1
|
Lee YJ, Jo DH. Retinal Organoids from Induced Pluripotent Stem Cells of Patients with Inherited Retinal Diseases: A Systematic Review. Stem Cell Rev Rep 2025; 21:167-197. [PMID: 39422807 PMCID: PMC11762450 DOI: 10.1007/s12015-024-10802-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Currently, most inherited retinal diseases lack curative interventions, and available treatment modalities are constrained to symptomatic approaches. Retinal organoid technology has emerged as a method for treating inherited retinal diseases, with growing academic interest in recent years. The purpose of this review was to systematically organize the current protocols for generating retinal organoids using induced pluripotent stem cells from patients with inherited retinal disease and to investigate the application of retinal organoids in inherited retinal disease research. METHODS Data were collected from the PubMed, Scopus, and Web of Science databases using a keyword search. The main search term used was "retinal organoid," accompanied by secondary keywords such as "optic cup," "three-dimensional," and "self-organizing." The final search was conducted on October 2, 2024. RESULTS Of the 2,129 studies retrieved, 130 were included in the qualitative synthesis. The protocols for the generation of retinal organoids in inherited retinal disease research use five major approaches, categorized into 3D and a combination of 2D/3D approaches, implemented with modifications. Disease phenotypes have been successfully reproduced via the generation of retinal organoids from the induced pluripotent stem cells of individuals with inherited retinal diseases, facilitating the progression of research into novel therapeutic developments. Cells have been obtained from retinal organoids for cell therapy, and progress toward their potential integration into clinical practice is underway. Considering their potential applications, retinal organoid technology has shown promise across various domains. CONCLUSION In this systematic review, we organized protocols for generating retinal organoids using induced pluripotent stem cells from patients with inherited retinal diseases. Retinal organoid technology has various applications including disease modeling, screening for novel therapies, and cell replacement therapy. Further advancements would make this technology a clinically significant tool for patients with inherited retinal diseases.
Collapse
Affiliation(s)
- Yoo Jin Lee
- Department of Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong Hyun Jo
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
2
|
Cerna-Chavez R, Ortega-Gasco A, Baig HMA, Ehrenreich N, Metais T, Scandura MJ, Bujakowska K, Pierce EA, Garita-Hernandez M. Optimized Prime Editing of Human Induced Pluripotent Stem Cells to Efficiently Generate Isogenic Models of Mendelian Diseases. Int J Mol Sci 2024; 26:114. [PMID: 39795970 PMCID: PMC11719581 DOI: 10.3390/ijms26010114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Prime editing (PE) is a CRISPR-based tool for genome engineering that can be applied to generate human induced pluripotent stem cell (hiPSC)-based disease models. PE technology safely introduces point mutations, small insertions, and deletions (indels) into the genome. It uses a Cas9-nickase (nCas9) fused to a reverse transcriptase (RT) as an editor and a PE guide RNA (pegRNA), which introduces the desired edit with great precision without creating double-strand breaks (DSBs). PE leads to minimal off-targets or indels when introducing single-strand breaks (SSB) in the DNA. Low efficiency can be an obstacle to its use in hiPSCs, especially when the genetic context precludes the screening of multiple pegRNAs, and other strategies must be employed to achieve the desired edit. We developed a PE platform to efficiently generate isogenic models of Mendelian disorders. We introduced the c.25G>A (p.V9M) mutation in the NMNAT1 gene with over 25% efficiency by optimizing the PE workflow. Using our optimized system, we generated other isogenic models of inherited retinal diseases (IRDs), including the c.1481C>T (p.T494M) mutation in PRPF3 and the c.6926A>C (p.H2309P) mutation in PRPF8. We modified several determinants of the hiPSC PE procedure, such as plasmid concentrations, PE component ratios, and delivery method settings, showing that our improved workflow increased the hiPSC editing efficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Marcela Garita-Hernandez
- Ocular Genomics Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA; (R.C.-C.); (A.O.-G.); (H.M.A.B.); (N.E.); (T.M.); (M.J.S.); (K.B.); (E.A.P.)
| |
Collapse
|
3
|
Rajendran Nair DS, Gupta A, Iseri E, Wei T, Phuong Quach LT, Seiler MJ, Lazzi G, Thomas BB. Extrinsic electric field modulates neuronal development and increases photoreceptor population in retinal organoids. Front Neurosci 2024; 18:1438903. [PMID: 39678532 PMCID: PMC11639233 DOI: 10.3389/fnins.2024.1438903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/24/2024] [Indexed: 12/17/2024] Open
Abstract
Introduction Considering the significant role played by both intrinsic and extrinsic electric fields in the growth and maturation of the central nervous system, the impact of short exposure to external electric fields on the development and differentiation of retinal organoids was investigated. Methods Retinal organoids derived from human embryonic stem cells were used at day 80, a key stage in their differentiation. A single 60-minute exposure to a biphasic electrical field was administered to assess its influence on retinal cell populations and maturation markers. Immunohistochemistry, qPCR, and RNA sequencing were employed to evaluate cell type development and gene expression changes. Results Electrical stimulation significantly enhanced neuronal development and increased the population of photoreceptors within the organoids. RNA sequencing data showed upregulated expression of genes related to rod photoreceptors, Müller cells, horizontal cells, and amacrine cells, while genes associated with retinal pigment epithelium and retinal ganglion cells were downregulated. Variations in development and maturation were observed depending on the specific parameters of the applied electric field. Discussion These findings highlight the significant impact of extrinsic electrical fields on early retinal development and suggest that optimizing electrical field parameters could effectively address certain limitations in retinal organoid technology, potentially reducing the reliance on chemicals and small molecules.
Collapse
Affiliation(s)
- Deepthi S. Rajendran Nair
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Anika Gupta
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Ege Iseri
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Tianyuan Wei
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Le Tam Phuong Quach
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
| | - Magdalene J. Seiler
- Departments of Physical Medicine and Rehabilitation; Ophthalmology; Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Stem Cell Research Center, University of California, Irvine, Irvine, CA, United States
| | - Gianluca Lazzi
- Department of Electrical and Computer Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| | - Biju B. Thomas
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA, United States
- USC Ginsburg Institute for Biomedical Therapeutics, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
4
|
Galgani G, Bray G, Martelli A, Calderone V, Citi V. In Vitro Models of Diabetes: Focus on Diabetic Retinopathy. Cells 2024; 13:1864. [PMID: 39594613 PMCID: PMC11592768 DOI: 10.3390/cells13221864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetic retinopathy is a major eye complication in patients with diabetes mellitus, and it is the leading cause of blindness and visual impairment in the world. Chronic hyperglycemia induces endothelial damage with consequent vascular lesions, resulting in global vasculitis, which affects the small vessels of the retina. These vascular lesions cause ischemic conditions in certain areas of the retina, with a consequent increase in the release of pro-angiogenic mediators. In addition to pharmacological interventions for controlling the blood glycaemic level, the main strategies for treating diabetic retinopathy are the intravitreal injections of drugs, surgical treatments, and vitrectomies. The complexity of diabetic retinopathy is due to its close interactions with different cell types (endothelial cells, astrocytes, and Müller cells). The evaluation of the efficacy of novel pharmacological strategies is mainly performed through in vivo models. However, the use of different animal species leads to heterogenic results and ethical concerns. For these reasons, the development of new and reliable in vitro models, such as cell co-cultures and eye organoids, represents an urgent need in this area of research. This review features an overview of the in vitro models used to date and highlights the advances in technology used to study this pathology.
Collapse
Affiliation(s)
- Giulia Galgani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Giorgia Bray
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (G.G.); (A.M.); (V.C.); (V.C.)
- Interuniversity Centre for the Promotion of the 3R Principles in Teaching and Research, Italy
| |
Collapse
|
5
|
Matsushita T, Onishi A, Matsuyama T, Masuda T, Ogino Y, Kageyama M, Takahashi M, Uchiumi F. Rapid and efficient generation of mature retinal organoids derived from human pluripotent stem cells via optimized pharmacological modulation of Sonic hedgehog, activin A, and retinoic acid signal transduction. PLoS One 2024; 19:e0308743. [PMID: 39121095 PMCID: PMC11315325 DOI: 10.1371/journal.pone.0308743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/29/2024] [Indexed: 08/11/2024] Open
Abstract
Human retinal organoids have become indispensable tools for retinal disease modeling and drug screening. Despite its versatile applications, the long timeframe for their differentiation and maturation limits the throughput of such research. Here, we successfully shortened this timeframe by accelerating human retinal organoid development using unique pharmacological approaches. Our method comprised three key steps: 1) a modified self-formed ectodermal autonomous multizone (SEAM) method, including dual SMAD inhibition and bone morphogenetic protein 4 treatment, for initial neural retinal induction; 2) the concurrent use of a Sonic hedgehog agonist SAG, activin A, and all-trans retinoic acid for rapid retinal cell specification; and 3) switching to SAG treatment alone for robust retinal maturation and lamination. The generated retinal organoids preserved typical morphological features of mature retinal organoids, including hair-like surface structures and well-organized outer layers. These features were substantiated by the spatial immunostaining patterns of several retinal cell markers, including rhodopsin and L/M opsin expression in the outermost layer, which was accompanied by reduced ectopic cone photoreceptor generation. Importantly, our method required only 90 days for retinal organoid maturation, which is approximately two-thirds the time necessary for other conventional methods. These results indicate that thoroughly optimized pharmacological interventions play a pivotal role in rapid and precise photoreceptor development during human retinal organoid differentiation and maturation. Thus, our present method may expedite human retinal organoid research, eventually contributing to the development of better treatment options for various degenerative retinal diseases.
Collapse
Affiliation(s)
- Tokiyoshi Matsushita
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Akishi Onishi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Takahiro Matsuyama
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Tomohiro Masuda
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Yoko Ogino
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
| | - Masaaki Kageyama
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Fumiaki Uchiumi
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
6
|
Eintracht J, Owen N, Harding P, Moosajee M. Disruption of common ocular developmental pathways in patient-derived optic vesicle models of microphthalmia. Stem Cell Reports 2024; 19:839-858. [PMID: 38821055 PMCID: PMC11390689 DOI: 10.1016/j.stemcr.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
Genetic perturbations influencing early eye development can result in microphthalmia, anophthalmia, and coloboma (MAC). Over 100 genes are associated with MAC, but little is known about common disease mechanisms. In this study, we generated induced pluripotent stem cell (iPSC)-derived optic vesicles (OVs) from two unrelated microphthalmia patients and healthy controls. At day 20, 35, and 50, microphthalmia patient OV diameters were significantly smaller, recapitulating the "small eye" phenotype. RNA sequencing (RNA-seq) analysis revealed upregulation of apoptosis-initiating and extracellular matrix (ECM) genes at day 20 and 35. Western blot and immunohistochemistry revealed increased expression of lumican, nidogen, and collagen type IV, suggesting ECM overproduction. Increased apoptosis was observed in microphthalmia OVs with reduced phospho-histone 3 (pH3+) cells confirming decreased cell proliferation at day 35. Pharmacological inhibition of caspase-8 activity with Z-IETD-FMK decreased apoptosis in one patient model, highlighting a potential therapeutic approach. These data reveal shared pathophysiological mechanisms contributing to a microphthalmia phenotype.
Collapse
Affiliation(s)
| | | | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; Moorfields Eye Hospital NHS Foundation Trust, London EC1V 9EL, UK; Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
7
|
Kawai K, Ho MT, Ueno Y, Abdo D, Xue C, Nonaka H, Nishida H, Honma Y, Wallace VA, Shoichet MS. Hyaluronan improves photoreceptor differentiation and maturation in human retinal organoids. Acta Biomater 2024; 181:117-132. [PMID: 38705224 DOI: 10.1016/j.actbio.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
Human stem cell-derived organoids enable both disease modeling and serve as a source of cells for transplantation. Human retinal organoids are particularly important as a source of human photoreceptors; however, the long differentiation period required and lack of vascularization in the organoid often results in a necrotic core and death of inner retinal cells before photoreceptors are fully mature. Manipulating the in vitro environment of differentiating retinal organoids through the incorporation of extracellular matrix components could influence retinal development. We investigated the addition of hyaluronan (HA), a component of the interphotoreceptor matrix, as an additive to promote long-term organoid survival and enhance retinal maturation. HA treatment had a significant reduction in the proportion of proliferating (Ki67+) cells and increase in the proportion of photoreceptors (CRX+), suggesting that HA accelerated photoreceptor commitment in vitro. HA significantly upregulated genes specific to photoreceptor maturation and outer segment development. Interestingly, prolonged HA-treatment significantly decreased the length of the brush border layer compared to those in control retinal organoids, where the photoreceptor outer segments reside; however, HA-treated organoids also had more mature outer segments with organized discs structures, as revealed by transmission electron microscopy. The brush border layer length was inversely proportional to the molar mass and viscosity of the hyaluronan added. This is the first study to investigate the role of exogenous HA, viscosity, and polymer molar mass on photoreceptor maturation, emphasizing the importance of material properties on organoid culture. STATEMENT OF SIGNIFICANCE: Retinal organoids are a powerful tool to study retinal development in vitro, though like many other organoid systems, can be highly variable. In this work, Shoichet and colleagues investigated the use of hyaluronan (HA), a native component of the interphotoreceptor matrix, to improve photoreceptor maturation in developing human retinal organoids. HA promoted human photoreceptor differentiation leading to mature outer segments with disc formation and more uniform and healthy retinal organoids. These findings highlight the importance of adding components native to the developing retina to generate more physiologically relevant photoreceptors for cell therapy and in vitro models to drive drug discovery and uncover novel disease mechanisms.
Collapse
Affiliation(s)
- Kotoe Kawai
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Margaret T Ho
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Yui Ueno
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada
| | - Dhana Abdo
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Chang Xue
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Hidenori Nonaka
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Hiroyuki Nishida
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Yoichi Honma
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Valerie A Wallace
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Canada
| | - Molly S Shoichet
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Department of Chemistry, University of Toronto, Canada.
| |
Collapse
|
8
|
Moyo MTG, Adali T, Tulay P. Exploring gellan gum-based hydrogels for regenerating human embryonic stem cells in age-related macular degeneration therapy: A literature review. Regen Ther 2024; 26:235-250. [PMID: 38966602 PMCID: PMC11222715 DOI: 10.1016/j.reth.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/15/2024] [Accepted: 05/26/2024] [Indexed: 07/06/2024] Open
Abstract
Age-related macular degeneration (AMD) is a progressive ocular disease marked by the deterioration of retinal photoreceptor cells, leading to central vision decline, predominantly affecting the elderly population worldwide. Current treatment modalities, such as anti-VEGF agents, laser therapy, and photodynamic therapy, aim to manage the condition, with emerging strategies like stem cell replacement therapy showing promise. However, challenges like immune rejection and cell survival hinder the efficacy of stem cell interventions. Regenerative medicine faces obstacles in maximizing stem cell potential due to limitations in mimicking the dynamic cues of the extracellular matrix (ECM) crucial for guiding stem cell behaviour. Innovative biomaterials like gellan gum hydrogels offer tailored microenvironments conducive to enhancing stem cell culture efficacy and tissue regeneration. Gellan gum-based hydrogels, renowned for biocompatibility and customizable mechanical properties, provide crucial support for cell viability, differentiation, and controlled release of therapeutic factors, making them an ideal platform for culturing human embryonic stem cells (hESCs). These hydrogels mimic native tissue mechanics, promoting optimal hESC differentiation while minimizing immune responses and facilitating localized delivery. This review explores the potential of Gellan Gum-Based Hydrogels in regenerative AMD therapy, emphasizing their role in enhancing hESC regeneration and addressing current status, treatment limitations, and future directions.
Collapse
Affiliation(s)
- Mthabisi Talent George Moyo
- Near East University, Faculty of Engineering, Department of Biomedical Engineering, P.O. Box: 99138, Nicosia, Cyprus, Mersin 10, Turkey
- Girne American University, Faculty of Medicine, Department of Medical Biochemistry, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, Cyprus, Mersin 10, Turkey
- Girne American University, Research and Application Center of Biomedical Sciences, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, North Cyprus, Mersin 10, Turkey
| | - Terin Adali
- Girne American University, Faculty of Medicine, Department of Medical Biochemistry, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, Cyprus, Mersin 10, Turkey
- Girne American University, Research and Application Center of Biomedical Sciences, PO Box 99428, Karmi Campus, Karaoglanoglu, Kyrenia, North Cyprus, Mersin 10, Turkey
| | - Pinar Tulay
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus, Mersin 10, Turkey
- Near East University, DESAM Research Institute, Nicosia, Cyprus, Mersin 10, Turkey
| |
Collapse
|
9
|
Dorgau B, Collin J, Rozanska A, Boczonadi V, Moya-Molina M, Unsworth A, Hussain R, Coxhead J, Dhanaseelan T, Armstrong L, Queen R, Lako M. Deciphering the spatiotemporal transcriptional and chromatin accessibility of human retinal organoid development at the single-cell level. iScience 2024; 27:109397. [PMID: 38510120 PMCID: PMC10952046 DOI: 10.1016/j.isci.2024.109397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/28/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Molecular information on the early stages of human retinal development remains scarce due to limitations in obtaining early human eye samples. Pluripotent stem cell-derived retinal organoids (ROs) provide an unprecedented opportunity for studying early retinogenesis. Using a combination of single cell RNA-seq and spatial transcriptomics we present for the first-time a single cell spatiotemporal transcriptome of RO development. Our data demonstrate that ROs recapitulate key events of retinogenesis including optic vesicle/cup formation, presence of a putative ciliary margin zone, emergence of retinal progenitor cells and their orderly differentiation to retinal neurons. Combining the scRNA- with scATAC-seq data, we were able to reveal cell-type specific transcription factor binding motifs on accessible chromatin at each stage of organoid development, and to show that chromatin accessibility is highly correlated to the developing human retina, but with some differences in the temporal emergence and abundance of some of the retinal neurons.
Collapse
Affiliation(s)
- Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Agata Rozanska
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Veronika Boczonadi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Marina Moya-Molina
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
- Newcells Biotech, Newcastle upon Tyne NE4 5BX, UK
| | - Adrienne Unsworth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Tamil Dhanaseelan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
10
|
Woronkowicz M, Roberts H, Skopiński P. The Role of Insulin-like Growth Factor (IGF) System in the Corneal Epithelium Homeostasis-From Limbal Epithelial Stem Cells to Therapeutic Applications. BIOLOGY 2024; 13:144. [PMID: 38534414 DOI: 10.3390/biology13030144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024]
Abstract
The corneal epithelium, comprising three layers of cells, represents the outermost portion of the eye and functions as a vital protective barrier while concurrently serving as a critical refractive structure. Maintaining its homeostasis involves a complex regenerative process facilitated by the functions of the lacrimal gland, tear film, and corneal nerves. Crucially, limbal epithelial stem cells located in the limbus (transitional zone between the cornea and the conjunctiva) are instrumental for the corneal epithelium integrity by replenishing and renewing cells. Re-epithelialization failure results in persistent defects, often associated with various ocular conditions including diabetic keratopathy. The insulin-like growth factor (IGF) system is a sophisticated network of insulin and other proteins essential for numerous physiological processes. This review examines its role in maintaining the corneal epithelium homeostasis, with a special focus on the interplay with corneal limbal stem cells and the potential therapeutic applications of the system components.
Collapse
Affiliation(s)
- Małgorzata Woronkowicz
- NDDH, Royal Devon University Healthcare NHS Foundation Trust, Barnstaple EX31 4JB, UK
- Moorfields Eye Hospital NHS Foundation Trust, 162 City Road, London EC1V 2PD, UK
| | - Harry Roberts
- West of England Eye Unit, Royal Devon University Healthcare NHS Foundation Trust, Exeter EX2 5DW, UK
- University of Exeter Medical School, Exeter EX1 2HZ, UK
| | - Piotr Skopiński
- Department of Ophthalmology, SPKSO Ophthalmic University Hospital, Medical University of Warsaw, 00-576 Warsaw, Poland
- Department of Histology and Embryology, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
11
|
Wang Y, Yin N, Yang R, Zhao M, Li S, Zhang S, Zhao Y, Faiola F. Development of a simplified human embryonic stem cell-based retinal pre-organoid model for toxicity evaluations of common pollutants. Cutan Ocul Toxicol 2023; 42:264-272. [PMID: 37602871 DOI: 10.1080/15569527.2023.2249988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/20/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
OBJECTIVE To explore the retinal toxicity of pharmaceuticals and personal care products (PPCPs), flame retardants, bisphenols, phthalates, and polycyclic aromatic hydrocarbons (PAHs) on human retinal progenitor cells (RPCs) and retinal pigment epithelial (RPE) cells, which are the primary cell types at the early stages of retinal development, vital for subsequent functional cell type differentiation, and closely related to retinal diseases. MATERIALS AND METHODS After 23 days of differentiation, human embryonic stem cell (hESC)-based retinal pre-organoids, containing RPCs and RPE cells, were exposed to 10, 100, and 1000 nM pesticides (butachlor, terbutryn, imidacloprid, deltamethrin, pendimethalin, and carbaryl), flame retardants (PFOS, TBBPA, DBDPE, and TDCIPP), PPCPs (climbazole and BHT), and other typical pollutants (phenanthrene, DCHP, and BPA) for seven days. Then, mRNA expression changes were monitored and compared. RESULTS (1) The selected pollutants did not show strong effects at environmental and human-relevant concentrations, although the effects of flame retardants were more potent than those of other categories of chemicals. Surprisingly, some pollutants with distinct structures showed similar adverse effects. (2) Exposure to pollutants induced different degrees of cell detachment, probably due to alterations in extracellular matrix and/or cell adhesion. CONCLUSIONS In this study, we established a retinal pre-organoid model suitable for evaluating multiple pollutants' effects, and pointed out the potential retinal toxicity of flame retardants, among other pollutants. Nevertheless, the potential mechanisms of toxicity and the effects on cell detachment are still unclear and deserve further exploration. Additionally, this model holds promise for screening interventions aimed at mitigating the detrimental effects of these pollutants.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Renjun Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Miaomiao Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Shichang Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Shuxian Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Yanyi Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Francesco Faiola
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, P.R. China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, P.R. China
| |
Collapse
|
12
|
Wadkin LE, Makarenko I, Parker NG, Shukurov A, Figueiredo FC, Lako M. Human Stem Cells for Ophthalmology: Recent Advances in Diagnostic Image Analysis and Computational Modelling. CURRENT STEM CELL REPORTS 2023; 9:57-66. [PMID: 38145008 PMCID: PMC10739444 DOI: 10.1007/s40778-023-00229-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 12/26/2023]
Abstract
Purpose of Review To explore the advances and future research directions in image analysis and computational modelling of human stem cells (hSCs) for ophthalmological applications. Recent Findings hSCs hold great potential in ocular regenerative medicine due to their application in cell-based therapies and in disease modelling and drug discovery using state-of-the-art 2D and 3D organoid models. However, a deeper characterisation of their complex, multi-scale properties is required to optimise their translation to clinical practice. Image analysis combined with computational modelling is a powerful tool to explore mechanisms of hSC behaviour and aid clinical diagnosis and therapy. Summary Many computational models draw on a variety of techniques, often blending continuum and discrete approaches, and have been used to describe cell differentiation and self-organisation. Machine learning tools are having a significant impact in model development and improving image classification processes for clinical diagnosis and treatment and will be the focus of much future research.
Collapse
Affiliation(s)
- L. E. Wadkin
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - I. Makarenko
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - N. G. Parker
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - A. Shukurov
- School of Mathematics, Statistics and Physics, Newcastle University, Newcastle upon Tyne, UK
| | - F. C. Figueiredo
- Department of Ophthalmology, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - M. Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
13
|
Beaver D, Limnios IJ. A treatment within sight: challenges in the development of stem cell-derived photoreceptor therapies for retinal degenerative diseases. FRONTIERS IN TRANSPLANTATION 2023; 2:1130086. [PMID: 38993872 PMCID: PMC11235385 DOI: 10.3389/frtra.2023.1130086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 09/07/2023] [Indexed: 07/13/2024]
Abstract
Stem cell therapies can potentially treat various retinal degenerative diseases, including age-related macular degeneration (AMD) and inherited retinal diseases like retinitis pigmentosa. For these diseases, transplanted cells may include stem cell-derived retinal pigmented epithelial (RPE) cells, photoreceptors, or a combination of both. Although stem cell-derived RPE cells have progressed to human clinical trials, therapies using photoreceptors and other retinal cell types are lagging. In this review, we discuss the potential use of human pluripotent stem cell (hPSC)-derived photoreceptors for the treatment of retinal degeneration and highlight the progress and challenges for their efficient production and clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Davinia Beaver
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| | - Ioannis Jason Limnios
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| |
Collapse
|
14
|
Lima Cunha D, Sarkar H, Eintracht J, Harding P, Zhou JH, Moosajee M. Restoration of functional PAX6 in aniridia patient iPSC-derived ocular tissue models using repurposed nonsense suppression drugs. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:240-253. [PMID: 37483273 PMCID: PMC10362734 DOI: 10.1016/j.omtn.2023.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/22/2023] [Indexed: 07/25/2023]
Abstract
Congenital aniridia is a rare, pan-ocular disease causing severe sight loss, with only symptomatic intervention offered to patients. Approximately 40% of aniridia patients present with heterozygous nonsense variants in PAX6, resulting in haploinsufficiency. Translational readthrough-inducing drugs (TRIDs) have the ability to weaken the recognition of in-frame premature termination codons (PTCs), permitting full-length protein to be translated. We established induced pluripotent stem cell (iPSC)-derived 3D optic cups and 2D limbal epithelial stem cell (LESC) models from two aniridia patients with prevalent PAX6 nonsense mutations. Both in vitro models show reduced PAX6 protein levels, mimicking the disease. The repurposed TRIDs amlexanox and 2,6-diaminopurine (DAP) and the positive control compounds ataluren and G418 were tested for their efficiency. Amlexanox was identified as the most promising TRID, increasing full-length PAX6 levels in both models and rescuing the disease phenotype through normalization of VSX2 and cell proliferation in the optic cups and reduction of ABCG2 protein and SOX10 expression in LESCs. This study highlights the significance of patient iPSC-derived cells as a new model system for aniridia and proposes amlexanox as a new putative treatment for nonsense-mediated aniridia.
Collapse
Affiliation(s)
- Dulce Lima Cunha
- UCL Institute of Ophthalmology, London, UK
- Radboud Institute of Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Hajrah Sarkar
- UCL Institute of Ophthalmology, London, UK
- The Francis Crick Institute, London, UK
| | | | | | - Jo Huiqing Zhou
- Radboud Institute of Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Mariya Moosajee
- UCL Institute of Ophthalmology, London, UK
- The Francis Crick Institute, London, UK
- Moorfields Eye Hospital, London, UK
| |
Collapse
|
15
|
Watson A, Lako M. Retinal organoids provide unique insights into molecular signatures of inherited retinal disease throughout retinogenesis. J Anat 2023; 243:186-203. [PMID: 36177499 PMCID: PMC10335378 DOI: 10.1111/joa.13768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022] Open
Abstract
The demand for induced pluripotent stem cells (iPSC)-derived retinal organoid and retinal pigment epithelium (RPE) models for the modelling of inherited retinopathies has increased significantly in the last decade. These models are comparable with foetal retinas up until the later stages of retinogenesis, expressing all of the key neuronal markers necessary for retinal function. These models have proven to be invaluable in the understanding of retinogenesis, particular in the context of patient-specific diseases. Inherited retinopathies are infamously described as clinically and phenotypically heterogeneous, such that developing gene/mutation-specific animal models in each instance of retinal disease is not financially or ethically feasible. Further to this, many animal models are insufficient in the study of disease pathogenesis due to anatomical differences and failure to recapitulate human disease phenotypes. In contrast, iPSC-derived retinal models provide a high throughput platform which is physiologically relevant for studying human health and disease. They also serve as a platform for drug screening, gene therapy approaches and in vitro toxicology of novel therapeutics in pre-clinical studies. One unique characteristic of stem cell-derived retinal models is the ability to mimic in vivo retinogenesis, providing unparalleled insights into the effects of pathogenic mutations in cells of the developing retina, in a highly accessible way. This review aims to give the reader an overview of iPSC-derived retinal organoids and/or RPE in the context of disease modelling of several inherited retinopathies including Retinitis Pigmentosa, Stargardt disease and Retinoblastoma. We describe the ability of each model to recapitulate in vivo disease phenotypes, validate previous findings from animal models and identify novel pathomechanisms that underpin individual IRDs.
Collapse
Affiliation(s)
- Avril Watson
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Majlinda Lako
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
16
|
Hockney S, Parker J, Turner JE, Todd X, Todryk S, Gieling RG, Hilgen G, Simoes DCM, Pal D. Next generation organoid engineering to replace animals in cancer drug testing. Biochem Pharmacol 2023; 213:115586. [PMID: 37164297 DOI: 10.1016/j.bcp.2023.115586] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/12/2023]
Abstract
Cancer therapies have several clinical challenges associated with them, namely treatment toxicity, treatment resistance and relapse. Due to factors ranging from patient profiles to the tumour microenvironment (TME), there are several hurdles to overcome in developing effective treatments that have low toxicity that can mitigate emergence of resistance and occurrence of relapse. De novo cancer development has the highest drug attrition rates with only 1 in 10,000 preclinical candidates reaching the market. To alleviate this high attrition rate, more mimetic and sustainable preclinical models that can capture the disease biology as in the patient, are required. Organoids and next generation 3D tissue engineering is an emerging area that aims to address this problem. Advancement of three-dimensional (3D) in vitro cultures into complex organoid models incorporating multiple cell types alongside acellular aspects of tissue microenvironments can provide a system for therapeutic testing. Development of microfluidic technologies have furthermore increased the biomimetic nature of these models. Additionally, 3D bio-printing facilitates generation of tractable ex vivo models in a controlled, scalable and reproducible manner. In this review we highlight some of the traditional preclinical models used in cancer drug testing and debate how next generation organoids are being used to replace not only animal models, but also some of the more elementary in vitro approaches, such as cell lines. Examples of applications of the various models will be appraised alongside the future challenges that still need to be overcome.
Collapse
Affiliation(s)
- Sean Hockney
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Jessica Parker
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Jasmin E Turner
- Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 4EP, UK
| | - Xanthea Todd
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Stephen Todryk
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Roben Ger Gieling
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Gerrit Hilgen
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK; Biosciences Institute, Newcastle University, International Centre for Life, Newcastle Upon Tyne NE1 4EP, UK
| | - Davina Camargo Madeira Simoes
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Deepali Pal
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK; Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE1 7RU, UK.
| |
Collapse
|
17
|
Kang J, Gong J, Yang C, Lin X, Yan L, Gong Y, Xu H. Application of Human Stem Cell Derived Retinal Organoids in the Exploration of the Mechanisms of Early Retinal Development. Stem Cell Rev Rep 2023:10.1007/s12015-023-10553-x. [PMID: 37269529 DOI: 10.1007/s12015-023-10553-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 06/05/2023]
Abstract
The intricate neural circuit of retina extracts salient features of the natural world and forms bioelectric impulse as the origin of vision. The early development of retina is a highly complex and coordinated process in morphogenesis and neurogenesis. Increasing evidence indicates that stem cells derived human retinal organoids (hROs) in vitro faithfully recapitulates the embryonic developmental process of human retina no matter in the transcriptome, cellular biology and histomorphology. The emergence of hROs greatly deepens on the understanding of early development of human retina. Here, we reviewed the events of early retinal development both in animal embryos and hROs studies, which mainly comprises the formation of optic vesicle and optic cup shape, differentiation of retinal ganglion cells (RGCs), photoreceptor cells (PRs) and its supportive retinal pigment epithelium cells (RPE). We also discussed the classic and frontier molecular pathways up to date to decipher the underlying mechanisms of early development of human retina and hROs. Finally, we summarized the application prospect, challenges and cutting-edge techniques of hROs for uncovering the principles and mechanisms of retinal development and related developmental disorder. hROs is a priori selection for studying human retinal development and function and may be a fundamental tool for unlocking the unknown insight into retinal development and disease.
Collapse
Affiliation(s)
- Jiahui Kang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Cao Yang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xi Lin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Lijuan Yan
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yu Gong
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
- Department of Ophthalmology, Medical Sciences Research Center, University-Town Hospital of Chongqing Medical University, Chongqing, China.
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
18
|
Bohrer LR, Stone NE, Mullin NK, Voigt AP, Anfinson KR, Fick JL, Luangphakdy V, Hittle B, Powell K, Muschler GF, Mullins RF, Stone EM, Tucker BA. Automating iPSC generation to enable autologous photoreceptor cell replacement therapy. J Transl Med 2023; 21:161. [PMID: 36855199 PMCID: PMC9976478 DOI: 10.1186/s12967-023-03966-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/03/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Inherited retinal degeneration is a leading cause of incurable vision loss in the developed world. While autologous iPSC mediated photoreceptor cell replacement is theoretically possible, the lack of commercially available technologies designed to enable high throughput parallel production of patient specific therapeutics has hindered clinical translation. METHODS In this study, we describe the use of the Cell X precision robotic cell culture platform to enable parallel production of clinical grade patient specific iPSCs. The Cell X is housed within an ISO Class 5 cGMP compliant closed aseptic isolator (Biospherix XVivo X2), where all procedures from fibroblast culture to iPSC generation, clonal expansion and retinal differentiation were performed. RESULTS Patient iPSCs generated using the Cell X platform were determined to be pluripotent via score card analysis and genetically stable via karyotyping. As determined via immunostaining and confocal microscopy, iPSCs generated using the Cell X platform gave rise to retinal organoids that were indistinguishable from organoids derived from manually generated iPSCs. In addition, at 120 days post-differentiation, single-cell RNA sequencing analysis revealed that cells generated using the Cell X platform were comparable to those generated under manual conditions in a separate laboratory. CONCLUSION We have successfully developed a robotic iPSC generation platform and standard operating procedures for production of high-quality photoreceptor precursor cells that are compatible with current good manufacturing practices. This system will enable clinical grade production of iPSCs for autologous retinal cell replacement.
Collapse
Affiliation(s)
- Laura R Bohrer
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicholas E Stone
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nathaniel K Mullin
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Andrew P Voigt
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kristin R Anfinson
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Jessica L Fick
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Viviane Luangphakdy
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Cell X Technologies Inc, Cleveland, OH, USA
| | - Bradley Hittle
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Kimerly Powell
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - George F Muschler
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Orthopaedic Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Robert F Mullins
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Edwin M Stone
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Budd A Tucker
- Institute for Vision Research, Carver College of Medicine, University of Iowa, 375 Newton Road, Iowa City, IA, 52242, USA.
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
19
|
Watari K, Yamasaki S, Tu HY, Shikamura M, Kamei T, Adachi H, Tochitani T, Kita Y, Nakamura A, Ueyama K, Ono K, Morinaga C, Matsuyama T, Sho J, Nakamura M, Fujiwara M, Hori Y, Tanabe A, Hirai R, Terai O, Ohno O, Ohara H, Hayama T, Ikeda A, Nukaya D, Matsushita K, Takahashi M, Kishino A, Kimura T, Kawamata S, Mandai M, Kuwahara A. Self-organization, quality control, and preclinical studies of human iPSC-derived retinal sheets for tissue-transplantation therapy. Commun Biol 2023; 6:164. [PMID: 36765170 PMCID: PMC9918541 DOI: 10.1038/s42003-023-04543-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/31/2023] [Indexed: 02/12/2023] Open
Abstract
Three-dimensional retinal organoids (3D-retinas) are a promising graft source for transplantation therapy. We previously developed self-organizing culture for 3D-retina generation from human pluripotent stem cells (hPSCs). Here we present a quality control method and preclinical studies for tissue-sheet transplantation. Self-organizing hPSCs differentiated into both retinal and off-target tissues. Gene expression analyses identified the major off-target tissues as eye-related, cortex-like, and spinal cord-like tissues. For quality control, we developed a qPCR-based test in which each hPSC-derived neuroepithelium was dissected into two tissue-sheets: inner-central sheet for transplantation and outer-peripheral sheet for qPCR to ensure retinal tissue selection. During qPCR, tissue-sheets were stored for 3-4 days using a newly developed preservation method. In a rat tumorigenicity study, no transplant-related adverse events were observed. In retinal degeneration model rats, retinal transplants differentiated into mature photoreceptors and exhibited light responses in electrophysiology assays. These results demonstrate our rationale toward self-organizing retinal sheet transplantation therapy.
Collapse
Affiliation(s)
- Kenji Watari
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Suguru Yamasaki
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan ,grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Hung-Ya Tu
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Masayuki Shikamura
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Tatsuya Kamei
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Hideki Adachi
- grid.417741.00000 0004 1797 168XPreclinical Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Konohana-ku, Osaka 554-0022 Japan
| | - Tomoaki Tochitani
- grid.417741.00000 0004 1797 168XPreclinical Research Unit, Research Division, Sumitomo Pharma Co., Ltd., Konohana-ku, Osaka 554-0022 Japan
| | - Yasuyuki Kita
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Aya Nakamura
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Kazuki Ueyama
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Keiichi Ono
- grid.417741.00000 0004 1797 168XTechnology Research & Development Division, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Chikako Morinaga
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan ,grid.7597.c0000000094465255RIKEN Program for Drug Discovery and Medical Technology Platforms, RIKEN Cluster for Science, Technology and Innovation Hub., Saitama, 351-0198 Japan
| | - Take Matsuyama
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Junki Sho
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Miyuki Nakamura
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Masayo Fujiwara
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Yoriko Hori
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Anna Tanabe
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Rina Hirai
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Orie Terai
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Osamu Ohno
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Hidetaka Ohara
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Tetsuya Hayama
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Atsushi Ikeda
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Daiki Nukaya
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Keizo Matsushita
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan ,grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Masayo Takahashi
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan
| | - Akiyoshi Kishino
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Toru Kimura
- grid.417741.00000 0004 1797 168XRegenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe 650-0047 Japan
| | - Shin Kawamata
- grid.417982.10000 0004 0623 246XResearch & Development Center for Cell Therapy, Foundation for Biomedical Research and Innovation at Kobe, Chuo-ku, Kobe 650-0047 Japan
| | - Michiko Mandai
- grid.508743.dLaboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe 650-0047 Japan ,grid.7597.c0000000094465255RIKEN Program for Drug Discovery and Medical Technology Platforms, RIKEN Cluster for Science, Technology and Innovation Hub., Saitama, 351-0198 Japan
| | - Atsushi Kuwahara
- Regenerative & Cellular Medicine Kobe Center, Sumitomo Pharma Co., Ltd., Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
20
|
Cardozo MJ, Sánchez-Bustamante E, Bovolenta P. Optic cup morphogenesis across species and related inborn human eye defects. Development 2023; 150:dev200399. [PMID: 36714981 PMCID: PMC10110496 DOI: 10.1242/dev.200399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The vertebrate eye is shaped as a cup, a conformation that optimizes vision and is acquired early in development through a process known as optic cup morphogenesis. Imaging living, transparent teleost embryos and mammalian stem cell-derived organoids has provided insights into the rearrangements that eye progenitors undergo to adopt such a shape. Molecular and pharmacological interference with these rearrangements has further identified the underlying molecular machineries and the physical forces involved in this morphogenetic process. In this Review, we summarize the resulting scenarios and proposed models that include common and species-specific events. We further discuss how these studies and those in environmentally adapted blind species may shed light on human inborn eye malformations that result from failures in optic cup morphogenesis, including microphthalmia, anophthalmia and coloboma.
Collapse
Affiliation(s)
- Marcos J. Cardozo
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Elena Sánchez-Bustamante
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| | - Paola Bovolenta
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), c/ Nicolás Cabrera 1, Cantoblanco, Madrid 28049, Spain
| |
Collapse
|
21
|
Li M, Gao L, Zhao L, Zou T, Xu H. Toward the next generation of vascularized human neural organoids. Med Res Rev 2023; 43:31-54. [PMID: 35993813 DOI: 10.1002/med.21922] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/22/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Thanks to progress in the development of three-dimensional (3D) culture technologies, human central nervous system (CNS) development and diseases have been gradually deciphered by using organoids derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs). Selforganized neural organoids (NOs) have been used to mimic morphogenesis and functions of specific organs in vitro. Many NOs have been reproduced in vitro, such as those mimicking the human brain, retina, and spinal cord. However, NOs fail to capitulate to the maturation and complexity of in vivo neural tissues. The persistent issues with current NO cultivation protocols are inadequate oxygen supply and nutrient diffusion due to the absence of vascular networks. In vivo, the developing CNS is interpenetrated by vasculature that not only supplies oxygen and nutrients but also provides a structural template for neuronal growth. To address these deficiencies, recent studies have begun to couple NO culture with bioengineering techniques and methodologies, including genetic engineering, coculture, multidifferentiation, microfluidics and 3D bioprinting, and transplantation, which might promote NO maturation and create more functional NOs. These cutting-edge methods could generate an ever more reliable NO model in vitro for deciphering the codes of human CNS development, disease progression, and translational application. In this review, we will summarize recent technological advances in culture strategies to generate vascularized NOs (vNOs), with a special focus on cerebral- and retinal-organoid models.
Collapse
Affiliation(s)
- Minghui Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Lixiong Gao
- Department of Ophthalmology, Third Medical Center of PLA General Hospital, Beijing, China
| | - Ling Zhao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, China
| |
Collapse
|
22
|
Leong YC, Sowden JC. Modeling Retinitis Pigmentosa with Patient-Derived iPSCs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:555-563. [PMID: 37440086 DOI: 10.1007/978-3-031-27681-1_81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Retinitis pigmentosa (RP) causes blindness in 1 out of 3000-4000 individuals worldwide. Understanding the disease mechanism underlying the death of photoreceptors in RP patient is crucial for the discovery and development of therapies to prevent and stop the progression of retinal degeneration. Despite having provided valuable insight into RP pathology, several shortcomings of animal models warrant the need for a better modeling system. This review discusses the current use of patient-derived induced pluripotent stem cells (iPSCs) to model RP and its advantages over animal models. Further improvement to enhance the representativeness of iPSC RP models is also discussed.
Collapse
Affiliation(s)
- Yeh Chwan Leong
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK
| | - Jane C Sowden
- Stem Cells and Regenerative Medicine Section, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK.
| |
Collapse
|
23
|
Insulin-like growth factor-1 stimulates retinal cell proliferation via activation of multiple signaling pathways. CURRENT RESEARCH IN NEUROBIOLOGY 2022; 4:100068. [PMID: 36589675 PMCID: PMC9800307 DOI: 10.1016/j.crneur.2022.100068] [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: 07/17/2022] [Revised: 09/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays critical roles in the development of the central nervous system (CNS), including the retina, regulating cell proliferation, differentiation, and survival. Here, we investigated the role of IGF-1 on retinal cell proliferation using primary cultures from rat neural retina. Our data show that IGF-1 stimulates retinal cell proliferation and regulates the expression of neurotrophic factors, such as interleukin-4 and brain-derived neurotrophic factor. In addition, our results indicates that IGF-1-induced retinal cell proliferation requires activation of multiple signaling pathways, including phosphatidylinositol 3-kinase, protein kinase Src, phospholipase-C, protein kinase C delta, and mitogen-activated protein kinase pathways. We further show that activation of matrix metalloproteinases and epidermal growth factor receptor is also necessary for IGF-1 enhancing retinal cell proliferation. Overall, these results unveil potential mechanisms by which IGF-1 ensures retinal cell proliferation and support the notion that manipulation of IGF-1 signaling may be beneficial in CNS disorders associated with abnormal cell proliferation.
Collapse
|
24
|
Cvekl A, Camerino MJ. Generation of Lens Progenitor Cells and Lentoid Bodies from Pluripotent Stem Cells: Novel Tools for Human Lens Development and Ocular Disease Etiology. Cells 2022; 11:3516. [PMID: 36359912 PMCID: PMC9658148 DOI: 10.3390/cells11213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into specialized tissues and organs represents a powerful approach to gain insight into those cellular and molecular mechanisms regulating human development. Although normal embryonic eye development is a complex process, generation of ocular organoids and specific ocular tissues from pluripotent stem cells has provided invaluable insights into the formation of lineage-committed progenitor cell populations, signal transduction pathways, and self-organization principles. This review provides a comprehensive summary of recent advances in generation of adenohypophyseal, olfactory, and lens placodes, lens progenitor cells and three-dimensional (3D) primitive lenses, "lentoid bodies", and "micro-lenses". These cells are produced alone or "community-grown" with other ocular tissues. Lentoid bodies/micro-lenses generated from human patients carrying mutations in crystallin genes demonstrate proof-of-principle that these cells are suitable for mechanistic studies of cataractogenesis. Taken together, current and emerging advanced in vitro differentiation methods pave the road to understand molecular mechanisms of cataract formation caused by the entire spectrum of mutations in DNA-binding regulatory genes, such as PAX6, SOX2, FOXE3, MAF, PITX3, and HSF4, individual crystallins, and other genes such as BFSP1, BFSP2, EPHA2, GJA3, GJA8, LIM2, MIP, and TDRD7 represented in human cataract patients.
Collapse
Affiliation(s)
- Aleš Cvekl
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael John Camerino
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
25
|
Diacou R, Nandigrami P, Fiser A, Liu W, Ashery-Padan R, Cvekl A. Cell fate decisions, transcription factors and signaling during early retinal development. Prog Retin Eye Res 2022; 91:101093. [PMID: 35817658 PMCID: PMC9669153 DOI: 10.1016/j.preteyeres.2022.101093] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/30/2022]
Abstract
The development of the vertebrate eyes is a complex process starting from anterior-posterior and dorso-ventral patterning of the anterior neural tube, resulting in the formation of the eye field. Symmetrical separation of the eye field at the anterior neural plate is followed by two symmetrical evaginations to generate a pair of optic vesicles. Next, reciprocal invagination of the optic vesicles with surface ectoderm-derived lens placodes generates double-layered optic cups. The inner and outer layers of the optic cups develop into the neural retina and retinal pigment epithelium (RPE), respectively. In vitro produced retinal tissues, called retinal organoids, are formed from human pluripotent stem cells, mimicking major steps of retinal differentiation in vivo. This review article summarizes recent progress in our understanding of early eye development, focusing on the formation the eye field, optic vesicles, and early optic cups. Recent single-cell transcriptomic studies are integrated with classical in vivo genetic and functional studies to uncover a range of cellular mechanisms underlying early eye development. The functions of signal transduction pathways and lineage-specific DNA-binding transcription factors are dissected to explain cell-specific regulatory mechanisms underlying cell fate determination during early eye development. The functions of homeodomain (HD) transcription factors Otx2, Pax6, Lhx2, Six3 and Six6, which are required for early eye development, are discussed in detail. Comprehensive understanding of the mechanisms of early eye development provides insight into the molecular and cellular basis of developmental ocular anomalies, such as optic cup coloboma. Lastly, modeling human development and inherited retinal diseases using stem cell-derived retinal organoids generates opportunities to discover novel therapies for retinal diseases.
Collapse
Affiliation(s)
- Raven Diacou
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Prithviraj Nandigrami
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Andras Fiser
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ruth Ashery-Padan
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Ales Cvekl
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, 10461, USA; Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
26
|
Looking into the Eyes—In Vitro Models for Ocular Research. Int J Mol Sci 2022; 23:ijms23169158. [PMID: 36012421 PMCID: PMC9409455 DOI: 10.3390/ijms23169158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Animal research undoubtedly provides scientists with virtually unlimited data but inflicts pain and suffering on animals. Currently, legislators and scientists alike are promoting alternative in vitro approaches allowing for an accurate evaluation of processes occurring in the body without animal sacrifice. Historically, one of the most infamous animal tests is the Draize test, mainly performed on rabbits. Even though this test was considered the gold standard for around 50 years, the Draize test fails to mimic human response mainly due to human and rabbit eye physiological differences. Therefore, many alternative assays were developed to evaluate ocular toxicity and drug effectiveness accurately. Here we review recent achievements in tissue engineering of in vitro 2D, 2.5D, 3D, organoid and organ-on-chip ocular models, as well as in vivo and ex vivo models in terms of their advantages and limitations.
Collapse
|
27
|
Xue Y, Lin B, Chen JT, Tang WC, Browne AW, Seiler MJ. The Prospects for Retinal Organoids in Treatment of Retinal Diseases. Asia Pac J Ophthalmol (Phila) 2022; 11:314-327. [PMID: 36041146 PMCID: PMC9966053 DOI: 10.1097/apo.0000000000000538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/22/2022] [Indexed: 12/28/2022] Open
Abstract
Retinal degeneration (RD) is a significant cause of incurable blindness worldwide. Photoreceptors and retinal pigmented epithelium are irreversibly damaged in advanced RD. Functional replacement of photoreceptors and/or retinal pigmented epithelium cells is a promising approach to restoring vision. This paper reviews the current status and explores future prospects of the transplantation therapy provided by pluripotent stem cell-derived retinal organoids (ROs). This review summarizes the status of rodent RD disease models and discusses RO culture and analytical tools to evaluate RO quality and function. Finally, we review and discuss the studies in which RO-derived cells or sheets were transplanted. In conclusion, methods to derive ROs from pluripotent stem cells have significantly improved and become more efficient in recent years. Meanwhile, more novel technologies are applied to characterize and validate RO quality. However, opportunity remains to optimize tissue differentiation protocols and achieve better RO reproducibility. In order to screen high-quality ROs for downstream applications, approaches such as noninvasive and label-free imaging and electrophysiological functional testing are promising and worth further investigation. Lastly, transplanted RO-derived tissues have allowed improvements in visual function in several RD models, showing promises for clinical applications in the future.
Collapse
Affiliation(s)
- Yuntian Xue
- Biomedical Engineering, University of California, Irvine, CA
- Stem Cell Research Center, University of California, Irvine, CA
| | - Bin Lin
- Stem Cell Research Center, University of California, Irvine, CA
| | - Jacqueline T. Chen
- Stem Cell Research Center, University of California, Irvine, CA
- Gavin Herbert Eye Institute Ophthalmology, University of California, Irvine, CA
| | - William C. Tang
- Biomedical Engineering, University of California, Irvine, CA
| | - Andrew W. Browne
- Biomedical Engineering, University of California, Irvine, CA
- Gavin Herbert Eye Institute Ophthalmology, University of California, Irvine, CA
- Institute for Clinical and Translational Science, University of California, Irvine, CA
| | - Magdalene J. Seiler
- Stem Cell Research Center, University of California, Irvine, CA
- Gavin Herbert Eye Institute Ophthalmology, University of California, Irvine, CA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA
| |
Collapse
|
28
|
Grigoryan EN. Self-Organization of the Retina during Eye Development, Retinal Regeneration In Vivo, and in Retinal 3D Organoids In Vitro. Biomedicines 2022; 10:1458. [PMID: 35740479 PMCID: PMC9221005 DOI: 10.3390/biomedicines10061458] [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: 03/30/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 11/23/2022] Open
Abstract
Self-organization is a process that ensures histogenesis of the eye retina. This highly intricate phenomenon is not sufficiently studied due to its biological complexity and genetic heterogeneity. The review aims to summarize the existing central theories and ideas for a better understanding of retinal self-organization, as well as to address various practical problems of retinal biomedicine. The phenomenon of self-organization is discussed in the spatiotemporal context and illustrated by key findings during vertebrate retina development in vivo and retinal regeneration in amphibians in situ. Described also are histotypic 3D structures obtained from the disaggregated retinal progenitor cells of birds and retinal 3D organoids derived from the mouse and human pluripotent stem cells. The review highlights integral parts of retinal development in these conditions. On the cellular level, these include competence, differentiation, proliferation, apoptosis, cooperative movements, and migration. On the physical level, the focus is on the mechanical properties of cell- and cell layer-derived forces and on the molecular level on factors responsible for gene regulation, such as transcription factors, signaling molecules, and epigenetic changes. Finally, the self-organization phenomenon is discussed as a basis for the production of retinal organoids, a promising model for a wide range of basic scientific and medical applications.
Collapse
Affiliation(s)
- Eleonora N Grigoryan
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| |
Collapse
|
29
|
O’Hara-Wright M, Mobini S, Gonzalez-Cordero A. Bioelectric Potential in Next-Generation Organoids: Electrical Stimulation to Enhance 3D Structures of the Central Nervous System. Front Cell Dev Biol 2022; 10:901652. [PMID: 35656553 PMCID: PMC9152151 DOI: 10.3389/fcell.2022.901652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived organoid models of the central nervous system represent one of the most exciting areas in in vitro tissue engineering. Classically, organoids of the brain, retina and spinal cord have been generated via recapitulation of in vivo developmental cues, including biochemical and biomechanical. However, a lesser studied cue, bioelectricity, has been shown to regulate central nervous system development and function. In particular, electrical stimulation of neural cells has generated some important phenotypes relating to development and differentiation. Emerging techniques in bioengineering and biomaterials utilise electrical stimulation using conductive polymers. However, state-of-the-art pluripotent stem cell technology has not yet merged with this exciting area of bioelectricity. Here, we discuss recent findings in the field of bioelectricity relating to the central nervous system, possible mechanisms, and how electrical stimulation may be utilised as a novel technique to engineer “next-generation” organoids.
Collapse
Affiliation(s)
- Michelle O’Hara-Wright
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM + CSIC), Madrid, Spain
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
- *Correspondence: Anai Gonzalez-Cordero,
| |
Collapse
|
30
|
Gasparini SJ, Tessmer K, Reh M, Wieneke S, Carido M, Völkner M, Borsch O, Swiersy A, Zuzic M, Goureau O, Kurth T, Busskamp V, Zeck G, Karl MO, Ader M. Transplanted human cones incorporate and function in a murine cone degeneration model. J Clin Invest 2022; 132:154619. [PMID: 35482419 PMCID: PMC9197520 DOI: 10.1172/jci154619] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 04/26/2022] [Indexed: 11/17/2022] Open
Abstract
Once human photoreceptors die, they do not regenerate, thus, photoreceptor transplantation has emerged as a potential treatment approach for blinding diseases. Improvements in transplant organization, donor cell maturation, and synaptic connectivity to the host will be critical in advancing this technology for use in clinical practice. Unlike the unstructured grafts of prior cell-suspension transplantations into end-stage degeneration models, we describe the extensive incorporation of induced pluripotent stem cell (iPSC) retinal organoid–derived human photoreceptors into mice with cone dysfunction. This incorporative phenotype was validated in both cone-only as well as pan-photoreceptor transplantations. Rather than forming a glial barrier, Müller cells extended throughout the graft, even forming a series of adherens junctions between mouse and human cells, reminiscent of an outer limiting membrane. Donor-host interaction appeared to promote polarization as well as the development of morphological features critical for light detection, namely the formation of inner and well-stacked outer segments oriented toward the retinal pigment epithelium. Putative synapse formation and graft function were evident at both structural and electrophysiological levels. Overall, these results show that human photoreceptors interacted readily with a partially degenerated retina. Moreover, incorporation into the host retina appeared to be beneficial to graft maturation, polarization, and function.
Collapse
Affiliation(s)
| | - Karen Tessmer
- Ader Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Miriam Reh
- Department of Neurophysics, NMI Natural and Medical Sciences Institute at the University Tübingen, Reutlingen, Germany
| | - Stephanie Wieneke
- Karl Lab, Center for Regenerative Therapies TU Dresden and German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Madalena Carido
- Ader Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Manuela Völkner
- Karl Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Oliver Borsch
- Ader Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Anka Swiersy
- Busskamp Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Marta Zuzic
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Olivier Goureau
- Institut de la Vision, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Thomas Kurth
- Center for Molecular and Cellular Biology, Technische Universität (TU) Dresden, Dresden, Germany
| | - Volker Busskamp
- Department of Ophthalmology, University of Bonn, Bonn, Germany
| | - Günther Zeck
- Department of Neurophysics, NMI Natural and Medical Sciences Institute at the University Tübingen, Reutlingen, Germany
| | - Mike O Karl
- Karl Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| | - Marius Ader
- Ader Lab, Center for Regenerative Therapies TU Dresden, Dresden, Germany
| |
Collapse
|
31
|
Rashidi H, Leong YC, Venner K, Pramod H, Fei QZ, Jones OJR, Moulding D, Sowden JC. Generation of 3D retinal tissue from human pluripotent stem cells using a directed small molecule-based serum-free microwell platform. Sci Rep 2022; 12:6646. [PMID: 35459774 PMCID: PMC9033780 DOI: 10.1038/s41598-022-10540-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/08/2022] [Indexed: 11/09/2022] Open
Abstract
Retinal degenerative diseases are a leading cause of blindness worldwide with debilitating life-long consequences for the affected individuals. Cell therapy is considered a potential future clinical intervention to restore and preserve sight by replacing lost photoreceptors and/or retinal pigment epithelium. Development of protocols to generate retinal tissue from human pluripotent stem cells (hPSC), reliably and at scale, can provide a platform to generate photoreceptors for cell therapy and to model retinal disease in vitro. Here, we describe an improved differentiation platform to generate retinal organoids from hPSC at scale and free from time-consuming manual microdissection steps. The scale up was achieved using an agarose mould platform enabling generation of uniform self-assembled 3D spheres from dissociated hPSC in microwells. Subsequent retinal differentiation was efficiently achieved via a stepwise differentiation protocol using a number of small molecules. To facilitate clinical translation, xeno-free approaches were developed by substituting Matrigel™ and foetal bovine serum with recombinant laminin and human platelet lysate, respectively. Generated retinal organoids exhibited important features reminiscent of retinal tissue including correct site-specific localisation of proteins involved in phototransduction.
Collapse
Affiliation(s)
- Hassan Rashidi
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Yeh Chwan Leong
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Kerrie Venner
- UCL Institute of Neurology, Queens Square, University College London, London, UK
| | - Hema Pramod
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Qi-Zhen Fei
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Owen J R Jones
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Dale Moulding
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK
| | - Jane C Sowden
- Stem Cells and Regenerative Medicine Section, Birth Defects Research Centre, UCL Great Ormond Street Institute of Child Health, University College London and NIHR Great Ormond Street Hospital Biomedical Research Centre, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
32
|
One-step induction of photoreceptor-like cells from human iPSCs by delivering transcription factors. iScience 2022; 25:103987. [PMID: 35330684 PMCID: PMC8938283 DOI: 10.1016/j.isci.2022.103987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 01/10/2022] [Accepted: 02/24/2022] [Indexed: 11/22/2022] Open
|
33
|
Eintracht J, Harding P, Lima Cunha D, Moosajee M. Efficient embryoid-based method to improve generation of optic vesicles from human induced pluripotent stem cells. F1000Res 2022; 11:324. [PMID: 35811797 PMCID: PMC9218590 DOI: 10.12688/f1000research.108829.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/08/2022] [Indexed: 01/02/2023] Open
Abstract
Animal models have provided many insights into ocular development and disease, but they remain suboptimal for understanding human oculogenesis. Eye development requires spatiotemporal gene expression patterns and disease phenotypes can differ significantly between humans and animal models, with patient-associated mutations causing embryonic lethality reported in some animal models. The emergence of human induced pluripotent stem cell (hiPSC) technology has provided a new resource for dissecting the complex nature of early eye morphogenesis through the generation of three-dimensional (3D) cellular models. By using patient-specific hiPSCs to generate in vitro optic vesicle-like models, we can enhance the understanding of early developmental eye disorders and provide a pre-clinical platform for disease modelling and therapeutics testing. A major challenge of in vitro optic vesicle generation is the low efficiency of differentiation in 3D cultures. To address this, we adapted a previously published protocol of retinal organoid differentiation to improve embryoid body formation using a microwell plate. Established morphology, upregulated transcript levels of known early eye-field transcription factors and protein expression of standard retinal progenitor markers confirmed the optic vesicle/presumptive optic cup identity of in vitro models between day 20 and 50 of culture. This adapted protocol is relevant to researchers seeking a physiologically relevant model of early human ocular development and disease with a view to replacing animal models.
Collapse
Affiliation(s)
- Jonathan Eintracht
- Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Philippa Harding
- Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Dulce Lima Cunha
- Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London, EC1V 9EL, UK
- The Francis Crick Institute, London, NW1 1AT, UK
- Moorfields Eye Hospital NHS Foundation Trust, London, EC1V 2PD, UK
- Great Ormond Street Hospital for Children, London, WC1N 3JH, UK
| |
Collapse
|
34
|
Dorgau B, Georgiou M, Chaudhary A, Moya-Molina M, Collin J, Queen R, Hilgen G, Davey T, Hewitt P, Schmitt M, Kustermann S, Pognan F, Steel DH, Sernagor E, Armstrong L, Lako M. Human Retinal Organoids Provide a Suitable Tool for Toxicological Investigations: A Comprehensive Validation Using Drugs and Compounds Affecting the Retina. Stem Cells Transl Med 2022; 11:159-177. [PMID: 35298655 PMCID: PMC8929478 DOI: 10.1093/stcltm/szab010] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/13/2021] [Indexed: 12/04/2022] Open
Abstract
Retinal drug toxicity screening is essential for the development of safe treatment strategies for a large number of diseases. To this end, retinal organoids derived from human pluripotent stem cells (hPSCs) provide a suitable screening platform due to their similarity to the human retina and the ease of generation in large-scale formats. In this study, two hPSC cell lines were differentiated to retinal organoids, which comprised all key retinal cell types in multiple nuclear and synaptic layers. Single-cell RNA-Seq of retinal organoids indicated the maintenance of retinal ganglion cells and development of bipolar cells: both cell types segregated into several subtypes. Ketorolac, digoxin, thioridazine, sildenafil, ethanol, and methanol were selected as key compounds to screen on retinal organoids because of their well-known retinal toxicity profile described in the literature. Exposure of the hPSC-derived retinal organoids to digoxin, thioridazine, and sildenafil resulted in photoreceptor cell death, while digoxin and thioridazine additionally affected all other cell types, including Müller glia cells. All drug treatments caused activation of astrocytes, indicated by dendrites sprouting into neuroepithelium. The ability to respond to light was preserved in organoids although the number of responsive retinal ganglion cells decreased after drug exposure. These data indicate similar drug effects in organoids to those reported in in vivo models and/or in humans, thus providing the first robust experimental evidence of their suitability for toxicological studies.
Collapse
Affiliation(s)
- Birthe Dorgau
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcells Biotech, Biosphere, Newcastle Helix, Newcastle upon Tyne, UK
| | - Maria Georgiou
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Alexander Chaudhary
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Marina Moya-Molina
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcells Biotech, Biosphere, Newcastle Helix, Newcastle upon Tyne, UK
| | - Joseph Collin
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Rachel Queen
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Gerrit Hilgen
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Northumbria University, Applied Sciences, Faculty of Health and Life Science, Newcastle upon Tyne, UK
| | - Tracey Davey
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Electron Microscopy Research Services, Newcastle University, Newcastle upon Tyne, UK
| | | | | | - Stefan Kustermann
- Pharmaceutical Sciences, F. Hoffmann-La Roche, Pharma Research and Early Development, Roche Innovation Center Basel, Switzerland
| | | | - David H Steel
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Evelyne Sernagor
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| | - Lyle Armstrong
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
- Newcells Biotech, Biosphere, Newcastle Helix, Newcastle upon Tyne, UK
| | - Majlinda Lako
- Newcastle University, Biosciences Institute, Faculty of Medical Sciences, Newcastle upon Tyne, UK
| |
Collapse
|
35
|
Bellapianta A, Cetkovic A, Bolz M, Salti A. Retinal Organoids and Retinal Prostheses: An Overview. Int J Mol Sci 2022; 23:2922. [PMID: 35328339 PMCID: PMC8953078 DOI: 10.3390/ijms23062922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 01/27/2023] Open
Abstract
Despite the progress of modern medicine in the last decades, millions of people diagnosed with retinal dystrophies (RDs), such as retinitis pigmentosa, or age-related diseases, such as age-related macular degeneration, are suffering from severe visual impairment or even legal blindness. On the one hand, the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) and the progress of three-dimensional (3D) retinal organoids (ROs) technology provide a great opportunity to study, understand, and even treat retinal diseases. On the other hand, research advances in the field of electronic retinal prosthesis using inorganic photovoltaic polymers and the emergence of organic semiconductors represent an encouraging therapeutical strategy to restore vision to patients at the late onset of the disease. This review will provide an overview of the latest advancement in both fields. We first describe the retina and the photoreceptors, briefly mention the most used RD animal models, then focus on the latest RO differentiation protocols, carry out an overview of the current technology on inorganic and organic retinal prostheses to restore vision, and finally summarize the potential utility and applications of ROs.
Collapse
Affiliation(s)
| | | | | | - Ahmad Salti
- Center for Medical Research, Faculty of Medicine, University Clinic for Ophthalmology and Optometry, Johannes Kepler University Linz, 4020 Linz, Austria; (A.B.); (A.C.); (M.B.)
| |
Collapse
|
36
|
Isla-Magrané H, Veiga A, García-Arumí J, Duarri A. Multiocular organoids from human induced pluripotent stem cells displayed retinal, corneal, and retinal pigment epithelium lineages. Stem Cell Res Ther 2021; 12:581. [PMID: 34809716 PMCID: PMC8607587 DOI: 10.1186/s13287-021-02651-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/05/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recently, great efforts have been made to design protocols for obtaining ocular cells from human stem cells to model diseases or for regenerative purposes. Current protocols generally focus on isolating retinal cells, retinal pigment epithelium (RPE), or corneal cells and fail to recapitulate the complexity of the tissue during eye development. Here, the generation of more advanced in vitro multiocular organoids from human induced pluripotent stem cells (hiPSCs) is demonstrated. METHODS A 2-step method was established to first obtain self-organized multizone ocular progenitor cells (mzOPCs) from 2D hiPSC cultures within three weeks. Then, after the cells were manually isolated and grown in suspension, 3D multiocular organoids were generated to model important cellular features of developing eyes. RESULTS In the 2D culture, self-formed mzOPCs spanned the neuroectoderm, surface ectoderm, neural crest, and RPE, mimicking early stages of eye development. After lifting, mzOPCs developed into different 3D multiocular organoids composed of multiple cell lineages including RPE, retina, and cornea, and interactions between the different cell types and regions of the eye system were observed. Within these organoids, the retinal regions exhibited correct layering and contained all major retinal cell subtypes as well as retinal morphological cues, whereas the corneal regions closely resembled the transparent ocular-surface epithelium and contained of corneal, limbal, and conjunctival epithelial cells. The arrangement of RPE cells also formed organoids composed of polarized pigmented epithelial cells at the surface that were completely filled with collagen matrix. CONCLUSIONS This approach clearly demonstrated the advantages of the combined 2D-3D construction tissue model as it provided a more ocular native-like cellular environment than that of previous models. In this complex preparations, multiocular organoids may be used to model the crosstalk between different cell types in eye development and disease.
Collapse
Affiliation(s)
- Helena Isla-Magrané
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Veiga
- Regenerative Medicine Program IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - José García-Arumí
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
- Department of Ophthalmology, Vall d'Hebron Hospital Universitari, Barcelona, Spain
- Department of Ophthalmology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Anna Duarri
- Ophthalmology Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
| |
Collapse
|
37
|
Strachan EL, Mac White-Begg D, Crean J, Reynolds AL, Kennedy BN, O’Sullivan NC. The Role of Mitochondria in Optic Atrophy With Autosomal Inheritance. Front Neurosci 2021; 15:784987. [PMID: 34867178 PMCID: PMC8634724 DOI: 10.3389/fnins.2021.784987] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Optic atrophy (OA) with autosomal inheritance is a form of optic neuropathy characterized by the progressive and irreversible loss of vision. In some cases, this is accompanied by additional, typically neurological, extra-ocular symptoms. Underlying the loss of vision is the specific degeneration of the retinal ganglion cells (RGCs) which form the optic nerve. Whilst autosomal OA is genetically heterogenous, all currently identified causative genes appear to be associated with mitochondrial organization and function. However, it is unclear why RGCs are particularly vulnerable to mitochondrial aberration. Despite the relatively high prevalence of this disorder, there are currently no approved treatments. Combined with the lack of knowledge concerning the mechanisms through which aberrant mitochondrial function leads to RGC death, there remains a clear need for further research to identify the underlying mechanisms and develop treatments for this condition. This review summarizes the genes known to be causative of autosomal OA and the mitochondrial dysfunction caused by pathogenic mutations. Furthermore, we discuss the suitability of available in vivo models for autosomal OA with regards to both treatment development and furthering the understanding of autosomal OA pathology.
Collapse
Affiliation(s)
- Elin L. Strachan
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Delphi Mac White-Begg
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - John Crean
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Diabetes Complications Research Centre, Conway Institute of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Alison L. Reynolds
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Breandán N. Kennedy
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Niamh C. O’Sullivan
- UCD Conway Institute, University College Dublin, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
38
|
Zhang X, Wang W, Jin ZB. Retinal organoids as models for development and diseases. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:33. [PMID: 34719743 PMCID: PMC8557999 DOI: 10.1186/s13619-021-00097-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
The evolution of pluripotent stem cell-derived retinal organoids (ROs) has brought remarkable opportunities for developmental studies while also presenting new therapeutic avenues for retinal diseases. With a clear understanding of how well these models mimic native retinas, such preclinical models may be crucial tools that are widely used for the more efficient translation of studies into novel treatment strategies for retinal diseases. Genetic modifications or patient-derived ROs can allow these models to simulate the physical microenvironments of the actual disease process. However, we are currently at the beginning of the three-dimensional (3D) RO era, and a general quantitative technology for analyzing ROs derived from numerous differentiation protocols is still missing. Continued efforts to improve the efficiency and stability of differentiation, as well as understanding the disparity between the artificial retina and the native retina and advancing the current treatment strategies, will be essential in ensuring that these scientific advances can benefit patients with retinal disease. Herein, we briefly discuss RO differentiation protocols, the current applications of RO as a disease model and the treatments for retinal diseases by using RO modeling, to have a clear view of the role of current ROs in retinal development and diseases.
Collapse
Affiliation(s)
- Xiao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Wen Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Science Key Laboratory, Beijing, 100730, China.
| |
Collapse
|
39
|
Li J, Chen Y, Ouyang S, Ma J, Sun H, Luo L, Chen S, Liu Y. Generation and Staging of Human Retinal Organoids Based on Self-Formed Ectodermal Autonomous Multi-Zone System. Front Cell Dev Biol 2021; 9:732382. [PMID: 34631711 PMCID: PMC8493070 DOI: 10.3389/fcell.2021.732382] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/03/2021] [Indexed: 01/12/2023] Open
Abstract
Methods for stem cell-derived, three-dimensional retinal organoids induction have been established and shown great potential for retinal development modeling and drug screening. Herein, we reported an exogenous-factors-free and robust method to generate retinal organoids based on “self-formed ectodermal autonomous multi-zone” (SEAM) system, a two-dimensional induction scheme that can synchronously generate multiple ocular cell lineages. Characterized by distinct morphological changes, the differentiation of the obtained retinal organoids could be staged into the early and late differentiation phases. During the early differentiation stage, retinal ganglion cells, cone photoreceptor cells (PRs), amacrine cells, and horizontal cells developed; whereas rod PRs, bipolar cells, and Müller glial cells were generated in the late differentiation phase, resembling early-phase and late-phase retinogenesis in vivo. Additionally, we modified the maintenance strategy for the retinal organoids and successfully promoted their long-term survival. Using 3D immunofluorescence image reconstruction and transmission electron microscopy, the substantial mature PRs with outer segment, inner segment and ribbon synapse were demonstrated. Besides, the retinal pigment epithelium (RPE) was induced with distinct boundary and the formation of ciliary margin was observed by co-suspending retina organoids with the zone containing RPE. The obtained RPE could be expanded and displayed similar marker expression, ultrastructural feature and functional phagocytosis to native RPE. Thus, this research described a simple and robust system which enabled generation of retina organoids with substantial mature PRs, RPE and the ciliary margin without the need of exogenous factors, providing a new platform for research of retinogenesis and retinal translational application.
Collapse
Affiliation(s)
- Jinyan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yijia Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shuai Ouyang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jingyu Ma
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Hui Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lixia Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
40
|
Afanasyeva TAV, Corral-Serrano JC, Garanto A, Roepman R, Cheetham ME, Collin RWJ. A look into retinal organoids: methods, analytical techniques, and applications. Cell Mol Life Sci 2021; 78:6505-6532. [PMID: 34420069 PMCID: PMC8558279 DOI: 10.1007/s00018-021-03917-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 12/15/2022]
Abstract
Inherited retinal diseases (IRDs) cause progressive loss of light-sensitive photoreceptors in the eye and can lead to blindness. Gene-based therapies for IRDs have shown remarkable progress in the past decade, but the vast majority of forms remain untreatable. In the era of personalised medicine, induced pluripotent stem cells (iPSCs) emerge as a valuable system for cell replacement and to model IRD because they retain the specific patient genome and can differentiate into any adult cell type. Three-dimensional (3D) iPSCs-derived retina-like tissue called retinal organoid contains all major retina-specific cell types: amacrine, bipolar, horizontal, retinal ganglion cells, Müller glia, as well as rod and cone photoreceptors. Here, we describe the main applications of retinal organoids and provide a comprehensive overview of the state-of-art analysis methods that apply to this model system. Finally, we will discuss the outlook for improvements that would bring the cellular model a step closer to become an established system in research and treatment development of IRDs.
Collapse
Affiliation(s)
- Tess A V Afanasyeva
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands
| | | | - Alejandro Garanto
- Department of Pediatrics, Amalia Children's Hospital and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ronald Roepman
- Department of Human Genetics and Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michael E Cheetham
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Rob W J Collin
- Department of Human Genetics and Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA, Nijmegen, The Netherlands.
| |
Collapse
|
41
|
Retinal Organoid Technology: Where Are We Now? Int J Mol Sci 2021; 22:ijms221910244. [PMID: 34638582 PMCID: PMC8549701 DOI: 10.3390/ijms221910244] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022] Open
Abstract
It is difficult to regenerate mammalian retinal cells once the adult retina is damaged, and current clinical approaches to retinal damages are very limited. The introduction of the retinal organoid technique empowers researchers to study the molecular mechanisms controlling retinal development, explore the pathogenesis of retinal diseases, develop novel treatment options, and pursue cell/tissue transplantation under a certain genetic background. Here, we revisit the historical background of retinal organoid technology, categorize current methods of organoid induction, and outline the obstacles and potential solutions to next-generation retinal organoids. Meanwhile, we recapitulate recent research progress in cell/tissue transplantation to treat retinal diseases, and discuss the pros and cons of transplanting single-cell suspension versus retinal organoid sheet for cell therapies.
Collapse
|
42
|
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: 0.8] [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
|
43
|
Yang C, Georgiou M, Atkinson R, Collin J, Al-Aama J, Nagaraja-Grellscheid S, Johnson C, Ali R, Armstrong L, Mozaffari-Jovin S, Lako M. Pre-mRNA Processing Factors and Retinitis Pigmentosa: RNA Splicing and Beyond. Front Cell Dev Biol 2021; 9:700276. [PMID: 34395430 PMCID: PMC8355544 DOI: 10.3389/fcell.2021.700276] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Retinitis pigmentosa (RP) is the most common inherited retinal disease characterized by progressive degeneration of photoreceptors and/or retinal pigment epithelium that eventually results in blindness. Mutations in pre-mRNA processing factors (PRPF3, 4, 6, 8, 31, SNRNP200, and RP9) have been linked to 15–20% of autosomal dominant RP (adRP) cases. Current evidence indicates that PRPF mutations cause retinal specific global spliceosome dysregulation, leading to mis-splicing of numerous genes that are involved in a variety of retina-specific functions and/or general biological processes, including phototransduction, retinol metabolism, photoreceptor disk morphogenesis, retinal cell polarity, ciliogenesis, cytoskeleton and tight junction organization, waste disposal, inflammation, and apoptosis. Importantly, additional PRPF functions beyond RNA splicing have been documented recently, suggesting a more complex mechanism underlying PRPF-RPs driven disease pathogenesis. The current review focuses on the key RP-PRPF genes, depicting the current understanding of their roles in RNA splicing, impact of their mutations on retinal cell’s transcriptome and phenome, discussed in the context of model species including yeast, zebrafish, and mice. Importantly, information on PRPF functions beyond RNA splicing are discussed, aiming at a holistic investigation of PRPF-RP pathogenesis. Finally, work performed in human patient-specific lab models and developing gene and cell-based replacement therapies for the treatment of PRPF-RPs are thoroughly discussed to allow the reader to get a deeper understanding of the disease mechanisms, which we believe will facilitate the establishment of novel and better therapeutic strategies for PRPF-RP patients.
Collapse
Affiliation(s)
- Chunbo Yang
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Maria Georgiou
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Robert Atkinson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jumana Al-Aama
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Colin Johnson
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, United Kingdom
| | - Robin Ali
- King's College London, London, United Kingdom
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Sina Mozaffari-Jovin
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Cellular Biochemistry, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
44
|
Wagstaff EL, Heredero Berzal A, Boon CJF, Quinn PMJ, ten Asbroek ALMA, Bergen AA. The Role of Small Molecules and Their Effect on the Molecular Mechanisms of Early Retinal Organoid Development. Int J Mol Sci 2021; 22:7081. [PMID: 34209272 PMCID: PMC8268497 DOI: 10.3390/ijms22137081] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Early in vivo embryonic retinal development is a well-documented and evolutionary conserved process. The specification towards eye development is temporally controlled by consecutive activation or inhibition of multiple key signaling pathways, such as the Wnt and hedgehog signaling pathways. Recently, with the use of retinal organoids, researchers aim to manipulate these pathways to achieve better human representative models for retinal development and disease. To achieve this, a plethora of different small molecules and signaling factors have been used at various time points and concentrations in retinal organoid differentiations, with varying success. Additions differ from protocol to protocol, but their usefulness or efficiency has not yet been systematically reviewed. Interestingly, many of these small molecules affect the same and/or multiple pathways, leading to reduced reproducibility and high variability between studies. In this review, we make an inventory of the key signaling pathways involved in early retinogenesis and their effect on the development of the early retina in vitro. Further, we provide a comprehensive overview of the small molecules and signaling factors that are added to retinal organoid differentiation protocols, documenting the molecular and functional effects of these additions. Lastly, we comparatively evaluate several of these factors using our established retinal organoid methodology.
Collapse
Affiliation(s)
- Ellie L. Wagstaff
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center—New York-Presbyterian Hospital, New York, NY 10032, USA;
| | | | - Arthur A. Bergen
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Netherlands Institute for Neuroscience (NIN-KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
45
|
Singh D, Chen X, Xia T, Ghiassi-Nejad M, Tainsh L, Adelman RA, Rizzolo LJ. Partially Differentiated Neuroretinal Cells Promote Maturation of the Retinal Pigment Epithelium. Invest Ophthalmol Vis Sci 2021; 61:9. [PMID: 33151282 PMCID: PMC7671856 DOI: 10.1167/iovs.61.13.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Purpose Many studies have demonstrated the ability of the retinal pigment epithelium (RPE) to foster the maturation of the developing retina. Few studies have examined the reciprocal effects of developing retina on the RPE. Methods RPE isolated from human fetal RPE or differentiated from human stem cells was cultured on Transwell filter inserts. Retinal progenitor cells (RPCs) were differentiated from human stem cells and cultured on a planar scaffold composed of gelatin, chondroitin sulfate, hyaluronic acid, and laminin-521. Cultures were analyzed by quantitative RT-PCR, immunofluorescence, immunoblotting, and transepithelial electrical resistance (TER). Results RPCs initially differentiated into several retina-like cell types that segregated from one another and formed loosely organized layers or zones. With time, the presumptive photoreceptor and ganglion cell layers persisted, but the intervening zone became dominated by cells that expressed glial markers with no evidence of bipolar cells or interneurons. Co-culture of this underdeveloped retinoid with the RPE resulted in a thickened layer of recoverin-positive cells but did not prevent the loss of interneuron markers in the intervening zone. Although photoreceptor inner and outer segments were not observed, immunoblots revealed that co-culture increased expression of rhodopsin and red/green opsin. Co-culture of the RPE with this underdeveloped retinal culture increased the TER of the RPE and the expression of RPE signature genes. Conclusions These studies indicated that an immature neurosensory retina can foster maturation of the RPE; however, the ability of RPE alone to foster maturation of the neurosensory retina is limited.
Collapse
Affiliation(s)
- Deepti Singh
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| | - Xiaoyu Chen
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tina Xia
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| | - Maryam Ghiassi-Nejad
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| | - Laurel Tainsh
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| | - Ron A Adelman
- Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| | - Lawrence J Rizzolo
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, Connecticut, United States.,Department of Ophthalmology and Visual Sciences, Yale School of Medicine, Yale University, New Haven, Connecticut, United States
| |
Collapse
|
46
|
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: 24] [Impact Index Per Article: 6.0] [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
|
47
|
Cuevas E, Holder DL, Alshehri AH, Tréguier J, Lakowski J, Sowden JC. NRL -/- gene edited human embryonic stem cells generate rod-deficient retinal organoids enriched in S-cone-like photoreceptors. Stem Cells 2021; 39:414-428. [PMID: 33400844 PMCID: PMC8438615 DOI: 10.1002/stem.3325] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/27/2022]
Abstract
Organoid cultures represent a unique tool to investigate the developmental complexity of tissues like the human retina. NRL is a transcription factor required for the specification and homeostasis of mammalian rod photoreceptors. In Nrl-deficient mice, photoreceptor precursor cells do not differentiate into rods, and instead follow a default photoreceptor specification pathway to generate S-cone-like cells. To investigate whether this genetic switch mechanism is conserved in humans, we used CRISPR/Cas9 gene editing to engineer an NRL-deficient embryonic stem cell (ESC) line (NRL-/- ), and differentiated it into retinal organoids. Retinal organoids self-organize and resemble embryonic optic vesicles (OVs) that recapitulate the natural histogenesis of rods and cone photoreceptors. NRL-/- OVs develop comparably to controls, and exhibit a laminated, organized retinal structure with markers of photoreceptor synaptogenesis. Using immunohistochemistry and quantitative polymerase chain reaction (qPCR), we observed that NRL-/- OVs do not express NRL, or other rod photoreceptor markers directly or indirectly regulated by NRL. On the contrary, they show an abnormal number of photoreceptors positive for S-OPSIN, which define a primordial subtype of cone, and overexpress other cone genes indicating a conserved molecular switch in mammals. This study represents the first evidence in a human in vitro ESC-derived organoid system that NRL is required to define rod identity, and that in its absence S-cone-like cells develop as the default photoreceptor cell type. It shows how gene edited retinal organoids provide a useful system to investigate human photoreceptor specification, relevant for efforts to generate cells for transplantation in retinal degenerative diseases.
Collapse
Affiliation(s)
- Elisa Cuevas
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Daniel L. Holder
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Ashwak H. Alshehri
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Julie Tréguier
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| | - Jörn Lakowski
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
- Centre for Human Development, Stem Cells and RegenerationUniversity of SouthamptonSouthamptonUK
| | - Jane C. Sowden
- UCL Great Ormond Street Institute of Child HealthUniversity College London and NIHR Great Ormond Street Hospital Biomedical Research CentreLondonUK
| |
Collapse
|
48
|
Zerti D, Molina MM, Dorgau B, Mearns S, Bauer R, Al-Aama J, Lako M. IGFBPs mediate IGF-1's functions in retinal lamination and photoreceptor development during pluripotent stem cell differentiation to retinal organoids. Stem Cells 2021; 39:458-466. [PMID: 33442906 DOI: 10.1002/stem.3331] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/06/2020] [Accepted: 12/10/2020] [Indexed: 02/02/2023]
Abstract
Development of the retina is regulated by growth factors, such as insulin-like growth factors 1 and 2 (IGF-1/2), which coordinate proliferation, differentiation, and maturation of the neuroepithelial precursors cells. In the circulation, IGF-1/2 are transported by the insulin growth factor binding proteins (IGFBPs) family members. IGFBPs can impact positively and negatively on IGF-1, by making it available or sequestering IGF-1 to or from its receptor. In this study, we investigated the expression of IGFBPs and their role in the generation of human retinal organoids from human pluripotent stem cells, showing a dynamic expression pattern suggestive of different IGFBPs being used in a stage-specific manner to mediate IGF-1 functions. Our data show that IGF-1 addition to culture media facilitated the generation of retinal organoids displaying the typical laminated structure and photoreceptor maturation. The organoids cultured in the absence of IGF-1, lacked the typical laminated structure at the early stages of differentiation and contained significantly less photoreceptors and more retinal ganglion cells at the later stages of differentiation, confirming the positive effects of IGF-1 on retinal lamination and photoreceptor development. The organoids cultured with the IGFBP inhibitor (NBI-31772) and IGF-1 showed lack of retinal lamination at the early stages of differentiation, an increased propensity to generate horizontal cells at mid-stages of differentiation and reduced photoreceptor development at the later stages of differentiation. Together these data suggest that IGFBPs enable IGF-1's role in retinal lamination and photoreceptor development in a stage-specific manner.
Collapse
Affiliation(s)
- Darin Zerti
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Microscopy Centre and Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Marina Moya Molina
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Birthe Dorgau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Sarah Mearns
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Roman Bauer
- Department of Computer Science, University of Surrey, Guildford, UK
| | - Jumana Al-Aama
- Department of Genetic Medicine and Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
49
|
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: 2.0] [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
|
50
|
Zerti D, Hilgen G, Dorgau B, Collin J, Ader M, Armstrong L, Sernagor E, Lako M. Transplanted pluripotent stem cell-derived photoreceptor precursors elicit conventional and unusual light responses in mice with advanced retinal degeneration. STEM CELLS (DAYTON, OHIO) 2021; 39:882-896. [PMID: 33657251 DOI: 10.1002/stem.3365] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/19/2021] [Indexed: 11/09/2022]
Abstract
Retinal dystrophies often lead to blindness. Developing therapeutic interventions to restore vision is therefore of paramount importance. Here we demonstrate the ability of pluripotent stem cell-derived cone precursors to engraft and restore light responses in the Pde6brd1 mouse, an end-stage photoreceptor degeneration model. Our data show that up to 1.5% of precursors integrate into the host retina, differentiate into cones, and engraft in close apposition to the host bipolar cells. Half of the transplanted mice exhibited visual behavior and of these 33% showed binocular light sensitivity. The majority of retinal ganglion cells exhibited contrast-sensitive ON, OFF or ON-OFF light responses and even motion sensitivity; however, quite a few exhibited unusual responses (eg, light-induced suppression), presumably reflecting remodeling of the neural retina. Our data indicate that despite relatively low engraftment yield, pluripotent stem cell-derived cone precursors can elicit light responsiveness even at advanced degeneration stages. Further work is needed to improve engraftment yield and counteract retinal remodeling to achieve useful clinical applications.
Collapse
Affiliation(s)
- Darin Zerti
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Microscopy Centre and Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Gerrit Hilgen
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Birthe Dorgau
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Joseph Collin
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Marius Ader
- CRTD/Center for Regenerative Therapies Dresden, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Lyle Armstrong
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Evelyne Sernagor
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Majlinda Lako
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|