1
|
Yin Z, Kang J, Cheng X, Gao H, Huo S, Xu H. Investigating Müller glia reprogramming in mice: a retrospective of the last decade, and a look to the future. Neural Regen Res 2025; 20:946-959. [PMID: 38989930 PMCID: PMC11438324 DOI: 10.4103/nrr.nrr-d-23-01612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/05/2024] [Indexed: 07/12/2024] Open
Abstract
Müller glia, as prominent glial cells within the retina, plays a significant role in maintaining retinal homeostasis in both healthy and diseased states. In lower vertebrates like zebrafish, these cells assume responsibility for spontaneous retinal regeneration, wherein endogenous Müller glia undergo proliferation, transform into Müller glia-derived progenitor cells, and subsequently regenerate the entire retina with restored functionality. Conversely, Müller glia in the mouse and human retina exhibit limited neural reprogramming. Müller glia reprogramming is thus a promising strategy for treating neurodegenerative ocular disorders. Müller glia reprogramming in mice has been accomplished with remarkable success, through various technologies. Advancements in molecular, genetic, epigenetic, morphological, and physiological evaluations have made it easier to document and investigate the Müller glia programming process in mice. Nevertheless, there remain issues that hinder improving reprogramming efficiency and maturity. Thus, understanding the reprogramming mechanism is crucial toward exploring factors that will improve Müller glia reprogramming efficiency, and for developing novel Müller glia reprogramming strategies. This review describes recent progress in relatively successful Müller glia reprogramming strategies. It also provides a basis for developing new Müller glia reprogramming strategies in mice, including epigenetic remodeling, metabolic modulation, immune regulation, chemical small-molecules regulation, extracellular matrix remodeling, and cell-cell fusion, to achieve Müller glia reprogramming in mice.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | | | | | | | | | | |
Collapse
|
2
|
Yin Z, Kang J, Xu H, Huo S, Xu H. Recent progress of principal techniques used in the study of Müller glia reprogramming in mice. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:30. [PMID: 39663301 PMCID: PMC11635068 DOI: 10.1186/s13619-024-00211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024]
Abstract
In zebrafish, Müller glia (MG) cells retain the ability to proliferate and de-differentiate into retinal progenitor-like cells, subsequently differentiating into retinal neurons that can replace those damaged or lost due to retinal injury. In contrast, the reprogramming potential of MG in mammals has been lost, with these cells typically responding to retinal damage through gliosis. Considerable efforts have been dedicated to achieving the reprogramming of MG cells in mammals. Notably, significant advancements have been achieved in reprogramming MG cells in mice employing various methodologies. At the same time, some inevitable challenges have hindered identifying accurate MG cell reprogramming rather than the illusion, let alone improving the reprogramming efficiency and maturity of daughter cells. Recently, several strategies, including lineage tracking, multi-omics techniques, and functional analysis, have been developed to investigate the MG reprogramming process in mice. This review summarizes both the advantages and limitations of these novel strategies for analyzing MG reprogramming in mice, offering insights into enhancing the reliability and efficiency of MG reprogramming.
Collapse
Affiliation(s)
- Zhiyuan Yin
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Jiahui Kang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China
| | - Haoan Xu
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shujia Huo
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| | - Haiwei Xu
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, P.R. China.
| |
Collapse
|
3
|
Huang X, Luodan A, Gao H, He J, Ge L, Cha Z, Gong H, Lin X, Li H, Tang Y, Jiang D, Fan X, Xu H. Mitochondrial transfer between BMSCs and Müller promotes mitochondrial fusion and suppresses gliosis in degenerative retina. iScience 2024; 27:110309. [PMID: 39055937 PMCID: PMC11269791 DOI: 10.1016/j.isci.2024.110309] [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: 12/26/2023] [Revised: 04/19/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
Mitochondrial dysfunction and Müller cells gliosis are significant pathological characteristics of retinal degeneration (RD) and causing blinding. Stem cell therapy is a promising treatment for RD, the recently accepted therapeutic mechanism is cell fusion induced materials transfer. However, whether materials including mitochondrial transfer between grafted stem cells and recipient's cells contribute to suppressing gliosis and mechanism are unclear. In present study, we demonstrated that bone marrow mesenchymal stem cells (BMSCs) transferred mitochondria to Müller cells by cell fusion and tunneling nanotubes. BMSCs-derived mitochondria (BMSCs-mito) were integrated into mitochondrial network of Müller cells, improving mitochondrial function, reducing oxidative stress and gliosis, which protected visual function partially in the degenerative rat retina. RNA sequencing analysis revealed that BMSCs-mito increased mitochondrial DNA (mtDNA) content and facilitated mitochondrial fusion in damaged Müller cells. It suggests that mitochondrial transfer from BMSCs remodels Müller cells metabolism and suppresses gliosis; thus, delaying the degenerative progression of RD.
Collapse
Affiliation(s)
- Xiaona Huang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Luodan A
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Hui Gao
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Juncai He
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Zhe Cha
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Hong Gong
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Lin
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Huiting Li
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Yongping Tang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Dan Jiang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
4
|
Kumazaki T, Yonekawa C, Tsubouchi T. Microscopic Analysis of Cell Fate Alteration Induced by Cell Fusion. Cell Reprogram 2023; 25:251-259. [PMID: 37847898 DOI: 10.1089/cell.2023.0073] [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: 10/19/2023] Open
Abstract
In mammals, differentiated cells generally do not de-differentiate nor undergo cell fate alterations. However, they can be experimentally guided toward a different lineage. Cell fusion involving two different cell types has long been used to study this process, as this method induces cell fate alterations within hours to days in a subpopulation of fused cells, as evidenced by changes in gene-expression profiles. Despite the robustness of this system, its use has been restricted by low fusion rates and difficulty in eliminating unfused populations, thereby compromising resolution. In this study, we address these limitations by isolating fused cells using antibody-conjugated beads. This approach enables the microscopic tracking of fused cells starting as early as 5 hours after fusion. By taking advantage of species-specific FISH probes, we show that a small population of fused cells resulting from the fusion of mouse ES and human B cells, expresses OCT4 from human nuclei at levels comparable to human induced pluripotent stem cells (iPSCs) as early as 25 hours after fusion. We also show that this response can vary depending on the fusion partner. Our study broadens the usage of the cell fusion system for comprehending the mechanisms underlying cell fate alterations. These findings hold promise for diverse fields, including regenerative medicine and cancer.
Collapse
Affiliation(s)
- Taisei Kumazaki
- Laboratory of Stem Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Japan
- The Graduate University for Advanced Studies, SOKENDAI, Shonah Village, Hayama, 240-0193, Japan
| | - Chinatsu Yonekawa
- Laboratory of Stem Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Japan
| | - Tomomi Tsubouchi
- Laboratory of Stem Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Japan
- The Graduate University for Advanced Studies, SOKENDAI, Shonah Village, Hayama, 240-0193, Japan
| |
Collapse
|
5
|
Li R, Liu J, Yi P, Yang X, Chen J, Zhao C, Liao X, Wang X, Xu Z, Lu H, Li H, Zhang Z, Liu X, Xiang J, Hu K, Qi H, Yu J, Yang P, Hou S. Integrative Single-Cell Transcriptomics and Epigenomics Mapping of the Fetal Retina Developmental Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206623. [PMID: 37017569 DOI: 10.1002/advs.202206623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/24/2023] [Indexed: 06/04/2023]
Abstract
The underlying mechanisms that determine gene expression and chromatin accessibility in retinogenesis are poorly understood. Herein, single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing are performed on human embryonic eye samples obtained 9-26 weeks after conception to explore the heterogeneity of retinal progenitor cells (RPCs) and neurogenic RPCs. The differentiation trajectory from RPCs to 7 major types of retinal cells are verified. Subsequently, diverse lineage-determining transcription factors are identified and their gene regulatory networks are refined at the transcriptomic and epigenomic levels. Treatment of retinospheres, with the inhibitor of RE1 silencing transcription factor, X5050, induces more neurogenesis with the regular arrangement, and a decrease in Müller glial cells. The signatures of major retinal cells and their correlation with pathogenic genes associated with multiple ocular diseases, including uveitis and age-related macular degeneration are also described. A framework for the integrated exploration of single-cell developmental dynamics of the human primary retina is provided.
Collapse
Affiliation(s)
- Ruonan Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Jiangyi Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Xianli Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Jun Chen
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Chenyang Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Xingyun Liao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, P. R. China
| | - Xiaotang Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Zongren Xu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
| | - Huiping Lu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Hongshun Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Zhi Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Junjie Xiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Hongbo Qi
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jia Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, P. R. China
- The Key Laboratory of RNA and Hematopoietic Regulation, Chinese Academy of Medical Sciences, Beijing, 100005, P. R. China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, P. R. China
| |
Collapse
|
6
|
Magner E, Sandoval-Sanchez P, Kramer AC, Thummel R, Hitchcock PF, Taylor SM. Disruption of miR-18a Alters Proliferation, Photoreceptor Replacement Kinetics, Inflammatory Signaling, and Microglia/Macrophage Numbers During Retinal Regeneration in Zebrafish. Mol Neurobiol 2022; 59:2910-2931. [PMID: 35246819 PMCID: PMC9018604 DOI: 10.1007/s12035-022-02783-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/24/2022] [Indexed: 10/18/2022]
Abstract
In mammals, photoreceptor loss causes permanent blindness, but in zebrafish (Danio rerio), photoreceptor loss reprograms Müller glia to function as stem cells, producing progenitors that regenerate photoreceptors. MicroRNAs (miRNAs) regulate CNS neurogenesis, but the roles of miRNAs in injury-induced neuronal regeneration are largely unknown. In the embryonic zebrafish retina, miR-18a regulates photoreceptor differentiation. The purpose of the current study was to determine, in zebrafish, the function of miR-18a during injury-induced photoreceptor regeneration. RT-qPCR, in situ hybridization, and immunohistochemistry showed that miR-18a expression increases throughout the retina between 1 and 5 days post-injury (dpi). To test miR-18a function during photoreceptor regeneration, we used homozygous miR-18a mutants (miR-18ami5012), and knocked down miR-18a with morpholino oligonucleotides. During photoreceptor regeneration, miR-18ami5012 retinas have fewer mature photoreceptors than WT at 7 and 10 dpi, but there is no difference at 14 dpi, indicating that photoreceptor regeneration is delayed. Labeling dividing cells with 5-bromo-2'-deoxyuridine (BrdU) showed that at 7 and 10 dpi, there are excess dividing progenitors in both mutants and morphants, indicating that miR-18a negatively regulates injury-induced proliferation. Tracing 5-ethynyl-2'-deoxyuridine (EdU) and BrdU-labeled cells showed that in miR-18ami5012 retinas excess progenitors migrate to other retinal layers in addition to the photoreceptor layer. Inflammation is critical for photoreceptor regeneration, and RT-qPCR showed that in miR-18ami5012 retinas, inflammatory gene expression and microglia activation are prolonged. Suppressing inflammation with dexamethasone rescues the miR-18ami5012 phenotype. Together, these data show that in the injured zebrafish retina, disruption of miR-18a alters proliferation, inflammation, the microglia/macrophage response, and the timing of photoreceptor regeneration.
Collapse
Affiliation(s)
- Evin Magner
- Plant and Microbial Biology, University of Minnesota, 1479 Gortner Avenue, St. Paul, MN, 55108, USA
| | - Pamela Sandoval-Sanchez
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, FL, 32514, USA
| | - Ashley C Kramer
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Ryan Thummel
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Peter F Hitchcock
- Department of Ophthalmology and Visual Sciences, University of Michigan, W. K. Kellogg Eye Center, 1000 Wall Street, Ann Arbor, MI, 48105, USA
| | - Scott M Taylor
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, FL, 32514, USA.
| |
Collapse
|
7
|
Bonilla-Pons SÀ, Nakagawa S, Bahima EG, Fernández-Blanco Á, Pesaresi M, D'Antin JC, Sebastian-Perez R, Greco D, Domínguez-Sala E, Gómez-Riera R, Compte RIB, Dierssen M, Pulido NM, Cosma MP. Müller glia fused with adult stem cells undergo neural differentiation in human retinal models. EBioMedicine 2022; 77:103914. [PMID: 35278743 PMCID: PMC8917309 DOI: 10.1016/j.ebiom.2022.103914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 02/09/2022] [Accepted: 02/18/2022] [Indexed: 12/15/2022] Open
Abstract
Background Visual impairments are a critical medical hurdle to be addressed in modern society. Müller glia (MG) have regenerative potential in the retina in lower vertebrates, but not in mammals. However, in mice, in vivo cell fusion between MG and adult stem cells forms hybrids that can partially regenerate ablated neurons. Methods We used organotypic cultures of human retina and preparations of dissociated cells to test the hypothesis that cell fusion between human MG and adult stem cells can induce neuronal regeneration in human systems. Moreover, we established a microinjection system for transplanting human retinal organoids to demonstrate hybrid differentiation. Findings We first found that cell fusion occurs between MG and adult stem cells, in organotypic cultures of human retina as well as in cell cultures. Next, we showed that the resulting hybrids can differentiate and acquire a proto-neural electrophysiology profile when the Wnt/beta-catenin pathway is activated in the adult stem cells prior fusion. Finally, we demonstrated the engraftment and differentiation of these hybrids into human retinal organoids. Interpretation We show fusion between human MG and adult stem cells, and demonstrate that the resulting hybrid cells can differentiate towards neural fate in human model systems. Our results suggest that cell fusion-mediated therapy is a potential regenerative approach for treating human retinal dystrophies. Funding This work was supported by La Caixa Health (HR17-00231), Velux Stiftung (976a) and the Ministerio de Ciencia e Innovación, (BFU2017-86760-P) (AEI/FEDER, UE), AGAUR (2017 SGR 689, 2017 SGR 926).
Collapse
Affiliation(s)
- Sergi Àngel Bonilla-Pons
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat de Barcelona (UB), Barcelona, Spain
| | - Shoma Nakagawa
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain
| | - Elena Garreta Bahima
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Álvaro Fernández-Blanco
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Martina Pesaresi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Justin Christopher D'Antin
- Centro de Oftalmología Barraquer, Barcelona, Spain; Institut Universitari Barraquer, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Ruben Sebastian-Perez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Daniela Greco
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain
| | - Eduardo Domínguez-Sala
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain
| | - Raúl Gómez-Riera
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain
| | - Rafael Ignacio Barraquer Compte
- Centro de Oftalmología Barraquer, Barcelona, Spain; Institut Universitari Barraquer, Universitat Autónoma de Barcelona, Barcelona, Spain
| | - Mara Dierssen
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Madrid, Spain
| | - Nuria Montserrat Pulido
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, C/Dr. Aiguader 88, Barcelona 08003, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; ICREA, Pg. Lluis Companys 23, Barcelona 08010, Spain; Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China; CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell an Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Science, Guangzhou 510530, China.
| |
Collapse
|
8
|
Alfarhan M, Liu F, Shan S, Pichavaram P, Somanath PR, Narayanan SP. Pharmacological Inhibition of Spermine Oxidase Suppresses Excitotoxicity Induced Neuroinflammation in Mouse Retina. Int J Mol Sci 2022; 23:2133. [PMID: 35216248 PMCID: PMC8875684 DOI: 10.3390/ijms23042133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/10/2022] [Accepted: 02/11/2022] [Indexed: 02/07/2023] Open
Abstract
Polyamine oxidation plays a major role in neurodegenerative diseases. Previous studies from our laboratory demonstrated that spermine oxidase (SMOX, a member of the polyamine oxidase family) inhibition using MDL 72527 reduced neurodegeneration in models of retinal excitotoxicity and diabetic retinopathy. However, the mechanisms behind the neuroprotection offered by SMOX inhibition are not completely studied. Utilizing the experimental model of retinal excitotoxicity, the present study determined the impact of SMOX blockade in retinal neuroinflammation. Our results demonstrated upregulation in the number of cells positive for Iba-1 (ionized calcium-binding adaptor molecule 1), CD (Cluster Differentiation) 68, and CD16/32 in excitotoxicity-induced retinas, while MDL 72527 treatment reduced these changes, along with increases in the number of cells positive for Arginase1 and CD206. When retinal excitotoxicity upregulated several pro-inflammatory genes, MDL 72527 treatment reduced many of them and increased anti-inflammatory genes. Furthermore, SMOX inhibition upregulated antioxidant signaling (indicated by elevated Nrf2 and HO-1 levels) and reduced protein-conjugated acrolein in excitotoxic retinas. In vitro studies using C8-B4 cells showed changes in cellular morphology and increased reactive oxygen species formation in response to acrolein (a product of SMOX activity) treatment. Overall, our findings indicate that the inhibition SMOX pathway reduced neuroinflammation and upregulated antioxidant signaling in the retina.
Collapse
Affiliation(s)
- Moaddey Alfarhan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Fang Liu
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| | - Shengshuai Shan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| | | | - Payaningal R. Somanath
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics Program, Department of Clinical and Administrative Pharmacy, University of Georgia, Augusta, GA 30912, USA; (M.A.); (F.L.); (S.S.); (P.R.S.)
- Research Division, Charlie Norwood VA Medical Center, Augusta, GA 30901, USA
- Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA;
| |
Collapse
|
9
|
Ohno Y, Yako T, Satoh K, Nagase H, Shitara A, Hara H, Kashimata M. Retinal damage alters gene expression profile in lacrimal glands of mice. J Pharmacol Sci 2022; 149:20-26. [DOI: 10.1016/j.jphs.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 10/19/2022] Open
|
10
|
Tan S, Yao Y, Yang Q, Yuan XL, Cen LP, Ng TK. Diversified Treatment Options of Adult Stem Cells for Optic Neuropathies. Cell Transplant 2022; 31. [PMID: 36165292 PMCID: PMC9523835 DOI: 10.1177/09636897221123512] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 07/28/2022] [Accepted: 08/16/2022] [Indexed: 02/05/2023] Open
Abstract
Optic neuropathies refer to a group of ocular disorders with abnormalities or dysfunction of the optic nerve, sharing a common pathophysiology of retinal ganglion cell (RGC) death and axonal loss. RGCs, as the retinal neurons in the central nervous system, show limited capacity in regeneration or recovery upon diseases or after injuries. Critically, there is still no effective clinical treatment to cure most types of optic neuropathies. Recently, stem cell therapy was proposed as a potential treatment strategy for optic neuropathies. Adult stem cells, including mesenchymal stem cells and hematopoietic stem cells, have been applied in clinical trials based on their neuroprotective properties. In this article, the applications of adult stem cells on different types of optic neuropathies and the related mechanisms will be reviewed. Research updates on the strategies to enhance the neuroprotective effects of human adult stem cells will be summarized. This review article aims to enlighten the research scientists on the diversified functions of adult stem cells and consideration of adult stem cells as a potential treatment for optic neuropathies in future clinical practices.
Collapse
Affiliation(s)
- Shaoying Tan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong
- Research Centre for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Yao Yao
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Qichen Yang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| | - Xiang-Ling Yuan
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Ling-Ping Cen
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| | - Tsz Kin Ng
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
- Shantou University Medical College, Shantou, China
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, Kowloon, Hong Kong
| |
Collapse
|
11
|
Too LK, Simunovic MP. Retinal Stem/Progenitor Cells Derived From Adult Müller Glia for the Treatment of Retinal Degeneration. Front Cell Dev Biol 2021; 9:749131. [PMID: 34660607 PMCID: PMC8511496 DOI: 10.3389/fcell.2021.749131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/06/2021] [Indexed: 01/09/2023] Open
Abstract
Over the past two decades, progress in our understanding of glial function has been revolutionary. Within the retina, a subset of glial cells termed the “Müller glia (MG),” have been demonstrated to play key roles in retinal homeostasis, structure and metabolism. Additionally, MG have also been shown to possess the regenerative capacity that varies across species. In teleost fish, MG respond to injury by reprogramming into stem-like cells capable of regenerating lost tissue. The expression of stem/progenitor cell markers has been demonstrated broadly in mammalian MG, including human MG, but their in vivo regenerative capacity appears evolutionarily limited. Advances in stem cell therapy have progressively elucidated critical mechanisms underlying innate MG reprogramming in teleost fish, which have shown promising results when applied to rodents. Furthermore, when cultured ex vivo, MG from mammals can differentiate into several retina cell types. In this review, we will explore the reparative and regenerative potential of MG in cellular therapy approaches, and outline our current understanding of embryonic retinal development, the stem-cell potential of MG in adult vertebrate retina (including human), and microenvironmental cues that guide MG reprogramming.
Collapse
Affiliation(s)
- Lay Khoon Too
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Matthew P Simunovic
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Sydney Eye Hospital, Sydney, NSW, Australia
| |
Collapse
|
12
|
Nagashima M, Hitchcock PF. Inflammation Regulates the Multi-Step Process of Retinal Regeneration in Zebrafish. Cells 2021; 10:cells10040783. [PMID: 33916186 PMCID: PMC8066466 DOI: 10.3390/cells10040783] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/20/2022] Open
Abstract
The ability to regenerate tissues varies between species and between tissues within a species. Mammals have a limited ability to regenerate tissues, whereas zebrafish possess the ability to regenerate almost all tissues and organs, including fin, heart, kidney, brain, and retina. In the zebrafish brain, injury and cell death activate complex signaling networks that stimulate radial glia to reprogram into neural stem-like cells that repair the injury. In the retina, a popular model for investigating neuronal regeneration, Müller glia, radial glia unique to the retina, reprogram into stem-like cells and undergo a single asymmetric division to generate multi-potent retinal progenitors. Müller glia-derived progenitors then divide rapidly, numerically matching the magnitude of the cell death, and differentiate into the ablated neurons. Emerging evidence reveals that inflammation plays an essential role in this multi-step process of retinal regeneration. This review summarizes the current knowledge of the inflammatory events during retinal regeneration and highlights the mechanisms whereby inflammatory molecules regulate the quiescence and division of Müller glia, the proliferation of Müller glia-derived progenitors and the survival of regenerated neurons.
Collapse
|
13
|
Stem Cell Ophthalmology Treatment Study (SCOTS): Bone Marrow-Derived Stem Cells in the Treatment of Stargardt Disease. MEDICINES 2021; 8:medicines8020010. [PMID: 33546345 PMCID: PMC7913552 DOI: 10.3390/medicines8020010] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/23/2021] [Accepted: 01/25/2021] [Indexed: 12/15/2022]
Abstract
Background: Stargardt Disease is the most common inherited macular degeneration, typically resulting in progressive central vision loss and legal blindness at an early age. We report regarding 34 eyes with Stargardt Disease treated in the Stem Cell Ophthalmology Treatment Study (SCOTS and SCOTS2). Methods: Autologous bone marrow was processed, separating the stem cell fraction which was provided Arms using retrobulbar, subtenons, intravitreal or subretinal and intravenous. The follow-up period was one year. Results: Of the 34 treated eyes, 21 (61.8%) improved, 8 (23.5%) remained stable, and 5 (14.7%) showed continued progression of their disease. Results were statistically significant with p = 0.0004. The average central vision improvement following treatment was 17.96% (95%CI, 16.39–19.53%) and ranged up to 80.5%. Of 17 patients treated, 13 (76.5%) showed visual acuity improvement in one or both eyes, 3 patients (17.6%) showed no net loss, and 1 worsened as a consequence of disease progression; 94.1% of patients had improved vision or remained stable. There were no adverse events. Conclusions: Patients with Stargardt Disease may potentially benefit from autologous bone marrow-derived stem cells (BMSC) as provided in SCOTS. Improvement or stabilization of vision was found to occur for the vast majority of reported patients and findings were highly statistically significant.
Collapse
|
14
|
Gao H, A L, Huang X, Chen X, Xu H. Müller Glia-Mediated Retinal Regeneration. Mol Neurobiol 2021; 58:2342-2361. [PMID: 33417229 DOI: 10.1007/s12035-020-02274-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/22/2020] [Indexed: 12/18/2022]
Abstract
Müller glia originate from neuroepithelium and are the principal glial cells in the retina. During retinal development, Müller glia are one of the last cell types to be born. In lower vertebrates, such as zebrafish, Müller glia possess a remarkable capacity for retinal regeneration following various forms of injury through a reprogramming process in which endogenous Müller glia proliferate and differentiate into all types of retinal cells. In mammals, Müller glia become reactive in response to damage to protect or to further impair retinal function. Although mammalian Müller glia have regenerative potential, it is limited as far as repairing damaged retina. Lessons learned from zebrafish will help reveal the critical mechanisms involved in Müller glia reprogramming. Progress has been made in triggering Müller glia to reprogram and generate functional neurons to restore vision in mammals indicating that Müller glia reprogramming may be a promising therapeutic strategy for human retinal diseases. This review comprehensively summarizes the mechanisms related to retinal regeneration in model animals and the critical advanced progress made in Müller glia reprogramming in mammals.
Collapse
Affiliation(s)
- Hui Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Luodan A
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xiaona Huang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xi Chen
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
15
|
Digesting the Importance of Cell Fusion in the Intestine. Cell Mol Gastroenterol Hepatol 2020; 11:299-302. [PMID: 33068773 PMCID: PMC7768557 DOI: 10.1016/j.jcmgh.2020.07.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/15/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
|
16
|
Toms M, Pagarkar W, Moosajee M. Usher syndrome: clinical features, molecular genetics and advancing therapeutics. Ther Adv Ophthalmol 2020; 12:2515841420952194. [PMID: 32995707 PMCID: PMC7502997 DOI: 10.1177/2515841420952194] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/27/2020] [Indexed: 01/12/2023] Open
Abstract
Usher syndrome has three subtypes, each being clinically and genetically heterogeneous characterised by sensorineural hearing loss and retinitis pigmentosa (RP), with or without vestibular dysfunction. It is the most common cause of deaf–blindness worldwide with a prevalence of between 4 and 17 in 100 000. To date, 10 causative genes have been identified for Usher syndrome, with MYO7A accounting for >50% of type 1 and USH2A contributing to approximately 80% of type 2 Usher syndrome. Variants in these genes can also cause non-syndromic RP and deafness. Genotype–phenotype correlations have been described for several of the Usher genes. Hearing loss is managed with hearing aids and cochlear implants, which has made a significant improvement in quality of life for patients. While there is currently no available approved treatment for the RP, various therapeutic strategies are in development or in clinical trials for Usher syndrome, including gene replacement, gene editing, antisense oligonucleotides and small molecule drugs.
Collapse
Affiliation(s)
- Maria Toms
- UCL Institute of Ophthalmology, London, UK; The Francis Crick Institute, London, UK
| | - Waheeda Pagarkar
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK; University College London Hospitals NHS Foundation Trust, London, UK
| | - Mariya Moosajee
- Development, Ageing and Disease, UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| |
Collapse
|
17
|
Stem Cell Ophthalmology Treatment Study (SCOTS): Bone Marrow-Derived Stem Cells in the Treatment of Age-Related Macular Degeneration. MEDICINES 2020; 7:medicines7040016. [PMID: 32231088 PMCID: PMC7235782 DOI: 10.3390/medicines7040016] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/23/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
Background: Dry age-related macular degeneration (AMD) is one of the leading causes of vision loss in older patients. The macula accumulates drusen with loss of retinal pigment epithelial cells and photoreceptors. Abnormal subretinal neovascularization is absent. There is no effective drug therapy for dry AMD and a large proportion of patients progress to legal blindness from macular atrophy. The Stem Cell Ophthalmology Treatment Study (SCOTS) was conducted to assess the effect of bone marrow-derived stem cells (BMSCs) on dry AMD and other retinal and optic nerve diseases. Methods: Thirty-two eyes were treated with BMSC per the protocols in SCOTS. Provision of BMSCs in Arm 1 was via retrobulbar (RB), sub-tenons (ST) and intravenous (IV); Arm 2 via intravitreal, RB, ST and IV; Arm 3 via subretinal and IV. Patient age averaged 78 years old and ranged from 69 to 90. Visual acuity preoperatively ranged from counting fingers to 20/50-2 with an average preoperative LogMAR of 1.125. Results: Following treatment, 20 of 32 (63%) of eyes experienced improvement in visual acuity averaging 27.6% on LogMAR and ranging from 2.5% to 44.6%. The mean improvement in LogMAR was 0.963 with a standard deviation (SD) of 0.42. The visual acuity remained stable in 34% of treated eyes. One eye continued to worsen as a consequence of disease progression. The results showed high statistical significance with p ≤ 0.001. The procedures were conducted safely, and no complications were observed. Conclusion: Treatment of dry AMD with BMSC using the protocols developed in the SCOTS clinical trial has shown statistically significant clinical benefit improving visual acuity and potentially delaying visual loss in the disease.
Collapse
|
18
|
Ofenbauer A, Tursun B. Strategies for in vivo reprogramming. Curr Opin Cell Biol 2019; 61:9-15. [DOI: 10.1016/j.ceb.2019.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 06/14/2019] [Indexed: 02/08/2023]
|
19
|
Cellular Reprogramming as a Therapeutic Target in Cancer. Trends Cell Biol 2019; 29:623-634. [PMID: 31153655 DOI: 10.1016/j.tcb.2019.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 04/28/2019] [Accepted: 05/01/2019] [Indexed: 12/30/2022]
Abstract
Cancer heterogeneity has long been recognized as an important clinical determinant of patient outcomes and, thus, many new cancer treatments have been designed to target these different cells. Despite the short-term achievements of current therapies, including chemotherapy, antiangiogenesis therapy, radiotherapy, and immunotherapy, the long-term success of cancer regression remains poor. Therefore, researchers have investigated a new property, cellular reprogramming, in cancer that not only contributes to the classic hallmarks of cancer, but also suggests that cancer is a dynamic event rather than a static cellular entity. Here, we discuss the mechanisms by which the cellular reprogramming of cancer cells can explain some of the phenotypic and functional heterogeneity observed among cancer cells.
Collapse
|
20
|
Grigoryan EN. Endogenous Cell Sources for Eye Retina Regeneration in Vertebrate Animals and Humans. Russ J Dev Biol 2019. [DOI: 10.1134/s106236041901003x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
21
|
Pesaresi M, Sebastian-Perez R, Cosma MP. Dedifferentiation, transdifferentiation and cell fusion: in vivo reprogramming strategies for regenerative medicine. FEBS J 2018; 286:1074-1093. [PMID: 30103260 DOI: 10.1111/febs.14633] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 07/01/2018] [Accepted: 08/10/2018] [Indexed: 12/23/2022]
Abstract
Regenerative capacities vary enormously across the animal kingdom. In contrast to most cold-blooded vertebrates, mammals, including humans, have very limited regenerative capacity when it comes to repairing damaged or degenerating tissues. Here, we review the main mechanisms of tissue regeneration, underlying the importance of cell dedifferentiation and reprogramming. We discuss the significance of cell fate and identity changes in the context of regenerative medicine, with a particular focus on strategies aiming at the promotion of the body's self-repairing mechanisms. We also introduce some of the most recent advances that have resulted in complete reprogramming of cell identity in vivo. Lastly, we discuss the main challenges that need to be addressed in the near future to develop in vivo reprogramming approaches with therapeutic potential.
Collapse
Affiliation(s)
- Martina Pesaresi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Spain
| | - Ruben Sebastian-Perez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
22
|
Pesaresi M, Bonilla-Pons SA, Cosma MP. In vivo somatic cell reprogramming for tissue regeneration: the emerging role of the local microenvironment. Curr Opin Cell Biol 2018; 55:119-128. [PMID: 30071468 DOI: 10.1016/j.ceb.2018.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/01/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
The past few years have witnessed an exponential increase of interest in the reprogramming process. This has been motivated by the enthusiasm of unravelling key aspects not only of cell identity and dedifferentiation, but also of the endogenous regenerative capacities of mammalian organs. Here, we present the most recent advances in the field of reprogramming, stressing how they are re-defining the rules of cell fate and plasticity in vivo. Specifically, we focus on the emerging role of the tissue microenvironment, with particular emphasis on tissue damage, inflammation and senescence that can facilitate in vivo reprogramming and regeneration through cell-extrinsic mechanisms.
Collapse
Affiliation(s)
- Martina Pesaresi
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Sergi A Bonilla-Pons
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain
| | - Maria Pia Cosma
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain; Universitat Pompeu Fabra (UPF), Dr Aiguader 88, 08003 Barcelona, Spain; ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain.
| |
Collapse
|