1
|
Hameed SS, Sharma TP. Generation of Retinal Ganglion Cells from Reprogrammed Keratocytes of Non-Glaucoma and Glaucoma Donors. Curr Protoc 2025; 5:e70091. [PMID: 39781605 PMCID: PMC11713219 DOI: 10.1002/cpz1.70091] [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: 01/12/2025]
Abstract
Human induced pluripotent stem cell (hiPSC)-based disease modeling can be successfully recapitulated to mimic disease characteristics across various human pathologies. Glaucoma, a progressive optic neuropathy, primarily affects the retinal ganglion cells (RGCs). While multiple groups have successfully generated RGCs from non-diseased hiPSCs, producing RGCs from glaucomatous human samples holds significant promise for understanding disease pathology by revealing patient-specific disease signatures. Given that keratocytes originate from the neural crest and previous reports suggest that ocular fibroblasts from glaucomatous donors carry pathogenic signatures, it is highly plausible that these signatures imprinted within the keratocytes will also be present in the derived RGCs. Thus, we aimed to generate RGCs from both glaucomatous and non-glaucomatous donor keratocytes and validate disease-specific signatures in 3D retinal organoids and in isolated RGCs. Our protocol describes the generation of iPSCs from keratocytes of both glaucomatous and non-glaucomatous donors, followed by their differentiation into retinal organoids. Subsequent isolation and culturing of RGCs were performed. Disease signatures in the RGCs were validated in both 3D retinal organoids (ROs) and 2D RGC cultures, and glaucomatous RGCs in 3D and 2D cultures demonstrated increased cleaved CASP3 and significant RGC loss, indicating disease imprints in the hiPSC-derived RGCs. This model offers a venue and high throughput platform for studying glaucomatous disease pathology and holds significant potential for drug discovery using RGCs derived from human donors. © 2025 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Culturing of keratocytes from human cadaveric donors Basic Protocol 2: Reprogramming donor keratocytes into iPSCs Basic Protocol 3: Evaluation of chromosomal loss during reprogramming in iPSCs by karyotyping Basic Protocol 4: Generation of 3D ROs Basic Protocol 5: Dissociation and culturing of RGCs from 3D ROs Support Protocol 1: Immunostaining for phenotypic characterization of cells Support Protocol 2: Sectioning of 3D ROs and immunostaining Support Protocol 3: Western blotting for cleaved CASP3 and THY1.
Collapse
Affiliation(s)
- Shahna S. Hameed
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndiana
| | - Tasneem P. Sharma
- Department of OphthalmologyIndiana University School of MedicineIndianapolisIndiana
| |
Collapse
|
2
|
García-López M, Jiménez-Vicente L, González-Jabardo R, Dorado H, Gómez-Manjón I, Martín MÁ, Ayuso C, Arenas J, Gallardo ME. Creation of an Isogenic Human iPSC-Based RGC Model of Dominant Optic Atrophy Harboring the Pathogenic Variant c.1861C>T (p.Gln621Ter) in the OPA1 Gene. Int J Mol Sci 2024; 25:7240. [PMID: 39000346 PMCID: PMC11242102 DOI: 10.3390/ijms25137240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Autosomal dominant optic atrophy (ADOA) is a rare progressive disease mainly caused by mutations in OPA1, a nuclear gene encoding for a mitochondrial protein that plays an essential role in mitochondrial dynamics, cell survival, oxidative phosphorylation, and mtDNA maintenance. ADOA is characterized by the degeneration of retinal ganglion cells (RGCs). This causes visual loss, which can lead to legal blindness in many cases. Nowadays, there is no effective treatment for ADOA. In this article, we have established an isogenic human RGC model for ADOA using iPSC technology and the genome editing tool CRISPR/Cas9 from a previously generated iPSC line of an ADOA plus patient harboring the pathogenic variant NM_015560.3: c.1861C>T (p.Gln621Ter) in heterozygosis in OPA1. To this end, a protocol based on supplementing the iPSC culture media with several small molecules and defined factors trying to mimic embryonic development has been employed. Subsequently, the created model was validated, confirming the presence of a defect of intergenomic communication, impaired mitochondrial respiration, and an increase in apoptosis and ROS generation. Finally, we propose the analysis of OPA1 expression by qPCR as an easy read-out method to carry out future drug screening studies using the created RGC model. In summary, this model provides a useful platform for further investigation of the underlying pathophysiological mechanisms of ADOA plus and for testing compounds with potential pharmacological action.
Collapse
Affiliation(s)
- Marta García-López
- Grupo de Investigación Traslacional con Células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Lydia Jiménez-Vicente
- Grupo de Investigación Traslacional con Células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Raquel González-Jabardo
- Grupo de Investigación Traslacional con Células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Helena Dorado
- Grupo de Investigación Traslacional con Células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Irene Gómez-Manjón
- Servicio de Genética, Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Miguel Ángel Martín
- Servicio de Genética, Hospital 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Laboratorio de Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - Carmen Ayuso
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
- Department of Genetics & Genomics, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital, Universidad Autónoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Joaquín Arenas
- Laboratorio de Enfermedades Mitocondriales y Neuromusculares, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28029 Madrid, Spain
| | - María Esther Gallardo
- Grupo de Investigación Traslacional con Células iPS, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| |
Collapse
|
3
|
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
|
4
|
Aweidah H, Matsevich C, Khaner H, Idelson M, Ejzenberg A, Reubinoff B, Banin E, Obolensky A. Survival of Neural Progenitors Derived from Human Embryonic Stem Cells Following Subretinal Transplantation in Rodents. J Ocul Pharmacol Ther 2023. [PMID: 37140896 DOI: 10.1089/jop.2022.0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Purpose: To examine the survival of neural progenitors (NPs) cells derived from human embryonic stem cells (hESCs) following subretinal (SR) transplantation in rodents. Methods: hESCs engineered to express enhanced green fluorescent protein (eGFP) were differentiated in vitro toward an NP fate using a 4-week protocol. State of differentiation was characterized by quantitative-PCR. NPs in suspension (75,000/μl) were transplanted to the SR-space of Royal College of Surgeons (RCS) rats (n = 66), nude-RCS rats (n = 18), and NOD scid gamma (NSG) mice (n = 53). Success of engraftment was determined at 4 weeks post-transplant by in vivo visualization of GFP-expression using a properly filtered rodent fundus camera. Transplanted eyes were examined in vivo at set time points using the fundus camera, and in select cases, by optical coherence tomography imaging, and after enucleation, by retinal histology and immunohistochemistry. Results: In RCS rats, cell rejection was observed in 29% of eyes at 6 weeks, rising to 92% at 8 weeks. In the more immunodeficient nude-RCS rats, the rejection rate was still high reaching 62% of eyes at 6 weeks post-transplant. Following transplantation in highly immunodeficient NSG mice, survival of the hESC-derived NPs was much improved, with 100% survival at 9 weeks and 72% at 20 weeks. A small number of eyes that were followed past 20 weeks showed survival also at 22 weeks. Conclusions: Immune status of recipient animals influences transplant survival. Highly immunodeficient NSG mice provide a better model for studying long-term survival, differentiation, and possible integration of hESC-derived NPs. Clinical Trial Registration numbers: NCT02286089, NCT05626114.
Collapse
Affiliation(s)
- Hamzah Aweidah
- Center for Retinal and Macular Degenerations, Department of Ophthalmology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Chen Matsevich
- Center for Retinal and Macular Degenerations, Department of Ophthalmology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Hanita Khaner
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Masha Idelson
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Ayala Ejzenberg
- Center for Retinal and Macular Degenerations, Department of Ophthalmology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Benjamin Reubinoff
- Hadassah Stem Cell Research Center, Goldyne Savad Institute of Gene Therapy, Department of Obstetrics and Gynecology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Eyal Banin
- Center for Retinal and Macular Degenerations, Department of Ophthalmology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Alexey Obolensky
- Center for Retinal and Macular Degenerations, Department of Ophthalmology, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
5
|
Esmaeili M, Mead B. Differentiation of Human Embryonic/Induced-Pluripotent Stem Cells to Retinal Ganglion Cells. Methods Mol Biol 2023; 2708:41-48. [PMID: 37558958 DOI: 10.1007/978-1-0716-3409-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/11/2023]
Abstract
The generation of retinal ganglion cells (RGCs) differentiated from human embryonic stem cell (hESC) or induced-pluripotent stem cells (iPSC) could aid with understanding of human RGC development, neuronal biology, drug discovery, potential cell-based therapies, and gene regulation. Here, we present a protocol for differentiation of hESC to RGCs using a 40-day protocol, significantly shorter than typical retinal organoids while still yielding cells with RGC-enriched markers and show physiological and morphological properties typical of RGCs.
Collapse
Affiliation(s)
- Maryam Esmaeili
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Ben Mead
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
6
|
Message in a Scaffold: Natural Biomaterials for Three-Dimensional (3D) Bioprinting of Human Brain Organoids. Biomolecules 2022; 13:biom13010025. [PMID: 36671410 PMCID: PMC9855696 DOI: 10.3390/biom13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/07/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Brain organoids are invaluable tools for pathophysiological studies or drug screening, but there are still challenges to overcome in making them more reproducible and relevant. Recent advances in three-dimensional (3D) bioprinting of human neural organoids is an emerging approach that may overcome the limitations of self-organized organoids. It requires the development of optimal hydrogels, and a wealth of research has improved our knowledge about biomaterials both in terms of their intrinsic properties and their relevance on 3D culture of brain cells and tissue. Although biomaterials are rarely biologically neutral, few articles have reviewed their roles on neural cells. We here review the current knowledge on unmodified biomaterials amenable to support 3D bioprinting of neural organoids with a particular interest in their impact on cell homeostasis. Alginate is a particularly suitable bioink base for cell encapsulation. Gelatine is a valuable helper agent for 3D bioprinting due to its viscosity. Collagen, fibrin, hyaluronic acid and laminin provide biological support to adhesion, motility, differentiation or synaptogenesis and optimize the 3D culture of neural cells. Optimization of specialized hydrogels to direct differentiation of stem cells together with an increased resolution in phenotype analysis will further extend the spectrum of possible bioprinted brain disease models.
Collapse
|
7
|
Daniszewski M, Senabouth A, Liang HH, Han X, Lidgerwood GE, Hernández D, Sivakumaran P, Clarke JE, Lim SY, Lees JG, Rooney L, Gulluyan L, Souzeau E, Graham SL, Chan CL, Nguyen U, Farbehi N, Gnanasambandapillai V, McCloy RA, Clarke L, Kearns LS, Mackey DA, Craig JE, MacGregor S, Powell JE, Pébay A, Hewitt AW. Retinal ganglion cell-specific genetic regulation in primary open-angle glaucoma. CELL GENOMICS 2022; 2:100142. [PMID: 36778138 PMCID: PMC9903700 DOI: 10.1016/j.xgen.2022.100142] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 03/08/2021] [Accepted: 05/11/2022] [Indexed: 10/18/2022]
Abstract
To assess the transcriptomic profile of disease-specific cell populations, fibroblasts from patients with primary open-angle glaucoma (POAG) were reprogrammed into induced pluripotent stem cells (iPSCs) before being differentiated into retinal organoids and compared with those from healthy individuals. We performed single-cell RNA sequencing of a total of 247,520 cells and identified cluster-specific molecular signatures. Comparing the gene expression profile between cases and controls, we identified novel genetic associations for this blinding disease. Expression quantitative trait mapping identified a total of 4,443 significant loci across all cell types, 312 of which are specific to the retinal ganglion cell subpopulations, which ultimately degenerate in POAG. Transcriptome-wide association analysis identified genes at loci previously associated with POAG, and analysis, conditional on disease status, implicated 97 statistically significant retinal ganglion cell-specific expression quantitative trait loci. This work highlights the power of large-scale iPSC studies to uncover context-specific profiles for a genetically complex disease.
Collapse
Affiliation(s)
- Maciej Daniszewski
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Anne Senabouth
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Helena H. Liang
- Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Xikun Han
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Grace E. Lidgerwood
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Damián Hernández
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Priyadharshini Sivakumaran
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Jordan E. Clarke
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Shiang Y. Lim
- Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,O’Brien Institute Department of St Vincent’s Institute of Medical Research, Melbourne, Fitzroy, VIC 3065, Australia
| | - Jarmon G. Lees
- O’Brien Institute Department of St Vincent’s Institute of Medical Research, Melbourne, Fitzroy, VIC 3065, Australia,Department of Medicine, St Vincent’s Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Louise Rooney
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Lerna Gulluyan
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Emmanuelle Souzeau
- Department of Ophthalmology, Flinders University, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Stuart L. Graham
- Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW 2109, Australia
| | - Chia-Ling Chan
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Uyen Nguyen
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Nona Farbehi
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Vikkitharan Gnanasambandapillai
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Rachael A. McCloy
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Linda Clarke
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - Lisa S. Kearns
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia
| | - David A. Mackey
- Lions Eye Institute, Centre for Vision Sciences, University of Western Australia, Crawley, WA 6009, Australia,School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7005, Australia
| | - Jamie E. Craig
- Department of Ophthalmology, Flinders University, Flinders Medical Centre, Bedford Park, SA 5042, Australia
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Joseph E. Powell
- Garvan Weizmann Centre for Cellular Genomics, Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia,UNSW Cellular Genomics Futures Institute, University of New South Wales, Sydney, NSW 2052, Australia,Corresponding author
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia,Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia,Corresponding author
| | - Alex W. Hewitt
- Department of Surgery, The University of Melbourne, Parkville, VIC 3010, Australia,Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia,School of Medicine, Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS 7005, Australia,Corresponding author
| |
Collapse
|
8
|
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
|
9
|
Advances in Regeneration of Retinal Ganglion Cells and Optic Nerves. Int J Mol Sci 2021; 22:ijms22094616. [PMID: 33924833 PMCID: PMC8125313 DOI: 10.3390/ijms22094616] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma, the second leading cause of blindness worldwide, is an incurable neurodegenerative disorder due to the dysfunction of retinal ganglion cells (RGCs). RGCs function as the only output neurons conveying the detected light information from the retina to the brain, which is a bottleneck of vision formation. RGCs in mammals cannot regenerate if injured, and RGC subtypes differ dramatically in their ability to survive and regenerate after injury. Recently, novel RGC subtypes and markers have been uncovered in succession. Meanwhile, apart from great advances in RGC axon regeneration, some degree of experimental RGC regeneration has been achieved by the in vitro differentiation of embryonic stem cells and induced pluripotent stem cells or in vivo somatic cell reprogramming, which provides insights into the future therapy of myriad neurodegenerative disorders. Further approaches to the combination of different factors will be necessary to develop efficacious future therapeutic strategies to promote ultimate axon and RGC regeneration and functional vision recovery following injury.
Collapse
|
10
|
Ge Y, Wu F, Cheng M, Bard J, Mu X. Two new genetically modified mouse alleles labeling distinct phases of retinal ganglion cell development by fluorescent proteins. Dev Dyn 2020; 249:1514-1528. [PMID: 32741043 PMCID: PMC7855626 DOI: 10.1002/dvdy.233] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND During development, all retinal cell types arise from retinal progenitor cells (RPCs) in a step-wise fashion. Atoh7 and Pou4f2 mark, and function in, two phases of retinal ganglion cell (RGC) genesis; Atoh7 functions in a subpopulation of RPCs to render them competent for the RGC fate, whereas Pou4f2 participates in RGC fate specification and RGC differentiation. Despite extensive research on their roles, the properties of the two phases represented by these two factors have not been well studied, likely due to the retinal cellular heterogeneity. RESULTS In this report, we describe two novel knock-in mouse alleles, Atoh7zsGreenCreERT2 and Pou4f2FlagtdTomato , which labeled retinal cells in the two phases of RGC development by fluorescent proteins. Also, the Atoh7zsGreenCreERT2 allele allowed for indirect labeling of RGCs and other cell types upon tamoxifen induction in a dose-dependent manner. Further, these alleles could be used to purify retinal cells in the different phases by fluorescence assisted cell sorting (FACS). Single cell RNA-seq analysis of purified cells from Atoh7zsGreenCreERT2 retinas further validated that this allele labeled both transitional/competent RPCs and their progenies including RGCs. CONCLUSIONS Thus, these two alleles are very useful tools for studying the molecular and genetic mechanisms underlying RGC formation.
Collapse
Affiliation(s)
- Yichen Ge
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Fuguo Wu
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Mobin Cheng
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
| | - Jonathan Bard
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY
| | - Xiuqian Mu
- Department of Ophthalmology/Ross Eye Institute, University at Buffalo, Buffalo, NY
- New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
11
|
Subramaniam MD, Iyer M, Nair AP, Venkatesan D, Mathavan S, Eruppakotte N, Kizhakkillach S, Chandran MK, Roy A, Gopalakrishnan AV, Vellingiri B. Oxidative stress and mitochondrial transfer: A new dimension towards ocular diseases. Genes Dis 2020; 9:610-637. [PMID: 35782976 PMCID: PMC9243399 DOI: 10.1016/j.gendis.2020.11.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/18/2020] [Accepted: 11/27/2020] [Indexed: 12/12/2022] Open
Abstract
Ocular cells like, retinal pigment epithelium (RPE) is a highly specialized pigmented monolayer of post-mitotic cells, which is located in the posterior segment of the eye between neuro sensory retina and vascular choroid. It functions as a selective barrier and nourishes retinal visual cells. As a result of high-level oxygen consumption of retinal cells, RPE cells are vulnerable to chronic oxidative stress and an increased level of reactive oxygen species (ROS) generated from mitochondria. These oxidative stress and ROS generation in retinal cells lead to RPE degeneration. Various sources including mtDNA damage could be an important factor of oxidative stress in RPE. Gene therapy and mitochondrial transfer studies are emerging fields in ocular disease research. For retinal degenerative diseases stem cell-based transplantation methods are developed from basic research to preclinical and clinical trials. Translational research contributions of gene and cell therapy would be a new strategy to prevent, treat and cure various ocular diseases. This review focuses on the effect of oxidative stress in ocular cell degeneration and recent translational researches on retinal degenerative diseases to cure blindness.
Collapse
Affiliation(s)
- Mohana Devi Subramaniam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
- Corresponding author.
| | - Mahalaxmi Iyer
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
- Department of Zoology, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India
| | - Aswathy P. Nair
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
| | - Dhivya Venkatesan
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Sinnakaruppan Mathavan
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai 600006, Tamil Nadu, India
| | - Nimmisha Eruppakotte
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Soumya Kizhakkillach
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Manoj kumar Chandran
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Ayan Roy
- Department of Biotechnology, Lovely Professional University, Punjab 144411, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 600127, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
- Corresponding author. Human Molecular Cytogenetics and Stem Cell, Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India.Fax: +91 422 2422387.
| |
Collapse
|
12
|
Freude KK, Saruhanian S, McCauley A, Paterson C, Odette M, Oostenink A, Hyttel P, Gillies M, Haukedal H, Kolko M. Enrichment of retinal ganglion and Müller glia progenitors from retinal organoids derived from human induced pluripotent stem cells - possibilities and current limitations. World J Stem Cells 2020; 12:1171-1183. [PMID: 33178399 PMCID: PMC7596448 DOI: 10.4252/wjsc.v12.i10.1171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 08/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Retinal organoids serve as excellent human-specific disease models for conditions affecting otherwise inaccessible retinal tissue from patients. They permit the isolation of key cell types affected in various eye diseases including retinal ganglion cells (RGCs) and Müller glia.
AIM To refine human-induced pluripotent stem cells (hiPSCs) differentiated into three-dimensional (3D) retinal organoids to generate sufficient numbers of RGCs and Müller glia progenitors for downstream analyses.
METHODS In this study we described, evaluated, and refined methods with which to generate Müller glia and RGC progenitors, isolated them via magnetic-activated cell sorting, and assessed their lineage stability after prolonged 2D culture. Putative progenitor populations were characterized via quantitative PCR and immunocytochemistry, and the ultrastructural composition of retinal organoid cells was investigated.
RESULTS Our study confirms the feasibility of generating marker-characterized Müller glia and RGC progenitors within retinal organoids. Such retinal organoids can be dissociated and the Müller glia and RGC progenitor-like cells isolated via magnetic-activated cell sorting and propagated as monolayers.
CONCLUSION Enrichment of Müller glia and RGC progenitors from retinal organoids is a feasible method with which to study cell type-specific disease phenotypes and to potentially generate specific retinal populations for cell replacement therapies.
Collapse
Affiliation(s)
- Kristine Karla Freude
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Sarkis Saruhanian
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
| | - Alanna McCauley
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Colton Paterson
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Madeleine Odette
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Annika Oostenink
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Poul Hyttel
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Mark Gillies
- Save Sight Institute, South Block, Sydney Eye Hospital, Sydney 2000, Australia
| | - Henriette Haukedal
- Department of Veterinary and Animal Sciences, Group of Stem Cell Models for Studies of Neurodegenerative Diseases, Section for Pathobiological Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen 2100, Denmark
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet-Glostrup, Glostrup 2600, Denmark
| |
Collapse
|
13
|
Shen Y. Stem cell therapies for retinal diseases: from bench to bedside. J Mol Med (Berl) 2020; 98:1347-1368. [PMID: 32794020 DOI: 10.1007/s00109-020-01960-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/02/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022]
Abstract
As the human retina has no regenerative ability, stem cell interventions represent potential therapies for various blinding retinal diseases. This type of therapy has been extensively studied in the human eyes through decades of preclinical studies. The safety profiles shown in clinical trials thus far have indicated that these strategies should be further explored. There are still challenges with regard to cell source, cell delivery, immuno-related adverse events and long-term maintenance of the therapeutic effects. Retinal stem cell therapy is likely to be most successful with a combination of multiple technologies, such as gene therapy. The purpose of this review is to present a synthetical and systematic coverage of stem cell therapies that target retinal diseases from bench to bedside, intending to appeal to both junior specialists and the broader community of clinical investigators alike. This review will only focus on therapies that have already been studied in clinical trials. This review summarizes key concepts, highlights the main studies in human patients and discusses the current challenges and potential methods to reduce safety concerns while enhancing the therapeutic effects.
Collapse
Affiliation(s)
- Yuening Shen
- Institute of Ophthalmology, University College London , 11-43 Bath St, London, EC1V 9EL, UK. .,Department of Medical Retina, Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London, EC1V 2PD, UK.
| |
Collapse
|
14
|
Dual SMAD inhibition and Wnt inhibition enable efficient and reproducible differentiations of induced pluripotent stem cells into retinal ganglion cells. Sci Rep 2020; 10:11828. [PMID: 32678240 PMCID: PMC7366935 DOI: 10.1038/s41598-020-68811-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is a group of progressive optic neuropathies that share common biological and clinical characteristics including irreversible changes to the optic nerve and visual field loss caused by the death of retinal ganglion cells (RGCs). The loss of RGCs manifests as characteristic cupping or optic nerve degeneration, resulting in visual field loss in patients with Glaucoma. Published studies on in vitro RGC differentiation from stem cells utilized classical RGC signaling pathways mimicking retinal development in vivo. Although many strategies allowed for the generation of RGCs, increased variability between experiments and lower yield hampered the cross comparison between individual lines and between experiments. To address this critical need, we developed a reproducible chemically defined in vitro methodology for generating retinal progenitor cell (RPC) populations from iPSCs, that are efficiently directed towards RGC lineage. Using this method, we reproducibly differentiated iPSCs into RGCs with greater than 80% purity, without any genetic modifications. We used small molecules and peptide modulators to inhibit BMP, TGF-β (SMAD), and canonical Wnt pathways that reduced variability between iPSC lines and yielded functional and mature iPSC-RGCs. Using CD90.2 antibody and Magnetic Activated Cell Sorter (MACS) technique, we successfully purified Thy-1 positive RGCs with nearly 95% purity.
Collapse
|
15
|
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
|
16
|
Behtaj S, Öchsner A, Anissimov YG, Rybachuk M. Retinal Tissue Bioengineering, Materials and Methods for the Treatment of Glaucoma. Tissue Eng Regen Med 2020; 17:253-269. [PMID: 32390117 PMCID: PMC7260329 DOI: 10.1007/s13770-020-00254-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 03/17/2020] [Accepted: 03/19/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Glaucoma, a characteristic type of optic nerve degeneration in the posterior pole of the eye, is a common cause of irreversible vision loss and the second leading cause of blindness worldwide. As an optic neuropathy, glaucoma is identified by increasing degeneration of retinal ganglion cells (RGCs), with consequential vision loss. Current treatments only postpone the development of retinal degeneration, and there are as yet no treatments available for this disability. Recent studies have shown that replacing lost or damaged RGCs with healthy RGCs or RGC precursors, supported by appropriately designed bio-material scaffolds, could facilitate the development and enhancement of connections to ganglion cells and optic nerve axons. The consequence may be an improved retinal regeneration. This technique could also offer the possibility for retinal regeneration in treating other forms of optic nerve ailments through RGC replacement. METHODS In this brief review, we describe the innovations and recent developments in retinal regenerative medicine such as retinal organoids and gene therapy which are specific to glaucoma treatment and focus on the selection of appropriate bio-engineering principles, biomaterials and cell therapies that are presently employed in this growing research area. RESULTS Identification of optimal sources of cells, improving cell survival, functional integration upon transplantation, and developing techniques to deliver cells into the retinal space without provoking immune responses are the main challenges in retinal cell replacement therapies. CONCLUSION The restoration of visual function in glaucoma patients by the RGC replacement therapies requires appropriate protocols and biotechnology methods. Tissue-engineered scaffolds, the generation of retinal organoids, and gene therapy may help to overcome some of the challenges in the generation of clinically safe RGCs.
Collapse
Affiliation(s)
- Sanaz Behtaj
- School of Engineering and Built Environment, Griffith University, Engineering Drive, Southport, QLD, 4222, Australia
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- Department of Cell and Molecular Biology, Cell Science Research Centre, Royan Institute for Biotechnology, Isfahan, Iran
| | - Andreas Öchsner
- Faculty of Mechanical Engineering, Esslingen University of Applied Sciences, Kanalstrasse 33, 73728, Esslingen, Germany
| | - Yuri G Anissimov
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia
- School of Environment and Science, Griffith University, Parklands Drive, Southport, QLD, 4222, Australia
- Institute of Molecular Medicine, I.M. Sechenov First Moscow State Medical University, Moscow, 119146, Russia
| | - Maksym Rybachuk
- Queensland Micro- and Nanotechnology Centre, Griffith University, West Creek Road, Nathan, QLD, 4111, Australia.
- School of Engineering and Built Environment, Griffith University, 170 Kessels Road, Nathan, QLD, 4111, Australia.
| |
Collapse
|
17
|
Zhang X, Tenerelli K, Wu S, Xia X, Yokota S, Sun C, Galvao J, Venugopalan P, Li C, Madaan A, Goldberg JL, Chang KC. Cell transplantation of retinal ganglion cells derived from hESCs. Restor Neurol Neurosci 2020; 38:131-140. [PMID: 31815704 DOI: 10.3233/rnn-190941] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Glaucoma, the number one cause of irreversible blindness, is characterized by the loss of retinal ganglion cells (RGCs), which do not regenerate in humans or mammals after cell death. Cell transplantation provides an opportunity to restore vision in glaucoma, or other optic neuropathies. Since transplanting primary RGCs from deceased donor tissues may not be feasible, stem cell-derived RGCs could provide a plausible alternative source of donor cells for transplant. OBJECTIVE We define a robust chemically defined protocol to differentiate human embryonic stem cells (hESCs) into RGC-like neurons. METHODS Human embryonic stem cell lines (H7-A81 and H9) and induced pluripotent stem cell (iPSC) were used for RGC differentiation. RGC immaturity was measured by calcium imaging against muscimol. Cell markers were detected by immunofluorescence staining and qRT-PCR. RGC-like cells were intravitreally injected to rat eye, and co-stained with RBPMS and human nuclei markers. All experiments were conducted at least three times independently. Data were analyzed by ANOVA with Tukey's test with P value of <0.05 considered statistically significant. RESULTS We detected retinal progenitor markers Rx and Pax6 after 15 days of differentiation, and the expression of markers for RGC-specific differentiation (Brn3a and Brn3b), maturation (synaptophysin) and neurite growth (β-III-Tubulin) after an additional 15 days. We further examined the physiologic differentiation of these hESC-derived RGC-like progeny to those differentiated in vitro from primary rodent retinal progenitor cells (RPCs) with calcium imaging, and found that both populations demonstrate the immature RGC-like response to muscimol, a GABAA receptor agonist. By one week after transplant to the adult rat eye by intravitreal injection, the human RGC-like cells successfully migrated into the ganglion cell layer. CONCLUSIONS Our protocol provides a novel, short, and cost-effective approach for RGC differentiation from hESCs, and may broaden the scope for cell replacement therapy in RGC-related optic neuropathies such as glaucoma.
Collapse
Affiliation(s)
- Xiong Zhang
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Kevin Tenerelli
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Suqian Wu
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
- Department of Ophthalmology & Visual Science, Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, P.R. China
| | - Xin Xia
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Satoshi Yokota
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Catalina Sun
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Joana Galvao
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | | | - Chenyi Li
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Ankush Madaan
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
- Shiley Eye Institute, University of California San Diego, La Jolla, CA, USA
| | - Kun-Che Chang
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, USA
| |
Collapse
|
18
|
Chung SH, Shen W, Davidson KC, Pébay A, Wong RCB, Yau B, Gillies M. Differentiation of Retinal Glial Cells From Human Embryonic Stem Cells by Promoting the Notch Signaling Pathway. Front Cell Neurosci 2019; 13:527. [PMID: 31849614 PMCID: PMC6901827 DOI: 10.3389/fncel.2019.00527] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/12/2019] [Indexed: 01/21/2023] Open
Abstract
Dysfunction of retinal glial cells, particularly Müller cells, has been implicated in several retinal diseases. Despite their important contribution to retinal homeostasis, a specific way to differentiate retinal glial cells from human pluripotent stem cells has not yet been described. Here, we report a method to differentiate retinal glial cells from human embryonic stem cells (hESCs) through promoting the Notch signaling pathway. We first generated retinal progenitor cells (RPCs) from hESCs then promoted the Notch signaling pathway using Notch ligands, including Delta-like ligand 4 and Jagged-1. We validated glial cell differentiation with qRT-PCR, immunocytochemistry, western blots and fluorescence-activated cell sorting as we promoted Notch signaling in RPCs. We found that promoting Notch signaling in RPCs for 2 weeks led to upregulation of glial cell markers, including glial fibrillary acidic protein (GFAP), glutamine synthetase, vimentin and cellular retinaldehyde-binding protein (CRALBP). Of these markers, we found the greatest increase in expression of the pan glial cell marker, GFAP. Conversely, we also found that inhibition of Notch signaling in RPCs led to upregulation of retinal neuronal markers including cone-rod homeobox (CRX) and orthodenticle homeobox 2 (OTX2) but with little expression of GFAP. This retinal glial differentiation method will help advance the generation of stem cell disease models to study the pathogenesis of retinal diseases associated with glial dysfunction such as macular telangiectasia type 2. This method may also be useful for the development of future therapeutics such as drug screening and gene editing using patient-derived retinal glial cells.
Collapse
Affiliation(s)
- Sook Hyun Chung
- Save Sight Institute, Department of Clinical Ophthalmology and Eye Health, The University of Sydney, Sydney, NSW, Australia
| | - Weiyong Shen
- Save Sight Institute, Department of Clinical Ophthalmology and Eye Health, The University of Sydney, Sydney, NSW, Australia
| | - Kathryn C Davidson
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
| | - Alice Pébay
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia.,Department of Surgery, The University of Melbourne, Parkville, VIC, Australia
| | - Raymond C B Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia.,Department of Surgery, The University of Melbourne, Parkville, VIC, Australia.,Shenzhen Eye Hospital, Shenzhen, China
| | - Belinda Yau
- Save Sight Institute, Department of Clinical Ophthalmology and Eye Health, The University of Sydney, Sydney, NSW, Australia
| | - Mark Gillies
- Save Sight Institute, Department of Clinical Ophthalmology and Eye Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
19
|
Murali A, Ramlogan-Steel CA, Steel JC, Layton CJ. Characterisation and validation of the 8-fold quadrant dissected human retinal explant culture model for pre-clinical toxicology investigation. Toxicol In Vitro 2019; 63:104716. [PMID: 31706033 DOI: 10.1016/j.tiv.2019.104716] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/13/2019] [Accepted: 11/04/2019] [Indexed: 11/30/2022]
Abstract
One of the major challenges in studying ocular toxicology is a lack of clinically-relevant retinal experimental models. In this study we describe the use of an in vitro human retinal explant strategy to generate a reproducible experimental model with utility in neuro-toxicity retinal studies. A retinal dissection strategy, referred to as the 8 fold quadrant dissection, was developed by dissecting human donor retinas into 4 fragments through the fovea in order to obtain 8 experimentally reproducible retinal explants from a single donor. This quadrant dissection gave rise to equivalent proportions of CD73+ photoreceptors and CD90+ ganglion cells in 8 fragments from a single donor and this remained stable for up to 3 days in culture. Major retinal cell types continued to be observed after 8 weeks in culture, despite breakdown of the retinal layers, suggesting the potential to use this model in long-term studies where observation of individual cell types is possible. The utility of this system was examined in a proof of principle neuro-toxicology study. We showed reproducible induction of toxicity in photoreceptors and retinal ganglion cells by glutamate, cobalt chloride and hydrogen peroxide insults, and observed the therapeutic positive effects of the administration of memantine, formononetin and trolox. The quadrant dissected human retinal explants have the potential to be used in toxicology studies in human ocular diseases.
Collapse
Affiliation(s)
- Aparna Murali
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Australia
| | - Charmaine A Ramlogan-Steel
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Central Queensland University, School of Health, Medical and Applied Science, Rockhampton, QLD, Australia
| | - Jason C Steel
- Central Queensland University, School of Health, Medical and Applied Science, Rockhampton, QLD, Australia.
| | - Christopher J Layton
- LVF Ophthalmology Research Centre, Translational Research Institute, Woolloongabba, QLD, Australia; Greenslopes Clinical School, Faculty of Medicine, University of Queensland, Greenslopes Hospital, Australia.
| |
Collapse
|
20
|
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
|
21
|
Hsu CC, Chien KH, Yarmishyn AA, Buddhakosai W, Wu WJ, Lin TC, Chiou SH, Chen JT, Peng CH, Hwang DK, Chen SJ, Chang YL. Modulation of osmotic stress-induced TRPV1 expression rescues human iPSC-derived retinal ganglion cells through PKA. Stem Cell Res Ther 2019; 10:284. [PMID: 31547874 PMCID: PMC6755708 DOI: 10.1186/s13287-019-1363-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/25/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Background Transient receptor potential vanilloid 1 (TRPV1), recognized as a hyperosmolarity sensor, is a crucial ion channel involved in the pathogenesis of neural and glial signaling. Recently, TRPV1 was determined to play a role in retinal physiology and visual transmission. In this study, we sought to clarify the role of TRPV1 and the downstream pathway in the osmotic stress-related retina ganglion cell (RGC) damage. Methods First, we modified the RGC differentiation protocol to obtain a homogeneous RGC population from human induced pluripotent stem cells (hiPSCs). Subsequently, we induced high osmotic pressure in the hiPSC-derived RGCs by administering NaCl solution and observed the behavior of the TRPV1 channel and its downstream cascade. Results We obtained a purified RGC population from the heterogeneous retina cell population using our modified method. Our findings revealed that TRPV1 was activated after 24 h of NaCl treatment. Upregulation of TRPV1 was noted with autophagy and apoptosis induction. Downstream protein expression analysis indicated increased phosphorylation of CREB and downregulated brain-derived neurotrophic factor (BDNF). However, hyperosmolarity-mediated defective morphological change and apoptosis of RGCs, CREB phosphorylation, and BDNF downregulation were abrogated after concomitant treatment with the PKA inhibitor H89. Conclusion Collectively, our study results indicated that the TRPV1–PKA pathway contributed to cellular response under high levels of osmolarity stress; furthermore, the PKA inhibitor had a protective effect on RGCs exposed to this stress. Therefore, our findings may assist in the treatment of eye diseases involving RGC damage.
Collapse
Affiliation(s)
- Chih-Chien Hsu
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Ke-Hung Chien
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Aliaksandr A Yarmishyn
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Waradee Buddhakosai
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Wen-Ju Wu
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan
| | - Tai-Chi Lin
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Hwa Chiou
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,Department of Medical Research, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Jiann-Torng Chen
- Department of Ophthalmology, Tri-Service General Hospital and National Defense Medical Center, Taipei, 114, Taiwan
| | - Chi-Hsien Peng
- Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital and Fu-Jen Catholic University, Taipei, Taiwan
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| | - Yuh-Lih Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan. .,Department of Pharmacy, Taipei Veterans General Hospital; Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.
| |
Collapse
|
22
|
Polybenzyl Glutamate Biocompatible Scaffold Promotes the Efficiency of Retinal Differentiation toward Retinal Ganglion Cell Lineage from Human-Induced Pluripotent Stem Cells. Int J Mol Sci 2019; 20:ijms20010178. [PMID: 30621308 PMCID: PMC6337229 DOI: 10.3390/ijms20010178] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/27/2018] [Accepted: 12/29/2018] [Indexed: 02/06/2023] Open
Abstract
Optic neuropathy is one of the leading causes of irreversible blindness caused by retinal ganglion cell (RGC) degeneration. The development of induced pluripotent stem cell (iPSC)-based therapy opens a therapeutic window for RGC degeneration, and tissue engineering may further promote the efficiency of differentiation process of iPSCs. The present study was designed to evaluate the effects of a novel biomimetic polybenzyl glutamate (PBG) scaffold on culturing iPSC-derived RGC progenitors. The iPSC-derived neural spheres cultured on PBG scaffold increased the differentiated retinal neurons and promoted the neurite outgrowth in the RGC progenitor layer. Additionally, iPSCs cultured on PBG scaffold formed the organoid-like structures compared to that of iPSCs cultured on cover glass within the same culture period. With RNA-seq, we found that cells of the PBG group were differentiated toward retinal lineage and may be related to the glutamate signaling pathway. Further ontological analysis and the gene network analysis showed that the differentially expressed genes between cells of the PBG group and the control group were mainly associated with neuronal differentiation, neuronal maturation, and more specifically, retinal differentiation and maturation. The novel electrospinning PBG scaffold is beneficial for culturing iPSC-derived RGC progenitors as well as retinal organoids. Cells cultured on PBG scaffold differentiate effectively and shorten the process of RGC differentiation compared to that of cells cultured on coverslip. The new culture system may be helpful in future disease modeling, pharmacological screening, autologous transplantation, as well as narrowing the gap to clinical application.
Collapse
|
23
|
Bioactivity and gene expression profiles of hiPSC-generated retinal ganglion cells in MT-ND4 mutated Leber's hereditary optic neuropathy. Exp Cell Res 2018; 363:299-309. [PMID: 29366807 DOI: 10.1016/j.yexcr.2018.01.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 01/09/2018] [Accepted: 01/15/2018] [Indexed: 01/01/2023]
Abstract
Leber's hereditary optic neuropathy (LHON) is the maternally inherited mitochondrial disease caused by homoplasmic mutations in mitochondrial electron transport chain Complex I subunit genes. The mechanism of its incomplete penetrance is still largely unclear. In this study, we created the patient-specific human induced pluripotent stem cells (hiPSCs) from MT-ND4 mutated LHON-affected patient, asymptomatic mutation carrier and healthy control, and differentiated them into retinal ganglion cells (RGCs). We found the defective neurite outgrowth in affected RGCs, but not in the carrier RGCs which had significant expression of SNCG gene. We observed enhanced mitochondrial biogenesis in affected and carrier derived RGCs. Surprisingly, we observed increased NADH dehydrogenase enzymatic activity of Complex I in hiPSC-derived RGCs of asymptomatic carrier, but not of the affected patient. LHON mutation substantially decreased basal respiration in both affected and unaffected carrier hiPSCs, and had the same effect on spare respiratory capacity, which ensures normal function of mitochondria in conditions of increased energy demand or environmental stress. The expression of antioxidant enzyme catalase was decreased in affected and carrier patient hiPSC-derived RGCs as compared to the healthy control, which might indicate to higher oxidative stress-enriched environment in the LHON-specific RGCs. Microarray profiling demonstrated enhanced expression of cell cycle machinery and downregulation of neuronal specific genes.
Collapse
|
24
|
Jin ZB, Gao ML, Deng WL, Wu KC, Sugita S, Mandai M, Takahashi M. Stemming retinal regeneration with pluripotent stem cells. Prog Retin Eye Res 2018; 69:38-56. [PMID: 30419340 DOI: 10.1016/j.preteyeres.2018.11.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 08/09/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022]
Abstract
Cell replacement therapy is a promising treatment for irreversible retinal cell death in diverse diseases, such as age-related macular degeneration (AMD), Stargardt's disease, retinitis pigmentosa (RP) and glaucoma. These diseases are all characterized by the degeneration of one or two retinal cell types that cannot regenerate spontaneously in humans. Aberrant retinal pigment epithelial (RPE) cells can be observed through optical coherence tomography (OCT) in AMD patients. In RP patients, the morphological and functional abnormalities of RPE and photoreceptor layers are caused by a genetic abnormality. Stargardt's disease or juvenile macular degeneration, which is characterized by the loss of the RPE and photoreceptors in the macular area, causes central vision loss at an early age. Loss of retinal ganglion cells (RGCs) can be observed in patients with glaucoma. Once the retinal cell degeneration is triggered, no treatments can reverse it. Transplantation-based approaches have been proposed as a universal therapy to target patients with various concomitant diseases. Both the replacement of dead cells and neuroprotection are strategies used to rescue visual function in animal models of retinal degeneration. Diverse retinal cell types derived from pluripotent stem cells, including RPE cells, photoreceptors, RGCs and even retinal organoids with a layered structure, provide unlimited cell sources for transplantation. In addition, mesenchymal stem cells (MSCs) are multifunctional and protect degenerating retinal cells. The aim of this review is to summarize current findings from preclinical and clinical studies. We begin with a brief introduction to retinal degenerative diseases and cell death in diverse diseases, followed by methods for retinal cell generation. Preclinical and clinical studies are discussed, and future concerns about efficacy, safety and immunorejection are also addressed.
Collapse
Affiliation(s)
- Zi-Bing Jin
- Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory for Ophthalmology, Optometry & Visual Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, 325027, China.
| | - Mei-Ling Gao
- Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory for Ophthalmology, Optometry & Visual Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, 325027, China
| | - Wen-Li Deng
- Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory for Ophthalmology, Optometry & Visual Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, 325027, China
| | - Kun-Chao Wu
- Laboratory for Stem Cell & Retinal Regeneration, Institute of Stem Cell Research, Division of Ophthalmic Genetics, The Eye Hospital, Wenzhou Medical University, State Key Laboratory for Ophthalmology, Optometry & Visual Science, National Center for International Research in Regenerative Medicine and Neurogenetics, Wenzhou, 325027, China
| | - Sunao Sugita
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo, 650-0047, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo, 650-0047, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Developmental Biology, Kobe, Hyogo, 650-0047, Japan
| |
Collapse
|
25
|
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
|
26
|
Slembrouck-Brec A, Nanteau C, Sahel JA, Goureau O, Reichman S. Defined Xeno-free and Feeder-free Culture Conditions for the Generation of Human iPSC-derived Retinal Cell Models. J Vis Exp 2018. [PMID: 30247478 DOI: 10.3791/57795] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The production of specialized cells from pluripotent stem cells provides a powerful tool to develop new approaches for regenerative medicine. The use of human-induced pluripotent stem cells (iPSCs) is particularly attractive for neurodegenerative disease studies, including retinal dystrophies, where iPSC-derived retinal cell models mark a major step forward to understand and fight blindness. In this paper, we describe a simple and scalable protocol to generate, mature, and cryopreserve retinal organoids. Based on medium changing, the main advantage of this method is to avoid multiple and time-consuming steps commonly required in a guided differentiation of iPSCs. Mimicking the early phases of retinal development by successive changes of defined media on adherent human iPSC cultures, this protocol allows the simultaneous generation of self-forming neuroretinal structures and retinal pigmented epithelial (RPE) cells in a reproducible and efficient manner in 4 weeks. These structures containing retinal progenitor cells (RPCs) can be easily isolated for further maturation in a floating culture condition enabling the differentiation of RPCs into the seven retinal cell types present in the adult human retina. Additionally, we describe quick methods for the cryopreservation of retinal organoids and RPE cells for long-term storage. Combined together, the methods described here will be useful to produce and bank human iPSC-derived retinal cells or tissues for both basic and clinical research.
Collapse
Affiliation(s)
| | - Céline Nanteau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France
| | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012 Paris, France;
| |
Collapse
|
27
|
Chuang JH, Yarmishyn AA, Hwang DK, Hsu CC, Wang ML, Yang YP, Chien KH, Chiou SH, Peng CH, Chen SJ. Expression profiling of cell-intrinsic regulators in the process of differentiation of human iPSCs into retinal lineages. Stem Cell Res Ther 2018; 9:140. [PMID: 29751772 PMCID: PMC5948821 DOI: 10.1186/s13287-018-0848-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Differentiation of human induced pluripotent stem cells (hiPSCs) into retinal lineages offers great potential for medical application. Therefore, it is of crucial importance to know the key intrinsic regulators of differentiation and the specific biomarker signatures of cell lineages. METHODS In this study, we used microarrays to analyze transcriptomes of terminally differentiated retinal ganglion cell (RGC) and retinal pigment epithelium (RPE) lineages, as well as intermediate retinal progenitor cells of optic vesicles (OVs) derived from hiPSCs. In our analysis, we specifically focused on the classes of transcripts that encode intrinsic regulators of gene expression: the transcription factors (TFs) and epigenetic chromatin state regulators. We applied two criteria for the selection of potentially important regulators and markers: firstly, the magnitude of fold-change of upregulation; secondly, the contrasted pattern of differential expression between OV, RGC and RPE lineages. RESULTS We found that among the most highly overexpressed TF-encoding genes in the OV/RGC lineage were three members of the Collier/Olfactory-1/Early B-cell family: EBF1, EBF2 and EBF3. Knockdown of EBF1 led to significant impairment of differentiation of hiPSCs into RGCs. EBF1 was shown to act upstream of ISL1 and BRN3A, the well-characterized regulators of RGC lineage specification. TF-encoding genes DLX1, DLX2 and INSM1 were the most highly overexpressed genes in the OVs, indicating their important role in the early stages of retinal differentiation. Along with MITF, the two paralogs, BHLHE41 and BHLHE40, were the most robust TF markers of RPE cells. The markedly contrasted expression of ACTL6B, encoding the component of chromatin remodeling complex SWI/SNF, discriminated hiPSC-derived OV/RGC and RPE lineages. CONCLUSIONS We identified novel, potentially important intrinsic regulators of RGC and RPE cell lineage specification in the process of differentiation from hiPSCs. We demonstrated the crucial role played by EBF1 in differentiation of RGCs. We identified intrinsic regulator biomarker signatures of these two retinal cell types that can be applied with high confidence to confirm the cell lineage identities.
Collapse
Affiliation(s)
- Jen-Hua Chuang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Aliaksandr A Yarmishyn
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - De-Kuang Hwang
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Chien Hsu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Mong-Lien Wang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | - Ke-Hung Chien
- Department of Ophthalmology, Tri-Service General Hospital & National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Hsien Peng
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Ophthalmology, Shin Kong Wu Ho-Su Memorial Hospital and Fu Jen Catholic University, Taipei, Taiwan.
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei, Taiwan. .,Department of Ophthalmology, Tri-Service General Hospital & National Defense Medical Center, Taipei, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
28
|
Single cell RNA sequencing of stem cell-derived retinal ganglion cells. Sci Data 2018; 5:180013. [PMID: 29437159 PMCID: PMC5810423 DOI: 10.1038/sdata.2018.13] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 12/22/2017] [Indexed: 11/09/2022] Open
Abstract
We used single cell sequencing technology to characterize the transcriptomes of 1,174 human embryonic stem cell-derived retinal ganglion cells (RGCs) at the single cell level. The human embryonic stem cell line BRN3B-mCherry (A81-H7), was differentiated to RGCs using a guided differentiation approach. Cells were harvested at day 36 and prepared for single cell RNA sequencing. Our data indicates the presence of three distinct subpopulations of cells, with various degrees of maturity. One cluster of 288 cells showed increased expression of genes involved in axon guidance together with semaphorin interactions, cell-extracellular matrix interactions and ECM proteoglycans, suggestive of a more mature RGC phenotype.
Collapse
|
29
|
Apatoff MBL, Sengillo JD, White EC, Bakhoum MF, Bassuk AG, Mahajan VB, Tsang SH. Autologous stem cell therapy for inherited and acquired retinal disease. Regen Med 2018; 13:89-96. [PMID: 29360008 DOI: 10.2217/rme-2017-0089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The mammalian retina, derived from neural ectoderm, has little regenerative potential. For conditions where irreversible retinal pigment epithelium or photoreceptor cell loss occurs, advanced techniques are required to restore vision. Inherited retinal dystrophies and some acquired conditions, such as age-related macular degeneration, have a similar end result of photoreceptor cell death leading to debilitating vision loss. These diseases stand to benefit from future regenerative medicine as dietary recommendations and current pharmacologic therapy only seek to prevent further disease progression. Cell-based strategies, such as autologously derived induced pluripotent stem cells, have come a long way in overcoming previous technical and ethical concerns. Clinical trials for such techniques are already underway. These trials and the preceding preclinical studies will be discussed in the context of retinal disease.
Collapse
Affiliation(s)
- Mary Ben L Apatoff
- Jonas Children's Vision Care & Bernard & Shirlee Brown Glaucoma Laboratory, Columbia University, New York, NY 10032, USA.,Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Jesse D Sengillo
- Jonas Children's Vision Care & Bernard & Shirlee Brown Glaucoma Laboratory, Columbia University, New York, NY 10032, USA.,Department of Ophthalmology, Columbia University, New York, NY 10032, USA.,College of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Eugenia C White
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | - Mathieu F Bakhoum
- Department of Ophthalmology, Columbia University, New York, NY 10032, USA
| | | | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94304, USA.,Department of Ophthalmology, Palo Alto Veterans Administration, Palo Alto, CA 94304, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care & Bernard & Shirlee Brown Glaucoma Laboratory, Columbia University, New York, NY 10032, USA.,Department of Ophthalmology, Columbia University, New York, NY 10032, USA.,Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA.,Institute of Human Nutrition, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| |
Collapse
|
30
|
Effects of neuroactive agents on axonal growth and pathfinding of retinal ganglion cells generated from human stem cells. Sci Rep 2017; 7:16757. [PMID: 29196712 PMCID: PMC5711798 DOI: 10.1038/s41598-017-16727-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 11/16/2017] [Indexed: 12/22/2022] Open
Abstract
We recently established a novel method for generating functional human retinal ganglion cells (RGCs) from human induced pluripotent cells (hiPSCs). Here, we confirmed that RGCs can also be generated from human embryonic stem cells (hESCs). We investigated the usefulness of human RGCs with long axons for assessing the effects of chemical agents, such as the neurotrophic factor, nerve growth factor (NGF), and the chemorepellent factors, semaphorin 3 A (SEMA3A) and SLIT1. The effects of direct and local administration of each agent on axonal projection were evaluated by immunohistochemistry, real-time polymerase chain reaction (PCR), and real-time imaging, in which the filopodia of the growth cone served as an excellent marker. A locally sustained agent system showed that the axons elongate towards NGF, but were repelled by SEMA3A and SLIT1. Focally transplanted beads that released SLIT1 bent the pathfinding of axons, imitating normal retinal development. Our innovative system for assessing the effects of chemical compounds using human RGCs may facilitate development of novel drugs for the examination, prophylaxis, and treatment of diseases. It may also be useful for observing the physiology of the optic nerve in vitro, which might lead to significant progress in the science of human RGCs.
Collapse
|
31
|
The use of induced pluripotent stem cells for studying and treating optic neuropathies. Curr Opin Organ Transplant 2017; 21:484-9. [PMID: 27517502 DOI: 10.1097/mot.0000000000000348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The present review aims to provide an update of applications of induced pluripotent stem cells (iPSCs) for disease modeling, cell/gene therapy, and drug screening for optic neuropathies. RECENT FINDINGS Degeneration of retinal ganglion cells (RGCs) is a characteristic of optic neuropathies. Human iPSCs can serve as a model to investigate disease pathology and potential repair mechanisms. In recent years, significant progress has been made in generating RGCs from iPSCs. Various groups have reported the potential of iPSCs for modeling optic neuropathies, such as glaucoma. The literature also highlights the potential to use iPSC-derived cells for high-throughput drug and toxicity screening. SUMMARY The present review summarizes current work in the field of iPSCs in optic neuropathies. Future studies to characterize iPSC-derived RGCs in a more in-depth manner will help expand the use of iPSCs to model and treat optic neuropathic diseases. Furthermore, iPSC modeling can be used in drug development by offering a new avenue to test novel therapeutic drugs for optic neuropathies.
Collapse
|
32
|
Vetter ML, Hitchcock PF. Report on the National Eye Institute Audacious Goals Initiative: Replacement of Retinal Ganglion Cells from Endogenous Cell Sources. Transl Vis Sci Technol 2017; 6:5. [PMID: 28316878 PMCID: PMC5354473 DOI: 10.1167/tvst.6.2.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 12/22/2022] Open
Abstract
This report emerges from a workshop convened by the National Eye Institute (NEI) as part of the "Audacious Goals Initiative" (AGI). The workshop addressed the replacement of retinal ganglion cells (RGCs) from exogenous and endogenous sources, and sought to identify the gaps in our knowledge and barriers to progress in devising cellular replacement therapies for diseases where RGCs die. Here, we briefly review relevant literature regarding common diseases associated with RGC death, the genesis of RGCs in vivo, strategies for generating transplantable RGCs in vitro, and potential endogenous cellular sources to regenerate these cells. These topics provided the clinical and scientific context for the discussion among the workshop participants and are relevant to efforts that may lead to therapeutic approaches for replacing RGCs. This report also summarizes the content of the workshop discussion, which focused on: (1) cell sources for RGC replacement and regeneration, (2) optimizing integration, survival, and synaptogenesis of new RGCs, and (3) approaches for assessing the outcomes of RGC replacement therapies. We conclude this report with a summary of recommendations, based on the workshop discussions, which may guide vision scientists seeking to develop therapies for replacing RGCs in humans.
Collapse
Affiliation(s)
- Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA
| | - Peter F Hitchcock
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT, USA ; Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | | |
Collapse
|
33
|
Lopez Sanchez M, Crowston J, Mackey D, Trounce I. Emerging Mitochondrial Therapeutic Targets in Optic Neuropathies. Pharmacol Ther 2016; 165:132-52. [DOI: 10.1016/j.pharmthera.2016.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 12/14/2022]
|
34
|
Enriched retinal ganglion cells derived from human embryonic stem cells. Sci Rep 2016; 6:30552. [PMID: 27506453 PMCID: PMC4978994 DOI: 10.1038/srep30552] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 07/04/2016] [Indexed: 12/21/2022] Open
Abstract
Optic neuropathies are characterised by a loss of retinal ganglion cells (RGCs) that lead to vision impairment. Development of cell therapy requires a better understanding of the signals that direct stem cells into RGCs. Human embryonic stem cells (hESCs) represent an unlimited cellular source for generation of human RGCs in vitro. In this study, we present a 45-day protocol that utilises magnetic activated cell sorting to generate enriched population of RGCs via stepwise retinal differentiation using hESCs. We performed an extensive characterization of these stem cell-derived RGCs by examining the gene and protein expressions of a panel of neural/RGC markers. Furthermore, whole transcriptome analysis demonstrated similarity of the hESC-derived RGCs to human adult RGCs. The enriched hESC-RGCs possess long axons, functional electrophysiological profiles and axonal transport of mitochondria, suggestive of maturity. In summary, this RGC differentiation protocol can generate an enriched population of functional RGCs from hESCs, allowing future studies on disease modeling of optic neuropathies and development of cell therapies.
Collapse
|
35
|
Transitional Progenitors during Vertebrate Retinogenesis. Mol Neurobiol 2016; 54:3565-3576. [PMID: 27194297 DOI: 10.1007/s12035-016-9899-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 05/03/2016] [Indexed: 12/13/2022]
Abstract
The retina is a delicate neural tissue responsible for light signal capturing, modulating, and passing to mid-brain. The brain then translated the signals into three-dimensional vision. The mature retina is composed of more than 50 subtypes of cells, all of which are developed from a pool of early multipotent retinal progenitors, which pass through sequential statuses of oligopotent, bipotent, and unipotent progenitors, and finally become terminally differentiated retinal cells. A transitional progenitor model is proposed here to describe how intrinsic developmental programs, along with environmental cues, control the step-by-step differentiation during retinogenesis. The model could elegantly explain many current findings as well as predict roles of intrinsic factors during retinal development.
Collapse
|
36
|
Chamling X, Sluch VM, Zack DJ. The Potential of Human Stem Cells for the Study and Treatment of Glaucoma. Invest Ophthalmol Vis Sci 2016; 57:ORSFi1-6. [PMID: 27116666 PMCID: PMC5110236 DOI: 10.1167/iovs.15-18590] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/05/2016] [Indexed: 01/01/2023] Open
Abstract
PURPOSE Currently, the only available and approved treatments for glaucoma are various pharmacologic, laser-based, and surgical procedures that lower IOP. Although these treatments can be effective, they are not always sufficient, and they cannot restore vision that has already been lost. The goal of this review is to briefly assess current developments in the application of stem cell biology to the study and treatment of glaucoma and other forms of optic neuropathy. METHODS A combined literature review and summary of the glaucoma-related discussion at the 2015 "Sight Restoration Through Stem Cell Therapy" meeting that was sponsored by the Ocular Research Symposia Foundation (ORSF). RESULTS Ongoing advancements in basic and eye-related developmental biology have enabled researchers to direct murine and human stem cells along specific developmental paths and to differentiate them into a variety of ocular cell types of interest. The most advanced of these efforts involve the differentiation of stem cells into retinal pigment epithelial cells, work that has led to the initiation of several human trials. More related to the glaucoma field, there have been recent advances in developing protocols for differentiation of stem cells into trabecular meshwork and retinal ganglion cells. Additionally, efforts are being made to generate stem cell-derived cells that can be used to secrete neuroprotective factors. CONCLUSIONS Advancing stem cell technology provides opportunities to improve our understanding of glaucoma-related biology and develop models for drug development, and offers the possibility of cell-based therapies to restore sight to patients who have already lost vision.
Collapse
Affiliation(s)
- Xitiz Chamling
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Valentin M. Sluch
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Donald J. Zack
- Department of Ophthalmology Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
37
|
Sluch VM, Davis CHO, Ranganathan V, Kerr JM, Krick K, Martin R, Berlinicke CA, Marsh-Armstrong N, Diamond JS, Mao HQ, Zack DJ. Differentiation of human ESCs to retinal ganglion cells using a CRISPR engineered reporter cell line. Sci Rep 2015; 5:16595. [PMID: 26563826 PMCID: PMC4643248 DOI: 10.1038/srep16595] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/13/2015] [Indexed: 12/30/2022] Open
Abstract
Retinal ganglion cell (RGC) injury and cell death from glaucoma and other forms of optic nerve disease is a major cause of irreversible vision loss and blindness. Human pluripotent stem cell (hPSC)-derived RGCs could provide a source of cells for the development of novel therapeutic molecules as well as for potential cell-based therapies. In addition, such cells could provide insights into human RGC development, gene regulation, and neuronal biology. Here, we report a simple, adherent cell culture protocol for differentiation of hPSCs to RGCs using a CRISPR-engineered RGC fluorescent reporter stem cell line. Fluorescence-activated cell sorting of the differentiated cultures yields a highly purified population of cells that express a range of RGC-enriched markers and exhibit morphological and physiological properties typical of RGCs. Additionally, we demonstrate that aligned nanofiber matrices can be used to guide the axonal outgrowth of hPSC-derived RGCs for in vitro optic nerve-like modeling. Lastly, using this protocol we identified forskolin as a potent promoter of RGC differentiation.
Collapse
Affiliation(s)
- Valentin M Sluch
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine Baltimore, MD 21287
| | - Chung-ha O Davis
- Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, 21205
| | - Vinod Ranganathan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Justin M Kerr
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Kellin Krick
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Russ Martin
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Cynthia A Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| | - Nicholas Marsh-Armstrong
- Hugo W. Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, 21205.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205
| | - Jeffrey S Diamond
- Synaptic Physiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Hai-Quan Mao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,Department of Materials Science and Engineering, Whiting School of Engineering, and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218
| | - Donald J Zack
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine Baltimore, MD 21287.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287.,The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205.,Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287
| |
Collapse
|
38
|
Measurement of Systemic Mitochondrial Function in Advanced Primary Open-Angle Glaucoma and Leber Hereditary Optic Neuropathy. PLoS One 2015; 10:e0140919. [PMID: 26496696 PMCID: PMC4619697 DOI: 10.1371/journal.pone.0140919] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/01/2015] [Indexed: 11/18/2022] Open
Abstract
Primary Open Angle Glaucoma (POAG) is a common neurodegenerative disease characterized by the selective and gradual loss of retinal ganglion cells (RGCs). Aging and increased intraocular pressure (IOP) are glaucoma risk factors; nevertheless patients deteriorate at all levels of IOP, implying other causative factors. Recent evidence presents mitochondrial oxidative phosphorylation (OXPHOS) complex-I impairments in POAG. Leber Hereditary Optic Neuropathy (LHON) patients suffer specific and rapid loss of RGCs, predominantly in young adult males, due to complex-I mutations in the mitochondrial genome. This study directly compares the degree of OXPHOS impairment in POAG and LHON patients, testing the hypothesis that the milder clinical disease in POAG is due to a milder complex-I impairment. To assess overall mitochondrial capacity, cells can be forced to produce ATP primarily from mitochondrial OXPHOS by switching the media carbon source to galactose. Under these conditions POAG lymphoblasts grew 1.47 times slower than controls, whilst LHON lymphoblasts demonstrated a greater degree of growth impairment (2.35 times slower). Complex-I enzyme specific activity was reduced by 18% in POAG lymphoblasts and by 29% in LHON lymphoblasts. We also assessed complex-I ATP synthesis, which was 19% decreased in POAG patients and 17% decreased in LHON patients. This study demonstrates both POAG and LHON lymphoblasts have impaired complex-I, and in the majority of aspects the functional defects in POAG were milder than LHON, which could reflect the milder disease development of POAG. This new evidence places POAG in the spectrum of mitochondrial optic neuropathies and raises the possibility for new therapeutic targets aimed at improving mitochondrial function.
Collapse
|
39
|
Yu W, Niu W, Wang S, Chen X, Sun BO, Wang F, Sun Y. Co-culture with endometrial stromal cells enhances the differentiation of human embryonic stem cells into endometrium-like cells. Exp Ther Med 2015; 10:43-50. [PMID: 26170910 DOI: 10.3892/etm.2015.2490] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/08/2015] [Indexed: 11/06/2022] Open
Abstract
In vitro differentiation of human embryonic stem cells (hESCs) into endometrium-like cells may provide a useful tool for clinical treatment. The aim of the present study was to investigate the differentiation potential of hESCs into endometrium-like cells using three methods, which included induction by feeder cells, co-culture with endometrial stromal cells and induction with embryoid bodies. Following differentiation, the majority of cells positively expressed cytokeratin and epithelial cell adhesion molecule (EPCAM). Factors associated with endometrium cell function, namely the estrogen and progesterone receptors (ER and PR), were also detected. At day 21 following the induction of differentiation, the expression levels of cytokeratin, EPCAM, ER and PR were significantly increased in the co-culture method group, as compared with the other two methods. Furthermore, these cells became decidualized in response to progesterone and prolactin. In addition, the number of cytokeratin-positive or EPCAM-positive cells significantly increased following the induction of differentiation using the co-culture method, as compared with the other two methods. The mRNA expression levels of Wnt members that are associated with endometrial development were subsequently examined, and Wnt5a was found to be significantly upregulated in the differentiated cells induced by feeder cells and co-culture with endometrial stromal cells; however, Wnt4 and Wnt7a expression levels were unaffected. Additionally, the mRNA expression levels of Wnt5a in the differentiated cells co-cultured with endometrial stromal cells were higher when compared with those induced by feeder cells. In conclusion, the present findings indicated that the co-culture system is the optimal protocol for the induction of hESC differentiation into endometrium-like cells, and Wnt5a signaling may be involved in this process.
Collapse
Affiliation(s)
- Wenzhu Yu
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Wenbin Niu
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shuna Wang
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xuemei Chen
- Department of Human Anatomy, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - B O Sun
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fang Wang
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingpu Sun
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|