51
|
Retinal Organoid Technology: Where Are We Now? Int J Mol Sci 2021; 22:ijms221910244. [PMID: 34638582 PMCID: PMC8549701 DOI: 10.3390/ijms221910244] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
It is difficult to regenerate mammalian retinal cells once the adult retina is damaged, and current clinical approaches to retinal damages are very limited. The introduction of the retinal organoid technique empowers researchers to study the molecular mechanisms controlling retinal development, explore the pathogenesis of retinal diseases, develop novel treatment options, and pursue cell/tissue transplantation under a certain genetic background. Here, we revisit the historical background of retinal organoid technology, categorize current methods of organoid induction, and outline the obstacles and potential solutions to next-generation retinal organoids. Meanwhile, we recapitulate recent research progress in cell/tissue transplantation to treat retinal diseases, and discuss the pros and cons of transplanting single-cell suspension versus retinal organoid sheet for cell therapies.
Collapse
|
52
|
Andreazzoli M, Barravecchia I, De Cesari C, Angeloni D, Demontis GC. Inducible Pluripotent Stem Cells to Model and Treat Inherited Degenerative Diseases of the Outer Retina: 3D-Organoids Limitations and Bioengineering Solutions. Cells 2021; 10:cells10092489. [PMID: 34572137 PMCID: PMC8471616 DOI: 10.3390/cells10092489] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies.
Collapse
Affiliation(s)
| | - Ivana Barravecchia
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | | | - Debora Angeloni
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | - Gian Carlo Demontis
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Correspondence: (M.A.); (G.C.D.)
| |
Collapse
|
53
|
Xue Y, Seiler MJ, Tang WC, Wang JY, Delgado J, McLelland BT, Nistor G, Keirstead HS, Browne AW. Retinal organoids on-a-chip: a micro-millifluidic bioreactor for long-term organoid maintenance. LAB ON A CHIP 2021; 21:3361-3377. [PMID: 34236056 PMCID: PMC8387452 DOI: 10.1039/d1lc00011j] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Retinal degeneration is a leading cause of vision impairment and blindness worldwide and medical care for advanced disease does not exist. Stem cell-derived retinal organoids (RtOgs) became an emerging tool for tissue replacement therapy. However, existing RtOg production methods are highly heterogeneous. Controlled and predictable methodology and tools are needed to standardize RtOg production and maintenance. In this study, we designed a shear stress-free micro-millifluidic bioreactor for nearly labor-free retinal organoid maintenance. We used a stereolithography (SLA) 3D printer to fabricate a mold from which Polydimethylsiloxane (PDMS) was cast. We optimized the chip design using in silico simulations and in vitro evaluation to optimize mass transfer efficiency and concentration uniformity in each culture chamber. We successfully cultured RtOgs at three different differentiation stages (day 41, 88, and 128) on an optimized bioreactor chip for more than 1 month. We used different quantitative and qualitative techniques to fully characterize the RtOgs produced by static dish culture and bioreactor culture methods. By analyzing the results from phase contrast microscopy, single-cell RNA sequencing (scRNA seq), quantitative polymerase chain reaction (qPCR), immunohistology, and electron microscopy, we found that bioreactor-cultured RtOgs developed cell types and morphology comparable to static cultured ones and exhibited similar retinal genes expression levels. We also evaluated the metabolic activity of RtOgs in both groups using fluorescence lifetime imaging (FLIM), and found that the outer surface region of bioreactor cultured RtOgs had a comparable free/bound NADH ratio and overall lower long lifetime species (LLS) ratio than static cultured RtOgs during imaging. To summarize, we validated an automated micro-millifluidic device with significantly reduced shear stress to produce RtOgs of comparable quality to those maintained in conventional static culture.
Collapse
Affiliation(s)
- Yuntian Xue
- Biomedical Engineering, University of California, Irvine, Irvine, CA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Matsuyama T, Tu HY, Sun J, Hashiguchi T, Akiba R, Sho J, Fujii M, Onishi A, Takahashi M, Mandai M. Genetically engineered stem cell-derived retinal grafts for improved retinal reconstruction after transplantation. iScience 2021; 24:102866. [PMID: 34409267 PMCID: PMC8361135 DOI: 10.1016/j.isci.2021.102866] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/23/2021] [Accepted: 07/14/2021] [Indexed: 11/29/2022] Open
Abstract
ESC/iPSC-retinal sheet transplantation, which supplies photoreceptors as well as other retinal cells, has been shown to be able to restore visual function in mice with end-stage retinal degeneration. Here, by introducing a novel type of genetically engineered mouse ESC/iPSC-retinal sheet with reduced numbers of secondary retinal neurons but intact photoreceptor cell layer structure, we reinforced the evidence that ESC/iPSC-retinal sheet transplantation can establish synaptic connections with the host, restore light responsiveness, and reduce aberrant retinal ganglion cell spiking in mice. Furthermore, we show that genetically engineered grafts can substantially improve the outcome of the treatment by improving neural integration. We speculate that this leads to reduced spontaneous activity in the host which in turn contributes to a better visual recovery.
Collapse
Affiliation(s)
- Take Matsuyama
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Hyogo, Japan
| | - Hung-Ya Tu
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Osaka, Japan
| | - Jianan Sun
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Tomoyo Hashiguchi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Ryutaro Akiba
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Junki Sho
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Momo Fujii
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Akishi Onishi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Hyogo, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
- Department of Ophthalmology, Kobe City Eye Hospital, Kobe, Hyogo, Japan
| |
Collapse
|
55
|
Ludwig AL, Gamm DM. Outer Retinal Cell Replacement: Putting the Pieces Together. Transl Vis Sci Technol 2021; 10:15. [PMID: 34724034 PMCID: PMC8572485 DOI: 10.1167/tvst.10.10.15] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/17/2022] Open
Abstract
Retinal degenerative diseases (RDDs) affecting photoreceptors (PRs) are one of the most prevalent sources of incurable blindness worldwide. Due to a lack of endogenous repair mechanisms, functional cell replacement of PRs and/or retinal pigmented epithelium (RPE) cells are among the most anticipated approaches for restoring vision in advanced RDD. Human pluripotent stem cell (hPSC) technologies have accelerated development of outer retinal cell therapies as they provide a theoretically unlimited source of donor cells. Human PSC-RPE replacement therapies have progressed rapidly, with several completed and ongoing clinical trials. Although potentially more promising, hPSC-PR replacement therapies are still in their infancy. A first-in-human trial of hPSC-derived neuroretinal transplantation has recently begun, but a number of questions regarding survival, reproducibility, functional integration, and mechanism of action remain. The discovery of biomaterial transfer between donor and PR cells has highlighted the need for rigorous safety and efficacy studies of PR replacement. In this review, we briefly discuss the history of neuroretinal and PR cell transplantation to identify remaining challenges and outline a stepwise approach to address specific pieces of the outer retinal cell replacement puzzle.
Collapse
Affiliation(s)
- Allison L. Ludwig
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin–Madison, Madison, WI, USA
- School of Veterinary Medicine, University of Wisconsin–Madison, Madison, WI, USA
| | - David M. Gamm
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin–Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, WI, USA
| |
Collapse
|
56
|
Döpper H, Menges J, Bozet M, Brenzel A, Lohmann D, Steenpass L, Kanber D. Differentiation Protocol for 3D Retinal Organoids, Immunostaining and Signal Quantitation. ACTA ACUST UNITED AC 2021; 55:e120. [PMID: 32956559 DOI: 10.1002/cpsc.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Structures resembling whole organs, called organoids, are generated using pluripotent stem cells and 3D culturing methods. This relies on the ability of cells to self-reorganize after dissociation. In combination with certain supplemented factors, differentiation can be directed toward the formation of several organ-like structures. Here, a protocol for the generation of retinal organoids containing all seven retinal cell types is described. This protocol does not depend on Matrigel, and by keeping the organoids single and independent at all times, fusion is prevented and monitoring of differentiation is improved. Comprehensive phenotypic characterization of the in vitro-generated retinal organoids is achieved by the protocol for immunostaining outlined here. By comparing different stages of retinal organoids, the decrease and increase of certain cell populations can be determined. In order to be able to detect even small differences, it is necessary to quantify the immunofluorescent signals, for which we have provided a detailed protocol describing signal quantitation using the image-processing program Fiji. © 2020 The Authors. Basic Protocol 1: Differentiation protocol for 3D retinal organoids Basic Protocol 2: Immunostaining protocol for cryosections of retinal organoids Support Protocol: Embedding and sectioning protocol for 3D retinal organoids Basic Protocol 3: Quantitation protocol using Fiji.
Collapse
Affiliation(s)
- Hannah Döpper
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julia Menges
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Morgane Bozet
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Brenzel
- Institute for Experimental Immunology and Imaging, Imaging Center Essen (LMU), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dietmar Lohmann
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Laura Steenpass
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Present address: Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures GmbH, Braunschweig, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
57
|
Occelli LM, Marinho F, Singh RK, Binette F, Nasonkin IO, Petersen-Jones SM. Subretinal Transplantation of Human Embryonic Stem Cell-Derived Retinal Tissue in a Feline Large Animal Model. J Vis Exp 2021:10.3791/61683. [PMID: 34424232 PMCID: PMC10029721 DOI: 10.3791/61683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Retinal degenerative (RD) conditions associated with photoreceptor loss such as age-related macular degeneration (AMD), retinitis pigmentosa (RP) and Leber Congenital Amaurosis (LCA) cause progressive and debilitating vision loss. There is an unmet need for therapies that can restore vision once photoreceptors have been lost. Transplantation of human pluripotent stem cell (hPSC)-derived retinal tissue (organoids) into the subretinal space of an eye with advanced RD brings retinal tissue sheets with thousands of healthy mutation-free photoreceptors and has a potential to treat most/all blinding diseases associated with photoreceptor degeneration with one approved protocol. Transplantation of fetal retinal tissue into the subretinal space of animal models and people with advanced RD has been developed successfully but cannot be used as a routine therapy due to ethical concerns and limited tissue supply. Large eye inherited retinal degeneration (IRD) animal models are valuable for developing vision restoration therapies utilizing advanced surgical approaches to transplant retinal cells/tissue into the subretinal space. The similarities in globe size, and photoreceptor distribution (e.g., presence of macula-like region area centralis) and availability of IRD models closely recapitulating human IRD would facilitate rapid translation of a promising therapy to the clinic. Presented here is a surgical technique of transplanting hPSC-derived retinal tissue into the subretinal space of a large animal model allowing assessment of this promising approach in animal models.
Collapse
Affiliation(s)
- Laurence M Occelli
- College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Michigan State University
| | - Felipe Marinho
- College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Michigan State University
| | | | | | | | - Simon M Petersen-Jones
- College of Veterinary Medicine, Department of Small Animal Clinical Sciences, Michigan State University;
| |
Collapse
|
58
|
Deloria AJ, Haider S, Dietrich B, Kunihs V, Oberhofer S, Knofler M, Leitgeb R, Liu M, Drexler W, Haindl R. Ultra-High-Resolution 3D Optical Coherence Tomography Reveals Inner Structures of Human Placenta-Derived Trophoblast Organoids. IEEE Trans Biomed Eng 2021; 68:2368-2376. [PMID: 33201804 DOI: 10.1109/tbme.2020.3038466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE 3D optical coherence tomography (OCT) is used for analyses of human placenta organoids in situ without sample preparation. METHODS The trophoblast organoids analyzed were derived from primary human trophoblast. In this study a custom made ultra-high-resolution spectral domain OCT system with uniform spatial and axial resolution of 2.48 μm in organoid tissue was used. The obtained OCT results align to differentiation status tested via quantitative polymerase chain reaction, Western blot analyses, immunohistochemistry, and immunofluorescence of histological sections. RESULTS 3D OCT enables a more detailed placenta organoid monitoring compared to brightfield microscopy. Inner architecture with light scattering "bridges" surrounding cavities were visualized and quantified in situ for the first time. The formation of these bridges and cavities is congruent to differentiated trophoblast organoids having developed syncytiotrophoblasts. CONCLUSION Using 3D OCT in living placenta organoids is a fast tool to assess the differentiation status and resolve internal structures in situ, which is not possible with standard live cell imaging modality. SIGNIFICANCE Only recently human placenta-derived organoids were established, allowing to have a highly reproducible and stable in vitro model to investigate not only developmental but also physiological and pathophysiological processes during early pregnancy. To our knowledge, this work is the first to analyze living human placenta organoids using 3D OCT. Thereby, the rapid and especially non-endpoint OCT qualitative analyses align to the differentiation stage of organoids, which will aid future advancement in this field.
Collapse
|
59
|
Wagstaff EL, Heredero Berzal A, Boon CJF, Quinn PMJ, ten Asbroek ALMA, Bergen AA. The Role of Small Molecules and Their Effect on the Molecular Mechanisms of Early Retinal Organoid Development. Int J Mol Sci 2021; 22:7081. [PMID: 34209272 PMCID: PMC8268497 DOI: 10.3390/ijms22137081] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Early in vivo embryonic retinal development is a well-documented and evolutionary conserved process. The specification towards eye development is temporally controlled by consecutive activation or inhibition of multiple key signaling pathways, such as the Wnt and hedgehog signaling pathways. Recently, with the use of retinal organoids, researchers aim to manipulate these pathways to achieve better human representative models for retinal development and disease. To achieve this, a plethora of different small molecules and signaling factors have been used at various time points and concentrations in retinal organoid differentiations, with varying success. Additions differ from protocol to protocol, but their usefulness or efficiency has not yet been systematically reviewed. Interestingly, many of these small molecules affect the same and/or multiple pathways, leading to reduced reproducibility and high variability between studies. In this review, we make an inventory of the key signaling pathways involved in early retinogenesis and their effect on the development of the early retina in vitro. Further, we provide a comprehensive overview of the small molecules and signaling factors that are added to retinal organoid differentiation protocols, documenting the molecular and functional effects of these additions. Lastly, we comparatively evaluate several of these factors using our established retinal organoid methodology.
Collapse
Affiliation(s)
- Ellie L. Wagstaff
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center—New York-Presbyterian Hospital, New York, NY 10032, USA;
| | | | - Arthur A. Bergen
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Netherlands Institute for Neuroscience (NIN-KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
60
|
Li X, Zhang L, Tang F, Wei X. Retinal Organoids: Cultivation, Differentiation, and Transplantation. Front Cell Neurosci 2021; 15:638439. [PMID: 34276307 PMCID: PMC8282056 DOI: 10.3389/fncel.2021.638439] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/08/2021] [Indexed: 02/05/2023] Open
Abstract
Retinal organoids (ROs), which are derived from stem cells, can automatically form three-dimensional laminar structures that include all cell types and the ultrastructure of the retina. Therefore, they are highly similar to the retinal structure in the human body. The development of organoids has been a great technological breakthrough in the fields of transplantation therapy and disease modeling. However, the translation of RO applications into medical practice still has various deficiencies at the current stage, including the long culture process, insufficient yield, and great heterogeneity among ROs produced under different conditions. Nevertheless, many technological breakthroughs have been made in transplanting ROs for treatment of diseases such as retinal degeneration. This review discusses recent advances in the development of ROs, improvements of the culture protocol, and the latest developments in RO replacement therapy techniques.
Collapse
Affiliation(s)
- Xuying Li
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Tang
- Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| |
Collapse
|
61
|
Fathi M, Ross CT, Hosseinzadeh Z. Functional 3-Dimensional Retinal Organoids: Technological Progress and Existing Challenges. Front Neurosci 2021; 15:668857. [PMID: 33958988 PMCID: PMC8095320 DOI: 10.3389/fnins.2021.668857] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Stem cell scientists have developed methods for the self-formation of artificial organs, often referred to as organoids. Organoids can be used as model systems for research in multiple biological disciplines. Yoshiki Sasai’s innovation for deriving mammalian retinal tissue from in vitro stem cells has had a large impact on the study of the biology of vision. New developments in retinal organoid technology provide avenues for in vitro models of human retinal diseases, studies of pathological mechanisms, and development of therapies for retinal degeneration, including electronic retinal implants and gene therapy. Moreover, these innovations have played key roles in establishing models for large-scale drug screening, studying the stages of retinal development, and providing a human model for personalized therapeutic approaches, like cell transplants to replace degenerated retinal cells. Here, we first discuss the importance of human retinal organoids to the biomedical sciences. Then, we review various functional features of retinal organoids that have been developed. Finally, we highlight the current limitations of retinal organoid technologies.
Collapse
Affiliation(s)
- Meimanat Fathi
- Department of Cell Techniques and Applied Stem Cell Biology, Faculty of Medicine, Center for Biotechnology and Biomedicine (BBZ), University of Leipzig, Leipzig, Germany.,Physiology and Pathophysiology of the Retina Group, Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| | - Cody T Ross
- Department of Human Behavior, Ecology and Culture, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Zohreh Hosseinzadeh
- Physiology and Pathophysiology of the Retina Group, Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute of Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
62
|
Singh RK, Winkler PA, Binette F, Petersen-Jones SM, Nasonkin IO. Comparison of Developmental Dynamics in Human Fetal Retina and Human Pluripotent Stem Cell-Derived Retinal Tissue. Stem Cells Dev 2021; 30:399-417. [PMID: 33677999 DOI: 10.1089/scd.2020.0085] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Progressive vision loss, caused by retinal degenerative (RD) diseases such as age-related macular degeneration, retinitis pigmentosa, and Leber congenital amaurosis, severely impacts quality of life and affects millions of people. Finding efficient treatment for blinding diseases is among the greatest unmet clinical needs. The evagination of optic vesicles from developing pluripotent stem cell-derived neuroepithelium and self-organization, lamination, and differentiation of retinal tissue in a dish generated considerable optimism for developing innovative approaches for treating RD diseases, which previously were not feasible. Retinal organoids may be a limitless source of multipotential retinal progenitors, photoreceptors (PRs), and the whole retinal tissue, which are productive approaches for developing RD disease therapies. In this study we compared the distribution and expression level of molecular markers (genetic and epigenetic) in human fetal retina (age 8-16 weeks) and human embryonic stem cell (hESC)-derived retinal tissue (organoids) by immunohistochemistry, RNA-seq, flow cytometry, and mass-spectrometry (to measure methylated and hydroxymethylated cytosine level), with a focus on PRs to evaluate the clinical application of hESC-retinal tissue for vision restoration. Our results revealed high correlation in gene expression profiles and histological profiles between human fetal retina (age 8-13 weeks) and hESC-derived retinal tissue (10-12 weeks). The transcriptome signature of hESC-derived retinal tissue from retinal organoids maintained for 24 weeks in culture resembled the transcriptome of human fetal retina of more advanced developmental stages. The histological profiles of 24 week-old hESC-derived retinal tissue displayed mature PR immunophenotypes and presence of developing inner and outer segments. Collectively, our work highlights the similarity of hESC-derived retinal tissue at early stages of development (10 weeks), and human fetal retina (age 8-13 weeks) and it supports the development of regenerative medicine therapies aimed at using tissue from hESC-derived retinal organoids (hESC-retinal implants) for mitigating vision loss.
Collapse
Affiliation(s)
| | - Paige A Winkler
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | | | - Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | | |
Collapse
|
63
|
Ribeiro J, Procyk CA, West EL, O'Hara-Wright M, Martins MF, Khorasani MM, Hare A, Basche M, Fernando M, Goh D, Jumbo N, Rizzi M, Powell K, Tariq M, Michaelides M, Bainbridge JWB, Smith AJ, Pearson RA, Gonzalez-Cordero A, Ali RR. Restoration of visual function in advanced disease after transplantation of purified human pluripotent stem cell-derived cone photoreceptors. Cell Rep 2021; 35:109022. [PMID: 33882303 PMCID: PMC8065177 DOI: 10.1016/j.celrep.2021.109022] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/08/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022] Open
Abstract
Age-related macular degeneration and other macular diseases result in the loss of light-sensing cone photoreceptors, causing irreversible sight impairment. Photoreceptor replacement may restore vision by transplanting healthy cells, which must form new synaptic connections with the recipient retina. Despite recent advances, convincing evidence of functional connectivity arising from transplanted human cone photoreceptors in advanced retinal degeneration is lacking. Here, we show restoration of visual function after transplantation of purified human pluripotent stem cell-derived cones into a mouse model of advanced degeneration. Transplanted human cones elaborate nascent outer segments and make putative synapses with recipient murine bipolar cells (BCs), which themselves undergo significant remodeling. Electrophysiological and behavioral assessments demonstrate restoration of surprisingly complex light-evoked retinal ganglion cell responses and improved light-evoked behaviors in treated animals. Stringent controls exclude alternative explanations, including material transfer and neuroprotection. These data provide crucial validation for photoreceptor replacement therapy and for the potential to rescue cone-mediated vision.
Collapse
Affiliation(s)
- Joana Ribeiro
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Emma L West
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Monica F Martins
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Aura Hare
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Mark Basche
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Milan Fernando
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Debbie Goh
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Neeraj Jumbo
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Matteo Rizzi
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Kate Powell
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Menahil Tariq
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | | | - Alexander J Smith
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Rachael A Pearson
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Kellogg Eye Centre, University of Michigan, 1000 Wall St., Ann Arbor, MI 48105, USA.
| |
Collapse
|
64
|
Singh RK, Binette F, Seiler M, Petersen-Jones SM, Nasonkin IO. Pluripotent Stem Cell-Based Organoid Technologies for Developing Next-Generation Vision Restoration Therapies of Blindness. J Ocul Pharmacol Ther 2021; 37:147-156. [PMID: 33052761 PMCID: PMC8060716 DOI: 10.1089/jop.2020.0016] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 07/28/2020] [Indexed: 12/25/2022] Open
Abstract
Blindness, associated with death of retinal cells at the back of the eye, is caused by a number of conditions with high prevalence such as glaucoma, age-related macular degeneration, and diabetic retinopathy. In addition, a large number of orphan inherited (mostly monogenic) conditions, such as retinitis pigmentosa and Leber Congenital Amaurosis, add to the overall number of patients with blinding retinal degenerative diseases. Blindness caused by deterioration and loss of retina is so far incurable. Modern biomedical research leveraging molecular and regenerative medicine approaches had a number of groundbreaking discoveries and proof-of-principle treatments of blindness in animals. However, these methods are slow to be standardized and commercialized as therapies to benefit people losing their eyesight due to retinal degenerative conditions. In this review, we will outline major regenerative medicine approaches, which are emerging as promising for preserving or/and restoring vision. We discuss the potential of each of these approaches to reach commercialization step and be converted to treatments, which could at least ameliorate blindness caused by retinal cell death.
Collapse
Affiliation(s)
| | | | - Magdalene Seiler
- Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
- Department of Physical Medicine & Rehabilitation, University of California, Irvine, Irvine, California, USA
- Department of Ophthalmology, University of California, Irvine, Irvine, California, USA
- Department of Anatomy & Neurobiology, University of California, Irvine, Irvine, California, USA
| | - Simon M. Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | | |
Collapse
|
65
|
Garita-Hernandez M, Chaffiol A, Guibbal L, Routet F, Khabou H, Riancho L, Toualbi L, Picaud S, Sahel JA, Goureau O, Duebel J, Dalkara D. Control of Microbial Opsin Expression in Stem Cell Derived Cones for Improved Outcomes in Cell Therapy. Front Cell Neurosci 2021; 15:648210. [PMID: 33815066 PMCID: PMC8012682 DOI: 10.3389/fncel.2021.648210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Human-induced pluripotent stem cell (hiPSC) derived organoids have become increasingly used systems allowing 3D-modeling of human organ development, and disease. They are also a reliable source of cells for transplantation in cell therapy and an excellent model to validate gene therapies. To make full use of these systems, a toolkit of genetic modification techniques is necessary to control their activity in line with the downstream application. We have previously described adeno-associated viruse (AAV) vectors for efficient targeting of cells within human retinal organoids. Here, we describe biological restriction and enhanced gene expression in cone cells of such organoids thanks to the use of a 1.7-kb L-opsin promoter. We illustrate the usefulness of implementing such a promoter to enhance the expression of the red-shifted opsin Jaws in fusion with a fluorescent reporter gene, enabling cell sorting to enrich the desired cell population. Increased Jaws expression after transplantation improved light responses promising better therapeutic outcomes in a cell therapy setting. Our results point to the importance of promoter activity in restricting, improving, and controlling the kinetics of transgene expression during the maturation of hiPSC retinal derivatives. Differentiation requires mechanisms to initiate specific transcriptional changes and to reinforce those changes when mature cell states are reached. By employing a cell-type-specific promoter we put transgene expression under the new transcriptional program of mature cells.
Collapse
Affiliation(s)
| | | | - Laure Guibbal
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Fiona Routet
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Hanen Khabou
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Luisa Riancho
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Lyes Toualbi
- Institut de la Vision, Sorbonne Université, Paris, France
| | - Serge Picaud
- Institut de la Vision, Sorbonne Université, Paris, France
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, Paris, France
- CHNO des Quinze−Vingts, DHU Sight Restore, Paris, France
- Department of Ophthalmology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | | | - Jens Duebel
- Institut de la Vision, Sorbonne Université, Paris, France
- Department of Ophthalmology, University Medical Center Göttingen, Göttingen, Germany
| | - Deniz Dalkara
- Institut de la Vision, Sorbonne Université, Paris, France
| |
Collapse
|
66
|
Nair DSR, Seiler MJ, Patel KH, Thomas V, Camarillo JCM, Humayun MS, Thomas BB. Tissue Engineering Strategies for Retina Regeneration. APPLIED SCIENCES-BASEL 2021; 11. [PMID: 35251703 PMCID: PMC8896578 DOI: 10.3390/app11052154] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The retina is a complex and fragile photosensitive part of the central nervous system which is prone to degenerative diseases leading to permanent vision loss. No proven treatment strategies exist to treat or reverse the degenerative conditions. Recent investigations demonstrate that cell transplantation therapies to replace the dysfunctional retinal pigment epithelial (RPE) cells and or the degenerating photoreceptors (PRs) are viable options to restore vision. Pluripotent stem cells, retinal progenitor cells, and somatic stem cells are the main cell sources used for cell transplantation therapies. The success of retinal transplantation based on cell suspension injection is hindered by limited cell survival and lack of cellular integration. Recent advances in material science helped to develop strategies to grow cells as intact monolayers or as sheets on biomaterial scaffolds for transplantation into the eyes. Such implants are found to be more promising than the bolus injection approach. Tissue engineering techniques are specifically designed to construct biodegradable or non-degradable polymer scaffolds to grow cells as a monolayer and construct implantable grafts. The engineered cell construct along with the extracellular matrix formed, can hold the cells in place to enable easy survival, better integration, and improved visual function. This article reviews the advances in the use of scaffolds for transplantation studies in animal models and their application in current clinical trials.
Collapse
Affiliation(s)
- Deepthi S. Rajendran Nair
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Magdalene J. Seiler
- Departments of Physical Medicine & Rehabilitation, Ophthalmology, Anatomy & Neurobiology, Sue and Bill Gross Stem Cell Research Centre, University of California, Irvine, CA 92697-1705, USA
| | - Kahini H. Patel
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Vinoy Thomas
- Department of Physics, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Juan Carlos Martinez Camarillo
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
| | - Mark S. Humayun
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
| | - Biju B. Thomas
- Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA 90033, USA
- Correspondence:
| |
Collapse
|
67
|
Clinical Application of Human Induced Pluripotent Stem Cell-Derived Organoids as an Alternative to Organ Transplantation. Stem Cells Int 2021; 2021:6632160. [PMID: 33679987 PMCID: PMC7929656 DOI: 10.1155/2021/6632160] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/19/2021] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Transplantation is essential and crucial for individuals suffering from end-stage organ failure diseases. However, there are still many challenges regarding these procedures, such as high rates of organ rejection, shortage of organ donors, and long waiting lines. Thus, investments and efforts to develop laboratory-grown organs have increased over the past years, and with the recent progress in regenerative medicine, growing organs in vitro might be a reality within the next decades. One of the many different strategies to address this issue relies on organoid technology, a miniaturized and simplified version of an organ. Here, we address recent progress on organoid research, focusing on transplantation of intestine, retina, kidney, liver, pancreas, brain, lung, and heart organoids. Also, we discuss the main outcomes after organoid transplantation, common challenges faced by these promising regenerative medicine approaches, and future perspectives on the field.
Collapse
|
68
|
Humanization of Immunodeficient Animals for the Modeling of Transplantation, Graft Versus Host Disease, and Regenerative Medicine. Transplantation 2021; 104:2290-2306. [PMID: 32068660 PMCID: PMC7590965 DOI: 10.1097/tp.0000000000003177] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The humanization of animals is a powerful tool for the exploration of human disease pathogenesis in biomedical research, as well as for the development of therapeutic interventions with enhanced translational potential. Humanized models enable us to overcome biologic differences that exist between humans and other species, while giving us a platform to study human processes in vivo. To become humanized, an immune-deficient recipient is engrafted with cells, tissues, or organoids. The mouse is the most well studied of these hosts, with a variety of immunodeficient strains available for various specific uses. More recently, efforts have turned to the humanization of other animal species such as the rat, which offers some technical and immunologic advantages over mice. These advances, together with ongoing developments in the incorporation of human transgenes and additional mutations in humanized mouse models, have expanded our opportunities to replicate aspects of human allotransplantation and to assist in the development of immunotherapies. In this review, the immune and tissue humanization of various species is presented with an emphasis on their potential for use as models for allotransplantation, graft versus host disease, and regenerative medicine.
Collapse
|
69
|
The Future of Regenerative Medicine: Cell Therapy Using Pluripotent Stem Cells and Acellular Therapies Based on Extracellular Vesicles. Cells 2021; 10:cells10020240. [PMID: 33513719 PMCID: PMC7912181 DOI: 10.3390/cells10020240] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/13/2021] [Accepted: 01/23/2021] [Indexed: 12/11/2022] Open
Abstract
The rapid progress in the field of stem cell research has laid strong foundations for their use in regenerative medicine applications of injured or diseased tissues. Growing evidences indicate that some observed therapeutic outcomes of stem cell-based therapy are due to paracrine effects rather than long-term engraftment and survival of transplanted cells. Given their ability to cross biological barriers and mediate intercellular information transfer of bioactive molecules, extracellular vesicles are being explored as potential cell-free therapeutic agents. In this review, we first discuss the state of the art of regenerative medicine and its current limitations and challenges, with particular attention on pluripotent stem cell-derived products to repair organs like the eye, heart, skeletal muscle and skin. We then focus on emerging beneficial roles of extracellular vesicles to alleviate these pathological conditions and address hurdles and operational issues of this acellular strategy. Finally, we discuss future directions and examine how careful integration of different approaches presented in this review could help to potentiate therapeutic results in preclinical models and their good manufacturing practice (GMP) implementation for future clinical trials.
Collapse
|
70
|
Lin Y, Ren X, Chen Y, Chen D. Interaction Between Mesenchymal Stem Cells and Retinal Degenerative Microenvironment. Front Neurosci 2021; 14:617377. [PMID: 33551729 PMCID: PMC7859517 DOI: 10.3389/fnins.2020.617377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Retinal degenerative diseases (RDDs) are a group of diseases contributing to irreversible vision loss with yet limited therapies. Stem cell-based therapy is a promising novel therapeutic approach in RDD treatment. Mesenchymal stromal/stem cells (MSCs) have emerged as a leading cell source due to their neurotrophic and immunomodulatory capabilities, limited ethical concerns, and low risk of tumor formation. Several pre-clinical studies have shown that MSCs have the potential to delay retinal degeneration, and recent clinical trials have demonstrated promising safety profiles for the application of MSCs in retinal disease. However, some of the clinical-stage MSC therapies have been unable to meet primary efficacy end points, and severe side effects were reported in some retinal “stem cell” clinics. In this review, we provide an update of the interaction between MSCs and the RDD microenvironment and discuss how to balance the therapeutic potential and safety concerns of MSCs' ocular application.
Collapse
Affiliation(s)
- Yu Lin
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Ren
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongjiang Chen
- The School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Danian Chen
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
71
|
Marcos LF, Wilson SL, Roach P. Tissue engineering of the retina: from organoids to microfluidic chips. J Tissue Eng 2021; 12:20417314211059876. [PMID: 34917332 PMCID: PMC8669127 DOI: 10.1177/20417314211059876] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/28/2021] [Indexed: 12/29/2022] Open
Abstract
Despite advancements in tissue engineering, challenges remain for fabricating functional tissues that incorporate essential features including vasculature and complex cellular organisation. Monitoring of engineered tissues also raises difficulties, particularly when cell population maturity is inherent to function. Microfluidic, or lab-on-a-chip, platforms address the complexity issues of conventional 3D models regarding cell numbers and functional connectivity. Regulation of biochemical/biomechanical conditions can create dynamic structures, providing microenvironments that permit tissue formation while quantifying biological processes at a single cell level. Retinal organoids provide relevant cell numbers to mimic in vivo spatiotemporal development, where conventional culture approaches fail. Modern bio-fabrication techniques allow for retinal organoids to be combined with microfluidic devices to create anato-physiologically accurate structures or 'retina-on-a-chip' devices that could revolution ocular sciences. Here we present a focussed review of retinal tissue engineering, examining the challenges and how some of these have been overcome using organoids, microfluidics, and bioprinting technologies.
Collapse
Affiliation(s)
- Luis F Marcos
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK
| | - Samantha L Wilson
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Leicestershire, UK
| | - Paul Roach
- Department of Chemistry, School of Science, Loughborough University, Leicestershire, UK
| |
Collapse
|
72
|
Jemni-Damer N, Guedan-Duran A, Fuentes-Andion M, Serrano-Bengoechea N, Alfageme-Lopez N, Armada-Maresca F, Guinea GV, Perez-Rigueiro J, Rojo F, Gonzalez-Nieto D, Kaplan DL, Panetsos F. Biotechnology and Biomaterial-Based Therapeutic Strategies for Age-Related Macular Degeneration. Part II: Cell and Tissue Engineering Therapies. Front Bioeng Biotechnol 2020; 8:588014. [PMID: 33363125 PMCID: PMC7758210 DOI: 10.3389/fbioe.2020.588014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022] Open
Abstract
Age-related Macular Degeneration (AMD) is an up-to-date untreatable chronic neurodegenerative eye disease of multifactorial origin, and the main causes of blindness in over 65 y.o. people. It is characterized by a slow progression and the presence of a multitude of factors, highlighting those related to diet, genetic heritage and environmental conditions, present throughout each of the stages of the illness. Current therapeutic approaches, mainly consisting on intraocular drug delivery, are only used for symptoms relief and/or to decelerate the progression of the disease. Furthermore, they are overly simplistic and ignore the complexity of the disease and the enormous differences in the symptomatology between patients. Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, Due to the wide impact of the AMD and the up-to-date absence of clinical solutions, different treatment options have to be considered. Cell therapy is a very promising alternative to drug-based approaches for AMD treatment. Cells delivered to the affected tissue as a suspension have shown poor retention and low survival rate. A solution to these inconveniences has been the encapsulation of these cells on biomaterials, which contrive to their protection, gives them support, and favor their retention of the desired area. We offer a two-papers critical review of the available and under development AMD therapeutic approaches, from a biomaterials and biotechnological point of view. We highlight benefits and limitations and we forecast forthcoming alternatives based on novel biomaterials and biotechnology methods. In this second part we review the preclinical and clinical cell-replacement approaches aiming at the development of efficient AMD-therapies, the employed cell types, as well as the cell-encapsulation and cell-implant systems. We discuss their advantages and disadvantages and how they could improve the survival and integration of the implanted cells.
Collapse
Affiliation(s)
- Nahla Jemni-Damer
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Atocha Guedan-Duran
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - María Fuentes-Andion
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
| | - Nora Serrano-Bengoechea
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | - Nuria Alfageme-Lopez
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| | | | - Gustavo V. Guinea
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - José Perez-Rigueiro
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Francisco Rojo
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Department of Material Science, Civil Engineering Superior School, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - Daniel Gonzalez-Nieto
- Silk Biomed SL, Madrid, Spain
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcon, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine, Madrid, Spain
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Fivos Panetsos
- Neuro-computing and Neuro-robotics Research Group, Complutense University of Madrid, Madrid, Spain
- Innovation Group, Institute for Health Research San Carlos Clinical Hospital, Madrid, Spain
- Silk Biomed SL, Madrid, Spain
| |
Collapse
|
73
|
Ghareeb AE, Lako M, Steel DH. Coculture techniques for modeling retinal development and disease, and enabling regenerative medicine. Stem Cells Transl Med 2020; 9:1531-1548. [PMID: 32767661 PMCID: PMC7695644 DOI: 10.1002/sctm.20-0201] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/22/2020] [Accepted: 07/05/2020] [Indexed: 12/14/2022] Open
Abstract
Stem cell-derived retinal organoids offer the opportunity to cure retinal degeneration of wide-ranging etiology either through the study of in vitro models or the generation of tissue for transplantation. However, despite much work in animals and several human pilot studies, satisfactory therapies have not been developed. Two major challenges for retinal regenerative medicine are (a) physical cell-cell interactions, which are critical to graft function, are not formed and (b) the host environment does not provide suitable queues for development. Several strategies offer to improve the delivery, integration, maturation, and functionality of cell transplantation. These include minimally invasive delivery, biocompatible material vehicles, retinal cell sheets, and optogenetics. Optimizing several variables in animal models is practically difficult, limited by anatomical and disease pathology which is often different to humans, and faces regulatory and ethical challenges. High-throughput methods are needed to experimentally optimize these variables. Retinal organoids will be important to the success of these models. In their current state, they do not incorporate a representative retinal pigment epithelium (RPE)-photoreceptor interface nor vascular elements, which influence the neural retina phenotype directly and are known to be dysfunctional in common retinal diseases such as age-related macular degeneration. Advanced coculture techniques, which emulate the RPE-photoreceptor and RPE-Bruch's-choriocapillaris interactions, can incorporate disease-specific, human retinal organoids and overcome these drawbacks. Herein, we review retinal coculture models of the neural retina, RPE, and choriocapillaris. We delineate the scientific need for such systems in the study of retinal organogenesis, disease modeling, and the optimization of regenerative cell therapies for retinal degeneration.
Collapse
Affiliation(s)
- Ali E. Ghareeb
- Sunderland Eye Infirmary, South Tyneside and Sunderland NHS Foundation TrustSunderlandUK
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| | - Majlinda Lako
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| | - David H. Steel
- Sunderland Eye Infirmary, South Tyneside and Sunderland NHS Foundation TrustSunderlandUK
- Biosciences Institute, Newcastle UniversityNewcastle‐upon‐TyneUK
| |
Collapse
|
74
|
Manafi N, Shokri F, Achberger K, Hirayama M, Mohammadi MH, Noorizadeh F, Hong J, Liebau S, Tsuji T, Quinn PMJ, Mashaghi A. Organoids and organ chips in ophthalmology. Ocul Surf 2020; 19:1-15. [PMID: 33220469 DOI: 10.1016/j.jtos.2020.11.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Recent advances have driven the development of stem cell-derived, self-organizing, three-dimensional miniature organs, termed organoids, which mimic different eye tissues including the retina, cornea, and lens. Organoids and engineered microfluidic organ-on-chips (organ chips) are transformative technologies that show promise in simulating the architectural and functional complexity of native organs. Accordingly, they enable exploration of facets of human disease and development not accurately recapitulated by animal models. Together, these technologies will increase our understanding of the basic physiology of different eye structures, enable us to interrogate unknown aspects of ophthalmic disease pathogenesis, and serve as clinically-relevant surrogates for the evaluation of ocular therapeutics. Both the burden and prevalence of monogenic and multifactorial ophthalmic diseases, which can cause visual impairment or blindness, in the human population warrants a paradigm shift towards organoids and organ chips that can provide sensitive, quantitative, and scalable phenotypic assays. In this article, we review the current situation of organoids and organ chips in ophthalmology and discuss how they can be leveraged for translational applications.
Collapse
Affiliation(s)
- Navid Manafi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Fereshteh Shokri
- Department of Epidemiology, Erasmus Medical Center, 3000 CA, Rotterdam, the Netherlands
| | - Kevin Achberger
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Masatoshi Hirayama
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, Chiba, 272-8513, Japan; Department of Ophthalmology, School of Medicine, Keio University, Tokyo, 160-8582, Japan
| | - Melika Haji Mohammadi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands
| | | | - Jiaxu Hong
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands; Department of Ophthalmology and Visual Science, Eye, and ENT Hospital, Shanghai Medical College, Fudan University, 83 Fenyang Road, Shanghai, China; Key NHC Key Laboratory of Myopia (Fudan University), Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China; Key Laboratory of Myopia, National Health and Family Planning Commission, Shanghai, China
| | - Stefan Liebau
- Institute of Neuroanatomy & Developmental Biology (INDB), Eberhard Karls University Tübingen, Österbergstrasse 3, 72074, Tübingen, Germany
| | - Takashi Tsuji
- Laboratory for Organ Regeneration, RIKEN Center for Biosystems Dynamics Research, Hyogo, 650-0047, Japan; Organ Technologies Inc., Minato, Tokyo, 105-0001, Japan
| | - Peter M J Quinn
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University. New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center - New York-Presbyterian Hospital, New York, NY, USA.
| | - Alireza Mashaghi
- Medical Systems Biophysics and Bioengineering, The Leiden Academic Centre for Drug Research (LACDR), Leiden University, 2333CC, Leiden, the Netherlands.
| |
Collapse
|
75
|
Zhang CJ, Ma Y, Jin ZB. The road to restore vision with photoreceptor regeneration. Exp Eye Res 2020; 202:108283. [PMID: 33010290 DOI: 10.1016/j.exer.2020.108283] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 09/13/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022]
Abstract
Neuroretinal diseases are the predominant cause of irreversible blindness worldwide, mainly due to photoreceptor loss. Currently, there are no radical treatments to fully reverse the degeneration or even stop the disease progression. Thus, it is urgent to develop new biological therapeutics for these diseases on the clinical side. Stem cell-based treatments have become a promising therapeutic for neuroretinal diseases through the replacement of damaged cells with photoreceptors and some allied cells. To date, considerable efforts have been made to regenerate the diseased retina based on stem cell technology. In this review, we overview the current status of stem cell-based treatments for photoreceptor regeneration, including the major cell sources derived from different stem cells in pre-clinical or clinical trial stages. Additionally, we discuss herein the major challenges ahead for and potential new strategy toward photoreceptor regeneration.
Collapse
Affiliation(s)
- Chang-Jun Zhang
- Laboratory for Stem Cell & Retinal Regeneration, The Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ya Ma
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
76
|
Bell CM, Zack DJ, Berlinicke CA. Human Organoids for the Study of Retinal Development and Disease. Annu Rev Vis Sci 2020; 6:91-114. [DOI: 10.1146/annurev-vision-121219-081855] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent advances in stem cell engineering have led to an explosion in the use of organoids as model systems for studies in multiple biological disciplines. Together with breakthroughs in genome engineering and the various omics, organoid technology is making possible studies of human biology that were not previously feasible. For vision science, retinal organoids derived from human stem cells allow differentiating and mature human retinal cells to be studied in unprecedented detail. In this review, we examine the technologies employed to generate retinal organoids and how organoids are revolutionizing the fields of developmental and cellular biology as they pertain to the retina. Furthermore, we explore retinal organoids from a clinical standpoint, offering a new platform with which to study retinal diseases and degeneration, test prospective drugs and therapeutic strategies, and promote personalized medicine. Finally, we discuss the range of possibilities that organoids may bring to future retinal research and consider their ethical implications.
Collapse
Affiliation(s)
- Claire M. Bell
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
| | - Donald J. Zack
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Cynthia A. Berlinicke
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
77
|
Singh RK, Nasonkin IO. Limitations and Promise of Retinal Tissue From Human Pluripotent Stem Cells for Developing Therapies of Blindness. Front Cell Neurosci 2020; 14:179. [PMID: 33132839 PMCID: PMC7513806 DOI: 10.3389/fncel.2020.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022] Open
Abstract
The self-formation of retinal tissue from pluripotent stem cells generated a tremendous promise for developing new therapies of retinal degenerative diseases, which previously seemed unattainable. Together with use of induced pluripotent stem cells or/and CRISPR-based recombineering the retinal organoid technology provided an avenue for developing models of human retinal degenerative diseases "in a dish" for studying the pathology, delineating the mechanisms and also establishing a platform for large-scale drug screening. At the same time, retinal organoids, highly resembling developing human fetal retinal tissue, are viewed as source of multipotential retinal progenitors, young photoreceptors and just the whole retinal tissue, which may be transplanted into the subretinal space with a goal of replacing patient's degenerated retina with a new retinal "patch." Both approaches (transplantation and modeling/drug screening) were projected when Yoshiki Sasai demonstrated the feasibility of deriving mammalian retinal tissue from pluripotent stem cells, and generated a lot of excitement. With further work and testing of both approaches in vitro and in vivo, a major implicit limitation has become apparent pretty quickly: the absence of the uniform layer of Retinal Pigment Epithelium (RPE) cells, which is normally present in mammalian retina, surrounds photoreceptor layer and develops and matures first. The RPE layer polarize into apical and basal sides during development and establish microvilli on the apical side, interacting with photoreceptors, nurturing photoreceptor outer segments and participating in the visual cycle by recycling 11-trans retinal (bleached pigment) back to 11-cis retinal. Retinal organoids, however, either do not have RPE layer or carry patches of RPE mostly on one side, thus directly exposing most photoreceptors in the developing organoids to neural medium. Recreation of the critical retinal niche between the apical RPE and photoreceptors, where many retinal disease mechanisms originate, is so far unattainable, imposes clear limitations on both modeling/drug screening and transplantation approaches and is a focus of investigation in many labs. Here we dissect different retinal degenerative diseases and analyze how and where retinal organoid technology can contribute the most to developing therapies even with a current limitation and absence of long and functional outer segments, supported by RPE.
Collapse
|
78
|
Lin B, McLelland BT, Aramant RB, Thomas BB, Nistor G, Keirstead HS, Seiler MJ. Retina Organoid Transplants Develop Photoreceptors and Improve Visual Function in RCS Rats With RPE Dysfunction. Invest Ophthalmol Vis Sci 2020; 61:34. [PMID: 32945842 PMCID: PMC7509771 DOI: 10.1167/iovs.61.11.34] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 08/17/2020] [Indexed: 01/01/2023] Open
Abstract
Purpose To study if human embryonic stem cell-derived photoreceptors could survive and function without the support of retinal pigment epithelium (RPE) after transplantation into Royal College of Surgeons rats, a rat model of retinal degeneration caused by RPE dysfunction. Methods CSC14 human embryonic stem cells were differentiated into primordial eye structures called retinal organoids. Retinal organoids were analyzed by quantitative PCR and immunofluorescence and compared with human fetal retina. Retinal organoid sheets (30-70 day of differentiation) were transplanted into immunodeficient RCS rats, aged 44 to 56 days. The development of transplant organoids in vivo in relation to the host was examined by optical coherence tomography. Visual function was assessed by optokinetic testing, electroretinogram, and superior colliculus electrophysiologic recording. Cryostat sections were analyzed for various retinal, synaptic, and donor markers. Results Retinal organoids showed similar gene expression to human fetal retina transplanted rats demonstrated significant improvement in visual function compared with RCS nonsurgery and sham surgery controls by ERGs at 2 months after surgery (but not later), optokinetic testing (up to 6 months after surgery) and electrophysiologic superior colliculus recordings (6-8 months after surgery). The transplanted organoids survived more than 7 months; developed photoreceptors with inner and outer segments, and other retinal cells; and were well-integrated within the host. Conclusions This study, to our knowledge, is the first to show that transplanted photoreceptors survive and function even with host's dysfunctional RPE. Our findings suggest that transplantation of organoid sheets from stem cells may be a promising approach/therapeutic for blinding diseases.
Collapse
Affiliation(s)
- Bin Lin
- Physical Medicine & Rehabilitation, Sue & Bill Gross Stem Cell Research Center, University of California at Irvine, School of Medicine, Irvine, California, United States
| | - Bryce T. McLelland
- Physical Medicine & Rehabilitation, Sue & Bill Gross Stem Cell Research Center, University of California at Irvine, School of Medicine, Irvine, California, United States
| | - Robert B. Aramant
- Physical Medicine & Rehabilitation, Sue & Bill Gross Stem Cell Research Center, University of California at Irvine, School of Medicine, Irvine, California, United States
| | - Biju B. Thomas
- USC Roski Eye Institute, Department of Ophthalmology, University of Southern California, Los Angeles, California, United States
| | - Gabriel Nistor
- AIVITA Biomedical Inc., Irvine, California, United States
| | | | - Magdalene J. Seiler
- Physical Medicine & Rehabilitation, Sue & Bill Gross Stem Cell Research Center, University of California at Irvine, School of Medicine, Irvine, California, United States
- Ophthalmology, University of California at Irvine, School of Medicine, Irvine, California, United States
- Anatomy & Neurobiology, University of California at Irvine School of Medicine, Irvine, California, United States
| |
Collapse
|
79
|
West EL, Ribeiro J, Ali RR. Development of Stem Cell Therapies for Retinal Degeneration. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a035683. [PMID: 31818854 DOI: 10.1101/cshperspect.a035683] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Degenerative retinal disease is the major cause of sight loss in the developed world and currently there is a lack of effective treatments. As the loss of vision is directly the result of the loss of retinal cells, effective cell replacement through stem-cell-based therapies may have the potential to treat a great number of retinal diseases whatever their underlying etiology. The eye is an ideal organ to develop cell therapies as it is immune privileged, and modern surgical techniques enable precise delivery of cells to the retina. Furthermore, a range of noninvasive diagnostic tests and high-resolution imaging techniques facilitate the evaluation of any therapeutic intervention. In this review, we evaluate the progress to date of current cell therapy strategies for retinal repair, focusing on transplantation of pluripotent stem-cell-derived retinal pigment epithelium (RPE) and photoreceptor cells.
Collapse
Affiliation(s)
- Emma L West
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Joana Ribeiro
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom
| | - Robin R Ali
- Division of Molecular Therapy, UCL Institute of Ophthalmology, London EC1V 9EL, United Kingdom.,Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan 48105, USA
| |
Collapse
|
80
|
Morizur L, Herardot E, Monville C, Ben M'Barek K. Human pluripotent stem cells: A toolbox to understand and treat retinal degeneration. Mol Cell Neurosci 2020; 107:103523. [PMID: 32634576 DOI: 10.1016/j.mcn.2020.103523] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 05/24/2020] [Accepted: 06/30/2020] [Indexed: 12/13/2022] Open
Abstract
Age-related Macular Degeneration (AMD) and Retinitis Pigmentosa (RP) are retinal degenerative disorders that dramatically damage the retina. As there is no therapeutic option for the majority of patients, vision is progressively and irremediably lost. Owing to their unlimited renewal and potency to give rise to any cell type of the human adult body, human pluripotent stem cells (hPSCs) have been extensively studied in recent years to develop more physiologically relevant in vitro cellular models. Such models open new perspectives to investigate the pathological molecular mechanisms of AMD and RP but also in drug screening. Moreover, proof-of-concept of hPSC-derived retinal cell therapy in animal models have led to first clinical trials. This review outlines the recent advances in the use of hPSCs in pathological modeling of retinal degeneration and their use in regenerative medicine. We also address the associated limitations and challenges that need to be overcome when using hPSCs.
Collapse
Affiliation(s)
- Lise Morizur
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100 Corbeil-Essonnes, France; Université Paris-Saclay, Université d'Evry, U861, 91100 Corbeil-Essonnes, France; Centre d'Etude des Cellules Souches, 91100 Corbeil-Essonnes, France
| | - Elise Herardot
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100 Corbeil-Essonnes, France; Université Paris-Saclay, Université d'Evry, U861, 91100 Corbeil-Essonnes, France
| | - Christelle Monville
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100 Corbeil-Essonnes, France; Université Paris-Saclay, Université d'Evry, U861, 91100 Corbeil-Essonnes, France.
| | - Karim Ben M'Barek
- INSERM U861, I-Stem, AFM, Institute for Stem Cell Therapy and Exploration of Monogenic Diseases, 91100 Corbeil-Essonnes, France; Université Paris-Saclay, Université d'Evry, U861, 91100 Corbeil-Essonnes, France; Centre d'Etude des Cellules Souches, 91100 Corbeil-Essonnes, France.
| |
Collapse
|
81
|
Goureau O, Orieux G. [Photoreceptor cell transplantation for future treatment of retinitis pigmentosa]. Med Sci (Paris) 2020; 36:600-606. [PMID: 32614311 DOI: 10.1051/medsci/2020097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In inherited retinal diseases such retinitis pigmentosa, characterized by progressive loss of light sensitive neurons (photoreceptors), cell therapy is now considered as an attractive strategy. Photoreceptor cell replacement would be valuable for restoring function to retinas in a way that is independent from the cause of the disease. With advances in stem cell biology, considerable strides have been made towards the generation of retinal cells, in particular with the development of 3D culture systems allowing the generation of retinal organoids from pluripotent stem cells. In this review, we present a state-of-the art of preclinical strategies conducted in animal models for photoreceptor replacement from stem cell-derived photoreceptors and we discuss the important obstacles to overcome in the future.
Collapse
Affiliation(s)
- Olivier Goureau
- Institut de la Vision, Sorbonne Université, Inserm, CNRS, 17 rue Moreau, F-75012 Paris, France
| | - Gaël Orieux
- Institut de la Vision, Sorbonne Université, Inserm, CNRS, 17 rue Moreau, F-75012 Paris, France
| |
Collapse
|
82
|
Fligor CM, Huang KC, Lavekar SS, VanderWall KB, Meyer JS. Differentiation of retinal organoids from human pluripotent stem cells. Methods Cell Biol 2020; 159:279-302. [PMID: 32586447 DOI: 10.1016/bs.mcb.2020.02.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Human pluripotent stem cells (hPSCs) possess the remarkable ability to differentiate into any cell type of the body, including those of the retina. Through the differentiation of these cells as retinal organoids, it is now possible to model the spatial and temporal development of the human retina using hPSCs, in which retinal progenitor cells produce the entire repertoire of retinal cells, first differentiating into retinal ganglion cells and ending with mature photoreceptors, bipolar cells, and Müller glia. Importantly, retinal organoids self-assemble into laminated structures that recapitulate the layering of the human retina with a retinal ganglion cell layer lining the inner layer and a distinctly separate photoreceptor layer occupying the outer layers. This organoid technology has provided access to human tissue for developmental and disease modeling, as well as translational applications such as high throughput drug screening and cell replacement therapies. However, the differentiation of retinal organoids does require some expertise and multiple strategies produce inconsistent results. Here, we describe in detail a well-established and relatively simple method for the generation of retinal organoids.
Collapse
Affiliation(s)
- Clarisse M Fligor
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kang-Chieh Huang
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Sailee S Lavekar
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Kirstin B VanderWall
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Jason S Meyer
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Ophthalmology, Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
83
|
Chen F, Liu X, Chen Y, Liu JY, Lu H, Wang W, Lu X, Dean KC, Gao L, Kaplan HJ, Dean DC, Peng X, Liu Y. Sphere-induced reprogramming of RPE cells into dual-potential RPE stem-like cells. EBioMedicine 2020; 52:102618. [PMID: 31982829 PMCID: PMC6994567 DOI: 10.1016/j.ebiom.2019.102618] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/20/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022] Open
Abstract
Background The retinal pigment epithelium (RPE) has the potential to regenerate the entire neuroretina upon retinal injury in amphibians. In contrast, this regenerative capacity has been lost in mammals. The reprogramming of differentiated somatic cells into induced pluripotent stem cells (iPSCs) by viral transduction of exogenous stem cell factors has triggered a revolution in regenerative medicine. However, the risks of potential mutation(s) caused by random viral vector insertion in host genomes and tumor formation in recipients hamper its clinical application. One alternative is to immortalize adult stem cells with limited potential or to partially reprogram differentiated somatic cells into progenitor-like cells through non-integration protocols. Methods Sphere-induced RPE stem cells (iRPESCs) were generated from adult mouse RPE cells. Their stem cell functionality was studied in a mouse model of retinal degeneration. The molecular mechanism underlying the sphere-induced reprogramming was investigated using microarray and loss-of-function approaches. Findings We provide evidence that our sphere-induced reprogramming protocol can immortalize and transform mouse RPE cells into iRPESCs with dual potential to differentiate into cells that express either RPE or photoreceptor markers both in vitro and in vivo. When subretinally transplanted into mice with retinal degeneration, iRPESCs can integrate to the RPE and neuroretina, thereby delaying retinal degeneration in the model animals. Our molecular analyses indicate that the Hippo signaling pathway is important in iRPESC reprogramming. Interpretation The Hippo factor Yap1 is activated in the nuclei of cells at the borders of spheres. The factors Zeb1 and P300 downstream of the Hippo pathway are shown to bind to the promoters of the stemness genes Oct4, Klf4 and Sox2, thereby likely transactivate them to reprogram RPE cells into iRPESCs. Fund National Natural Science Foundation of China and the National Institute of Health USA.
Collapse
Affiliation(s)
- Fenghua Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Xiao Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yao Chen
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China
| | - John Y Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Huayi Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Wei Wang
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Xiaoqin Lu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Kevin C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Ling Gao
- Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Henry J Kaplan
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | - Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China.
| | - Yongqing Liu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, 301 E Muhammad Ali Blvd, Louisville, Kentucky 40202, USA; James Graham Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA; Birth Defects Center; University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| |
Collapse
|
84
|
Quinn PM, Wijnholds J. Retinogenesis of the Human Fetal Retina: An Apical Polarity Perspective. Genes (Basel) 2019; 10:E987. [PMID: 31795518 PMCID: PMC6947654 DOI: 10.3390/genes10120987] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022] Open
Abstract
The Crumbs complex has prominent roles in the control of apical cell polarity, in the coupling of cell density sensing to downstream cell signaling pathways, and in regulating junctional structures and cell adhesion. The Crumbs complex acts as a conductor orchestrating multiple downstream signaling pathways in epithelial and neuronal tissue development. These pathways lead to the regulation of cell size, cell fate, cell self-renewal, proliferation, differentiation, migration, mitosis, and apoptosis. In retinogenesis, these are all pivotal processes with important roles for the Crumbs complex to maintain proper spatiotemporal cell processes. Loss of Crumbs function in the retina results in loss of the stratified appearance resulting in retinal degeneration and loss of visual function. In this review, we begin by discussing the physiology of vision. We continue by outlining the processes of retinogenesis and how well this is recapitulated between the human fetal retina and human embryonic stem cell (ESC) or induced pluripotent stem cell (iPSC)-derived retinal organoids. Additionally, we discuss the functionality of in utero and preterm human fetal retina and the current level of functionality as detected in human stem cell-derived organoids. We discuss the roles of apical-basal cell polarity in retinogenesis with a focus on Leber congenital amaurosis which leads to blindness shortly after birth. Finally, we discuss Crumbs homolog (CRB)-based gene augmentation.
Collapse
Affiliation(s)
- Peter M.J. Quinn
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
85
|
Artero Castro A, Rodríguez Jimenez FJ, Jendelova P, Erceg S. Deciphering retinal diseases through the generation of three dimensional stem cell-derived organoids: Concise Review. Stem Cells 2019; 37:1496-1504. [PMID: 31617949 PMCID: PMC6915910 DOI: 10.1002/stem.3089] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 08/08/2019] [Indexed: 01/09/2023]
Abstract
Three‐dimensional (3D) retinal organoids, in vitro tissue structures derived from self‐organizing cultures of differentiating human embryonic stem cells or induced pluripotent stem cells, could recapitulate some aspects of the cytoarchitectural structure and function of the retina in vivo. 3D retinal organoids display huge potential for the investigation of the pathogenesis of monogenic hereditary eye diseases that are related to the malfunction or degeneration of photoreceptors or retinal ganglion cells by providing an effective in vitro tool with multiple applications. In combination with recent genome editing tools, 3D retinal organoids could also represent a reliable and renewable source of transplantable cells for personalized therapies. In this review, we describe the recent advances in human pluripotent stem cells‐derived retinal organoids, determination of their histoarchitecture, complexity, and maturity. We also discuss their application as a means to decipher the pathogenesis of retinal diseases, as well as the main drawbacks and challenges. stem cells2019;37:1496–1504
Collapse
Affiliation(s)
- Ana Artero Castro
- Stem Cells Therapies in Neurodegenerative Diseases Lab, Centro de Investigación Principe Felipe (CIPF), Valencia, Spain
| | | | - Pavla Jendelova
- Department of Tissue Cultures and Stem Cells, Czech Academy of Science, Institute of Experimental Medicine, Prague, Czech Republic
| | - Slaven Erceg
- Stem Cells Therapies in Neurodegenerative Diseases Lab, Centro de Investigación Principe Felipe (CIPF), Valencia, Spain.,Department of Tissue Cultures and Stem Cells, Czech Academy of Science, Institute of Experimental Medicine, Prague, Czech Republic.,National Stem Cell Bank-Valencia Node, Proteomics, Genotyping and Cell Line Platform, PRB3, ISCIII, Research Centre Principe Felipe, Valencia, Spain
| |
Collapse
|
86
|
Gamm DM, Clark E, Capowski EE, Singh R. The Role of FGF9 in the Production of Neural Retina and RPE in a Pluripotent Stem Cell Model of Early Human Retinal Development. Am J Ophthalmol 2019; 206:113-131. [PMID: 31078532 DOI: 10.1016/j.ajo.2019.04.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 04/11/2019] [Accepted: 04/18/2019] [Indexed: 12/20/2022]
Abstract
PURPOSE To investigate the role of fibroblast growth factors (FGFs) in the production of neural retina (NR) and retinal pigmented epithelium (RPE) in a human pluripotent stem cell model of early retinal development. METHODS Human induced pluripotent stem cell (hiPSC) lines from an individual with microphthalmia caused by a functional null mutation (R200Q) in visual system homeobox 2 (VSX2), a transcription factor involved in early NR progenitor cell (NRPC) production, and a normal sibling were differentiated along the retinal and forebrain lineages using an established protocol. Quantitative and global gene expression analyses (microarray and RNAseq) were used to investigate endogenous FGF expression profiles in these cultures over time. Based on these results, mutant and control hiPSC cultures were treated exogenously with selected FGFs and subjected to gene and protein expression analyses to determine their effects on RPE and NR production. RESULTS We found that FGF9 and FGF19 were selectively increased in early hiPSC-derived optic vesicles (OVs) when compared to isogenic cultures of hiPSC-derived forebrain neurospheres. Furthermore, these same FGFs were downregulated over time in (R200Q)VSX2 hiPSC-OVs relative to sibling control hiPSC-OVs. Interestingly, long-term supplementation with FGF9, but not FGF19, partially rescued the mutant retinal phenotype of the (R200Q)VSX2 hiPSC-OV model. However, antagonizing FGF9 in wild-type control hiPSCs did not alter OV development. CONCLUSIONS Our results show that FGF9 acts in concert with VSX2 to promote NR differentiation in hiPSC-OVs and has potential to be used to manipulate early retinogenesis and mitigate ocular defects caused by functional loss of VSX2 activity. NOTE: Publication of this article is sponsored by the American Ophthalmological Society.
Collapse
Affiliation(s)
- David M Gamm
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA; Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| | - Eric Clark
- Department of Cell Biology, Neurobiology, & Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | | | - Ruchira Singh
- Department of Ophthalmology, University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
87
|
Procyk CA, Allen AE, Martial FP, Lucas RJ. Visual responses in the dorsal lateral geniculate nucleus at early stages of retinal degeneration in rd1 PDE6β mice. J Neurophysiol 2019; 122:1753-1764. [PMID: 31461375 DOI: 10.1152/jn.00231.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Inherited retinal degenerations encompass a wide range of diseases that result in the death of rod and cone photoreceptors, eventually leading to irreversible blindness. Low vision survives at early stages of degeneration, at which point it could rely on residual populations of rod/cone photoreceptors as well as the inner retinal photoreceptor, melanopsin. To date, the impact of partial retinal degeneration on visual responses in the primary visual thalamus (dorsal lateral geniculate nucleus, dLGN) remains unknown, as does their relative reliance on surviving rod and cone photoreceptors vs. melanopsin. To answer these questions, we recorded visually evoked responses in the dLGN of anesthetized rd1 mice using in vivo electrophysiology at an age (3-5 wk) at which cones are partially degenerate and rods are absent. We found that excitatory (ON) responses to light had lower amplitude and longer latency in rd1 mice compared with age-matched visually intact controls; however, contrast sensitivity and spatial receptive field size were largely unaffected at this early stage of degeneration. Responses were retained when those wavelengths to which melanopsin is most sensitive were depleted, indicating that they were driven primarily by surviving cones. Inhibitory responses appeared absent in the rd1 thalamus, as did light-evoked gamma oscillations in firing. This description of fundamental features of the dLGN visual response at this intermediate stage of retinal degeneration provides a context for emerging attempts to restore vision by introducing ectopic photoreception to the degenerate retina.NEW & NOTEWORTHY This study provides new therapeutically relevant insights to visual responses in the dorsal lateral geniculate nucleus during progressive retinal degeneration. Using in vivo electrophysiology, we demonstrate that visual responses have lower amplitude and longer latency during degeneration, but contrast sensitivity and spatial receptive fields remain unaffected. Such visual responses are driven predominantly by surviving cones rather than melanopsin photoreceptors. The functional integrity of this visual pathway is encouraging for emerging attempts at visual restoration.
Collapse
Affiliation(s)
- Christopher A Procyk
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Annette E Allen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Franck P Martial
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Robert J Lucas
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
88
|
Singh RK, Occelli LM, Binette F, Petersen-Jones SM, Nasonkin IO. Transplantation of Human Embryonic Stem Cell-Derived Retinal Tissue in the Subretinal Space of the Cat Eye. Stem Cells Dev 2019; 28:1151-1166. [PMID: 31210100 PMCID: PMC6708274 DOI: 10.1089/scd.2019.0090] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
To develop biological approaches to restore vision, we developed a method of transplanting stem cell-derived retinal tissue into the subretinal space of a large-eye animal model (cat). Human embryonic stem cells (hESC) were differentiated to retinal organoids in a dish. hESC-derived retinal tissue was introduced into the subretinal space of wild-type cats following a pars plana vitrectomy. The cats were systemically immunosuppressed with either prednisolone or prednisolone plus cyclosporine A. The eyes were examined by fundoscopy and spectral-domain optical coherence tomography imaging for adverse effects due to the presence of the subretinal grafts. Immunohistochemistry was done with antibodies to retinal and human markers to delineate graft survival, differentiation, and integration into cat retina. We successfully delivered hESC-derived retinal tissue into the subretinal space of the cat eye. We observed strong infiltration of immune cells in the graft and surrounding tissue in the cats treated with prednisolone. In contrast, we showed better survival and low immune response to the graft in cats treated with prednisolone plus cyclosporine A. Immunohistochemistry with antibodies (STEM121, CALB2, DCX, and SMI-312) revealed large number of graft-derived fibers connecting the graft and the host. We also show presence of human-specific synaptophysin puncta in the cat retina. This work demonstrates feasibility of engrafting hESC-derived retinal tissue into the subretinal space of large-eye animal models. Transplanting retinal tissue in degenerating cat retina will enable rapid development of preclinical in vivo work focused on vision restoration.
Collapse
Affiliation(s)
- Ratnesh K Singh
- Lineage Cell Therapeutics, Inc. (formerly BioTime Inc.), Carlsbad, California
| | - Laurence M Occelli
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lasing, Michigan
| | - Francois Binette
- Lineage Cell Therapeutics, Inc. (formerly BioTime Inc.), Carlsbad, California
| | - Simon M Petersen-Jones
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lasing, Michigan
| | - Igor O Nasonkin
- Lineage Cell Therapeutics, Inc. (formerly BioTime Inc.), Carlsbad, California
| |
Collapse
|
89
|
Photoreceptor cell replacement in macular degeneration and retinitis pigmentosa: A pluripotent stem cell-based approach. Prog Retin Eye Res 2019; 71:1-25. [DOI: 10.1016/j.preteyeres.2019.03.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023]
|
90
|
Lorach H, Kang S, Bhuckory MB, Trouillet A, Dalal R, Marmor M, Palanker D. Transplantation of Mature Photoreceptors in Rodents With Retinal Degeneration. Transl Vis Sci Technol 2019; 8:30. [PMID: 31171997 PMCID: PMC6543858 DOI: 10.1167/tvst.8.3.30] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 04/01/2019] [Indexed: 11/24/2022] Open
Abstract
Purpose To demonstrate survival and integration of mature photoreceptors transplanted with the retinal pigment epithelium (RPE). Methods Full-thickness retina with attached RPE was harvested from healthy adult rats. Grafts were implanted into two rat models of retinal degeneration, Royal College of Surgeons (RCS) and S334ter-3. Survival of the host and transplanted retina was monitored using optical coherence tomography (OCT) for up to 6 months. The retinal structure and synaptogenesis between the host and transplant was assessed by histology and immunohistochemistry. Results OCT and histology demonstrated a well-preserved photoreceptor layer with inner and outer segments, while the inner retinal layers of the transplant largely disappeared. Grafts, including RPE, survived better than without and the transplanted RPE appeared as a monolayer integrated with the native one. Synaptogenesis was observed through sprouting of new dendrites from the host bipolar cells and synaptic connections forming with cells of the transplant. However, in many samples, a glial fibrillary acidic protein–positive membrane separated the host retina and the graft. Conclusions Presence of RPE in the graft improved the survival of transplanted photoreceptors. Functional integration between the transplant and the host retina is likely to be further enhanced if formation of a glial seal could be prevented. Transplantation of the mature photoreceptors with RPE may be a practical approach to restoration of sight in retinal degeneration. Translational Relevance This approach to restoration of sight in patients with photoreceptor degeneration can be rapidly advanced to clinical testing. In patients with central scotoma, autologous transplantation of the peripheral retina can be an option.
Collapse
Affiliation(s)
- Henri Lorach
- Hansen Experimental Physics Laboratory, Stanford University, CA, USA
| | - Seungbum Kang
- Hansen Experimental Physics Laboratory, Stanford University, CA, USA.,Department of Ophthalmology and Visual Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mohajeet B Bhuckory
- Hansen Experimental Physics Laboratory, Stanford University, CA, USA.,Department of Ophthalmology, Stanford University, CA, USA
| | - Alix Trouillet
- Department of Otolaryngology, Stanford University, CA, USA
| | - Roopa Dalal
- Department of Ophthalmology, Stanford University, CA, USA
| | - Michael Marmor
- Department of Ophthalmology, Stanford University, CA, USA
| | - Daniel Palanker
- Hansen Experimental Physics Laboratory, Stanford University, CA, USA.,Department of Ophthalmology, Stanford University, CA, USA
| |
Collapse
|
91
|
Pluripotent Stem Cells as Models of Retina Development. Mol Neurobiol 2019; 56:6056-6070. [DOI: 10.1007/s12035-019-1504-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/21/2019] [Indexed: 01/01/2023]
|
92
|
Léveillard T, Klipfel L. Mechanisms Underlying the Visual Benefit of Cell Transplantation for the Treatment of Retinal Degenerations. Int J Mol Sci 2019; 20:ijms20030557. [PMID: 30696106 PMCID: PMC6387096 DOI: 10.3390/ijms20030557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 12/13/2022] Open
Abstract
The transplantation of retinal cells has been studied in animals to establish proof of its potential benefit for the treatment of blinding diseases. Photoreceptor precursors have been grafted in animal models of Mendelian-inherited retinal degenerations, and retinal pigmented epithelial cells have been used to restore visual function in animal models of age-related macular degeneration (AMD) and recently in patients. Cell therapy over corrective gene therapy in inherited retinal degeneration can overcome the genetic heterogeneity by providing one treatment for all genetic forms of the diseases. In AMD, the existence of multiple risk alleles precludes a priori the use of corrective gene therapy. Mechanistically, the experiments of photoreceptor precursor transplantation reveal the importance of cytoplasmic material exchange between the grafted cells and the host cells for functional rescue, an unsuspected mechanism and novel concept. For transplantation of retinal pigmented epithelial cells, the mechanisms behind the therapeutic benefit are only partially understood, and clinical trials are ongoing. The fascinating studies that describe the development of methodologies to produce cells to be grafted and demonstrate the functional benefit for vision are reviewed.
Collapse
Affiliation(s)
- Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Laurence Klipfel
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
93
|
Yanai A, McNab P, Gregory-Evans K. Retinal therapy with induced pluripotent stem cells; leading the way to human clinical trials. EXPERT REVIEW OF OPHTHALMOLOGY 2019. [DOI: 10.1080/17469899.2019.1568872] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Anat Yanai
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Pia McNab
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Gregory-Evans
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
94
|
Capowski EE, Samimi K, Mayerl SJ, Phillips MJ, Pinilla I, Howden SE, Saha J, Jansen AD, Edwards KL, Jager LD, Barlow K, Valiauga R, Erlichman Z, Hagstrom A, Sinha D, Sluch VM, Chamling X, Zack DJ, Skala MC, Gamm DM. Reproducibility and staging of 3D human retinal organoids across multiple pluripotent stem cell lines. Development 2019; 146:dev171686. [PMID: 30567931 PMCID: PMC6340149 DOI: 10.1242/dev.171686] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022]
Abstract
Numerous protocols have been described for producing neural retina from human pluripotent stem cells (hPSCs), many of which are based on the culture of 3D organoids. Although nearly all such methods yield at least partial segments of retinal structure with a mature appearance, variabilities exist within and between organoids that can change over a protracted time course of differentiation. Adding to this complexity are potential differences in the composition and configuration of retinal organoids when viewed across multiple differentiations and hPSC lines. In an effort to understand better the current capabilities and limitations of these cultures, we generated retinal organoids from 16 hPSC lines and monitored their appearance and structural organization over time by light microscopy, immunocytochemistry, metabolic imaging and electron microscopy. We also employed optical coherence tomography and 3D imaging techniques to assess and compare whole or broad regions of organoids to avoid selection bias. Results from this study led to the development of a practical staging system to reduce inconsistencies in retinal organoid cultures and increase rigor when utilizing them in developmental studies, disease modeling and transplantation.
Collapse
Affiliation(s)
| | - Kayvan Samimi
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Steven J Mayerl
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - M Joseph Phillips
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Isabel Pinilla
- Aragon Institute for Health Research (IIS Aragón), Lozano Blesa University Hospital, Zaragoza 50009, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza 50009, Spain
| | - Sara E Howden
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria 3052, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jishnu Saha
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Alex D Jansen
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Lindsey D Jager
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Katherine Barlow
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Rasa Valiauga
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary Erlichman
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Anna Hagstrom
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Divya Sinha
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Valentin M Sluch
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xitiz Chamling
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Melissa C Skala
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - David M Gamm
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Ophthamology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
95
|
Aberrant hiPSCs-Derived from Human Keratinocytes Differentiates into 3D Retinal Organoids that Acquire Mature Photoreceptors. Cells 2019; 8:cells8010036. [PMID: 30634512 PMCID: PMC6356277 DOI: 10.3390/cells8010036] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/12/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived three-dimensional retinal organoids are a new platform for studying the organoidogenesis. However, recurrent genomic aberration, acquired during generation of hiPSCs, limit its biomedical application and/or aberrant hiPSCs has not been evaluated for generation of differentiated derivatives, such as organoids and retinal pigment epithelium (RPE). In this study, we efficiently differentiated mosaic hiPSCs into retinal organoids containing mature photoreceptors. The feeder-free hiPSCs were generated from the human epidermal keratinocytes that were rapid in process with improved efficiency over several passages and maintained pluripotency. But, hiPSCs were cytogenetically mosaic with normal and abnormal karyotypes, while copy number variation analysis revealed the loss of chromosome 8q. Despite this abnormality, the stepwise differentiation of hiPSCs to form retinal organoids was autonomous and led to neuronal lamination. Furthermore, the use of a Notch inhibitor, DAPT, at an early timepoint from days 29⁻42 of culture improved the specification of the retinal neuron and the use of retinoic acid at days 70⁻120 led to the maturation of photoreceptors. hiPSC-derived retinal organoids acquired all subtypes of photoreceptors, such as RHODOPSIN, B-OPSIN and R/G-OPSIN. Additionally, the advanced maturation of photoreceptors was observed, revealing the development of specific sensory cilia and the formation of the outer-segment disc. This report is the first to show that hiPSCs with abnormal chromosomal content are permissive to the generation of three-dimensional retinal organoids.
Collapse
|
96
|
Gasparini SJ, Llonch S, Borsch O, Ader M. Transplantation of photoreceptors into the degenerative retina: Current state and future perspectives. Prog Retin Eye Res 2018; 69:1-37. [PMID: 30445193 DOI: 10.1016/j.preteyeres.2018.11.001] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/29/2018] [Accepted: 11/06/2018] [Indexed: 12/12/2022]
Abstract
The mammalian retina displays no intrinsic regenerative capacities, therefore retinal degenerative diseases such as age-related macular degeneration (AMD) or retinitis pigmentosa (RP) result in a permanent loss of the light-sensing photoreceptor cells. The degeneration of photoreceptors leads to vision impairment and, in later stages, complete blindness. Several therapeutic strategies have been developed to slow down or prevent further retinal degeneration, however a definitive cure i.e. replacement of the lost photoreceptors, has not yet been established. Cell-based treatment approaches, by means of photoreceptor transplantation, have been studied in pre-clinical animal models over the last three decades. The introduction of pluripotent stem cell-derived retinal organoids represents, in principle, an unlimited source for the generation of transplantable human photoreceptors. However, safety, immunological and reproducibility-related issues regarding the use of such cells still need to be solved. Moreover, the recent finding of cytoplasmic material transfer between donor and host photoreceptors demands reinterpretation of several former transplantation studies. At the same time, material transfer between healthy donor and dysfunctional patient photoreceptors also offers a potential alternative strategy for therapeutic intervention. In this review we discuss the history and current state of photoreceptor transplantation, the techniques used to assess rescue of visual function, the prerequisites for effective transplantation as well as the main roadblocks, including safety and immune response to the graft, that need to be overcome for successful clinical translation of photoreceptor transplantation approaches.
Collapse
Affiliation(s)
- Sylvia J Gasparini
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Sílvia Llonch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Oliver Borsch
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstraße 105, 01307, Dresden, Germany.
| |
Collapse
|
97
|
Lin B, McLelland BT, Mathur A, Aramant RB, Seiler MJ. Sheets of human retinal progenitor transplants improve vision in rats with severe retinal degeneration. Exp Eye Res 2018; 174:13-28. [PMID: 29782826 DOI: 10.1016/j.exer.2018.05.017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 05/13/2018] [Accepted: 05/17/2018] [Indexed: 01/22/2023]
Abstract
Loss of photoreceptors and other retinal cells is a common endpoint in retinal degenerate (RD) diseases that cause blindness. Retinal transplantation is a potential therapy to replace damaged retinal cells and improve vision. In this study, we examined the development of human fetal retinal sheets with or without their retinal pigment epithelium (RPE) transplanted to immunodeficient retinal degenerate rho S334ter-3 rats. Sheets were dissected from fetal human eyes (11-15.7 weeks gestation) and then transplanted to the subretinal space of 24-31 d old RD nude rats. Every month post surgery, eyes were imaged by high-resolution spectral-domain optical coherence tomography (SD-OCT). SD-OCT showed that transplants were placed into the subretinal space and developed laminated areas or rosettes, with clear development of plexiform layers first seen in OCT at 3 months post surgery. Several months later, as could be expected by the much slower development of human cells compared to rat cells, transplant photoreceptors developed inner and later outer segments. Retinal sections were analyzed by immunohistochemistry for human and retinal markers and confirmed the formation of several retinal subtypes within the retinal layers. Transplant cells extended processes and a lot of the cells could also be seen migrating into the host retina. At 5.8-8.6 months post surgery, selected rats were exposed to light flashes and recorded for visual responses in superior colliculus, (visual center in midbrain). Four of seven rats with transplants showed responses to flashes of light in a limited area of superior colliculus. No response with the same dim light intensity was found in age-matched RD controls (non-surgery or sham surgery). In summary, our data showed that human fetal retinal sheets transplanted to the severely disturbed subretinal space of RD nude rats develop mature photoreceptors and other retinal cells, integrate with the host and induce vision improvement.
Collapse
Affiliation(s)
- Bin Lin
- Stem Cell Research Center, University of CalifoArnia, Irvine, United States
| | - Bryce T McLelland
- Stem Cell Research Center, University of CalifoArnia, Irvine, United States
| | - Anuradha Mathur
- Stem Cell Research Center, University of CalifoArnia, Irvine, United States
| | - Robert B Aramant
- Stem Cell Research Center, University of CalifoArnia, Irvine, United States
| | - Magdalene J Seiler
- Stem Cell Research Center, University of CalifoArnia, Irvine, United States; Department of Physical Medicine & Rehabilitation, University of California, Irvine, United States.
| |
Collapse
|