1
|
Zhang J, Wang J, Zhou Q, Chen Z, Zhuang J, Zhao X, Gan Z, Wang Y, Wang C, Molday RS, Yang YT, Li X, Zhao XM. Spatiotemporally resolved transcriptomics reveals the cellular dynamics of human retinal development. Nat Commun 2025; 16:2307. [PMID: 40055379 PMCID: PMC11889126 DOI: 10.1038/s41467-025-57625-9] [Citation(s) in RCA: 1] [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: 05/02/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
The morphogenesis and cellular interactions in developing retina are incompletely characterized. The full understanding needs a precise mapping of the gene expression with a single-cell spatial resolution. Here, we present a spatial transcriptomic (ST) resource for the developing human retina at six developmental stages. Combining the spatial and single-cell transcriptomic data enables characterization of the cell-type-specific expression profiles at distinct anatomical regions at each developmental stage, highlighting the spatiotemporal dynamics of cellular composition during retinal development. All the ST spots are catalogued into consensus spatial domains, which are further associated to their specific expression signatures and biological functions associated with neuron and eye development. We prioritize a set of critical regulatory genes for the transitions of spatial domains during retinal development. Differentially expressed genes from different spatial domains are associated with distinct retinal diseases, indicating the biological relevance and clinical significance of the spatially defined gene expression. Finally, we reconstruct the spatial cellular communication networks, and highlight critical ligand-receptor interactions during retinal development. Overall, our study reports the spatiotemporal dynamics of gene expression and cellular profiles during retinal development, and provides a rich resource for the future studies on retinogenesis.
Collapse
Affiliation(s)
- Jinglong Zhang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qiongjie Zhou
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zixin Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingzhong Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ziquan Gan
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yinan Wang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chunxiu Wang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Robert S Molday
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology & Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yucheng T Yang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Xiaotian Li
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
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
|
3
|
Aparicio JG, Hopp H, Harutyunyan N, Stewart C, Cobrinik D, Borchert M. Aberrant gene expression yet undiminished retinal ganglion cell genesis in iPSC-derived models of optic nerve hypoplasia. Ophthalmic Genet 2024; 45:1-15. [PMID: 37807874 PMCID: PMC10841193 DOI: 10.1080/13816810.2023.2253902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Optic nerve hypoplasia (ONH), the leading congenital cause of permanent blindness, is characterized by a retinal ganglion cell (RGC) deficit at birth. Multifactorial developmental events are hypothesized to underlie ONH and its frequently associated neurologic and endocrine abnormalities; however, environmental influences are unclear and genetic underpinnings are unexplored. This work investigates the genetic contribution to ONH RGC production and gene expression using patient induced pluripotent stem cell (iPSC)-derived retinal organoids (ROs). MATERIALS AND METHODS iPSCs produced from ONH patients and controls were differentiated to ROs. RGC genesis was assessed using immunofluorescence and flow cytometry. Flow-sorted BRN3+ cells were collected for RNA extraction for RNA-Sequencing. Differential gene expression was assessed using DESeq2 and edgeR. PANTHER was employed to identify statistically over-represented ontologies among the differentially expressed genes (DEGs). DEGs of high interest to ONH were distinguished by assessing function, mutational constraint, and prior identification in ONH, autism and neurodevelopmental disorder (NDD) studies. RESULTS RGC genesis and survival were similar in ONH and control ROs. Differential expression of 70 genes was identified in both DESeq2 and edgeR analyses, representing a ~ 4-fold higher percentage of DEGs than in randomized study participants. DEGs showed trends towards over-representation of validated NDD genes and ONH exome variant genes. Among the DEGs, RAPGEF4 and DMD had the greatest number of disease-relevant features. CONCLUSIONS ONH genetic background was not associated with impaired RGC genesis but was associated with DEGs exhibiting disease contribution potential. This constitutes some of the first evidence of a genetic contribution to ONH.
Collapse
Affiliation(s)
- Jennifer G. Aparicio
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Hanno Hopp
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Narine Harutyunyan
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Carly Stewart
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - David Cobrinik
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of
Medicine, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mark Borchert
- The Vision Center and The Saban Research Institute,
Children’s Hospital Los Angeles, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck
School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Zhao H, Yan F. Retinal Organoids: A Next-Generation Platform for High-Throughput Drug Discovery. Stem Cell Rev Rep 2024; 20:495-508. [PMID: 38079086 PMCID: PMC10837228 DOI: 10.1007/s12015-023-10661-8] [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] [Accepted: 11/28/2023] [Indexed: 02/03/2024]
Abstract
Retinal diseases are leading causes of blindness globally. Developing new drugs is of great significance for preventing vision loss. Current drug discovery relies mainly on two-dimensional in vitro models and animal models, but translation to human efficacy and safety is biased. In recent years, the emergence of retinal organoid technology platforms, utilizing three-dimensional microenvironments to better mimic retinal structure and function, has provided new platforms for exploring pathogenic mechanisms and drug screening. This review summarizes the latest advances in retinal organoid technology, emphasizing its application advantages in high-throughput drug screening, efficacy and toxicity evaluation, and translational medicine research. The review also prospects the combination of emerging technologies such as organ-on-a-chip, 3D bioprinting, single cell sequencing, gene editing with retinal organoid technology, which is expected to further optimize retinal organoid models and advance the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Hongkun Zhao
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| | - Fei Yan
- Department of Pathology and Pathophysiology, Faculty of Basic Medicine School, Kunming Medical University, 1168 Yuhua Street, Chunrong West Road, Chenggong District, Kunming, Yunnan, 650500, China.
| |
Collapse
|
5
|
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: 0.5] [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
|
6
|
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: 5] [Impact Index Per Article: 2.5] [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
|
7
|
Gong J, Gong Y, Zou T, Zeng Y, Yang C, Mo L, Kang J, Fan X, Xu H, Yang J. A controllable perfusion microfluidic chip for facilitating the development of retinal ganglion cells in human retinal organoids. LAB ON A CHIP 2023; 23:3820-3836. [PMID: 37496497 DOI: 10.1039/d3lc00054k] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Retinal organoids (ROs) derived from human pluripotent stem cells (hPSCs) have become a promising model in vitro to recapitulate human retinal development, which can be further employed to explore the mechanisms of retinal diseases. However, the current culture systems for ROs lack physiologically relevant microenvironments, such as controllable mechano-physiological cues and dynamic feedback between cells and the extracellular matrix (ECM), which limits the accurate control of RO development. Therefore, we designed a controllable perfusion microfluidic chip (CPMC) with the advantages of precisely controlling fluidic shear stress (FSS) and oxygen concentration distribution in a human embryonic stem cell (hESC)-derived RO culture system. We found that ROs cultured under this system allow for expanding the retinal progenitor cell (RPC) pool, orchestrating the retinal ganglion cell (RGC) specification, and axon growth without disturbing the spatial and temporal patterning events at the early stage of RO development. Furthermore, RNA sequencing data revealed that the activation of voltage-gated ion channels and the increased expression of ECM components synergistically improve the growth of ROs and facilitate the differentiation of RGCs. This study elaborates on the advantages of the designed CPMC to promote RO growth and provide a controllable and reliable platform for the efficient maturity of RGCs in the ROs, promising applications in modeling RGC-related disorders, drug screening, and cell transplantation.
Collapse
Affiliation(s)
- Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- 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.
| | - Yu Gong
- 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.
| | - Ting Zou
- 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.
| | - Yuxiao Zeng
- 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.
| | - Cao Yang
- 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.
| | - Lingyue Mo
- 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.
| | - Jiahui Kang
- 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.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 40038, 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.
| | - Jun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
8
|
Luis J, Eastlake K, Lamb WDB, Limb GA, Jayaram H, Khaw PT. Cell-Based Therapies for Glaucoma. Transl Vis Sci Technol 2023; 12:23. [PMID: 37494052 PMCID: PMC10383000 DOI: 10.1167/tvst.12.7.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Glaucomatous optic neuropathy (GON) is the major cause of irreversible visual loss worldwide and can result from a range of disease etiologies. The defining features of GON are retinal ganglion cell (RGC) degeneration and characteristic cupping of the optic nerve head (ONH) due to tissue remodeling, while intraocular pressure remains the only modifiable GON risk factor currently targeted by approved clinical treatment strategies. Efforts to understand the mechanisms that allow species such as the zebrafish to regenerate their retinal cells have greatly increased our understanding of regenerative signaling pathways. However, proper integration within the retina and projection to the brain by the newly regenerated neuronal cells remain major hurdles. Meanwhile, a range of methods for in vitro differentiation have been developed to derive retinal cells from a variety of cell sources, including embryonic and induced pluripotent stem cells. More recently, there has been growing interest in the implantation of glial cells as well as cell-derived products, including neurotrophins, microRNA, and extracellular vesicles, to provide functional support to vulnerable structures such as RGC axons and the ONH. These approaches offer the advantage of not relying upon the replacement of degenerated cells and potentially targeting earlier stages of disease pathogenesis. In order to translate these techniques into clinical practice, appropriate cell sourcing, robust differentiation protocols, and accurate implantation methods are crucial to the success of cell-based therapy in glaucoma. Translational Relevance: Cell-based therapies for glaucoma currently under active development include the induction of endogenous regeneration, implantation of exogenously derived retinal cells, and utilization of cell-derived products to provide functional support.
Collapse
Affiliation(s)
- Joshua Luis
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Karen Eastlake
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - William D. B. Lamb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| |
Collapse
|
9
|
Zhang ZY, Zuo ZY, Liang Y, Zhang SM, Zhang CX, Chi J, Fan B, Li GY. Promotion of axon regeneration and protection on injured retinal ganglion cells by rCXCL2. Inflamm Regen 2023; 43:31. [PMID: 37340465 DOI: 10.1186/s41232-023-00283-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 05/31/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND In addition to rescuing injured retinal ganglion cells (RGCs) by stimulating the intrinsic growth ability of damaged RGCs in various retinal/optic neuropathies, increasing evidence has shown that the external microenvironmental factors also play a crucial role in restoring the survival of RGCs by promoting the regrowth of RGC axons, especially inflammatory factors. In this study, we aimed to screen out the underlying inflammatory factor involved in the signaling of staurosporine (STS)-induced axon regeneration and verify its role in the protection of RGCs and the promotion of axon regrowth. METHODS We performed transcriptome RNA sequencing for STS induction models in vitro and analyzed the differentially expressed genes. After targeting the key gene, we verified the role of the candidate factor in RGC protection and promotion of axon regeneration in vivo with two RGC-injured animal models (optic nerve crush, ONC; retinal N-methyl-D-aspartate, NMDA damage) by using cholera toxin subunit B anterograde axon tracing and specific immunostaining of RGCs. RESULTS We found that a series of inflammatory genes expressed upregulated in the signaling of STS-induced axon regrowth and we targeted the candidate CXCL2 gene since the level of the chemokine CXCL2 gene elevated significantly among the top upregulated genes. We further demonstrated that intravitreal injection of rCXCL2 robustly promoted axon regeneration and significantly improved RGC survival in ONC-injured mice in vivo. However, different from its role in ONC model, the intravitreal injection of rCXCL2 was able to simply protect RGCs against NMDA-induced excitotoxicity in mouse retina and maintain the long-distance projection of RGC axons, yet failed to promote significant axon regeneration. CONCLUSIONS We provide the first in vivo evidence that CXCL2, as an inflammatory factor, is a key regulator in the axon regeneration and neuroprotection of RGCs. Our comparative study may facilitate deciphering the exact molecular mechanisms of RGC axon regeneration and developing high-potency targeted drugs.
Collapse
Affiliation(s)
- Zi-Yuan Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Zhao-Yang Zuo
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yang Liang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Si-Ming Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Chun-Xia Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Jing Chi
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| | - Guang-Yu Li
- Department of Ophthalmology, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
10
|
Mustafi D, Bharathan SP, Calderon R, Nagiel A. HUMAN CELLULAR MODELS FOR RETINAL DISEASE: From Induced Pluripotent Stem Cells to Organoids. Retina 2022; 42:1829-1835. [PMID: 35858274 PMCID: PMC10119785 DOI: 10.1097/iae.0000000000003571] [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: 03/14/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022]
Abstract
PURPOSE To provide a concise review of induced pluripotent stem cells (iPSCs) and retinal organoids as models for human retinal diseases and their role in gene discovery and treatment of inherited retinal diseases (IRDs). METHODS A PubMed literature review was performed for models of human retinal disease, including animal models and human pluripotent stem cell-derived models. RESULTS There is a growing body of research on retinal disease using human pluripotent stem cells. This is a significant change from just a decade ago when most research was performed on animal models. The advent of induced pluripotent stem cells has permitted not only the generation of two-dimensional human cell cultures such as RPE but also more recently the generation of three-dimensional retinal organoids that better reflect the multicellular laminar architecture of the human retina. CONCLUSION Modern stem cell techniques are improving our ability to model human retinal disease in vitro, especially with the use of patient-derived induced pluripotent stem cells. In the future, a personalized approach may be used in which the individual's unique genotype can be modeled in two-dimensional culture or three-dimensional organoids and then rescued with an optimized therapy before treating the patient.
Collapse
Affiliation(s)
- Debarshi Mustafi
- Department of Ophthalmology, Karalis Johnson Retina Center, University of Washington, Seattle, Washington
- Department of Ophthalmology, Seattle Children's Hospital, Seattle, Washington
- Brotman Baty Institute for Precision Medicine, Seattle, Washington
| | - Sumitha P Bharathan
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Rosanna Calderon
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Development, Stem Cells and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
- Department of Ophthalmology, Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
11
|
Comparison of the Response to the CXCR4 Antagonist AMD3100 during the Development of Retinal Organoids Derived from ES Cells and Zebrafish Retina. Int J Mol Sci 2022; 23:ijms23137088. [PMID: 35806093 PMCID: PMC9266567 DOI: 10.3390/ijms23137088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/10/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Retinal organoids generated from human embryonic stem cells or iPSCs recreate the key structural and functional features of mammalian retinal tissue in vitro. However, the differences in the development of retinal organoids and normal retina in vivo are not well defined. Thus, in the present study, we analyzed the development of retinal organoids and zebrafish retina after inhibition of CXCR4, a key role in neurogenesis and optic nerve development, with the antagonist AMD3100. Our data indicated that CXCR4 was mainly expressed in ganglion cells in retinal organoids and was rarely expressed in amacrine or photoreceptor cells. AMD3100 treatment reduced the retinal organoid generation ratio, impaired differentiation, and induced morphological changes. Ganglion cells, amacrine cells, and photoreceptors were decreased and abnormal locations were observed in organoids treated with AMD3100. Neuronal axon outgrowth was also damaged in retinal organoids. Similarly, a decrease of ganglion cells, amacrine cells, and photoreceptors and the distribution of neural outgrowth was induced by AMD3100 treatment in zebrafish retina. However, abnormal photoreceptor ensembles induced by AMD3100 treatment in the organoids were not detected in zebrafish retina. Therefore, our study suggests that although retinal organoids might provide a reliable model for reproducing a retinal developmental model, there is a difference between the organoids and the retina in vivo.
Collapse
|
12
|
Bellapianta A, Cetkovic A, Bolz M, Salti A. Retinal Organoids and Retinal Prostheses: An Overview. Int J Mol Sci 2022; 23:2922. [PMID: 35328339 PMCID: PMC8953078 DOI: 10.3390/ijms23062922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the progress of modern medicine in the last decades, millions of people diagnosed with retinal dystrophies (RDs), such as retinitis pigmentosa, or age-related diseases, such as age-related macular degeneration, are suffering from severe visual impairment or even legal blindness. On the one hand, the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) and the progress of three-dimensional (3D) retinal organoids (ROs) technology provide a great opportunity to study, understand, and even treat retinal diseases. On the other hand, research advances in the field of electronic retinal prosthesis using inorganic photovoltaic polymers and the emergence of organic semiconductors represent an encouraging therapeutical strategy to restore vision to patients at the late onset of the disease. This review will provide an overview of the latest advancement in both fields. We first describe the retina and the photoreceptors, briefly mention the most used RD animal models, then focus on the latest RO differentiation protocols, carry out an overview of the current technology on inorganic and organic retinal prostheses to restore vision, and finally summarize the potential utility and applications of ROs.
Collapse
Affiliation(s)
| | | | | | - Ahmad Salti
- Center for Medical Research, Faculty of Medicine, University Clinic for Ophthalmology and Optometry, Johannes Kepler University Linz, 4020 Linz, Austria; (A.B.); (A.C.); (M.B.)
| |
Collapse
|
13
|
Bharathan SP, Ferrario A, Stepanian K, Fernandez GE, Reid MW, Kim JS, Hutchens C, Harutyunyan N, Marks C, Thornton ME, Grubbs BH, Cobrinik D, Aparicio JG, Nagiel A. Characterization and staging of outer plexiform layer development in human retina and retinal organoids. Development 2021; 148:272710. [PMID: 34738615 DOI: 10.1242/dev.199551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/26/2021] [Indexed: 11/20/2022]
Abstract
The development of the first synapse of the visual system between photoreceptors and bipolar cells in the outer plexiform layer (OPL) of the human retina is critical for visual processing but poorly understood. By studying the maturation state and spatial organization of photoreceptors, depolarizing bipolar cells, and horizontal cells in the human fetal retina, we establish a pseudo-temporal staging system for OPL development that we term OPL-Stages 0 to 4. This was validated through quantification of increasingly precise subcellular localization of Bassoon to the OPL with each stage (p<0.0001). By applying these OPL staging criteria to human retinal organoids (HROs) derived from human embryonic and induced pluripotent stem cells, we observed comparable maturation from OPL-Stage 0 at day 100 in culture up to OPL-Stage 3 by day 160. Quantification of presynaptic protein localization confirmed progression from OPL-Stage 0 to 3 (p<0.0001). Overall, this study defines stages of human OPL development through mid-gestation and establishes HROs as a model system that recapitulates key aspects of human photoreceptor-bipolar cell synaptogenesis in vitro.
Collapse
Affiliation(s)
- Sumitha Prameela Bharathan
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Angela Ferrario
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Kayla Stepanian
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - G Esteban Fernandez
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Mark W Reid
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Justin S Kim
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Chloe Hutchens
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Narine Harutyunyan
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Carolyn Marks
- Core Center of Excellence in Nano Imaging, University of Southern California, Los Angeles, CA, USA
| | - Matthew E Thornton
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brendan H Grubbs
- Maternal-Fetal Medicine Division, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Cobrinik
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jennifer G Aparicio
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Aaron Nagiel
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA.,The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA.,Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Zhang X, Wang W, Jin ZB. Retinal organoids as models for development and diseases. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:33. [PMID: 34719743 PMCID: PMC8557999 DOI: 10.1186/s13619-021-00097-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
The evolution of pluripotent stem cell-derived retinal organoids (ROs) has brought remarkable opportunities for developmental studies while also presenting new therapeutic avenues for retinal diseases. With a clear understanding of how well these models mimic native retinas, such preclinical models may be crucial tools that are widely used for the more efficient translation of studies into novel treatment strategies for retinal diseases. Genetic modifications or patient-derived ROs can allow these models to simulate the physical microenvironments of the actual disease process. However, we are currently at the beginning of the three-dimensional (3D) RO era, and a general quantitative technology for analyzing ROs derived from numerous differentiation protocols is still missing. Continued efforts to improve the efficiency and stability of differentiation, as well as understanding the disparity between the artificial retina and the native retina and advancing the current treatment strategies, will be essential in ensuring that these scientific advances can benefit patients with retinal disease. Herein, we briefly discuss RO differentiation protocols, the current applications of RO as a disease model and the treatments for retinal diseases by using RO modeling, to have a clear view of the role of current ROs in retinal development and diseases.
Collapse
Affiliation(s)
- Xiao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Wen Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
15
|
Retinal Organoid Technology: Where Are We Now? Int J Mol Sci 2021; 22:ijms221910244. [PMID: 34638582 PMCID: PMC8549701 DOI: 10.3390/ijms221910244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
It is difficult to regenerate mammalian retinal cells once the adult retina is damaged, and current clinical approaches to retinal damages are very limited. The introduction of the retinal organoid technique empowers researchers to study the molecular mechanisms controlling retinal development, explore the pathogenesis of retinal diseases, develop novel treatment options, and pursue cell/tissue transplantation under a certain genetic background. Here, we revisit the historical background of retinal organoid technology, categorize current methods of organoid induction, and outline the obstacles and potential solutions to next-generation retinal organoids. Meanwhile, we recapitulate recent research progress in cell/tissue transplantation to treat retinal diseases, and discuss the pros and cons of transplanting single-cell suspension versus retinal organoid sheet for cell therapies.
Collapse
|
16
|
Liu J, Ottaviani D, Sefta M, Desbrousses C, Chapeaublanc E, Aschero R, Sirab N, Lubieniecki F, Lamas G, Tonon L, Dehainault C, Hua C, Fréneaux P, Reichman S, Karboul N, Biton A, Mirabal-Ortega L, Larcher M, Brulard C, Arrufat S, Nicolas A, Elarouci N, Popova T, Némati F, Decaudin D, Gentien D, Baulande S, Mariani O, Dufour F, Guibert S, Vallot C, Rouic LLL, Matet A, Desjardins L, Pascual-Pasto G, Suñol M, Catala-Mora J, Llano GC, Couturier J, Barillot E, Schaiquevich P, Gauthier-Villars M, Stoppa-Lyonnet D, Golmard L, Houdayer C, Brisse H, Bernard-Pierrot I, Letouzé E, Viari A, Saule S, Sastre-Garau X, Doz F, Carcaboso AM, Cassoux N, Pouponnot C, Goureau O, Chantada G, de Reyniès A, Aerts I, Radvanyi F. A high-risk retinoblastoma subtype with stemness features, dedifferentiated cone states and neuronal/ganglion cell gene expression. Nat Commun 2021; 12:5578. [PMID: 34552068 PMCID: PMC8458383 DOI: 10.1038/s41467-021-25792-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
Retinoblastoma is the most frequent intraocular malignancy in children, originating from a maturing cone precursor in the developing retina. Little is known on the molecular basis underlying the biological and clinical behavior of this cancer. Here, using multi-omics data, we demonstrate the existence of two retinoblastoma subtypes. Subtype 1, of earlier onset, includes most of the heritable forms. It harbors few genetic alterations other than the initiating RB1 inactivation and corresponds to differentiated tumors expressing mature cone markers. By contrast, subtype 2 tumors harbor frequent recurrent genetic alterations including MYCN-amplification. They express markers of less differentiated cone together with neuronal/ganglion cell markers with marked inter- and intra-tumor heterogeneity. The cone dedifferentiation in subtype 2 is associated with stemness features including low immune and interferon response, E2F and MYC/MYCN activation and a higher propensity for metastasis. The recognition of these two subtypes, one maintaining a cone-differentiated state, and the other, more aggressive, associated with cone dedifferentiation and expression of neuronal markers, opens up important biological and clinical perspectives for retinoblastomas.
Collapse
Affiliation(s)
- Jing Liu
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Daniela Ottaviani
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Meriem Sefta
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Céline Desbrousses
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Elodie Chapeaublanc
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Rosario Aschero
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Nanor Sirab
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Fabiana Lubieniecki
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Gabriela Lamas
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina
| | - Laurie Tonon
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Catherine Dehainault
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France
| | - Clément Hua
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Paul Fréneaux
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Narjesse Karboul
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Anne Biton
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France ,grid.428999.70000 0001 2353 6535Present Address: Institut Pasteur – Hub Bioinformatique et Biostatistique – C3BI, USR 3756 IP CNRS, 75015 Paris, France
| | - Liliana Mirabal-Ortega
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Magalie Larcher
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Céline Brulard
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.411777.30000 0004 1765 1563Present Address: INSERM U930, CHU Bretonneau, 37000 Tours, France
| | - Sandrine Arrufat
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - André Nicolas
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Nabila Elarouci
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Tatiana Popova
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Fariba Némati
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Didier Decaudin
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - David Gentien
- grid.418596.70000 0004 0639 6384Département de Recherche Translationnelle, Institut Curie, 75005 Paris, France
| | - Sylvain Baulande
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, NGS Platform, 75005 Paris, France
| | - Odette Mariani
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Florent Dufour
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Sylvain Guibert
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Céline Vallot
- grid.425132.3GeCo Genomics Consulting, Integragen, 91000 Evry, France
| | - Livia Lumbroso-Le Rouic
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Alexandre Matet
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Laurence Desjardins
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France
| | - Guillem Pascual-Pasto
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Mariona Suñol
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Pathology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jaume Catala-Mora
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Department of Ophthalmology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Genoveva Correa Llano
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Jérôme Couturier
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France
| | - Emmanuel Barillot
- grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U900, 75005 Paris, France ,Ecole des Mines ParisTech, 77305 Fontainebleau, France
| | - Paula Schaiquevich
- grid.414531.60000 0001 0695 6255Pathology Service, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Marion Gauthier-Villars
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Dominique Stoppa-Lyonnet
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Lisa Golmard
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France
| | - Claude Houdayer
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Service de Génétique, Institut Curie, 75005 Paris, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM U830, 75005 Paris, France ,grid.41724.34Present Address: Department of Genetics, Rouen University Hospital, 76000 Rouen, France
| | - Hervé Brisse
- grid.418596.70000 0004 0639 6384Département d’Imagerie Médicale, Institut Curie, 75005 Paris, France
| | - Isabelle Bernard-Pierrot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| | - Eric Letouzé
- grid.417925.cCentre de Recherche des Cordeliers, Sorbonne Universités, INSERM, 75006 Paris, France ,grid.508487.60000 0004 7885 7602Functional Genomics of Solid Tumors, équipe labellisée Ligue Contre le Cancer, Université de Paris, Université Paris 13, Paris, France
| | - Alain Viari
- grid.418116.b0000 0001 0200 3174Synergie Lyon Cancer, Plateforme de Bioinformatique “Gilles Thomas”, Centre Léon Bérard, 69008 Lyon, France
| | - Simon Saule
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Xavier Sastre-Garau
- grid.418596.70000 0004 0639 6384Département de Biologie des Tumeurs, Institut Curie, 75005 Paris, France ,grid.414145.10000 0004 1765 2136Present Address: Department of Pathology, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France
| | - François Doz
- grid.508487.60000 0004 7885 7602Université de Paris, Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - Angel M. Carcaboso
- grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Nathalie Cassoux
- grid.418596.70000 0004 0639 6384Département de Chirurgie, Service d’Ophtalmologie, Institut Curie, 75005 Paris, France ,grid.508487.60000 0004 7885 7602Université de Paris, Paris, France
| | - Celio Pouponnot
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR3347, PSL Research University, 91405 Orsay, France ,grid.418596.70000 0004 0639 6384Institut Curie, PSL Research University, INSERM, U1021, 91405 Orsay, France ,grid.460789.40000 0004 4910 6535Université Paris-Saclay, 91405 Orsay, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, 75012 Paris, France
| | - Guillermo Chantada
- grid.414531.60000 0001 0695 6255Precision Medicine, Hospital J.P. Garrahan, Buenos Aires, Argentina ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu, 08950 Barcelona, Spain ,grid.411160.30000 0001 0663 8628Pediatric Hematology and Oncology, Hospital Sant Joan de Déu, 08950 Barcelona, Spain ,grid.423606.50000 0001 1945 2152National Scientific and Technical Research Council, CONICET, Buenos Aires, Argentina
| | - Aurélien de Reyniès
- grid.452770.30000 0001 2226 6748Programme Cartes d’Identité des Tumeurs, Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Isabelle Aerts
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France ,grid.418596.70000 0004 0639 6384SIREDO Center (Care, Innovation and Research in Pediatric Adolescent and Young Adult Oncology), Institut Curie, 75005 Paris, France
| | - François Radvanyi
- grid.4444.00000 0001 2112 9282Institut Curie, CNRS, UMR144, Equipe Labellisée Ligue contre le Cancer, PSL Research University, 75005 Paris, France ,grid.462844.80000 0001 2308 1657Sorbonne Universités, UPMC Université Paris 06, CNRS, UMR144, 75005 Paris, France
| |
Collapse
|
17
|
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: 19] [Impact Index Per Article: 4.8] [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
|
18
|
Wagstaff EL, Heredero Berzal A, Boon CJF, Quinn PMJ, ten Asbroek ALMA, Bergen AA. The Role of Small Molecules and Their Effect on the Molecular Mechanisms of Early Retinal Organoid Development. Int J Mol Sci 2021; 22:7081. [PMID: 34209272 PMCID: PMC8268497 DOI: 10.3390/ijms22137081] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Early in vivo embryonic retinal development is a well-documented and evolutionary conserved process. The specification towards eye development is temporally controlled by consecutive activation or inhibition of multiple key signaling pathways, such as the Wnt and hedgehog signaling pathways. Recently, with the use of retinal organoids, researchers aim to manipulate these pathways to achieve better human representative models for retinal development and disease. To achieve this, a plethora of different small molecules and signaling factors have been used at various time points and concentrations in retinal organoid differentiations, with varying success. Additions differ from protocol to protocol, but their usefulness or efficiency has not yet been systematically reviewed. Interestingly, many of these small molecules affect the same and/or multiple pathways, leading to reduced reproducibility and high variability between studies. In this review, we make an inventory of the key signaling pathways involved in early retinogenesis and their effect on the development of the early retina in vitro. Further, we provide a comprehensive overview of the small molecules and signaling factors that are added to retinal organoid differentiation protocols, documenting the molecular and functional effects of these additions. Lastly, we comparatively evaluate several of these factors using our established retinal organoid methodology.
Collapse
Affiliation(s)
- Ellie L. Wagstaff
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center—New York-Presbyterian Hospital, New York, NY 10032, USA;
| | | | - Arthur A. Bergen
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Netherlands Institute for Neuroscience (NIN-KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
19
|
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
|
20
|
Li X, Zhang L, Tang F, Wei X. Retinal Organoids: Cultivation, Differentiation, and Transplantation. Front Cell Neurosci 2021; 15:638439. [PMID: 34276307 PMCID: PMC8282056 DOI: 10.3389/fncel.2021.638439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/08/2021] [Indexed: 02/05/2023] Open
Abstract
Retinal organoids (ROs), which are derived from stem cells, can automatically form three-dimensional laminar structures that include all cell types and the ultrastructure of the retina. Therefore, they are highly similar to the retinal structure in the human body. The development of organoids has been a great technological breakthrough in the fields of transplantation therapy and disease modeling. However, the translation of RO applications into medical practice still has various deficiencies at the current stage, including the long culture process, insufficient yield, and great heterogeneity among ROs produced under different conditions. Nevertheless, many technological breakthroughs have been made in transplanting ROs for treatment of diseases such as retinal degeneration. This review discusses recent advances in the development of ROs, improvements of the culture protocol, and the latest developments in RO replacement therapy techniques.
Collapse
Affiliation(s)
- Xuying Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Tang
- Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| |
Collapse
|
21
|
Oswald J, Kegeles E, Minelli T, Volchkov P, Baranov P. Transplantation of miPSC/mESC-derived retinal ganglion cells into healthy and glaucomatous retinas. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:180-198. [PMID: 33816648 PMCID: PMC7994731 DOI: 10.1016/j.omtm.2021.03.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/06/2021] [Indexed: 12/11/2022]
Abstract
Optic neuropathies, including glaucoma, are a group of neurodegenerative diseases, characterized by the progressive loss of retinal ganglion cells (RGCs), leading to irreversible vision loss. While previous studies demonstrated the potential to replace RGCs with primary neurons from developing mouse retinas, their use is limited clinically. We demonstrate successful transplantation of mouse induced pluripotent stem cell (miPSC)/mouse embryonic stem cell (mESC)-derived RGCs into healthy and glaucomatous mouse retinas, at a success rate exceeding 65% and a donor cell survival window of up to 12 months. Transplanted Thy1-GFP+ RGCs were able to polarize within the host retina and formed axonal processes that followed host axons along the retinal surface and entered the optic nerve head. RNA sequencing of donor RGCs re-isolated from host retinas at 24 h and 1 week post-transplantation showed upregulation of cellular pathways mediating axonal outgrowth, extension, and guidance. Additionally, we provide evidence of subtype-specific diversity within miPSC-derived RGCs prior to transplantation.
Collapse
Affiliation(s)
- Julia Oswald
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Evgenii Kegeles
- Life Sciences Research Center, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
| | - Tomas Minelli
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Pavel Volchkov
- Life Sciences Research Center, Moscow Institute of Physics and Technology, Dolgoprudniy 141700, Russia
- Research Institute of Personalized Medicine, National Center for Personalized Medicine of Endocrine Diseases, The National Medical Research Center for Endocrinology, Moscow 117036, Russia
| | - Petr Baranov
- The Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Corresponding author: Petr Baranov, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
22
|
Brodie-Kommit J, Clark BS, Shi Q, Shiau F, Kim DW, Langel J, Sheely C, Ruzycki PA, Fries M, Javed A, Cayouette M, Schmidt T, Badea T, Glaser T, Zhao H, Singer J, Blackshaw S, Hattar S. Atoh7-independent specification of retinal ganglion cell identity. SCIENCE ADVANCES 2021; 7:7/11/eabe4983. [PMID: 33712461 PMCID: PMC7954457 DOI: 10.1126/sciadv.abe4983] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/29/2021] [Indexed: 06/11/2023]
Abstract
Retinal ganglion cells (RGCs) relay visual information from the eye to the brain. RGCs are the first cell type generated during retinal neurogenesis. Loss of function of the transcription factor Atoh7, expressed in multipotent early neurogenic retinal progenitors leads to a selective and essentially complete loss of RGCs. Therefore, Atoh7 is considered essential for conferring competence on progenitors to generate RGCs. Despite the importance of Atoh7 in RGC specification, we find that inhibiting apoptosis in Atoh7-deficient mice by loss of function of Bax only modestly reduces RGC numbers. Single-cell RNA sequencing of Atoh7;Bax-deficient retinas shows that RGC differentiation is delayed but that the gene expression profile of RGC precursors is grossly normal. Atoh7;Bax-deficient RGCs eventually mature, fire action potentials, and incorporate into retinal circuitry but exhibit severe axonal guidance defects. This study reveals an essential role for Atoh7 in RGC survival and demonstrates Atoh7-dependent and Atoh7-independent mechanisms for RGC specification.
Collapse
Affiliation(s)
| | - Brian S Clark
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Qing Shi
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Fion Shiau
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Dong Won Kim
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennifer Langel
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Catherine Sheely
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Philip A Ruzycki
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Michel Fries
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, QC H3C 3J7, Canada
| | - Awais Javed
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, QC H3C 3J7, Canada
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montréal, QC H2W 1R7, Canada
- Molecular Biology Programs, Université de Montréal, QC H3C 3J7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0G4, Canada
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Tiffany Schmidt
- Department of Neurobiology, Northwestern University, Evanston, IL, USA
| | - Tudor Badea
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
- Research and Development Institute, Transylvania University of Brasov, School of Medicine, Brasov, Romania
| | - Tom Glaser
- Department of Cell Biology and Human Anatomy, University of California, Davis School of Medicine, Davis, CA, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Joshua Singer
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Samer Hattar
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
23
|
Liu Y, Lee RK. Cell transplantation to replace retinal ganglion cells faces challenges - the Switchboard Dilemma. Neural Regen Res 2021; 16:1138-1143. [PMID: 33269762 PMCID: PMC8224141 DOI: 10.4103/1673-5374.300329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The mammalian retina displays incomplete intrinsic regenerative capacities; therefore, retina degeneration is a major cause of irreversible blindness such as glaucoma, age-related macular degeneration and diabetic retinopathy. These diseases lead to the loss of retinal cells and serious vision loss in the late stage. Stem cell transplantation is a great promising novel treatment for these incurable retinal degenerative diseases and represents an exciting area of regenerative neurotherapy. Several suitable stem cell sources for transplantation including human embryonic stem cells, induced pluripotent stem cells and adult stem cells have been identified as promising target populations. However, the retina is an elegant neuronal complex composed of various types of cells with different functions. The replacement of these different types of cells by transplantation should be addressed separately. So far, retinal pigment epithelium transplantation has achieved the most advanced stage of clinical trials, while transplantation of retinal neurons such as retinal ganglion cells and photoreceptors has been mostly studied in pre-clinical animal models. In this review, we opine on the key problems that need to be addressed before stem cells transplantation, especially for replacing injured retinal ganglion cells, may be used practically for treatment. A key problem we have called the Switchboard Dilemma is a major block to have functional retinal ganglion cell replacement. We use the public switchboard telephone network as an example to illustrate different difficulties for replacing damaged components in the retina that allow for visual signaling. Retinal ganglion cell transplantation is confronted by significant hurdles, because retinal ganglion cells receive signals from different interneurons, integrate and send signals to the correct targets of the visual system, which functions similar to the switchboard in a telephone network – therefore the Switchboard Dilemma.
Collapse
Affiliation(s)
- Yuan Liu
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Richard K Lee
- Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
24
|
Manafi N, Shokri F, Achberger K, Hirayama M, Mohammadi MH, Noorizadeh F, Hong J, Liebau S, Tsuji T, Quinn PMJ, Mashaghi A. Organoids and organ chips in ophthalmology. Ocul Surf 2020; 19:1-15. [PMID: 33220469 DOI: 10.1016/j.jtos.2020.11.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Recent advances have driven the development of stem cell-derived, self-organizing, three-dimensional miniature organs, termed organoids, which mimic different eye tissues including the retina, cornea, and lens. Organoids and engineered microfluidic organ-on-chips (organ chips) are transformative technologies that show promise in simulating the architectural and functional complexity of native organs. Accordingly, they enable exploration of facets of human disease and development not accurately recapitulated by animal models. Together, these technologies will increase our understanding of the basic physiology of different eye structures, enable us to interrogate unknown aspects of ophthalmic disease pathogenesis, and serve as clinically-relevant surrogates for the evaluation of ocular therapeutics. Both the burden and prevalence of monogenic and multifactorial ophthalmic diseases, which can cause visual impairment or blindness, in the human population warrants a paradigm shift towards organoids and organ chips that can provide sensitive, quantitative, and scalable phenotypic assays. In this article, we review the current situation of organoids and organ chips in ophthalmology and discuss how they can be leveraged for translational applications.
Collapse
Affiliation(s)
- Navid Manafi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Fereshteh Shokri
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, the Netherlands
| | - Kevin Achberger
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Masatoshi Hirayama
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, 272-8513, Japan; Department of Ophthalmology, School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Melika Haji Mohammadi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands
| | | | - Jiaxu Hong
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Department of Ophthalmology and Visual Science, Eye, and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China; Key Laboratory of Myopia, National Health and Family Planning Commission, Shanghai, China
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Takashi Tsuji
- Laboratory for Organ Regeneration, RIKEN Center for Biosystems Dynamics Research, Hyogo, 650-0047, Japan; Organ Technologies Inc., Minato, Tokyo, 105-0001, Japan
| | - Peter M J Quinn
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University. New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center - New York-Presbyterian Hospital, New York, NY, USA.
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands.
| |
Collapse
|
25
|
Rabesandratana O, Chaffiol A, Mialot A, Slembrouck-Brec A, Joffrois C, Nanteau C, Rodrigues A, Gagliardi G, Reichman S, Sahel JA, Chédotal A, Duebel J, Goureau O, Orieux G. Generation of a Transplantable Population of Human iPSC-Derived Retinal Ganglion Cells. Front Cell Dev Biol 2020; 8:585675. [PMID: 33195235 PMCID: PMC7652757 DOI: 10.3389/fcell.2020.585675] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/24/2020] [Indexed: 12/18/2022] Open
Abstract
Optic neuropathies are a major cause of visual impairment due to retinal ganglion cell (RGC) degeneration. Human induced-pluripotent stem cells (iPSCs) represent a powerful tool for studying both human RGC development and RGC-related pathological mechanisms. Because RGC loss can be massive before the diagnosis of visual impairment, cell replacement is one of the most encouraging strategies. The present work describes the generation of functional RGCs from iPSCs based on innovative 3D/2D stepwise differentiation protocol. We demonstrate that targeting the cell surface marker THY1 is an effective strategy to select transplantable RGCs. By generating a fluorescent GFP reporter iPSC line to follow transplanted cells, we provide evidence that THY1-positive RGCs injected into the vitreous of mice with optic neuropathy can survive up to 1 month, intermingled with the host RGC layer. These data support the usefulness of iPSC-derived RGC exploration as a potential future therapeutic strategy for optic nerve regeneration.
Collapse
Affiliation(s)
| | - Antoine Chaffiol
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Antoine Mialot
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Corentin Joffrois
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Céline Nanteau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Amélie Rodrigues
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,CHNO des Quinze-Vingts, INSERM-DHOS CIC 1423, Paris, France.,Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Alain Chédotal
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Jens Duebel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Gael Orieux
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
26
|
Pereiro X, Miltner AM, La Torre A, Vecino E. Effects of Adult Müller Cells and Their Conditioned Media on the Survival of Stem Cell-Derived Retinal Ganglion Cells. Cells 2020; 9:E1759. [PMID: 32708020 PMCID: PMC7465792 DOI: 10.3390/cells9081759] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/16/2022] Open
Abstract
Retinal neurons, particularly retinal ganglion cells (RGCs), are susceptible to the degenerative damage caused by different inherited conditions and environmental insults, leading to irreversible vision loss and, ultimately, blindness. Numerous strategies are being tested in different models of degeneration to restore vision and, in recent years, stem cell technologies have offered novel avenues to obtain donor cells for replacement therapies. To date, stem cell-based transplantation in the retina has been attempted as treatment for photoreceptor degeneration, but the same tools could potentially be applied to other retinal cell types, including RGCs. However, RGC-like cells are not an abundant cell type in stem cell-derived cultures and, often, these cells degenerate over time in vitro. To overcome this limitation, we have taken advantage of the neuroprotective properties of Müller glia (one of the main glial cell types in the retina) and we have examined whether Müller glia and the factors they secrete could promote RGC-like cell survival in organoid cultures. Accordingly, stem cell-derived RGC-like cells were co-cultured with adult Müller cells or Müller cell-conditioned media was added to the cultures. Remarkably, RGC-like cell survival was substantially enhanced in both culture conditions, and we also observed a significant increase in their neurite length. Interestingly, Atoh7, a transcription factor required for RGC development, was up-regulated in stem cell-derived organoids exposed to conditioned media, suggesting that Müller cells may also enhance the survival of retinal progenitors and/or postmitotic precursor cells. In conclusion, Müller cells and the factors they release promote organoid-derived RGC-like cell survival, neuritogenesis, and possibly neuronal maturation.
Collapse
Affiliation(s)
- Xandra Pereiro
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, 48940 Vizcaya, Spain;
| | - Adam M. Miltner
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA; (A.M.M.); (A.L.T.)
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA 95616, USA; (A.M.M.); (A.L.T.)
| | - Elena Vecino
- Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, 48940 Vizcaya, Spain;
| |
Collapse
|
27
|
Hua ZQ, Liu H, Wang N, Jin ZB. Towards stem cell-based neuronal regeneration for glaucoma. PROGRESS IN BRAIN RESEARCH 2020; 257:99-118. [PMID: 32988476 DOI: 10.1016/bs.pbr.2020.05.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glaucoma is a neurodegenerative disease as a leading cause of global blindness. Retinal ganglion cell (RGC) apoptosis and optic nerve damage are the main pathological changes. Patients have elevated intraocular pressure and progressive visual field loss. Unfortunately, current treatments for glaucoma merely stay at delaying the disease progression. As a promising treatment, stem cell-based neuronal regeneration therapy holds potential for glaucoma, thereby great efforts have been paid on it. RGC regeneration and transplantation are key approaches for the future treatment of glaucoma. A line of studies have shown that a variety of cells can be used to regenerate RGCs, including embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), mesenchymal stem cells (MSCs), and retinal progenitor cells (RPCs). In this review, we overview the current progress on the regeneration of pluripotent stem cell-derived RGCs and outlook the perspective and challenges in this field.
Collapse
Affiliation(s)
- Zi-Qi Hua
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui Liu
- Laboratory of Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, The Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ningli Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 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, China.
| |
Collapse
|
28
|
Miltner AM, Mercado-Ayon Y, Cheema SK, Zhang P, Zawadzki RJ, La Torre A. A Novel Reporter Mouse Uncovers Endogenous Brn3b Expression. Int J Mol Sci 2019; 20:E2903. [PMID: 31197108 PMCID: PMC6627301 DOI: 10.3390/ijms20122903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/18/2022] Open
Abstract
Brn3b (Pou4f2) is a class-4 POU domain transcription factor known to play central roles in the development of different neuronal populations of the Central Nervous System, including retinal ganglion cells (RGCs), the neurons that connect the retina with the visual centers of the brain. Here, we have used CRISPR-based genetic engineering to generate a Brn3b-mCherry reporter mouse without altering the endogenous expression of Brn3b. In our mouse line, mCherry faithfully recapitulates normal Brn3b expression in the retina, the optic tracts, the midbrain tectum, and the trigeminal ganglia. The high sensitivity of mCherry also revealed novel expression of Brn3b in the neuroectodermal cells of the optic stalk during early stages of eye development. Importantly, the fluorescent intensity of Brn3b-mCherry in our reporter mice allows for noninvasive live imaging of RGCs using Scanning Laser Ophthalmoscopy (SLO), providing a novel tool for longitudinal monitoring of RGCs.
Collapse
Affiliation(s)
- Adam M Miltner
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA.
| | - Yesica Mercado-Ayon
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA.
| | - Simranjeet K Cheema
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA.
| | - Pengfei Zhang
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA.
- UC Davis EyePod Small Animal Ocular Imaging Laboratory, University of California-Davis, Davis, CA 95616, USA.
| | - Robert J Zawadzki
- UC Davis EyePod Small Animal Ocular Imaging Laboratory, University of California-Davis, Davis, CA 95616, USA.
- Department of Ophthalmology and Vision Science, University of California-Davis, Sacramento, CA 95817, USA.
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California-Davis, Davis, CA 95616, USA.
| |
Collapse
|
29
|
Miltner AM, Torre AL. Retinal Ganglion Cell Replacement: Current Status and Challenges Ahead. Dev Dyn 2019; 248:118-128. [PMID: 30242792 PMCID: PMC7141838 DOI: 10.1002/dvdy.24672] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
The neurons of the retina can be affected by a wide variety of inherited or environmental degenerations that can lead to vision loss and even blindness. Retinal ganglion cell (RGC) degeneration is the hallmark of glaucoma and other optic neuropathies that affect millions of people worldwide. Numerous strategies are being trialed to replace lost neurons in different degeneration models, and in recent years, stem cell technologies have opened promising avenues to obtain donor cells for retinal repair. Stem cell-based transplantation has been most frequently used for the replacement of rod photoreceptors, but the same tools could potentially be used for other retinal cell types, including RGCs. However, RGCs are not abundant in stem cell-derived cultures, and in contrast to the short-distance wiring of photoreceptors, RGC axons take a long and intricate journey to connect with numerous brain nuclei. Hence, a number of challenges still remain, such as the ability to scale up the production of RGCs and a reliable and functional integration into the adult diseased retina upon transplantation. In this review, we discuss the recent advancements in the development of replacement therapies for RGC degenerations and the challenges that we need to overcome before these technologies can be applied to the clinic. Developmental Dynamics 248:118-128, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Adam M. Miltner
- Department of Cell Biology and Human Anatomy, University of California Davis, U.S
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, U.S
| |
Collapse
|
30
|
Miesfeld JB, Glaser T, Brown NL. The dynamics of native Atoh7 protein expression during mouse retinal histogenesis, revealed with a new antibody. Gene Expr Patterns 2018; 27:114-121. [PMID: 29225067 PMCID: PMC5835195 DOI: 10.1016/j.gep.2017.11.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 11/17/2017] [Accepted: 11/28/2017] [Indexed: 12/19/2022]
Abstract
The Atoh7 transcription factor catalyzes the rate-limiting step in the specification of retinal ganglion cells (RGCs). As a tool to study vertebrate retinal development, we validate an antibody that recognizes human and mouse Atoh7 polypeptide, using informative knockout and transgenic mouse tissues and overexpression experiments. The transient features of Atoh7 protein expression during retinal neurogenesis match the expected pattern at the tissue and cellular level. Further, we compare endogenous Atoh7 to established RGC markers, reporter mouse lines and cell cycle markers, demonstrating the utility of the antibody to investigate molecular mechanisms of retinal histogenesis.
Collapse
Affiliation(s)
- Joel B Miesfeld
- Department of Cell Biology & Human Anatomy, University of California, Davis School of Medicine, One Shields Avenue, Davis, CA 95616, United States
| | - Tom Glaser
- Department of Cell Biology & Human Anatomy, University of California, Davis School of Medicine, One Shields Avenue, Davis, CA 95616, United States
| | - Nadean L Brown
- Department of Cell Biology & Human Anatomy, University of California, Davis School of Medicine, One Shields Avenue, Davis, CA 95616, United States.
| |
Collapse
|
31
|
Llonch S, Carido M, Ader M. Organoid technology for retinal repair. Dev Biol 2017; 433:132-143. [PMID: 29291970 DOI: 10.1016/j.ydbio.2017.09.028] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 09/05/2017] [Accepted: 09/21/2017] [Indexed: 02/07/2023]
Abstract
A major cause for vision impairment and blindness in industrialized countries is the loss of the light-sensing retinal tissue in the eye. Photoreceptor damage is one of the main characteristics found in retinal degeneration diseases, such as Retinitis Pigmentosa or age-related macular degeneration. The lack of effective therapies to stop photoreceptor loss together with the absence of significant intrinsic regeneration in the human retina converts such degenerative diseases into permanent conditions that are currently irreversible. Cell replacement by means of photoreceptor transplantation has been proposed as a potential approach to tackle cell loss in the retina. Since the first attempt of photoreceptor transplantation in humans, about twenty years ago, several research groups have focused in the development and improvement of technologies necessary to bring cell transplantation for retinal degeneration diseases to reality. Progress in recent years in the generation of human tissue derived from pluripotent stem cells (PSCs) has significantly improved our tools to study human development and disease in the dish. Particularly the availability of 3D culture systems for the generation of PSC-derived organoids, including the human retina, has dramatically increased access to human material for basic and medical research. In this review, we focus on important milestones towards the generation of transplantable photoreceptor precursors from PSC-derived retinal organoids and discuss recent pre-clinical transplantation studies using organoid-derived photoreceptors in context to related in vivo work using primary photoreceptors as donor material. Additionally, we summarize remaining challenges for developing photoreceptor transplantation towards clinical application.
Collapse
Affiliation(s)
- Sílvia Llonch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany
| | - Madalena Carido
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany; German Center for Neurodegenerative Diseases Dresden (DZNE), Arnoldstraße 18, 01307 Dresden, Germany
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307 Dresden, Germany.
| |
Collapse
|
32
|
Browne AW, Arnesano C, Harutyunyan N, Khuu T, Martinez JC, Pollack HA, Koos DS, Lee TC, Fraser SE, Moats RA, Aparicio JG, Cobrinik D. Structural and Functional Characterization of Human Stem-Cell-Derived Retinal Organoids by Live Imaging. Invest Ophthalmol Vis Sci 2017; 58:3311-3318. [PMID: 28672397 PMCID: PMC5495152 DOI: 10.1167/iovs.16-20796] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose Human pluripotent stem cell (hPSC)-derived retinal organoids are a platform for investigating retinal development, pathophysiology, and cellular therapies. In contrast to histologic analysis in which multiple specimens fixed at different times are used to reconstruct developmental processes, repeated analysis of the same living organoids provides a more direct means to characterize changes. New live imaging modalities can provide insights into retinal organoid structure and metabolic function during in vitro growth. This study employed live tissue imaging to characterize retinal organoid development, including metabolic changes accompanying photoreceptor differentiation. Methods Live hPSC-derived retinal organoids at different developmental stages were examined for microanatomic organization and metabolic function by phase contrast microscopy, optical coherence tomography (OCT), fluorescence lifetime imaging microscopy (FLIM), and hyperspectral imaging (HSpec). Features were compared to those revealed by histologic staining, immunostaining, and microcomputed tomography (micro-CT) of fixed organoid tissue. Results We used FLIM and HSpec to detect changes in metabolic activity as organoids differentiated into organized lamellae. FLIM detected increased glycolytic activity and HSpec detected retinol and retinoic acid accumulation in the organoid outer layer, coinciding with photoreceptor genesis. OCT enabled imaging of lamellae formed during organoid maturation. Micro-CT revealed three-dimensional structure, but failed to detect lamellae. Conclusions Live imaging modalities facilitate real-time and nondestructive imaging of retinal organoids as they organize into lamellar structures. FLIM and HSpec enable rapid detection of lamellar structure and photoreceptor metabolism. Live imaging techniques may aid in the continuous evaluation of retinal organoid development in diverse experimental and cell therapy settings.
Collapse
Affiliation(s)
- Andrew W Browne
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Cosimo Arnesano
- Translational Imaging Center, University of Southern California, Los Angeles, California, United States 3Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States
| | - Narine Harutyunyan
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Thien Khuu
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Juan Carlos Martinez
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Harvey A Pollack
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States 6Department of Radiology, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - David S Koos
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States 6Department of Radiology, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Thomas C Lee
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 4The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - Scott E Fraser
- Translational Imaging Center, University of Southern California, Los Angeles, California, United States 3Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California, United States 5The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States 7Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States
| | - Rex A Moats
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States 6Department of Radiology, Children's Hospital Los Angeles, Los Angeles, California, United States 7Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, United States
| | - Jennifer G Aparicio
- The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States
| | - David Cobrinik
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 4The Vision Center, Department of Surgery, Children's Hospital Los Angeles, Los Angeles, California, United States 5The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California, United States 8Department of Biochemistry & Molecular Medicine, and Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| |
Collapse
|