1
|
Hu B, Zhou S, Wang X, Zhang Z, Wang R, Kang Q. Differentiation of primary retinal progenitor cells into retinal ganglion-like cells using low dose cytarabine. Biochem Biophys Res Commun 2025; 748:151359. [PMID: 39832436 DOI: 10.1016/j.bbrc.2025.151359] [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: 12/09/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The death of retinal ganglion cells (RGCs) is a key factor in the pathophysiology of all forms of glaucoma. RGC culture serves as a simple system for establishing and testing candidate therapies. This study aimed to explore the differentiation of primary retinal progenitor cells (RPCs) into RGC-like cells induced by low-dose cytarabine (Ara-C). RPCs were isolated from the retina of newborn rats and cultured in vitro. Different concentrations of Ara-C were added to the culture medium to induce the differentiation of RPCs into RGC-like cells. Differentiation efficiency was assessed through immunofluorescence staining and cell counting. The addition of Ara-C significantly increased the number of Brn3a/RBPMS double-positive cells. The RPC-RGCs induced displayed characteristic features of RGCs, with roughly 80.9 % ± 6.2 % of the cells positive for both TuJ1/NeuN and 77.5 % ± 4.9 % for Brn3a/RBPMS. The study demonstrates that the addition of Ara-C to primary cultures of rat RPCs can enhance their differentiation into RGC-like cells, providing a simple and rapid method for obtaining RGC-like cells with a relatively high purity. This method shows considerable promise for advancing glaucoma research and potential therapeutic strategies to restore vision after RGC loss.
Collapse
Affiliation(s)
- Baoqi Hu
- Department of Ophthalmology, Xi'an No.1 Hospital, the First Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710002, China; Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi, 710002, China
| | - Sijia Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiou Wang
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhichao Zhang
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, China
| | - Rui Wang
- Department of Ophthalmology, Xi'an No.1 Hospital, the First Affiliated Hospital of Northwest University, Xi'an, Shaanxi, 710002, China; Shaanxi Institute of Ophthalmology, Xi'an, Shaanxi, 710002, China.
| | - Qianyan Kang
- Department of Ophthalmology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
2
|
Yang T, Zhang N, Yang N. Single-cell sequencing in diabetic retinopathy: progress and prospects. J Transl Med 2025; 23:49. [PMID: 39806376 PMCID: PMC11727737 DOI: 10.1186/s12967-024-06066-x] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic retinopathy is a major ocular complication of diabetes, characterized by progressive retinal microvascular damage and significant visual impairment in working-age adults. Traditional bulk RNA sequencing offers overall gene expression profiles but does not account for cellular heterogeneity. Single-cell RNA sequencing overcomes this limitation by providing transcriptomic data at the individual cell level and distinguishing novel cell subtypes, developmental trajectories, and intercellular communications. Researchers can use single-cell sequencing to draw retinal cell atlases and identify the transcriptomic features of retinal cells, enhancing our understanding of the pathogenesis and pathological changes in diabetic retinopathy. Additionally, single-cell sequencing is widely employed to analyze retinal organoids and single extracellular vesicles. Single-cell multi-omics sequencing integrates omics information, whereas stereo-sequencing analyzes gene expression and spatiotemporal data simultaneously. This review discusses the protocols of single-cell sequencing for obtaining single cells from retina and accurate sequencing data. It highlights the applications and advancements of single-cell sequencing in the study of normal retinas and the pathological changes associated with diabetic retinopathy. This underscores the potential of these technologies to deepen our understanding of the pathogenesis of diabetic retinopathy that may lead to the introduction of new therapeutic strategies.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China.
| |
Collapse
|
3
|
KIMURA K, NAGAKURA H, TSUKAMOTO M, YOSHIDA T, SUGISAKI H, SHISHIDA K, TACHI Y, SHIMASAKI S, SUGIURA K, HATOYA S. Canine induced pluripotent stem cells can be successfully maintained in weekend-free culture systems. J Vet Med Sci 2024; 86:247-257. [PMID: 38171744 PMCID: PMC10963097 DOI: 10.1292/jvms.23-0422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
Canine induced pluripotent stem cells (ciPSCs) can provide useful insights into novel therapies in both veterinary and medical fields. However, limited accessibility to the present culture medium and requirement of considerable time, effort, and cost for routine ciPSC maintenance restrict advancement in ciPSC research. In addition, it is unknown whether ciPSC culture conditions influence differentiation propensity. We investigated the availability of the common human pluripotent stem cells (hPSCs) culture systems for ciPSC maintenance and the differentiation propensities of the ciPSCs maintained in these culture systems. StemFlex and mTeSR Plus supported PSC-like colony formation and pluripotency markers expression in ciPSCs even after five passages. Additionally, ciPSCs were maintained under weekend-free culture conditions with a stable growth rate, pluripotency marker expression, and differentiation abilities using vitronectin (VTN-N) and Geltrex. Following maintenance of spontaneously differentiated ciPSCs under various conditions by embryoid body formation, there were few differences in the differentiation propensities of ciPSCs among the tested culture conditions. Thus, ciPSCs were successfully cultured under weekend-free conditions for ciPSC maintenance using StemFlex or mTeSR Plus with VTN-N or Geltrex. The present study offers simpler and more effort-, time-, and cost-saving options for ciPSC culture systems, which may lead to further development in research using ciPSCs.
Collapse
Affiliation(s)
- Kazuto KIMURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Hiroya NAGAKURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Masaya TSUKAMOTO
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Takumi YOSHIDA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Hiroko SUGISAKI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Kohei SHISHIDA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Yuta TACHI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Shoko SHIMASAKI
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Kikuya SUGIURA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| | - Shingo HATOYA
- Department of Advanced Pathobiology, Graduate School of Veterinary Science, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
4
|
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
|
5
|
Soucy JR, Aguzzi EA, Cho J, Gilhooley MJ, Keuthan C, Luo Z, Monavarfeshani A, Saleem MA, Wang XW, Wohlschlegel J, Baranov P, Di Polo A, Fortune B, Gokoffski KK, Goldberg JL, Guido W, Kolodkin AL, Mason CA, Ou Y, Reh TA, Ross AG, Samuels BC, Welsbie D, Zack DJ, Johnson TV. Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium. Mol Neurodegener 2023; 18:64. [PMID: 37735444 PMCID: PMC10514988 DOI: 10.1186/s13024-023-00655-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/07/2023] [Indexed: 09/23/2023] Open
Abstract
Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.
Collapse
Affiliation(s)
- Jonathan R Soucy
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Erika A Aguzzi
- The Institute of Ophthalmology, University College London, London, England, UK
| | - Julie Cho
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Michael James Gilhooley
- The Institute of Ophthalmology, University College London, London, England, UK
- Moorfields Eye Hospital, London, England, UK
| | - Casey Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ziming Luo
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Aboozar Monavarfeshani
- Center for Brain Science and Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA
| | - Meher A Saleem
- Bascom Palmer Eye Institute, University of Miami Health System, Miami, FL, USA
| | - Xue-Wei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Petr Baranov
- Department of Ophthalmology, Schepens Eye Research Institute of Mass. Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
- University of Montreal Hospital Research Centre, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Health, Portland, OR, USA
| | - Kimberly K Gokoffski
- Department of Ophthalmology, Roski Eye Institute, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - William Guido
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
| | - Alex L Kolodkin
- The Solomon H Snyder, Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Yvonne Ou
- Department of Ophthalmology, University of California, San Francisco, CA, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, Seattle, WA, USA
| | - Ahmara G Ross
- Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA, USA
| | - Brian C Samuels
- Department of Ophthalmology and Visual Sciences, Callahan Eye Hospital, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Derek Welsbie
- Shiley Eye Institute and Viterbi Family Department of Ophthalmology, University of California, San Diego, CA, USA
| | - Donald J Zack
- Glaucoma Center of Excellence, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas V Johnson
- Departments of Neuroscience, Molecular Biology & Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Cellular & Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, 21287 MD, USA.
| |
Collapse
|
6
|
Ren H, Zhou X, Yang J, Kou K, Chen T, Pu Z, Ye K, Fan X, Zhang D, Kang X, Fan Z, Lei M, Sun T, Tan X, Ou X. Single-cell RNA sequencing of murine hearts for studying the development of the cardiac conduction system. Sci Data 2023; 10:577. [PMID: 37666871 PMCID: PMC10477280 DOI: 10.1038/s41597-023-02333-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 06/26/2023] [Indexed: 09/06/2023] Open
Abstract
The development of the cardiac conduction system (CCS) is essential for correct heart function. However, critical details on the cell types populating the CCS in the mammalian heart during the development remain to be resolved. Using single-cell RNA sequencing, we generated a large dataset of transcriptomes of ~0.5 million individual cells isolated from murine hearts at six successive developmental corresponding to the early, middle and late stages of heart development. The dataset provides a powerful library for studying the development of the heart's CCS and other cardiac components. Our initial analysis identified distinct cell types between 20 to 26 cell types across different stages, of which ten are involved in forming the CCS. Our dataset allows researchers to reuse the datasets for data mining and a wide range of analyses. Collectively, our data add valuable transcriptomic resources for further study of cardiac development, such as gene expression, transcriptional regulation and functional gene activity in developing hearts, particularly the CCS.
Collapse
Affiliation(s)
- Huiying Ren
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiaolin Zhou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Jun Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Kun Kou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Tangting Chen
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Zhaoli Pu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Kejun Ye
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Xuehui Fan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Dan Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Xinjiang Kang
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ming Lei
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, United Kingdom.
| | - Tianyi Sun
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, United Kingdom.
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
7
|
Tang Q, Li W, Huang J, Wu Y, Ma C, Tu Y, Zhu Q, Lu J, Xie J, Liu Y, Mao X, Wu W. Single-cell sequencing analysis of peripheral blood in patients with moyamoya disease. Orphanet J Rare Dis 2023; 18:174. [PMID: 37400835 DOI: 10.1186/s13023-023-02781-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 06/18/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND At present, the etiology of moyamoya disease is not clear, and it is necessary to explore the mechanism of its occurrence and development. Although some bulk sequencing data have previously revealed transcriptomic changes in Moyamoya disease, single-cell sequencing data has been lacking. METHODS Two DSA(Digital Subtraction Angiography)-diagnosed patients with moyamoya disease were recruited between January 2021 and December 2021. Their peripheral blood samples were single-cell sequenced. CellRanger(10 x Genomics, version 3.0.1) was used to process the raw data, demultiplex cellular barcodes, map reads to the transcriptome, and dowm-sample reads(as required to generate normalized aggregate data across samples). There were 4 normal control samples, including two normal samples GSM5160432 and GSM5160434 of GSE168732, and two normal samples of GSE155698, namely GSM4710726 and GSM4710727. Weighted co-expression network analysis was used to explore the gene sets associated with moyamoya disease. GO analysis and KEGG analysis were used to explore gene enrichment pathways. Pseudo-time series analysis and cell interaction analysis were used to explore cell differentiation and cell interaction. RESULTS For the first time, we present a peripheral blood single cell sequencing landscape of Moyamoya disease, revealing cellular heterogeneity and gene expression heterogeneity. In addition, by combining with WGCNA analysis in public database and taking intersection, the key genes in moyamoya disease were obtained. namely PTP4A1, SPINT2, CSTB, PLA2G16, GPX1, HN1, LGALS3BP, IFI6, NDRG1, GOLGA2, LGALS3. Moreover, pseudo-time series analysis and cell interaction analysis revealed the differentiation of immune cells and the relationship between immune cells in Moyamoya disease. CONCLUSIONS Our study can provide information for the diagnosis and treatment of moyamoya disease.
Collapse
Affiliation(s)
- Qikai Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Wenjun Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Jie Huang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Yuting Wu
- Department of pharmacy, university of Southern California, Los Angeles, CA, USA
| | - Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Yiming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Qianmiao Zhu
- Department of Neurosurgery, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, P.R. China
| | - Jiacheng Lu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Jiaheng Xie
- Department of Burn and Plastic Surgery, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yu Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Xiaoman Mao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China
| | - Wei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
8
|
Liu X, Zhang S, Mao Y, Lin S, Wu H, Ou S. Optimization of method for achieving a single-cell suspension from mouse corneas. Exp Eye Res 2023:109544. [PMID: 37336469 DOI: 10.1016/j.exer.2023.109544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 05/28/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
The single-cell RNA-sequencing (scRNA-seq) technique is used to explore the biological characteristics of tissues under pathological and physiological conditions that include certain chronic eye diseases. Harvesting of single-cell suspensions is one challenge inherent to scRNA-seq procedures. This study aimed to use an optimized method to digest a whole mouse cornea to harvest single-cell suspensions. We utilized five different mouse cornea digestion methods to obtain single-cell suspensions: (1) 5 dissected mouse corneas were cut into pieces (∼0.5 mm) and digested in trypsin for 10 min, and this digestion was repeated for 10 cycles; (2) 5 dissected mouse corneas were cut into pieces and incubated with 5 mg/ml collagenase A at 37 °C for 1h and then further digested in trypsin at 37 °C for 10 min; (3) used the same approach as that used in method 2, but the second digestion step was performed in TrypLE for 20 min; (4) used the same approach as that used in method 2, but the concentration of collagenase A was 2 mg/ml and the incubation time was 2h; (5) used the same approach as that used in method 3, but the corneas were incubated in 2 mg/ml collagenase A at 37 °C for 2h. Trypan blue staining was used to calculate the cell viability and agglomeration rate. The cell types and percentages were determined using immunofluorescence staining. RNA integrity number (RIN) was measured by Agilent 2100. Method 1 showed the lowest cell yield (0.375 × 105), epithelial cell percentage, and less than 70% cell viability, thus not a proper protocol. Method 2 showed the highest cell viability (over 90%), percentage of single-cell (89.53%), and high cell quantity (1.05 × 105). Method 3 had a significantly lower cell viability (55.30%). Cell agglomeration rates of method 4 and 5 reached up to 20% and 13%, and with lower cell viability (72.51%, 59.87%, respectively) and decreased epithelial cell rate compared to method 2 and 3. The results suggest that method 2 (5 mg/ml collagenase A and trypsin) is a preferred protocol for digesting mouse cornea to obtain single-cell suspension which achieves the criterion of single-cell RNA sequencing.
Collapse
Affiliation(s)
- Xiaodong Liu
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China; School of Medicine, Xiamen University, Xiamen, Fujian, 361002, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian, 361002, China
| | - Shengpeng Zhang
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China; School of Medicine, Xiamen University, Xiamen, Fujian, 361002, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian, 361002, China
| | - Yi Mao
- The Affiliated Hospital of Guizhou Medical University, Guiyang, 350000, Guizhou, China; Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China; School of Medicine, Xiamen University, Xiamen, Fujian, 361002, China
| | - Sijie Lin
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China
| | - Huping Wu
- Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China; School of Medicine, Xiamen University, Xiamen, Fujian, 361002, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian, 361002, China.
| | - Shangkun Ou
- The Affiliated Hospital of Guizhou Medical University, Guiyang, 350000, Guizhou, China; Eye Institute and Affiliated Xiamen Eye Center of Xiamen University, Fujian Provincial Key Laboratory of Cornea & Ocular Surface Diseases, Xiamen, Fujian, 361002, China; School of Medicine, Xiamen University, Xiamen, Fujian, 361002, China; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Xiamen, Fujian, 361002, China.
| |
Collapse
|
9
|
Theodoris CV, Xiao L, Chopra A, Chaffin MD, Al Sayed ZR, Hill MC, Mantineo H, Brydon EM, Zeng Z, Liu XS, Ellinor PT. Transfer learning enables predictions in network biology. Nature 2023; 618:616-624. [PMID: 37258680 PMCID: PMC10949956 DOI: 10.1038/s41586-023-06139-9] [Citation(s) in RCA: 242] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/27/2023] [Indexed: 06/02/2023]
Abstract
Mapping gene networks requires large amounts of transcriptomic data to learn the connections between genes, which impedes discoveries in settings with limited data, including rare diseases and diseases affecting clinically inaccessible tissues. Recently, transfer learning has revolutionized fields such as natural language understanding1,2 and computer vision3 by leveraging deep learning models pretrained on large-scale general datasets that can then be fine-tuned towards a vast array of downstream tasks with limited task-specific data. Here, we developed a context-aware, attention-based deep learning model, Geneformer, pretrained on a large-scale corpus of about 30 million single-cell transcriptomes to enable context-specific predictions in settings with limited data in network biology. During pretraining, Geneformer gained a fundamental understanding of network dynamics, encoding network hierarchy in the attention weights of the model in a completely self-supervised manner. Fine-tuning towards a diverse panel of downstream tasks relevant to chromatin and network dynamics using limited task-specific data demonstrated that Geneformer consistently boosted predictive accuracy. Applied to disease modelling with limited patient data, Geneformer identified candidate therapeutic targets for cardiomyopathy. Overall, Geneformer represents a pretrained deep learning model from which fine-tuning towards a broad range of downstream applications can be pursued to accelerate discovery of key network regulators and candidate therapeutic targets.
Collapse
Affiliation(s)
- Christina V Theodoris
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA.
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School Genetics Training Program, Boston, USA.
| | - Ling Xiao
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Anant Chopra
- Precision Cardiology Laboratory, Bayer US LLC, Cambridge, MA, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zeina R Al Sayed
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew C Hill
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Helene Mantineo
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Zexian Zeng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - X Shirley Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative and Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
10
|
Boal AM, McGrady NR, Chamling X, Kagitapalli BS, Zack DJ, Calkins DJ, Risner ML. Microfluidic Platforms Promote Polarization of Human-Derived Retinal Ganglion Cells That Model Axonopathy. Transl Vis Sci Technol 2023; 12:1. [PMID: 37010860 PMCID: PMC10080917 DOI: 10.1167/tvst.12.4.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023] Open
Abstract
Purpose Axons depend on long-range transport of proteins and organelles which increases susceptibility to metabolic stress in disease. The axon initial segment (AIS) is particularly vulnerable due to the high bioenergetic demand of action potential generation. Here, we prepared retinal ganglion cells derived from human embryonic stem cells (hRGCs) to probe how axonal stress alters AIS morphology. Methods hRGCs were cultured on coverslips or microfluidic platforms. We assayed AIS specification and morphology by immunolabeling against ankyrin G (ankG), an axon-specific protein, and postsynaptic density 95 (PSD-95), a dendrite-specific protein. Using microfluidic platforms that enable fluidic isolation, we added colchicine to the axon compartment to lesion axons. We verified axonopathy by measuring the anterograde axon transport of cholera toxin subunit B and immunolabeling against cleaved caspase 3 (CC3) and phosphorylated neurofilament H (SMI-34). We determined the influence of axon injury on AIS morphology by immunolabeling samples against ankG and measuring AIS distance from soma and length. Results Based on measurements of ankG and PSD-95 immunolabeling, microfluidic platforms promote the formation and separation of distinct somatic-dendritic versus axonal compartments in hRGCs compared to coverslip cultures. Chemical lesioning of axons by colchicine reduced hRGC anterograde axon transport, increased varicosity density, and enhanced expression of CC3 and SMI-34. Interestingly, we found that colchicine selectively affected hRGCs with axon-carrying dendrites by reducing AIS distance from somas and increasing length, thus suggesting reduced capacity to maintain excitability. Conclusions Thus, microfluidic platforms promote polarized hRGCs that enable modeling of axonopathy. Translational Relevance Microfluidic platforms may be used to assay compartmentalized degeneration that occurs during glaucoma.
Collapse
Affiliation(s)
- Andrew M. Boal
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nolan R. McGrady
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xitiz Chamling
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bhanu S. Kagitapalli
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Donald J. Zack
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J. Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael L. Risner
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
11
|
Hatit MZC, Dobrowolski CN, Lokugamage MP, Loughrey D, Ni H, Zurla C, Da Silva Sanchez AJ, Radmand A, Huayamares SG, Zenhausern R, Paunovska K, Peck HE, Kim J, Sato M, Feldman JI, Rivera MA, Cristian A, Kim Y, Santangelo PJ, Dahlman JE. Nanoparticle stereochemistry-dependent endocytic processing improves in vivo mRNA delivery. Nat Chem 2023; 15:508-515. [PMID: 36864143 PMCID: PMC11831600 DOI: 10.1038/s41557-023-01138-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 01/13/2023] [Indexed: 03/04/2023]
Abstract
Stereochemistry can alter small-molecule pharmacokinetics, safety and efficacy. However, it is unclear whether the stereochemistry of a single compound within a multicomponent colloid such as a lipid nanoparticle (LNP) can influence its activity in vivo. Here we report that LNPs containing stereopure 20α-hydroxycholesterol (20α) delivered mRNA to liver cells up to 3-fold more potently than LNPs containing a mixture of both 20α- and 20β-hydroxycholesterols (20mix). This effect was not driven by LNP physiochemical traits. Instead, in vivo single-cell RNA sequencing and imaging revealed that 20mix LNPs were sorted into phagocytic pathways more than 20α LNPs, resulting in key differences between LNP biodistribution and subsequent LNP functional delivery. These data are consistent with the fact that nanoparticle biodistribution is necessary, but not sufficient, for mRNA delivery, and that stereochemistry-dependent interactions between LNPs and target cells can improve mRNA delivery.
Collapse
Affiliation(s)
- Marine Z C Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Curtis N Dobrowolski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Melissa P Lokugamage
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Huanzhen Ni
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Chiara Zurla
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Alejandro J Da Silva Sanchez
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Afsane Radmand
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Ryan Zenhausern
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Hannah E Peck
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Jinwhan Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- School of Electrical & Computer Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Manaka Sato
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Jacob I Feldman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Michael-Alexander Rivera
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Ana Cristian
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - YongTae Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
12
|
Riordan SM, Aladdad AM, McLoughlin KJ, Kador KE. Purification of Retinal Ganglion Cells from Differentiation Through Adult via Immunopanning and Low-Pressure Flow Cytometry. Methods Mol Biol 2023; 2708:11-24. [PMID: 37558955 DOI: 10.1007/978-1-0716-3409-7_2] [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] [Indexed: 08/11/2023]
Abstract
The isolation and culturing of rodent retinal ganglion cells (RGC) is a key step in studying the function and cellular response of this crucial cell type. Typical methods used for isolation of RGCs include immunopanning or magnetic bead separation with antibodies targeting RGC specific protein markers. However, in developmental research, many of the most common markers, such as Thy-1, are not expressed in early stages of development. To help study these crucial early stage RGCs, we have developed a novel method that utilizes a transgenic mouse with a GFP tag on the protein BRN3 and a low-pressure fluorescence-activated cell sorter (FACS) system.
Collapse
Affiliation(s)
- Sean M Riordan
- Department of Ophthalmology and Department of Biomedical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Afnan M Aladdad
- Department of Ophthalmology and Department of Biomedical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Kiran J McLoughlin
- Department of Ophthalmology and Department of Biomedical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA
- Wellcome Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Karl E Kador
- Department of Ophthalmology and Department of Biomedical Sciences, University of Missouri-Kansas City, Kansas City, MO, USA.
| |
Collapse
|
13
|
Croteau LP, Risner ML, Wareham LK, McGrady NR, Chamling X, Zack DJ, Calkins DJ. Ex Vivo Integration of Human Stem Retinal Ganglion Cells into the Mouse Retina. Cells 2022; 11:cells11203241. [PMID: 36291110 PMCID: PMC9600680 DOI: 10.3390/cells11203241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/04/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Cell replacement therapies may be key in achieving functional recovery in neurodegenerative optic neuropathies diseases such as glaucoma. One strategy that holds promise in this regard is the use of human embryonic stem cell and induced pluripotent stem-derived retinal ganglion cells (hRGCs). Previous hRGC transplantation studies have shown modest success. This is in part due to the low survival and integration of the transplanted cells in the host retina. The field is further challenged by mixed assays and outcome measurements that probe and determine transplantation success. Thefore, we have devised a transplantation assay involving hRGCs and mouse retina explants that bypasses physical barriers imposed by retinal membranes. We show that hRGC neurites and somas are capable of invading mouse explants with a subset of hRGC neurites being guided by mouse RGC axons. Neonatal mouse retina explants, and to a lesser extent, adult explants, promote hRGC integrity and neurite outgrowth. Using this assay, we tested whether suppmenting cultures with brain derived neurotrophic factor (BDNF) and the adenylate cyclase activator, forskolin, enhances hRGC neurite integration, neurite outgrowth, and integrity. We show that supplementing cultures with a combination BDNF and forskolin strongly favors hRGC integrity, increasing neurite outgrowth and complexity as well as the invasion of mouse explants. The transplantation assay presented here is a practical tool for investigating strategies for testing and optimizing the integration of donor cells into host tissues.
Collapse
Affiliation(s)
- Louis-Philippe Croteau
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Michael L. Risner
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Lauren K. Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Nolan R. McGrady
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Donald J. Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David J. Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
14
|
Madadi Y, Sun J, Chen H, Williams R, Yousefi S. Detecting retinal neural and stromal cell classes and ganglion cell subtypes based on transcriptome data with deep transfer learning. Bioinformatics 2022; 38:4321-4329. [PMID: 35876552 PMCID: PMC9991888 DOI: 10.1093/bioinformatics/btac514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/11/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION To develop and assess the accuracy of deep learning models that identify different retinal cell types, as well as different retinal ganglion cell (RGC) subtypes, based on patterns of single-cell RNA sequencing (scRNA-seq) in multiple datasets. RESULTS Deep domain adaptation models were developed and tested using three different datasets. The first dataset included 44 808 single retinal cells from mice (39 cell types) with 24 658 genes, the second dataset included 6225 single RGCs from mice (41 subtypes) with 13 616 genes and the third dataset included 35 699 single RGCs from mice (45 subtypes) with 18 222 genes. We used four loss functions in the learning process to align the source and target distributions, reduce misclassification errors and maximize robustness. Models were evaluated based on classification accuracy and confusion matrix. The accuracy of the model for correctly classifying 39 different retinal cell types in the first dataset was ∼92%. Accuracy in the second and third datasets reached ∼97% and 97% in correctly classifying 40 and 45 different RGCs subtypes, respectively. Across a range of seven different batches in the first dataset, the accuracy of the lead model ranged from 74% to nearly 100%. The lead model provided high accuracy in identifying retinal cell types and RGC subtypes based on scRNA-seq data. The performance was reasonable based on data from different batches as well. The validated model could be readily applied to scRNA-seq data to identify different retinal cell types and subtypes. AVAILABILITY AND IMPLEMENTATION The code and datasets are available on https://github.com/DM2LL/Detecting-Retinal-Cell-Classes-and-Ganglion-Cell-Subtypes. We have also added the class labels of all samples to the datasets. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yeganeh Madadi
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
- University of Tehran, Tehran, Iran
| | - Jian Sun
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert Williams
- Department of Genetics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Siamak Yousefi
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Genetics and Informatics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
15
|
Menassa DA, Muntslag TAO, Martin-Estebané M, Barry-Carroll L, Chapman MA, Adorjan I, Tyler T, Turnbull B, Rose-Zerilli MJJ, Nicoll JAR, Krsnik Z, Kostovic I, Gomez-Nicola D. The spatiotemporal dynamics of microglia across the human lifespan. Dev Cell 2022; 57:2127-2139.e6. [PMID: 35977545 PMCID: PMC9616795 DOI: 10.1016/j.devcel.2022.07.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/22/2022] [Accepted: 07/26/2022] [Indexed: 11/07/2022]
Abstract
Microglia, the brain's resident macrophages, shape neural development and are key neuroimmune hubs in the pathological signatures of neurodevelopmental disorders. Despite the importance of microglia, their development has not been carefully examined in the human brain, and most of our knowledge derives from rodents. We aimed to address this gap in knowledge by establishing an extensive collection of 97 post-mortem tissues in order to enable quantitative, sex-matched, detailed analysis of microglia across the human lifespan. We identify the dynamics of these cells in the human telencephalon, describing waves in microglial density across gestation, infancy, and childhood, controlled by a balance of proliferation and apoptosis, which track key neurodevelopmental milestones. These profound changes in microglia are also observed in bulk RNA-seq and single-cell RNA-seq datasets. This study provides a detailed insight into the spatiotemporal dynamics of microglia across the human lifespan and serves as a foundation for elucidating how microglia contribute to shaping neurodevelopment in humans.
Collapse
Affiliation(s)
- David A Menassa
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom; Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Tim A O Muntslag
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Maria Martin-Estebané
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Liam Barry-Carroll
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Mark A Chapman
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Istvan Adorjan
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Teadora Tyler
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Bethany Turnbull
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | | | - James A R Nicoll
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Zeljka Krsnik
- Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Ivica Kostovic
- Croatian Institute for Brain Research, University of Zagreb Medical School, Zagreb, Croatia
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom.
| |
Collapse
|
16
|
Guy B, Zhang JS, Duncan LH, Johnston RJ. Human neural organoids: Models for developmental neurobiology and disease. Dev Biol 2021; 478:102-121. [PMID: 34181916 PMCID: PMC8364509 DOI: 10.1016/j.ydbio.2021.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/08/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022]
Abstract
Human organoids stand at the forefront of basic and translational research, providing experimentally tractable systems to study human development and disease. These stem cell-derived, in vitro cultures can generate a multitude of tissue and organ types, including distinct brain regions and sensory systems. Neural organoid systems have provided fundamental insights into molecular mechanisms governing cell fate specification and neural circuit assembly and serve as promising tools for drug discovery and understanding disease pathogenesis. In this review, we discuss several human neural organoid systems, how they are generated, advances in 3D imaging and bioengineering, and the impact of organoid studies on our understanding of the human nervous system.
Collapse
Affiliation(s)
- Brian Guy
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Jingliang Simon Zhang
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
17
|
Gautam P, Hamashima K, Chen Y, Zeng Y, Makovoz B, Parikh BH, Lee HY, Lau KA, Su X, Wong RCB, Chan WK, Li H, Blenkinsop TA, Loh YH. Multi-species single-cell transcriptomic analysis of ocular compartment regulons. Nat Commun 2021; 12:5675. [PMID: 34584087 PMCID: PMC8478974 DOI: 10.1038/s41467-021-25968-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 09/07/2021] [Indexed: 11/23/2022] Open
Abstract
The retina is a widely profiled tissue in multiple species by single-cell RNA sequencing studies. However, integrative research of the retina across species is lacking. Here, we construct the first single-cell atlas of the human and porcine ocular compartments and study inter-species differences in the retina. In addition to that, we identify putative adult stem cells present in the iris tissue. We also create a disease map of genes involved in eye disorders across compartments of the eye. Furthermore, we probe the regulons of different cell populations, which include transcription factors and receptor-ligand interactions and reveal unique directional signalling between ocular cell types. In addition, we study conservation of regulons across vertebrates and zebrafish to identify common core factors. Here, we show perturbation of KLF7 gene expression during retinal ganglion cells differentiation and conclude that it plays a significant role in the maturation of retinal ganglion cells. A comprehensive analysis of the ocular networks among various tissues is necessary to understand eye physiology in health and disease. Here the authors present a multi-species single-cell transcriptomic atlas consisting of cells of the cornea, iris, ciliary body, neural retina, retinal pigmented epithelium, and choroid.
Collapse
Affiliation(s)
- Pradeep Gautam
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore
| | - Kiyofumi Hamashima
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore
| | - Ying Chen
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore.,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore.,Integrative Sciences and Engineering Programme (ISEP), NUS Graduate School, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077, Singapore
| | - Yingying Zeng
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Bar Makovoz
- Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Bhav Harshad Parikh
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Translational Retinal Research Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore
| | - Hsin Yee Lee
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore
| | - Katherine Anne Lau
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore
| | - Xinyi Su
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Translational Retinal Research Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore.,Singapore Eye Research Institute, 11 Third Hospital Avenue, Singapore, 168751, Singapore
| | - Raymond C B Wong
- Centre for Eye Research Australia, Melbourne, Vic, Australia.,Ophthalmology, Department of Surgery, University of Melbourne, Melbourne, Vic, Australia.,Shenzhen Eye Hospital, Shenzhen University School of Medicine, Shenzhen, China
| | - Woon-Khiong Chan
- Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore.,Integrative Sciences and Engineering Programme (ISEP), NUS Graduate School, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077, Singapore
| | - Hu Li
- Center for Individualized Medicine, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| | | | - Yuin-Han Loh
- Cell Fate Engineering and Therapeutics Laboratory, A*STAR Institute of Molecular and Cell Biology, Singapore, 138673, Singapore. .,Department of Biological Sciences, National University of Singapore, Singapore, 117543, Singapore. .,Integrative Sciences and Engineering Programme (ISEP), NUS Graduate School, National University of Singapore, 21 Lower Kent Ridge Road, Singapore, 119077, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
18
|
Risner ML, Pasini S, Chamling X, McGrady NR, Goldberg JL, Zack DJ, Calkins DJ. Intrinsic Morphologic and Physiologic Development of Human Derived Retinal Ganglion Cells In Vitro. Transl Vis Sci Technol 2021; 10:1. [PMID: 34383881 PMCID: PMC8362626 DOI: 10.1167/tvst.10.10.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Purpose Human retinal ganglion cells (hRGC) derived from human pluripotent stem cells are promising candidates to model, protect, and replace degenerating RGCs. Here, we examined intrinsic morphologic and physiologic development of hRGCs. Methods We used CRISPR-Cas9 to selectively express tdTomato under the RGC-specific promoter, BRN3B. Human pluripotent stem cells were chemically differentiated into hRGCs and cultured up to 7 weeks. We measured soma area, neurite complexity, synaptic protein, axon-related messenger RNA and protein, and voltage-dependent responses. Results Soma area, neurite complexity, and postsynaptic density protein 95 increased over time. Soma area and neurite complexity increased proportionally week to week, and this relationship was dynamic, strengthening between 2 and 3 weeks and diminishing by 4 weeks. Postsynaptic density 95 localization was dependent on culture duration. After 1 to 2 weeks, postsynaptic density 95 localized within somas but redistributed along neurites after 3 to 4 weeks. Axon initial segment scaffolding protein, Ankyrin G, expression also increased over time, and by 7 weeks, Ankyrin G often localized within putative axons. Voltage-gated inward currents progressively developed, but outward currents matured by 4 weeks. Current-induced spike generation increased over time but limited by depolarization block. Conclusions Human RGCs develop up to 7 weeks after culture. Thus, the state of hRGC maturation should be accounted for in designing models and treatments for optic neuropathies. Translational Relevance We characterized hRGC morphologic and physiologic development towards identifying key time points when hRGCs express mechanisms that may be harnessed to enhance the efficacy of neuroprotective and cell replacement therapies.
Collapse
Affiliation(s)
- Michael L Risner
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Silvia Pasini
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xitiz Chamling
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nolan R McGrady
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey L Goldberg
- Byers Eye Institute, Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Donald J Zack
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J Calkins
- Vanderbilt Eye Institute, Department of Ophthalmology & Visual Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
19
|
Mullin NK, Voigt AP, Cooke JA, Bohrer LR, Burnight ER, Stone EM, Mullins RF, Tucker BA. Patient derived stem cells for discovery and validation of novel pathogenic variants in inherited retinal disease. Prog Retin Eye Res 2021; 83:100918. [PMID: 33130253 PMCID: PMC8559964 DOI: 10.1016/j.preteyeres.2020.100918] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/22/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Our understanding of inherited retinal disease has benefited immensely from molecular genetic analysis over the past several decades. New technologies that allow for increasingly detailed examination of a patient's DNA have expanded the catalog of genes and specific variants that cause retinal disease. In turn, the identification of pathogenic variants has allowed the development of gene therapies and low-cost, clinically focused genetic testing. Despite this progress, a relatively large fraction (at least 20%) of patients with clinical features suggestive of an inherited retinal disease still do not have a molecular diagnosis today. Variants that are not obviously disruptive to the codon sequence of exons can be difficult to distinguish from the background of benign human genetic variations. Some of these variants exert their pathogenic effect not by altering the primary amino acid sequence, but by modulating gene expression, isoform splicing, or other transcript-level mechanisms. While not discoverable by DNA sequencing methods alone, these variants are excellent targets for studies of the retinal transcriptome. In this review, we present an overview of the current state of pathogenic variant discovery in retinal disease and identify some of the remaining barriers. We also explore the utility of new technologies, specifically patient-derived induced pluripotent stem cell (iPSC)-based modeling, in further expanding the catalog of disease-causing variants using transcriptome-focused methods. Finally, we outline bioinformatic analysis techniques that will allow this new method of variant discovery in retinal disease. As the knowledge gleaned from previous technologies is informing targets for therapies today, we believe that integrating new technologies, such as iPSC-based modeling, into the molecular diagnosis pipeline will enable a new wave of variant discovery and expanded treatment of inherited retinal disease.
Collapse
Affiliation(s)
- Nathaniel K Mullin
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Andrew P Voigt
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jessica A Cooke
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Laura R Bohrer
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Erin R Burnight
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Edwin M Stone
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Robert F Mullins
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Budd A Tucker
- The Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
20
|
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
|
21
|
Chen T, Li J, Jia Y, Wang J, Sang R, Zhang Y, Rong R. Single-cell Sequencing in the Field of Stem Cells. Curr Genomics 2021; 21:576-584. [PMID: 33414679 PMCID: PMC7770636 DOI: 10.2174/1389202921999200624154445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 11/24/2022] Open
Abstract
Variation and heterogeneity between cells are the basic characteristics of stem cells. Traditional sequencing analysis methods often cover up this difference. Single-cell sequencing technology refers to the technology of high-throughput sequencing analysis of genomes at the single-cell level. It can effectively analyze cell heterogeneity and identify a small number of cell populations. With the continuous progress of cell sorting, nucleic acid extraction and other technologies, single-cell sequencing technology has also made great progress. Encouraging new discoveries have been made in stem cell research, including pluripotent stem cells, tissue-specific stem cells and cancer stem cells. In this review, we discuss the latest progress and future prospects of single-cell sequencing technology in the field of stem cells.
Collapse
Affiliation(s)
- Tian Chen
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiawei Li
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yichen Jia
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Jiyan Wang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ruirui Sang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yi Zhang
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Ruiming Rong
- 1Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 2Shanghai Key Laboratory of Organ Transplantation, Shanghai, P.R. China; 3Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai, P.R. China; 4Department of Urology, Shanghai Public Health Clinical Center, Shanghai, P.R. China; 5Department of Transfusion, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
22
|
Role of the Internal Limiting Membrane in Structural Engraftment and Topographic Spacing of Transplanted Human Stem Cell-Derived Retinal Ganglion Cells. Stem Cell Reports 2020; 16:149-167. [PMID: 33382979 PMCID: PMC7897583 DOI: 10.1016/j.stemcr.2020.12.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/30/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Retinal ganglion cell (RGC) replacement holds potential for restoring vision lost to optic neuropathy. Transplanted RGCs must undergo neuroretinal integration to receive afferent visual signals for processing and efferent transmission. To date, retinal integration following RGC transplantation has been limited. We sought to overcome key barriers to transplanted human stem cell-derived RGC integration. Following co-culture ex vivo on organotypic mouse retinal explants, human RGCs cluster and extend bundled neurites that remain superficial to the neuroretina, hindering afferent synaptogenesis. To enhance integration, we increased the cellular permeability of the internal limiting membrane (ILM). Extracellular matrix digestion using proteolytic enzymes achieved ILM disruption while minimizing retinal toxicity and preserving glial reactivity. ILM disruption is associated with dispersion rather than clustering of co-cultured RGC bodies and neurites, and increased parenchymal neurite ingrowth. The ILM represents a significant obstacle to transplanted RGC connectivity and its circumvention may be necessary for functional RGC replacement.
Collapse
|
23
|
Single-cell RNA sequencing in vision research: Insights into human retinal health and disease. Prog Retin Eye Res 2020; 83:100934. [PMID: 33383180 DOI: 10.1016/j.preteyeres.2020.100934] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 01/03/2023]
Abstract
Gene expression provides valuable insight into cell function. As such, vision researchers have frequently employed gene expression studies to better understand retinal physiology and disease. With the advent of single-cell RNA sequencing, expression experiments provide an unparalleled resolution of information. Instead of studying aggregated gene expression across all cells in a heterogenous tissue, single-cell technology maps RNA to an individual cell, which facilitates grouping of retinal and choroidal cell types for further study. Single-cell RNA sequencing has been quickly adopted by both basic and translational vision researchers, and single-cell level gene expression has been studied in the visual systems of animal models, retinal organoids, and primary human retina, RPE, and choroid. These experiments have generated detailed atlases of gene expression and identified new retinal cell types. Likewise, single-cell RNA sequencing investigations have characterized how gene expression changes in the setting of many retinal diseases, including how choroidal endothelial cells are altered in age-related macular degeneration. In addition, this technology has allowed vision researchers to discover drivers of retinal development and model rare retinal diseases with induced pluripotent stem cells. In this review, we will overview the growing number of single-cell RNA sequencing studies in the field of vision research. We will summarize experimental considerations for designing single-cell RNA sequencing experiments and highlight important advancements in retinal, RPE, choroidal, and retinal organoid biology driven by this technology. Finally, we generalize these findings to genes involved in retinal degeneration and outline the future of single-cell expression experiments in studying retinal disease.
Collapse
|
24
|
Young MD, Behjati S. SoupX removes ambient RNA contamination from droplet-based single-cell RNA sequencing data. Gigascience 2020; 9:giaa151. [PMID: 33367645 PMCID: PMC7763177 DOI: 10.1093/gigascience/giaa151] [Citation(s) in RCA: 704] [Impact Index Per Article: 140.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 10/13/2020] [Accepted: 11/27/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Droplet-based single-cell RNA sequence analyses assume that all acquired RNAs are endogenous to cells. However, any cell-free RNAs contained within the input solution are also captured by these assays. This sequencing of cell-free RNA constitutes a background contamination that confounds the biological interpretation of single-cell transcriptomic data. RESULTS We demonstrate that contamination from this "soup" of cell-free RNAs is ubiquitous, with experiment-specific variations in composition and magnitude. We present a method, SoupX, for quantifying the extent of the contamination and estimating "background-corrected" cell expression profiles that seamlessly integrate with existing downstream analysis tools. Applying this method to several datasets using multiple droplet sequencing technologies, we demonstrate that its application improves biological interpretation of otherwise misleading data, as well as improving quality control metrics. CONCLUSIONS We present SoupX, a tool for removing ambient RNA contamination from droplet-based single-cell RNA sequencing experiments. This tool has broad applicability, and its application can improve the biological utility of existing and future datasets.
Collapse
Affiliation(s)
- Matthew D Young
- Wellcome Trust Sanger Institute, Cellular Genetics, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
| | - Sam Behjati
- Wellcome Trust Sanger Institute, Cellular Genetics, Wellcome Genome Campus, Hinxton, CB10 1SA, UK
- Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge, CB2 0QQ, UK
- University of Cambridge, Department of Paediatrics, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| |
Collapse
|
25
|
O'Hara-Wright M, Gonzalez-Cordero A. Retinal organoids: a window into human retinal development. Development 2020; 147:147/24/dev189746. [PMID: 33361444 PMCID: PMC7774906 DOI: 10.1242/dev.189746] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Retinal development and maturation are orchestrated by a series of interacting signalling networks that drive the morphogenetic transformation of the anterior developing brain. Studies in model organisms continue to elucidate these complex series of events. However, the human retina shows many differences from that of other organisms and the investigation of human eye development now benefits from stem cell-derived organoids. Retinal differentiation methods have progressed from simple 2D adherent cultures to self-organising micro-physiological systems. As models of development, these have collectively offered new insights into the previously unexplored early development of the human retina and informed our knowledge of the key cell fate decisions that govern the specification of light-sensitive photoreceptors. Although the developmental trajectories of other retinal cell types remain more elusive, the collation of omics datasets, combined with advanced culture methodology, will enable modelling of the intricate process of human retinogenesis and retinal disease in vitro. Summary: Retinal organoid systems derived from human pluripotent stem cells are micro-physiological systems that offer new insights into previously unexplored human retina development.
Collapse
Affiliation(s)
- Michelle O'Hara-Wright
- Stem Cell Medicine Group, Children's Medical Research Institute, University of Sydney, Westmead, 2145, NSW, Australia.,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145, NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, University of Sydney, Westmead, 2145, NSW, Australia .,School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145, NSW, Australia
| |
Collapse
|
26
|
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
|
27
|
Fadl BR, Brodie SA, Malasky M, Boland JF, Kelly MC, Kelley MW, Boger E, Fariss R, Swaroop A, Campello L. An optimized protocol for retina single-cell RNA sequencing. Mol Vis 2020; 26:705-717. [PMID: 33088174 PMCID: PMC7553720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/08/2020] [Indexed: 10/25/2022] Open
Abstract
Purpose Single-cell RNA sequencing (scRNA-seq) is a powerful technique used to explore gene expression at the single cell level. However, appropriate preparation of samples is essential to obtain the most information out of this transformative technology. Generating high-quality single-cell suspensions from the retina is critical to preserve the native expression profile that will ensure meaningful transcriptome data analysis. Methods We modified the conditions for rapid and optimal dissociation of retina sample preparation. We also included additional filtering steps in data analysis for retinal scRNA-seq. Results We report a gentle method for dissociation of the mouse retina that minimizes cell death and preserves cell morphology. This protocol also results in detection of higher transcriptional complexity. In addition, the modified computational pipeline leads to better-quality single-cell RNA-sequencing data in retina samples. We also demonstrate the advantages and limitations of using fresh versus frozen retinas to prepare cell or nuclei suspensions for scRNA-seq. Conclusions We provide a simple yet robust and reproducible protocol for retinal scRNA-seq analysis, especially for comparative studies.
Collapse
Affiliation(s)
- Benjamin R. Fadl
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Seth A. Brodie
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health & NCI-Frederick, Leidos Biomedical Research Inc., Frederick, MD
| | - Michael Malasky
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health & NCI-Frederick, Leidos Biomedical Research Inc., Frederick, MD
| | - Joseph F. Boland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health & NCI-Frederick, Leidos Biomedical Research Inc., Frederick, MD
| | - Michael C. Kelly
- Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD
| | - Matthew W. Kelley
- Laboratory of Cochlear Development, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Erich Boger
- Genomics and Computational Biology Core, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD
| | - Robert Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD
| | - Laura Campello
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Palfi A, Yesmambetov A, Humphries P, Hokamp K, Farrar GJ. Non-photoreceptor Expression of Tulp1 May Contribute to Extensive Retinal Degeneration in Tulp1-/- Mice. Front Neurosci 2020; 14:656. [PMID: 32655363 PMCID: PMC7325604 DOI: 10.3389/fnins.2020.00656] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
Mutations in tubby like protein 1 gene (TULP1) are causative of early-onset recessive inherited retinal degenerations (IRDs); similarly, the Tulp1-/- mouse is also characterized by a rapid IRD. Tulp1 mRNA and protein expression was analyzed in wild type mouse retinas and expression data sets (NCBI) during early postnatal development. Comparative histology was undertaken in Tulp1-/-, rhodopsin-/- (Rho-/-) and retinal degeneration slow-/- (Rds-/-) mouse retinas. Bioinformatic analysis of predicted TULP1 interactors and IRD genes was performed. Peak expression of Tulp1 in healthy mouse retinas was detected at p8; of note, TULP1 was detected in both the outer and inner retina. Bioinformatic analysis indicated Tulp1 expression in retinal progenitor, photoreceptor and non-photoreceptor cells. While common features of photoreceptor degeneration were detected in Tulp1-/-, Rho-/-, and Rds-/- retinas, other alterations in bipolar, amacrine and ganglion cells were specific to Tulp1-/- mice. Additionally, predicted TULP1 interactors differed in various retinal cell types and new functions for TULP1 were suggested. A pilot bioinformatic analysis indicated that in a similar fashion to Tulp1, many other IRD genes were expressed in both inner and outer retinal cells at p4-p7. Our data indicate that expression of Tulp1 extends to multiple retinal cell types; lack of TULP1 may lead to primary degeneration not only of photoreceptor but also non-photoreceptor cells. Predicted interactors suggest widespread retinal functions for TULP1. Early and widespread expression of TULP1 and some other IRD genes in both the inner and outer retina highlights potential hurdles in the development of treatments for these IRDs.
Collapse
Affiliation(s)
- Arpad Palfi
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | - Pete Humphries
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Karsten Hokamp
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| | - G Jane Farrar
- Department of Genetics, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
31
|
Zerti D, Collin J, Queen R, Cockell SJ, Lako M. Understanding the complexity of retina and pluripotent stem cell derived retinal organoids with single cell RNA sequencing: current progress, remaining challenges and future prospective. Curr Eye Res 2020; 45:385-396. [PMID: 31794277 PMCID: PMC7034531 DOI: 10.1080/02713683.2019.1697453] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022]
Abstract
Single-cell sequencing technologies have emerged as a revolutionary tool with transformative new methods to profile genetic, epigenetic, spatial, and lineage information in individual cells. Single-cell RNA sequencing (scRNA-Seq) allows researchers to collect large datasets detailing the transcriptomes of individual cells in space and time and is increasingly being applied to reveal cellular heterogeneity in retinal development, normal physiology, and disease, and provide new insights into cell-type specific markers and signaling pathways. In recent years, scRNA-Seq datasets have been generated from retinal tissue and pluripotent stem cell-derived retinal organoids. Their cross-comparison enables staging of retinal organoids, identification of specific cells in developing and adult human neural retina and provides deeper insights into cell-type sub-specification and geographical differences. In this article, we review the recent rapid progress in scRNA-Seq analyses of retina and retinal organoids, the questions that remain unanswered and the technical challenges that need to be overcome to achieve consistent results that reflect the complexity, functionality, and interactions of all retinal cell types.
Collapse
Affiliation(s)
- Darin Zerti
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Joseph Collin
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| | - Rachel Queen
- Bioinformatics Core Facility, Newcastle University, Newcastle upon Tyne, UK
| | - Simon J. Cockell
- Bioinformatics Core Facility, Newcastle University, Newcastle upon Tyne, UK
| | - Majlinda Lako
- Institute of Genetic Medicine, Newcastle University, Newcastle, UK
| |
Collapse
|
32
|
Ohlemacher SK, Langer KB, Fligor CM, Feder EM, Edler MC, Meyer JS. Advances in the Differentiation of Retinal Ganglion Cells from Human Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:121-140. [PMID: 31654388 DOI: 10.1007/978-3-030-28471-8_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cell (hPSC) technology has revolutionized the field of biology through the unprecedented ability to study the differentiation of human cells in vitro. In the past decade, hPSCs have been applied to study development, model disease, develop drugs, and devise cell replacement therapies for numerous biological systems. Of particular interest is the application of this technology to study and treat optic neuropathies such as glaucoma. Retinal ganglion cells (RGCs) are the primary cell type affected in these diseases, and once lost, they are unable to regenerate in adulthood. This necessitates the development of strategies to study the mechanisms of degeneration as well as develop translational therapeutic approaches to treat early- and late-stage disease progression. Numerous protocols have been established to derive RGCs from hPSCs, with the ability to generate large populations of human RGCs for translational applications. In this review, the key applications of hPSCs within the retinal field are described, including the use of these cells as developmental models, disease models, drug development, and finally, cell replacement therapies. In greater detail, the current report focuses on the differentiation of hPSC-derived RGCs and the many unique characteristics associated with these cells in vitro including their genetic identifiers, their electrophysiological activity, and their morphological maturation. Also described is the current progress in the use of patient-specific hPSCs to study optic neuropathies affecting RGCs, with emphasis on the use of these RGCs for studying disease mechanisms and pathogenesis, drug screening, and cell replacement therapies in future studies.
Collapse
Affiliation(s)
- Sarah K Ohlemacher
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Kirstin B Langer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Clarisse M Fligor
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Elyse M Feder
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Michael C Edler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Jason S Meyer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA. .,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA. .,Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
33
|
Lidgerwood GE, Hewitt AW, Pébay A. Human pluripotent stem cells for the modelling of diseases of the retina and optic nerve: toward a retina in a dish. Curr Opin Pharmacol 2019; 48:114-119. [PMID: 31590110 DOI: 10.1016/j.coph.2019.09.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 01/08/2023]
Abstract
Human pluripotent stem cells can be differentiated into specific, relevant cell types of interest including the cells of the retina and optic nerve. These cells can then be used to study fundamental biology as well as disease modelling and subsequent screening of potential treatments. Many models of differentiation and modelling have relied on two-dimensional monocultures of specific cell types, which are not representative of the complexity of the human retina and optic nerve. Hence, more complex models of the human retina and optic nerve are required. Three-dimensional organoids and emerging cell culture methods may provide more physiologically relevant models to study developmental biology and pathology of the retina and optic nerve.
Collapse
Affiliation(s)
- Grace E Lidgerwood
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Australia; Department of Surgery, The University of Melbourne, Parkville, Australia; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia
| | - Alex W Hewitt
- Department of Surgery, The University of Melbourne, Parkville, Australia; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, Australia; School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Tasmania, Australia
| | - Alice Pébay
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Australia; Department of Surgery, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
34
|
Senabouth A, Lukowski SW, Hernandez JA, Andersen SB, Mei X, Nguyen QH, Powell JE. ascend: R package for analysis of single-cell RNA-seq data. Gigascience 2019; 8:giz087. [PMID: 31505654 PMCID: PMC6735844 DOI: 10.1093/gigascience/giz087] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/30/2019] [Accepted: 06/27/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Recent developments in single-cell RNA sequencing (scRNA-seq) platforms have vastly increased the number of cells typically assayed in an experiment. Analysis of scRNA-seq data is multidisciplinary in nature, requiring careful consideration of the application of statistical methods with respect to the underlying biology. Few analysis packages exist that are at once robust, are computationally fast, and allow flexible integration with other bioinformatics tools and methods. FINDINGS ascend is an R package comprising tools designed to simplify and streamline the preliminary analysis of scRNA-seq data, while addressing the statistical challenges of scRNA-seq analysis and enabling flexible integration with genomics packages and native R functions, including fast parallel computation and efficient memory management. The package incorporates both novel and established methods to provide a framework to perform cell and gene filtering, quality control, normalization, dimension reduction, clustering, differential expression, and a wide range of visualization functions. CONCLUSIONS ascend is designed to work with scRNA-seq data generated by any high-throughput platform and includes functions to convert data objects between software packages. The ascend workflow is simple and interactive, as well as suitable for implementation by a broad range of users, including those with little programming experience.
Collapse
Affiliation(s)
- Anne Senabouth
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, Australia 2010
| | - Samuel W Lukowski
- Institute of Molecular Bioscience, 306 Carmody Road, St Lucia, University of Queensland, Brisbane, Australia 4072
| | - Jose Alquicira Hernandez
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, Australia 2010
- Institute of Molecular Bioscience, 306 Carmody Road, St Lucia, University of Queensland, Brisbane, Australia 4072
| | - Stacey B Andersen
- Institute of Molecular Bioscience, 306 Carmody Road, St Lucia, University of Queensland, Brisbane, Australia 4072
| | - Xin Mei
- Institute of Molecular Bioscience, 306 Carmody Road, St Lucia, University of Queensland, Brisbane, Australia 4072
- South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Quan H Nguyen
- Institute of Molecular Bioscience, 306 Carmody Road, St Lucia, University of Queensland, Brisbane, Australia 4072
| | - Joseph E Powell
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, Australia 2010
- School of Medical Sciences, 18 High Street, University of New South Wales, Kensington, Sydney, Australia, 2052
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, Australia, 2010
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Lafzi A, Moutinho C, Picelli S, Heyn H. Tutorial: guidelines for the experimental design of single-cell RNA sequencing studies. Nat Protoc 2018; 13:2742-2757. [DOI: 10.1038/s41596-018-0073-y] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
38
|
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
|
39
|
Stern JH, Tian Y, Funderburgh J, Pellegrini G, Zhang K, Goldberg JL, Ali RR, Young M, Xie Y, Temple S. Regenerating Eye Tissues to Preserve and Restore Vision. Cell Stem Cell 2018; 22:834-849. [PMID: 29859174 PMCID: PMC6492284 DOI: 10.1016/j.stem.2018.05.013] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ocular regenerative therapies are on track to revolutionize treatment of numerous blinding disorders, including corneal disease, cataract, glaucoma, retinitis pigmentosa, and age-related macular degeneration. A variety of transplantable products, delivered as cell suspensions or as preformed 3D structures combining cells and natural or artificial substrates, are in the pipeline. Here we review the status of clinical and preclinical studies for stem cell-based repair, covering key eye tissues from front to back, from cornea to retina, and including bioengineering approaches that advance cell product manufacturing. While recognizing the challenges, we look forward to a deep portfolio of sight-restoring, stem cell-based medicine. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Jeffrey H Stern
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Yangzi Tian
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - James Funderburgh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Graziella Pellegrini
- Centre for Regenerative Medicine, University of Modena and Reggio Emilia, via G.Gottardi 100, 41125 Modena, Italy
| | - Kang Zhang
- Shiley Eye Institute and Institute for Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University and Guangzhou Regenerative Medicine and Health Laboratory, Guangzhou 510060, China
| | - Jeffrey L Goldberg
- Byers Eye Institute at Stanford University, 2452 Watson Court, Palo Alto, CA 94303, USA
| | - Robin R Ali
- Department of Genetics, University College London Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, City Road, London EC1V 2PD, UK; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Michael Young
- The Schepens Eye Research Institute, Massachusetts Eye and Ear, an affiliate of Harvard Medical School, Boston, MA 02114, USA
| | - Yubing Xie
- Colleges of Nanoscale Science and Engineering, SUNY Polytechnic Institute, 257 Fuller Road, Albany, NY 12203, USA
| | - Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA; Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA.
| |
Collapse
|