1
|
Howard L, Ishikawa Y, Katayama T, Park SJ, Hill MJ, Blake DJ, Nishida K, Hayashi R, Quantock AJ. Single-cell transcriptomics reveals the molecular basis of human iPS cell differentiation into ectodermal ocular lineages. Commun Biol 2024; 7:1495. [PMID: 39532995 PMCID: PMC11557866 DOI: 10.1038/s42003-024-07130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
The generation of a self-formed, ectodermal, autonomous multi-zone (SEAM) from human induced pluripotent stem cells (hiPSCs) offers a unique perspective to study the dynamics of ocular cell differentiation over time. Here, by utilising single-cell transcriptomics, we have (i) identified, (ii) molecularly characterised and (iii) ascertained the developmental trajectories of ectodermally-derived ocular cell populations which emerge within SEAMs as they form. Our analysis reveals interdependency between tissues of the early eye and delineates the sequential formation and maturation of distinct cell types over a 12-week period. We demonstrate a progression from pluripotency through to tissue specification and differentiation which encompasses both surface ectodermal and neuroectodermal ocular lineages and the generation of iPSC-derived components of the developing cornea, conjunctiva, lens, and retina. Our findings not only advance the understanding of ocular development in a stem cell-based system of human origin, but also establish a robust methodological paradigm for exploring cellular and molecular dynamics during SEAM formation at single-cell resolution and highlight the potential of hiPSC-derived systems as powerful platforms for modelling human eye development and disease.
Collapse
Affiliation(s)
- Laura Howard
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Yuki Ishikawa
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohiko Katayama
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sung-Joon Park
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Matthew J Hill
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Derek J Blake
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | - Ryuhei Hayashi
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Andrew J Quantock
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
2
|
Hammer J, Röppenack P, Yousuf S, Machate A, Fischer M, Hans S, Brand M. Blind But Alive - Congenital Loss of atoh7 Disrupts the Visual System of Adult Zebrafish. Invest Ophthalmol Vis Sci 2024; 65:42. [PMID: 39565303 PMCID: PMC11583992 DOI: 10.1167/iovs.65.13.42] [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: 04/29/2024] [Accepted: 09/30/2024] [Indexed: 11/21/2024] Open
Abstract
Purpose Vision is the predominant sense in most animal species. Loss of vision can be caused by a multitude of factors resulting in anatomic as well as behavioral changes. In mice and zebrafish, atoh7 mutants are completely blind as they fail to generate retinal ganglion cells (RGCs) during development. In contrast to mice, raising blind zebrafish to adulthood is challenging and this important model is currently missing in the field. Here, we report the phenotype of homozygous mutant adult zebrafish atoh7 mutants that have been raised using adjusted feeding and holding conditions. Methods The phenotype of adult mutants was characterized using classical histology and immunohistochemistry as well as optical coherence tomography. In addition, the optokinetic response was characterized. Results Adult atoh7 mutants display dark body pigmentation and significantly reduced body length. They fail to form RGCs, the resulting nerve fiber layer as well as the optic nerve, and consequently behave completely blindly. In contrast, increased amounts of other retinal neurons and Müller glia are formed. In addition, the optic tectum is anatomically reduced in size, presumably due to the missing retinal input. Conclusions Taken together, we provide a comprehensive characterization of a completely blind adult zebrafish mutant with focus on retinal and tectal morphology, as a useful model for glaucoma and optic nerve aplasia.
Collapse
Affiliation(s)
- Juliane Hammer
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Paul Röppenack
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Sarah Yousuf
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Anja Machate
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Marika Fischer
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Stefan Hans
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
| | - Michael Brand
- Center for Regenerative Therapies at TU Dresden(CRTD), Dresden, Germany
- Cluster of Excellence Physics of Life (PoL), TU Dresden, Dresden, Germany
| |
Collapse
|
3
|
Wong NK, Yip SP, Huang CL. Establishing Functional Retina in a Dish: Progress and Promises of Induced Pluripotent Stem Cell-Based Retinal Neuron Differentiation. Int J Mol Sci 2023; 24:13652. [PMID: 37686457 PMCID: PMC10487913 DOI: 10.3390/ijms241713652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
The human eye plays a critical role in vision perception, but various retinal degenerative diseases such as retinitis pigmentosa (RP), glaucoma, and age-related macular degeneration (AMD) can lead to vision loss or blindness. Although progress has been made in understanding retinal development and in clinical research, current treatments remain inadequate for curing or reversing these degenerative conditions. Animal models have limited relevance to humans, and obtaining human eye tissue samples is challenging due to ethical and legal considerations. Consequently, researchers have turned to stem cell-based approaches, specifically induced pluripotent stem cells (iPSCs), to generate distinct retinal cell populations and develop cell replacement therapies. iPSCs offer a novel platform for studying the key stages of human retinogenesis and disease-specific mechanisms. Stem cell technology has facilitated the production of diverse retinal cell types, including retinal ganglion cells (RGCs) and photoreceptors, and the development of retinal organoids has emerged as a valuable in vitro tool for investigating retinal neuron differentiation and modeling retinal diseases. This review focuses on the protocols, culture conditions, and techniques employed in differentiating retinal neurons from iPSCs. Furthermore, it emphasizes the significance of molecular and functional validation of the differentiated cells.
Collapse
Affiliation(s)
- Nonthaphat Kent Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China;
- Centre for Eye and Vision Research (CEVR), Hong Kong Science Park, Hong Kong, China
| |
Collapse
|
4
|
Yin Y, Wu S, Niu L, Huang S. Atonal homolog 7 (ATOH7) confers neuroprotection for photoreceptor cells in glaucoma via inhibition of the notch pathway. J Neurochem 2023; 166:847-861. [PMID: 37526008 DOI: 10.1111/jnc.15905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/13/2023] [Accepted: 06/20/2023] [Indexed: 08/02/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) technologies enable the profiling and analysis of the transcriptomes of single cells and hold promise for clarifying gene mechanisms at single-cell resolution. We based this study on scRNA-seq data to reveal glaucoma-related genes and downstream pathways with neuroprotection effects. The scRNA-seq datasets related to glaucoma of retinal tissue samples of human beings and Atonal Homolog 7 (ATOH7)-null mice were obtained from the GEO database. The 74 top marker genes and 20 cell clusters were obtained in human retinal tissue samples. The key gene ATOH7 was found after the intersection with genes from GeneCards data. In the ATOH7-null mouse retinal tissue samples, pseudotime inference demonstrated significant changes in cell differentiation. Moreover, mouse retinal photoreceptor cells (PRCs) were cultured and treated with lentivirus carrying oe-ATOH7 alone or in combination with Notch signaling pathway activator Jagged-1/FC, after which cell biological functions were determined. The involvement of ATOH7 in glaucoma was identified through regulating PRCs. Furthermore, ATOH7 conferred neuroprotection in PRCs in glaucoma by mediating the Notch signaling pathway. In vitro data confirmed that ATOH7 overexpression promoted the differentiation of PRCs and inhibited their apoptosis by suppressing the Notch signaling pathway. The evidence provided by our study highlighted the involvement of ATOH7 in the blockade of the Notch signaling pathway, resulting in the neuroprotection for PRCs in glaucoma.
Collapse
Affiliation(s)
- Yuan Yin
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Shuai Wu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| | - Lingzhi Niu
- Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Shiwei Huang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
5
|
Esmaeili M, Mead B. Differentiation of Human Embryonic/Induced-Pluripotent Stem Cells to Retinal Ganglion Cells. Methods Mol Biol 2023; 2708:41-48. [PMID: 37558958 DOI: 10.1007/978-1-0716-3409-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
The generation of retinal ganglion cells (RGCs) differentiated from human embryonic stem cell (hESC) or induced-pluripotent stem cells (iPSC) could aid with understanding of human RGC development, neuronal biology, drug discovery, potential cell-based therapies, and gene regulation. Here, we present a protocol for differentiation of hESC to RGCs using a 40-day protocol, significantly shorter than typical retinal organoids while still yielding cells with RGC-enriched markers and show physiological and morphological properties typical of RGCs.
Collapse
Affiliation(s)
- Maryam Esmaeili
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Ben Mead
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
6
|
Luo Z, Chang KC, Wu S, Sun C, Xia X, Nahmou M, Bian M, Wen RR, Zhu Y, Shah S, Tanasa B, Wernig M, Goldberg JL. Directly induced human retinal ganglion cells mimic fetal RGCs and are neuroprotective after transplantation in vivo. Stem Cell Reports 2022; 17:2690-2703. [PMID: 36368332 PMCID: PMC9768574 DOI: 10.1016/j.stemcr.2022.10.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
Retinal ganglion cell (RGC) replacement therapy could restore vision in glaucoma and other optic neuropathies. We developed a rapid protocol for directly induced RGC (iRGC) differentiation from human stem cells, leveraging overexpression of NGN2. Neuronal morphology and neurite growth were observed within 1 week of induction; characteristic RGC-specific gene expression confirmed identity. Calcium imaging demonstrated γ-aminobutyric acid (GABA)-induced excitation characteristic of immature RGCs. Single-cell RNA sequencing showed more similarities between iRGCs and early-stage fetal human RGCs than retinal organoid-derived RGCs. Intravitreally transplanted iRGCs survived and migrated into host retinas independent of prior optic nerve trauma, but iRGCs protected host RGCs from neurodegeneration. These data demonstrate rapid iRGC generation in vitro into an immature cell with high similarity to human fetal RGCs and capacity for retinal integration after transplantation and neuroprotective function after optic nerve injury. The simplicity of this system may benefit translational studies on human RGCs.
Collapse
Affiliation(s)
- Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Kun-Che Chang
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Department of Ophthalmology and Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Suqian Wu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Shanghai Key Laboratory of Visual Impairment and Restoration, Department of Ophthalmology and Vision Science, Eye, Ear, Nose & Throat Hospital, Fudan University, Shanghai 200031, China
| | - Catalina Sun
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Xin Xia
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Michael Nahmou
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Minjuan Bian
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Rain R. Wen
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Ying Zhu
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Sahil Shah
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Bogdan Tanasa
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Marius Wernig
- Department of Pathology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA 94304, USA,Corresponding author
| |
Collapse
|
7
|
Behtaj S, Öchsner A, Anissimov YG, Rybachuk M. Retinal Tissue Bioengineering, Materials and Methods for the Treatment of Glaucoma. Tissue Eng Regen Med 2020; 17:253-269. [PMID: 32390117 PMCID: PMC7260329 DOI: 10.1007/s13770-020-00254-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glaucoma, a characteristic type of optic nerve degeneration in the posterior pole of the eye, is a common cause of irreversible vision loss and the second leading cause of blindness worldwide. As an optic neuropathy, glaucoma is identified by increasing degeneration of retinal ganglion cells (RGCs), with consequential vision loss. Current treatments only postpone the development of retinal degeneration, and there are as yet no treatments available for this disability. Recent studies have shown that replacing lost or damaged RGCs with healthy RGCs or RGC precursors, supported by appropriately designed bio-material scaffolds, could facilitate the development and enhancement of connections to ganglion cells and optic nerve axons. The consequence may be an improved retinal regeneration. This technique could also offer the possibility for retinal regeneration in treating other forms of optic nerve ailments through RGC replacement. METHODS In this brief review, we describe the innovations and recent developments in retinal regenerative medicine such as retinal organoids and gene therapy which are specific to glaucoma treatment and focus on the selection of appropriate bio-engineering principles, biomaterials and cell therapies that are presently employed in this growing research area. RESULTS Identification of optimal sources of cells, improving cell survival, functional integration upon transplantation, and developing techniques to deliver cells into the retinal space without provoking immune responses are the main challenges in retinal cell replacement therapies. CONCLUSION The restoration of visual function in glaucoma patients by the RGC replacement therapies requires appropriate protocols and biotechnology methods. Tissue-engineered scaffolds, the generation of retinal organoids, and gene therapy may help to overcome some of the challenges in the generation of clinically safe RGCs.
Collapse
Affiliation(s)
- Sanaz Behtaj
- School of Engineering and Built Environment, Griffith University, Engineering Drive, Southport, QLD, 4222, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- Department of Cell and Molecular Biology, Cell Science Research Centre, Royan Institute for Biotechnology, Isfahan, Iran
| | - Andreas Öchsner
- Faculty of Mechanical Engineering, Esslingen University of Applied Sciences, Kanalstrasse 33, 73728, Esslingen, Germany
| | - Yuri G Anissimov
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- School of Environment and Science, Griffith University, Parklands Drive, Southport, QLD, 4222, Australia
- Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Maksym Rybachuk
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia.
- School of Engineering and Built Environment, Griffith University, 170 Kessels Road, Nathan, QLD, 4111, Australia.
| |
Collapse
|
8
|
Ji SL, Tang SB. Differentiation of retinal ganglion cells from induced pluripotent stem cells: a review. Int J Ophthalmol 2019; 12:152-160. [PMID: 30662854 DOI: 10.18240/ijo.2019.01.22] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 09/06/2018] [Indexed: 01/06/2023] Open
Abstract
Glaucoma is a common optic neuropathy that is characterized by the progressive degeneration of axons and the loss of retinal ganglion cells (RGCs). Glaucoma is one of the leading causes of irreversible blindness worldwide. Current glaucoma treatments only slow the progression of RGCs loss. Induced pluripotent stem cells (iPSCs) are capable of differentiating into all three germ layer cell lineages. iPSCs can be patient-specific, making iPSC-derived RGCs a promising candidate for cell replacement. In this review, we focus on discussing the detailed approaches used to differentiate iPSCs into RGCs.
Collapse
Affiliation(s)
- Shang-Li Ji
- Aier Eye Institute, Changsha 410015, Hunan Province, China
| | - Shi-Bo Tang
- Aier School of Ophthalmology, Central South University, Changsha 410015, Hunan Province, China
| |
Collapse
|
9
|
Jin C, Ou Q, Li Z, Wang J, Zhang J, Tian H, Xu JY, Gao F, Lu L, Xu GT. The combination of bFGF and CHIR99021 maintains stable self-renewal of mouse adult retinal progenitor cells. Stem Cell Res Ther 2018; 9:346. [PMID: 30545413 PMCID: PMC6292077 DOI: 10.1186/s13287-018-1091-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 11/13/2018] [Accepted: 11/26/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Millions of people are affected with retinal diseases that eventually cause blindness, and retinal progenitor cell (RPC) transplantation is a promising therapeutic avenue. However, RPC expansion and the underlying regulation mechanisms remain elusive. METHODS Adult mouse neural RPCs (mNRPCs) were isolated and amplified with the combination of basic fibroblast growth factor (bFGF) and glycogen synthase kinase 3 (GSK3) inhibitor CHIR99021. The progenitor characteristics were evaluated with RT-PCR, immunocytochemistry (ICC), western blot, flow cytometry, and transcriptome analysis prior to transplantation. By treating cells with or without bFGF and CHIR99021 at different time points, the mechanism for mNRPCs' self-renewal was investigated by transcriptome analysis and western blot assay. RESULTS mNRPCs were self-renewing in the presence of bFGF and CHIR99021 and showed prominent RPC characteristics. bFGF was essential in promoting cell cycle by facilitating G1/S and G2/M transitions. bFGF combined with CHIR99021 activated the non-canonical Wnt5A/Ca2+ pathway and form a calcium homeostasis. In addition, the self-renewing mNRPCs could differentiate into rod photoreceptor-like cells and retinal pigment epithelium (RPE)-like cells by in vitro induction. When green fluorescent protein (GFP)-labeled cells were transplanted into the subretinal space (SRS) of Pde6b (rd1) mice (also known as RD1 mice, or rodless mice), the cells survived for more than 12 weeks and migrated into the retina. Parts of the recipient retina showed positive expression of photoreceptor marker rhodopsin. Transplanted cells can migrate into the retina, mainly into the inner cell layer (INL) and ganglion cell layer (GCL). Some cells can differentiate into astrocytes and amacrine cells. Cultured mNRPCs did not form tumors after transplanted into NOD/SCID mice for 6 months. CONCLUSIONS Present study developed an approach to maintain long-term self-renewal of RPCs from adult retinal tissues and revealed that activation of the non-canonical Wnt5A/Ca2+ pathway may participate in regulating RPC self-renewal in vitro. This study presents a very promising platform to expand RPCs for future therapeutic application.
Collapse
Affiliation(s)
- Caixia Jin
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Qingjian Ou
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Zongyi Li
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong Academy of Medical Sciences, Qingdao, 266071, China
| | - Juan Wang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jieping Zhang
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Haibin Tian
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Jing-Ying Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Furong Gao
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China.,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China.,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China
| | - Lixia Lu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China. .,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China. .,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China.
| | - Guo-Tong Xu
- Department of Ophthalmology of Shanghai Tenth People's Hospital, and Tongji Eye Institute, Tongji University School of Medicine, Shanghai, 200072, China. .,Department of Regenerative Medicine and Stem Cell Research Center, Tongji University School of Medicine, Shanghai, 200092, China. .,Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, China. .,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
10
|
Rabesandratana O, Goureau O, Orieux G. Pluripotent Stem Cell-Based Approaches to Explore and Treat Optic Neuropathies. Front Neurosci 2018; 12:651. [PMID: 30294255 PMCID: PMC6158340 DOI: 10.3389/fnins.2018.00651] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Sight is a major sense for human and visual impairment profoundly affects quality of life, especially retinal degenerative diseases which are the leading cause of irreversible blindness worldwide. As for other neurodegenerative disorders, almost all retinal dystrophies are characterized by the specific loss of one or two cell types, such as retinal ganglion cells, photoreceptor cells, or retinal pigmented epithelial cells. This feature is a critical point when dealing with cell replacement strategies considering that the preservation of other cell types and retinal circuitry is a prerequisite. Retinal ganglion cells are particularly vulnerable to degenerative process and glaucoma, the most common optic neuropathy, is a frequent retinal dystrophy. Cell replacement has been proposed as a potential approach to take on the challenge of visual restoration, but its application to optic neuropathies is particularly challenging. Many obstacles need to be overcome before any clinical application. Beyond their survival and differentiation, engrafted cells have to reconnect with both upstream synaptic retinal cell partners and specific targets in the brain. To date, reconnection of retinal ganglion cells with distal central targets appears unrealistic since central nervous system is refractory to regenerative processes. Significant progress on the understanding of molecular mechanisms that prevent central nervous system regeneration offer hope to overcome this obstacle in the future. At the same time, emergence of reprogramming of human somatic cells into pluripotent stem cells has facilitated both the generation of new source of cells with therapeutic potential and the development of innovative methods for the generation of transplantable cells. In this review, we discuss the feasibility of stem cell-based strategies applied to retinal ganglion cells and optic nerve impairment. We present the different strategies for the generation, characterization and the delivery of transplantable retinal ganglion cells derived from pluripotent stem cells. The relevance of pluripotent stem cell-derived retinal organoid and retinal ganglion cells for disease modeling or drug screening will be also introduced in the context of optic neuropathies.
Collapse
Affiliation(s)
| | - Olivier Goureau
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Gaël Orieux
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| |
Collapse
|
11
|
Luo Z, Zhong X, Li K, Xie B, Liu Y, Ye M, Li K, Xu C, Ge J. An Optimized System for Effective Derivation of Three-Dimensional Retinal Tissue via Wnt Signaling Regulation. Stem Cells 2018; 36:1709-1722. [DOI: 10.1002/stem.2890] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 06/17/2018] [Accepted: 06/25/2018] [Indexed: 01/04/2023]
Affiliation(s)
- Ziming Luo
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Xiufeng Zhong
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Kaijing Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Bingbing Xie
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Yuchun Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Meifang Ye
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Kang Li
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Chaochao Xu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| | - Jian Ge
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center; Sun Yat-sen University; Guangzhou Guangdong People's Republic of China
| |
Collapse
|
12
|
Zhang XM, Hashimoto T, Tang R, Yang XJ. Elevated expression of human bHLH factor ATOH7 accelerates cell cycle progression of progenitors and enhances production of avian retinal ganglion cells. Sci Rep 2018; 8:6823. [PMID: 29717171 PMCID: PMC5931526 DOI: 10.1038/s41598-018-25188-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 04/03/2018] [Indexed: 12/13/2022] Open
Abstract
The production of vertebrate retinal projection neurons, retinal ganglion cells (RGCs), is regulated by cell-intrinsic determinants and cell-to-cell signaling events. The basic-helix-loop-helix (bHLH) protein Atoh7 is a key neurogenic transcription factor required for RGC development. Here, we investigate whether manipulating human ATOH7 expression among uncommitted progenitors can promote RGC fate specification and thus be used as a strategy to enhance RGC genesis. Using the chicken retina as a model, we show that cell autonomous expression of ATOH7 is sufficient to induce precocious RGC formation and expansion of the neurogenic territory. ATOH7 overexpression among neurogenic progenitors significantly enhances RGC production at the expense of reducing the progenitor pool. Furthermore, forced expression of ATOH7 leads to a minor increase of cone photoreceptors. We provide evidence that elevating ATOH7 levels accelerates cell cycle progression from S to M phase and promotes cell cycle exit. We also show that ATOH7-induced ectopic RGCs often exhibit aberrant axonal projection patterns and are correlated with increased cell death during the period of retinotectal connections. These results demonstrate the high potency of human ATOH7 in promoting early retinogenesis and specifying the RGC differentiation program, thus providing insight for manipulating RGC production from stem cell-derived retinal organoids.
Collapse
Affiliation(s)
- Xiang-Mei Zhang
- Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Takao Hashimoto
- Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Ronald Tang
- Stein Eye Institute, University of California, Los Angeles, CA, USA
| | - Xian-Jie Yang
- Stein Eye Institute, University of California, Los Angeles, CA, USA. .,Molecular Biology Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
13
|
Yu-Wai-Man P, Newman NJ. Inherited eye-related disorders due to mitochondrial dysfunction. Hum Mol Genet 2017; 26:R12-R20. [PMID: 28481993 DOI: 10.1093/hmg/ddx182] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/05/2017] [Indexed: 12/25/2022] Open
Abstract
Genetic disorders due to mitochondrial dysfunction are not uncommon and the majority of these patients will have eye-related manifestations, including visual loss from the optic nerve and retinal disease, visual field loss from retrochiasmal visual pathway damage, and ptosis and ocular dysmotility from extraocular muscle involvement. Defects in both the nuclear and mitochondrial genomes cause mitochondrial dysfunction via several mechanisms, including impaired mitochondrial energy production, oxidative stress, mitochondrial DNA instability, abnormalities in the regulation of mitochondrial dynamics and mitochondrial quality control, and disturbed cellular interorganellar communication. Advances in our understanding of the molecular genetic basis of mitochondrial disease have not only improved genetic diagnosis, but they have provided important insights into the pathophysiologic basis of these disorders and potential therapeutic targets. In parallel, more sophisticated techniques for genetic manipulation are facilitating the development of animal and in vitro models that should prove powerful and versatile tools for disease modelling and therapeutic experimentation. Effective therapies for mitochondrial disorders are beginning to translate from bench to bedside along the paths of neuroprotection, gene replacement and stem cell-based regenerative paradigms. Additionally, preventing the transmission of pathogenic mtDNA mutations from mother to child is now a reality with in vitro fertilization mitochondrial replacement techniques.
Collapse
Affiliation(s)
- Patrick Yu-Wai-Man
- Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Cambridge, UK.,NIHR Biomedical Research Centre at Moorfields Eye Hospital and UCL Institute of Ophthalmology, London, UK.,Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - Nancy J Newman
- Departments of Ophthalmology, Neurology and Neurological Surgery, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
14
|
The use of induced pluripotent stem cells for studying and treating optic neuropathies. Curr Opin Organ Transplant 2017; 21:484-9. [PMID: 27517502 DOI: 10.1097/mot.0000000000000348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The present review aims to provide an update of applications of induced pluripotent stem cells (iPSCs) for disease modeling, cell/gene therapy, and drug screening for optic neuropathies. RECENT FINDINGS Degeneration of retinal ganglion cells (RGCs) is a characteristic of optic neuropathies. Human iPSCs can serve as a model to investigate disease pathology and potential repair mechanisms. In recent years, significant progress has been made in generating RGCs from iPSCs. Various groups have reported the potential of iPSCs for modeling optic neuropathies, such as glaucoma. The literature also highlights the potential to use iPSC-derived cells for high-throughput drug and toxicity screening. SUMMARY The present review summarizes current work in the field of iPSCs in optic neuropathies. Future studies to characterize iPSC-derived RGCs in a more in-depth manner will help expand the use of iPSCs to model and treat optic neuropathic diseases. Furthermore, iPSC modeling can be used in drug development by offering a new avenue to test novel therapeutic drugs for optic neuropathies.
Collapse
|
15
|
Sluch VM, Chamling X, Liu MM, Berlinicke CA, Cheng J, Mitchell KL, Welsbie DS, Zack DJ. Enhanced Stem Cell Differentiation and Immunopurification of Genome Engineered Human Retinal Ganglion Cells. Stem Cells Transl Med 2017; 6:1972-1986. [PMID: 29024560 PMCID: PMC6430043 DOI: 10.1002/sctm.17-0059] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022] Open
Abstract
Human pluripotent stem cells have the potential to promote biological studies and accelerate drug discovery efforts by making possible direct experimentation on a variety of human cell types of interest. However, stem cell cultures are generally heterogeneous and efficient differentiation and purification protocols are often lacking. Here, we describe the generation of clustered regularly‐interspaced short palindromic repeats(CRISPR)‐Cas9 engineered reporter knock‐in embryonic stem cell lines in which tdTomato and a unique cell‐surface protein, THY1.2, are expressed under the control of the retinal ganglion cell (RGC)‐enriched gene BRN3B. Using these reporter cell lines, we greatly improved adherent stem cell differentiation to the RGC lineage by optimizing a novel combination of small molecules and established an anti‐THY1.2‐based protocol that allows for large‐scale RGC immunopurification. RNA‐sequencing confirmed the similarity of the stem cell‐derived RGCs to their endogenous human counterparts. Additionally, we developed an in vitro axonal injury model suitable for studying signaling pathways and mechanisms of human RGC cell death and for high‐throughput screening for neuroprotective compounds. Using this system in combination with RNAi‐based knockdown, we show that knockdown of dual leucine kinase (DLK) promotes survival of human RGCs, expanding to the human system prior reports that DLK inhibition is neuroprotective for murine RGCs. These improvements will facilitate the development and use of large‐scale experimental paradigms that require numbers of pure RGCs that were not previously obtainable. Stem Cells Translational Medicine2017;6:1972–1986
Collapse
Affiliation(s)
- Valentin M Sluch
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Melissa M Liu
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Cynthia A Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jie Cheng
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Katherine L Mitchell
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Derek S Welsbie
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Shiley Eye Institute, University of California, San Diego, La Jolla, California, USA
| | - Donald J Zack
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Stem cells in regenerative medicine - from laboratory to clinical application - the eye. Cent Eur J Immunol 2017; 42:173-180. [PMID: 28860936 PMCID: PMC5573891 DOI: 10.5114/ceji.2017.69360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/25/2016] [Indexed: 12/18/2022] Open
Abstract
Stem cells are currently one of the most researched and explored subject in science. They consstitue a very promising part of regenerative medicine and have many potential clinical applications. Harnessing their ability to replicate and differentiate into many cell types can enable successful treatment of diseases that were incurable until now. There are numerous types of stem cells (e.g. ESCs, FSCs, ASCs, iPSCs) and many different methods of deriving and cultivating them in order to obtain viable material. The eye is one of the most interesting targets for stem cell therapies. In this article we summarise different aspects of stem cells, discussing their characteristics, sources and methods of culture. We also demonstrate the most recent clinical applications in ophthalmology based on an extensive current literature review. Tissue engineering techniques developed for corneal limbal stem cell deficiency, age-related macular degeneration (AMD) and glaucoma are among those presented. Both laboratory and clinical aspects of stem cells are discussed.
Collapse
|
17
|
Homma K, Usui S, Kaneda M. Knock-in strategy at 3′-end ofCrxgene by CRISPR/Cas9 system shows the gene expression profiles during human photoreceptor differentiation. Genes Cells 2017; 22:250-264. [DOI: 10.1111/gtc.12472] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/22/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Kohei Homma
- Department of Physiology; Nippon Medical School; 1-25-16 Nezu Bunkyo-ku Tokyo 113-0031 Japan
| | - Sumiko Usui
- Department of Physiology; Nippon Medical School; 1-25-16 Nezu Bunkyo-ku Tokyo 113-0031 Japan
| | - Makoto Kaneda
- Department of Physiology; Nippon Medical School; 1-25-16 Nezu Bunkyo-ku Tokyo 113-0031 Japan
| |
Collapse
|
18
|
Wang J, Galvao J, Beach KM, Luo W, Urrutia RA, Goldberg JL, Otteson DC. Novel Roles and Mechanism for Krüppel-like Factor 16 (KLF16) Regulation of Neurite Outgrowth and Ephrin Receptor A5 (EphA5) Expression in Retinal Ganglion Cells. J Biol Chem 2016; 291:18084-95. [PMID: 27402841 DOI: 10.1074/jbc.m116.732339] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Indexed: 11/06/2022] Open
Abstract
Regenerative medicine holds great promise for the treatment of degenerative retinal disorders. Krüppel-like factors (KLFs) are transcription factors that have recently emerged as key tools in regenerative medicine because some of them can function as epigenetic reprogrammers in stem cell biology. Here, we show that KLF16, one of the least understood members of this family, is a POU4F2 independent transcription factor in retinal ganglion cells (RGCs) as early as embryonic day 15. When overexpressed, KLF16 inhibits RGC neurite outgrowth and enhances RGC growth cone collapse in response to exogenous ephrinA5 ligands. Ephrin/EPH signaling regulates RGC connectivity. The EphA5 promoter contains multiple GC- and GT-rich KLF-binding sites, which, as shown by ChIP-assays, bind KLF16 in vivo In electrophoretic mobility shift assays, KLF16 binds specifically to a single KLF site near the EphA5 transcription start site that is required for KLF16 transactivation. Interestingly, methylation of only six of 98 CpG dinucleotides within the EphA5 promoter blocks its transactivation by KLF16 but enables transactivation by KLF2 and KLF15. These data demonstrate a role for KLF16 in regulation of RGC neurite outgrowth and as a methylation-sensitive transcriptional regulator of EphA5 expression. Together, these data identify differential low level methylation as a novel mechanism for regulating KLF16-mediated EphA5 expression across the retina. Because of the critical role of ephrin/EPH signaling in patterning RGC connectivity, understanding the role of KLFs in regulating neurite outgrowth and Eph receptor expression will be vital for successful restoration of functional vision through optic nerve regenerative therapies.
Collapse
Affiliation(s)
- Jianbo Wang
- From the Departments of Physiological Optics and Vision Science and
| | - Joana Galvao
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Krista M Beach
- From the Departments of Physiological Optics and Vision Science and
| | - Weijia Luo
- Biology and Biochemistry, University of Houston, Houston, Texas 77204
| | - Raul A Urrutia
- the Laboratory of Epigenetics and Chromatin Dynamics, Gastroenterology Research Unit, Epigenomics Translational Program, Center for Individualized Medicine, Departments of Medicine, Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Jeffrey L Goldberg
- the Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, California 94303, the Shiley Eye Institute, University of California San Diego, La Jolla, California 92093, and
| | - Deborah C Otteson
- From the Departments of Physiological Optics and Vision Science and Biology and Biochemistry, University of Houston, Houston, Texas 77204,
| |
Collapse
|
19
|
Chamling X, Sluch VM, Zack DJ. The Potential of Human Stem Cells for the Study and Treatment of Glaucoma. Invest Ophthalmol Vis Sci 2016; 57:ORSFi1-6. [PMID: 27116666 PMCID: PMC5110236 DOI: 10.1167/iovs.15-18590] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Currently, the only available and approved treatments for glaucoma are various pharmacologic, laser-based, and surgical procedures that lower IOP. Although these treatments can be effective, they are not always sufficient, and they cannot restore vision that has already been lost. The goal of this review is to briefly assess current developments in the application of stem cell biology to the study and treatment of glaucoma and other forms of optic neuropathy. METHODS A combined literature review and summary of the glaucoma-related discussion at the 2015 "Sight Restoration Through Stem Cell Therapy" meeting that was sponsored by the Ocular Research Symposia Foundation (ORSF). RESULTS Ongoing advancements in basic and eye-related developmental biology have enabled researchers to direct murine and human stem cells along specific developmental paths and to differentiate them into a variety of ocular cell types of interest. The most advanced of these efforts involve the differentiation of stem cells into retinal pigment epithelial cells, work that has led to the initiation of several human trials. More related to the glaucoma field, there have been recent advances in developing protocols for differentiation of stem cells into trabecular meshwork and retinal ganglion cells. Additionally, efforts are being made to generate stem cell-derived cells that can be used to secrete neuroprotective factors. CONCLUSIONS Advancing stem cell technology provides opportunities to improve our understanding of glaucoma-related biology and develop models for drug development, and offers the possibility of cell-based therapies to restore sight to patients who have already lost vision.
Collapse
Affiliation(s)
- Xitiz Chamling
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Valentin M. Sluch
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Donald J. Zack
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
20
|
Sluch VM, Davis CHO, Ranganathan V, Kerr JM, Krick K, Martin R, Berlinicke CA, Marsh-Armstrong N, Diamond JS, Mao HQ, Zack DJ. Differentiation of human ESCs to retinal ganglion cells using a CRISPR engineered reporter cell line. Sci Rep 2015; 5:16595. [PMID: 26563826 PMCID: PMC4643248 DOI: 10.1038/srep16595] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022] Open
Abstract
Retinal ganglion cell (RGC) injury and cell death from glaucoma and other forms of optic nerve disease is a major cause of irreversible vision loss and blindness. Human pluripotent stem cell (hPSC)-derived RGCs could provide a source of cells for the development of novel therapeutic molecules as well as for potential cell-based therapies. In addition, such cells could provide insights into human RGC development, gene regulation, and neuronal biology. Here, we report a simple, adherent cell culture protocol for differentiation of hPSCs to RGCs using a CRISPR-engineered RGC fluorescent reporter stem cell line. Fluorescence-activated cell sorting of the differentiated cultures yields a highly purified population of cells that express a range of RGC-enriched markers and exhibit morphological and physiological properties typical of RGCs. Additionally, we demonstrate that aligned nanofiber matrices can be used to guide the axonal outgrowth of hPSC-derived RGCs for in vitro optic nerve-like modeling. Lastly, using this protocol we identified forskolin as a potent promoter of RGC differentiation.
Collapse
Affiliation(s)
- Valentin M Sluch
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine Baltimore, MD 21287
| | - Chung-ha O Davis
- Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, 21205
| | - Vinod Ranganathan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Justin M Kerr
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kellin Krick
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Russ Martin
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Cynthia A Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Nicholas Marsh-Armstrong
- Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, 21205.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Jeffrey S Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Donald J Zack
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine Baltimore, MD 21287.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205.,Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
21
|
Maekawa Y, Onishi A, Matsushita K, Koide N, Mandai M, Suzuma K, Kitaoka T, Kuwahara A, Ozone C, Nakano T, Eiraku M, Takahashi M. Optimized Culture System to Induce Neurite Outgrowth From Retinal Ganglion Cells in Three-Dimensional Retinal Aggregates Differentiated From Mouse and Human Embryonic Stem Cells. Curr Eye Res 2015; 41:558-68. [PMID: 25880804 DOI: 10.3109/02713683.2015.1038359] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To establish a practical research tool for studying the pathogenesis of retinal ganglion cell (RGC) diseases, we optimized culture procedures to induce neurite outgrowth from three-dimensional self-organizing optic vesicles (3D-retinas) differentiated in vitro from mouse and human embryonic stem cells (ESCs). MATERIALS AND METHODS The developing 3D-retinas isolated at various time points were placed on Matrigel-coated plates and cultured in media on the basis of the 3D-retinal culture or the retinal organotypic culture protocol. The number, length, and morphology of the neurites in each culture condition were compared. RESULTS First, we confirmed that Venus-positive cells were double-labeled with a RGC marker, Brn3a, in the 3D-retina differentiated from Fstl4::Venus mouse ESCs, indicating specific RGC-subtype differentiation. Second, Venus-positive neurites grown from these RGC subsets were positive for beta-III tubulin and SMI312 by immunohistochemistry. Enhanced neurite outgrowth was observed in the B27-supplemented Neurobasal-A medium on Matrigel-coated plates from the optic vesicles isolated after 14 days of differentiation from mouse ESCs. For the differentiated RGCs from human ESCs, we obtained neurite extension of >4 mm by modifying Matrigel coating and the culture medium from the mouse RGC culture. CONCLUSION We successfully optimized the culture conditions to enhance lengthy and high-frequency neurite outgrowth in mouse and human models. The procedure would be useful for not only developmental studies of RGCs, including maintenance and projection, but also clinical, pathological, and pharmacological studies of human RGC diseases.
Collapse
Affiliation(s)
- Yuki Maekawa
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan .,b Department of Ophthalmology and Visual Science , Graduate School of Biomedical Science, Nagasaki University , Kobe , Japan
| | - Akishi Onishi
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan
| | - Keizo Matsushita
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan .,c Regenerative and Cellular Medicine Office, Sumitomo Dainippon Phama Co., Ltd , Kobe , Japan
| | - Naoshi Koide
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan
| | - Michiko Mandai
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan
| | - Kiyoshi Suzuma
- b Department of Ophthalmology and Visual Science , Graduate School of Biomedical Science, Nagasaki University , Kobe , Japan
| | - Takashi Kitaoka
- b Department of Ophthalmology and Visual Science , Graduate School of Biomedical Science, Nagasaki University , Kobe , Japan
| | - Atsushi Kuwahara
- d Laboratory for Organogenesis and Neurogenesis , RIKEN Center for Developmental Biology , Kobe , Japan .,e Environmental Health Science Laboratory , Sumitomo Chemical Co., Ltd. , Osaka , Japan , and
| | - Chikafumi Ozone
- d Laboratory for Organogenesis and Neurogenesis , RIKEN Center for Developmental Biology , Kobe , Japan
| | - Tokushige Nakano
- d Laboratory for Organogenesis and Neurogenesis , RIKEN Center for Developmental Biology , Kobe , Japan .,e Environmental Health Science Laboratory , Sumitomo Chemical Co., Ltd. , Osaka , Japan , and
| | - Mototsugu Eiraku
- f Laboratory for in vitro Histogenesis , RIKEN Center for Developmental Biology , Kobe , Japan
| | - Masayo Takahashi
- a Laboratory for Retinal Regeneration , RIKEN Center for Developmental Biology , Kobe , Japan
| |
Collapse
|
22
|
Generation of retinal ganglion cells with functional axons from human induced pluripotent stem cells. Sci Rep 2015; 5:8344. [PMID: 25666360 PMCID: PMC4322369 DOI: 10.1038/srep08344] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/15/2015] [Indexed: 12/21/2022] Open
Abstract
We generated self-induced retinal ganglion cells (RGCs) with functional axons from human induced pluripotent stem cells. After development of the optic vesicle from the induced stem cell embryoid body in three-dimensional culture, conversion to two-dimensional culture, achieved by supplementation with BDNF, resulted in differentiation of RGCs at a rate of nearly 90% as indicated by a marginal subregion of an extruded clump of cells, suggesting the formation of an optic vesicle. Axons extended radially from the margin of the clump. Induced RGCs expressed specific markers, such as Brn3b and Math5, as assessed using by quantitative PCR and immunohistochemistry. The long, prominent axons contained neurofilaments and tau and exhibited anterograde axonal transport and sodium-dependent action potentials. The ability to generate RGCs with functional axons uniformly and at a high rate may contribute to both basic and clinical science, including embryology, neurology, pathognomy, and treatment of various optic nerve diseases that threaten vision.
Collapse
|