RGC replacement therapy in the repair of optic neuropathy for the restoration of visual function. STATEMENT OF SIGNIFICANCE: In the present study, we generated a human retinal sheet by seeding hiPSC-retinal organoid-derived RGCs on a biodegradable PLGA scaffold. We transplanted this retinal sheet onto the inner surface of the rhesus monkey retina. With scaffold support, donor cells survive, migrate and project their axons into the host optic nerve. Furthermore, an effective cryopreservation strategy for retinal organoids was developed, and the thawed organoids were also observed to survive and show cell migration after transplantation.
Collapse
|
42
|
Wu YR, Hashiguchi T, Sho J, Chiou SH, Takahashi M, Mandai M. Transplanted Mouse Embryonic Stem Cell-Derived Retinal Ganglion Cells Integrate and Form Synapses in a Retinal Ganglion Cell-Depleted Mouse Model. Invest Ophthalmol Vis Sci 2021; 62:26. [PMID: 34705025 PMCID: PMC8556558 DOI: 10.1167/iovs.62.13.26] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Purpose Retinal ganglion cell (RGC) transplantation is a therapeutic approach to replace irreversibly degenerated RGCs in diseases such as glaucoma. However, the application of primary RGCs is limited by the availability of tissues. The goal of this study was to evaluate whether transplanted mouse embryonic stem cell (mESC)-derived RGCs can integrate into the host retina and form cell connectivity with host cells. Methods In this study, we prepared small retinal fragments containing RGC as THY1-enhanced green fluorescent protein (EGFP)+ cells from mESCs and placed them near the retinal surface in the air-injected mouse eyes with or without N-methyl-d-aspartate (NMDA)-induced RGC depletion. After transplantation, THY1-EGFP+ cell integration was observed in whole-mounts and with immunostaining for synaptic markers. Results Transplanted THY1-EGFP+ cells survived for 12 weeks and extended neurites into the inner plexiform layer (IPL) of the host retina. Presumptive synapse formation was identified between grafted RGCs and host bipolar cells. The ratio of transplanted eyes with integration of THY1-EGFP+ neurites in the host IPL was higher in RGC-injured mice compared with healthy controls. Conclusions This report shows the potential for therapeutic use of pluripotent cell–derived RGCs by grafting the cells in healthy conditions and with an appropriate technical approach.
Collapse
Affiliation(s)
- You-Ren Wu
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tomoyo Hashiguchi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Junki Sho
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Shih-Hwa Chiou
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Vision Care Cell Therapy, Inc., Kobe, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
43
|
Rohiwal SS, Ellederová Z, Ardan T, Klima J. Advancement in Nanostructure-Based Tissue-Engineered Biomaterials for Retinal Degenerative Diseases. Biomedicines 2021; 9:biomedicines9081005. [PMID: 34440209 PMCID: PMC8393745 DOI: 10.3390/biomedicines9081005] [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: 07/08/2021] [Revised: 08/07/2021] [Accepted: 08/10/2021] [Indexed: 12/20/2022] Open
Abstract
The review intends to overview a wide range of nanostructured natural, synthetic and biological membrane implants for tissue engineering to help in retinal degenerative diseases. Herein, we discuss the transplantation strategies and the new development of material in combination with cells such as induced pluripotent stem cells (iPSC), mature retinal cells, adult stem cells, retinal progenitors, fetal retinal cells, or retinal pigment epithelial (RPE) sheets, etc. to be delivered into the subretinal space. Retinitis pigmentosa and age-related macular degeneration (AMD) are the most common retinal diseases resulting in vision impairment or blindness by permanent loss in photoreceptor cells. Currently, there are no therapies that can repair permanent vision loss, and the available treatments can only delay the advancement of retinal degeneration. The delivery of cell-based nanostructure scaffolds has been presented to enrich cell survival and direct cell differentiation in a range of retinal degenerative models. In this review, we sum up the research findings on different types of nanostructure scaffolds/substrate or material-based implants, with or without cells, used to deliver into the subretinal space for retinal diseases. Though, clinical and pre-clinical trials are still needed for these transplants to be used as a clinical treatment method for retinal degeneration.
Collapse
|
44
|
Qian C, Dong B, Wang XY, Zhou FQ. In vivo glial trans-differentiation for neuronal replacement and functional recovery in central nervous system. FEBS J 2021; 288:4773-4785. [PMID: 33351267 PMCID: PMC8217397 DOI: 10.1111/febs.15681] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/04/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
The adult mammalian central nervous system (CNS) is deficient in intrinsic machineries to replace neurons lost in injuries or progressive degeneration. Various types of these neurons constitute neural circuitries wired to support vital sensory, motor, and cognitive functions. Based on the pioneer studies in cell lineage conversion, one promising strategy is to convert in vivo glial cells into neural progenitors or directly into neurons that can be eventually rewired for functional recovery. We first briefly summarize the well-studied regeneration-capable CNS in the zebrafish, focusing on their postinjury spontaneous reprogramming of the retinal Müller glia (MG). We then compare the signaling transductions, and transcriptional and epigenetic regulations in the zebrafish MGs with their mammalian counterparts, which perpetuate certain barriers against proliferation and neurogenesis and thus fail in MG-to-progenitor conversion. Next, we discuss emerging evidence from mouse studies, in which the in vivo glia-to-neuron conversion could be achieved with sequential or one-step genetic manipulations, such as the conversions from retinal MGs to interneurons, photoreceptors, or retinal ganglion cells (RGCs), as well as the conversions from midbrain astrocytes to dopaminergic or GABAergic neurons. Some of these in vivo studies showed considerable coverage of subtypes in the newly induced neurons and partial reestablishment in neural circuits and functions. Importantly, we would like to point out some crucial technical concerns that need to be addressed to convincingly show successful glia-to-neuron conversion. Finally, we present challenges and future directions in the field for better neural function recovery.
Collapse
Affiliation(s)
- Cheng Qian
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Bryan Dong
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Xu-Yang Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Feng-Quan Zhou
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21287,The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, MD 21287,Corresponding author: Feng-Quan Zhou, Ph.D., , Phone: 443-287-5649, Address: The John G. Rangos Building, Room 291, 855 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
45
|
Abstract
The damage or loss of retinal ganglion cells (RGCs) and their axons accounts for the visual functional defects observed after traumatic injury, in degenerative diseases such as glaucoma, or in compressive optic neuropathies such as from optic glioma. By using optic nerve crush injury models, recent studies have revealed the cellular and molecular logic behind the regenerative failure of injured RGC axons in adult mammals and suggested several strategies with translational potential. This review summarizes these findings and discusses challenges for developing clinically applicable neural repair strategies.
Collapse
Affiliation(s)
- Philip R Williams
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Larry I Benowitz
- Laboratories for Neuroscience Research in Neurosurgery, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Neurosurgery, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Palo Alto, California 94303, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, Massachusetts 02115, USA; .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115, USA.,Department of Neurology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
46
|
Do JL, Allahwerdy S, David RCC, Weinreb RN, Tuszynski MH, Welsbie DS. Optic Nerve Engraftment of Neural Stem Cells. Invest Ophthalmol Vis Sci 2021; 62:30. [PMID: 34283208 PMCID: PMC8300061 DOI: 10.1167/iovs.62.9.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Purpose To evaluate the integrative potential of neural stem cells (NSCs) with the visual system and characterize effects on the survival and axonal regeneration of axotomized retinal ganglion cells (RGCs). Methods For in vitro studies, primary, postnatal rat RGCs were directly cocultured with human NSCs or cultured in NSC-conditioned media before their survival and neurite outgrowth were assessed. For in vivo studies, human NSCs were transplanted into the transected rat optic nerve, and immunohistology of the retina and optic nerve was performed to evaluate RGC survival, RGC axon regeneration, and NSC integration with the injured visual system. Results Increased neurite outgrowth was observed in RGCs directly cocultured with NSCs. NSC-conditioned media demonstrated a dose-dependent effect on RGC survival and neurite outgrowth in culture. NSCs grafted into the lesioned optic nerve modestly improved RGC survival following an optic nerve transection (593 ± 164 RGCs/mm2 vs. 199 ± 58 RGCs/mm2; P < 0.01). Additionally, RGC axonal regeneration following an optic nerve transection was modestly enhanced by NSCs transplanted at the lesion site (61.6 ± 8.5 axons vs. 40.3 ± 9.1 axons, P < 0.05). Transplanted NSCs also differentiated into neurons, received synaptic inputs from regenerating RGC axons, and extended axons along the transected optic nerve to incorporate with the visual system. Conclusions Human NSCs promote the modest survival and axonal regeneration of axotomized RGCs that is partially mediated by diffusible NSC-derived factors. Additionally, NSCs integrate with the injured optic nerve and have the potential to form neuronal relays to restore retinofugal connections.
Collapse
Affiliation(s)
- Jiun L Do
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Salam Allahwerdy
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Ryan Caezar C David
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Robert N Weinreb
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| | - Mark H Tuszynski
- Department of Neurosciences, University of California San Diego, La Jolla, California, United States.,Veterans Administration Medical Center, San Diego, California, United States
| | - Derek S Welsbie
- Hamilton Glaucoma Center, Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
47
|
Zhang J, Wu S, Jin ZB, Wang N. Stem Cell-Based Regeneration and Restoration for Retinal Ganglion Cell: Recent Advancements and Current Challenges. Biomolecules 2021; 11:biom11070987. [PMID: 34356611 PMCID: PMC8301853 DOI: 10.3390/biom11070987] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/25/2022] Open
Abstract
Glaucoma is a group of irreversible blinding eye diseases characterized by the progressive loss of retinal ganglion cells (RGCs) and their axons. Currently, there is no effective method to fundamentally resolve the issue of RGC degeneration. Recent advances have revealed that visual function recovery could be achieved with stem cell-based therapy by replacing damaged RGCs with cell transplantation, providing nutritional factors for damaged RGCs, and supplying healthy mitochondria and other cellular components to exert neuroprotective effects and mediate transdifferentiation of autologous retinal stem cells to accomplish endogenous regeneration of RGC. This article reviews the recent research progress in the above-mentioned fields, including the breakthroughs in the fields of in vivo transdifferentiation of retinal endogenous stem cells and reversal of the RGC aging phenotype, and discusses the obstacles in the clinical translation of the stem cell therapy.
Collapse
Affiliation(s)
- Jingxue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing 100730, China; (J.Z.); (S.W.)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing 100730, China; (J.Z.); (S.W.)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing 100730, China; (J.Z.); (S.W.)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
- Correspondence: (Z.-B.J.); (N.W.)
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing 100730, China; (J.Z.); (S.W.)
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China
- Correspondence: (Z.-B.J.); (N.W.)
| |
Collapse
|
48
|
Abstract
BACKGROUND Restoration of vision in patients blinded by advanced optic neuropathies requires technologies that can either 1) salvage damaged and prevent further degeneration of retinal ganglion cells (RGCs), or 2) replace lost RGCs. EVIDENCE ACQUISITION Review of scientific literature. RESULTS In this article, we discuss the different barriers to cell-replacement based strategies for optic nerve regeneration and provide an update regarding what progress that has been made to overcome them. We also provide an update on current stem cell-based therapies for optic nerve regeneration. CONCLUSIONS As neuro-regenerative and cell-transplantation based strategies for optic nerve regeneration continue to be refined, researchers and clinicians will need to work together to determine who will be a good candidate for such therapies.
Collapse
|
49
|
Trans-synaptic degeneration in the visual pathway: Neural connectivity, pathophysiology, and clinical implications in neurodegenerative disorders. Surv Ophthalmol 2021; 67:411-426. [PMID: 34146577 DOI: 10.1016/j.survophthal.2021.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 12/13/2022]
Abstract
There is a strong interrelationship between eye and brain diseases. It has been shown that neurodegenerative changes can spread bidirectionally in the visual pathway along neuronal projections. For example, damage to retinal ganglion cells in the retina leads to degeneration of the visual cortex (anterograde degeneration) and vice versa (retrograde degeneration). The underlying mechanisms of this process, known as trans-synaptic degeneration (TSD), are unknown, but TSD contributes to the progression of numerous neurodegenerative disorders, leading to clinical and functional deterioration. The hierarchical structure of the visual system comprises of a strong topographic connectivity between the retina and the visual cortex and therefore serves as an ideal model to study the cellular effect, clinical manifestations, and deterioration extent of TSD. With this review we provide comprehensive information about the neural connectivity, synapse function, molecular changes, and pathophysiology of TSD in visual pathways. We then discuss its bidirectional nature and clinical implications in neurodegenerative diseases. A thorough understanding of TSD in the visual pathway can provide insights into progression of neurodegenerative disorders and its potential as a therapeutic target.
Collapse
|
50
|
Nuzzi R, Vitale A. Cerebral Modifications in Glaucoma and Macular Degeneration: Analysis of Current Evidence in Literature and Their Implications on Therapeutic Perspectives. Eye Brain 2021; 13:159-173. [PMID: 34168513 PMCID: PMC8216745 DOI: 10.2147/eb.s307551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 04/14/2021] [Indexed: 01/19/2023] Open
Abstract
Glaucoma and macular degeneration are leading causes of irreversible blindness, significantly compromising the quality of life and having a high economic and social impact. Promising therapeutic approaches aimed at regenerating or bypassing the damaged anatomical-functional components are currently under development: these approaches have generated great expectations, but to be effective require a visual network that, despite the pathology, maintains its integrity up to the higher brain areas. In the light of this, the existing findings concerning how the central nervous system modifies its connections following the pathological damage caused by glaucoma and macular degeneration acquire great interest. This review aims to examine the scientific literature concerning the morphological and functional changes affecting the central nervous system in these pathological conditions, summarizing the evidence in an analytical way, discussing their possible causes and highlighting the potential repercussions on the current therapeutic perspectives.
Collapse
Affiliation(s)
- Raffaele Nuzzi
- Eye Clinic, Department of Surgical Sciences, University of Turin, Turin, 10126, Italy
| | - Alessio Vitale
- Eye Clinic, Department of Surgical Sciences, University of Turin, Turin, 10126, Italy
| |
Collapse
|