1
|
Huang Y, Ye Q, Wang J, Zhu K, Yang H, Jiang X, Shen M. Recent progress in the identification and in vitro culture of skin organoids. Regen Ther 2025; 29:341-351. [PMID: 40242086 PMCID: PMC12000699 DOI: 10.1016/j.reth.2025.01.001] [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: 09/07/2024] [Revised: 11/13/2024] [Accepted: 01/04/2025] [Indexed: 04/18/2025] Open
Abstract
An organoid is a cell-based structure that shows organ-specific properties and shares a similar spatial organization as the corresponding organ. Organoids possess powerful capability to reproduce the key functions of the associated organ structures, and their similarity to the organs makes them physiologically relevant systems. The primary challenge associated with the development of skin organoids is the complexity of the human skin architecture, which encompasses the epidermis and the dermis as well as accessory structures, including hair follicles, sweat glands, and sebaceous glands, that perform various functions such as thermoregulation. The ultimate objectives of developing skin organoids are to regenerate the complete skin structure in vitro and reconstruct the skin in vivo. Consequently, safety, reliability, and the fidelity of the tissue interfaces are key considerations in this process. For this purpose, the present article reviews the most recent advances in this field, focusing on the cell sources, culture methods, culture conditions, and biomarkers for identifying the structure and function of skin organoids developed in vitro or in vivo. The subsequent sections summarize the recent applications of skin organoids in related disease diagnosis and treatments, and discuss the future prospects of these organoids in terms of clinical applications. This review of skin organoids can provide an important foundation for studies on human skin development, disease modeling, and reconstructive surgery, with broad utility for promising future opportunities in both biomedical research and clinical practice.
Collapse
Affiliation(s)
- Yanan Huang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Qing Ye
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | | | - Kaimin Zhu
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Haojie Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, China
| | - Xiaoping Jiang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Meihua Shen
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| |
Collapse
|
2
|
Gómez-Escribano AP, García-García G, Pérez-Santamarina E, Aller-Mañas E, Vázquez-Manrique RP, Millán-Salvador JM. Innovative therapies for inherited retinal dystrophies: navigating DNA, RNA, and protein approaches. EBioMedicine 2025; 116:105751. [PMID: 40367639 DOI: 10.1016/j.ebiom.2025.105751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/30/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
Gene therapy has become a promising treatment for inherited retinal dystrophies (IRDs), but understanding the genetic mechanisms involved is essential for its success. Various approaches, such as gene augmentation and DNA/RNA-based therapies, have shown effectiveness in some clinical trials. However, gene augmentation is not effective for some dominant mutations and genome editing produces off-target effects. Here, we review safer and viable alternatives that are being evaluated in preclinical models and clinical trials to address this challenge. We propose a novel perspective based on protein-targeting therapies, which although promising, remain unexplored. We suggest that frameshift variants, which produce novel epitopes, may allow for the development of mutant protein targeting agents for selective protein degradation. This approach could be useful for dominant variants, where gene replacement is ineffective. By examining these approaches, we aim to guide more targeted and effective gene therapies for IRDs, offering potential treatments where current methods fall short.
Collapse
Affiliation(s)
- Ana Pilar Gómez-Escribano
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| | - Gema García-García
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | | | - Elena Aller-Mañas
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - Rafael Pascual Vázquez-Manrique
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain
| | - José María Millán-Salvador
- Laboratory of Molecular, Cellular and Genomic Biomedicine, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain; Joint Unit for Rare Diseases IIS La Fe-CIPF, Valencia, Spain.
| |
Collapse
|
3
|
Chen Y, Xu H, Xiao L, Zhang M, Yan N. Single-cell RNA sequencing in the study of human retinal organoids. Exp Eye Res 2025; 256:110417. [PMID: 40320034 DOI: 10.1016/j.exer.2025.110417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/26/2025] [Accepted: 05/01/2025] [Indexed: 05/10/2025]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed the study of retinal development and diseases by enabling a detailed analysis of cellular diversity within retinal organoids (ROs). ROs generated from pluripotent stem cells mimic the essential characteristics of the human retina and provide a valuable in vitro model for investigating retinal development, cell interactions, and disease mechanisms. This review summarizes the application of scRNA-seq on RO research, emphasizing its capacity to identify distinct cell populations, uncover developmental trajectories, and reveal the molecular signatures of retinal diseases. scRNA-seq provides new insights into retinal neurogenesis, cellular diversity, and the pathophysiology of retinal degenerative diseases. This technology has enabled the identification of novel biomarkers and potential therapeutic targets. Integrating scRNA-seq with other technologies, such as spatial transcriptomics and CRISPR-based screening, can further deepen our understanding of retinal biology and improve treatment strategies.
Collapse
Affiliation(s)
- Yi Chen
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hanyue Xu
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Lirong Xiao
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Naihong Yan
- Department of Ophthalmology and Research Laboratory of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
4
|
Méjécase C, Zhou Y, Owen N, Soro-Barrio P, Cheloni R, Nair N, Sarkar H, Toualbi L, Moosajee M. Dominant RDH12-retinitis pigmentosa impairs photoreceptor development and implicates cone involvement in retinal organoids. Front Cell Dev Biol 2025; 13:1511066. [PMID: 40365019 PMCID: PMC12069300 DOI: 10.3389/fcell.2025.1511066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Retinal dehydrogenase 12 (RDH12) is a photoreceptor NADPH-dependent retinal reductase enzyme, converting all-trans-retinal to all-trans-retinol. Heterozygous variants in RDH12 cause a rare autosomal dominant (AD) retinitis pigmentosa. Methods As no disease models exist, we generated human induced pluripotent stem cell-derived retinal organoids (RO) from an RDH12-AD patient (with pathogenic variant c.759del, p.Phe254Leufs*24) alongside a healthy, unaffected control. Results RDH12-AD RO exhibited correct localization of RDH12 to the photoreceptor inner segments up to week 44; however, transmission electron microscopy at week 37 showed that photoreceptors were less abundant and shorter in length. Visual cone function, retinol biosynthesis, and the vitamin A pathway were also highly disrupted at week 44. Discussion Our study is the first to describe an RDH12-AD disease model with pathology at later stages of photoreceptor differentiation, in keeping with the milder disease course seen in humans. It provides insights into the etiology and possible targets for future therapeutic development.
Collapse
Affiliation(s)
- Cécile Méjécase
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London, United Kingdom
| | - Ya Zhou
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Nicholas Owen
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
| | - Pablo Soro-Barrio
- Bioinformatics and Biostatistics Science Technology Platform, The Francis Crick Institute, London, United Kingdom
| | - Riccardo Cheloni
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
- Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Neelima Nair
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
| | - Hajrah Sarkar
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London, United Kingdom
| | - Lyes Toualbi
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London, United Kingdom
| | - Mariya Moosajee
- Development, Ageing and Disease, UCL Institute of Ophthalmology, London, United Kingdom
- Ocular Genomics and Therapeutics, The Francis Crick Institute, London, United Kingdom
- Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
5
|
Zhang P, Xu Z. The advancements in precision medicine for Leber congenital amaurosis: Breakthroughs from genetic diagnosis to therapy. Surv Ophthalmol 2025:S0039-6257(25)00070-0. [PMID: 40311816 DOI: 10.1016/j.survophthal.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Leber congenital amaurosis (LCA) is a hereditary retinal disease, typically manifesting as severe vision impairment in infancy. With the advancement of precision medicine, genetic diagnosis and targeted therapies offer new hope for LCA patients, significantly improving both diagnostic accuracy and therapeutic efficacy. We summarize the epidemiological characteristics, clinical manifestations, and molecular genetics underlying LCA. It also highlights recent developments in precision treatment strategies, including gene replacement therapy, CRISPR/Cas9-mediated gene editing, and antisense oligonucleotide therapies. In addition, we discuss the applications of induced pluripotent stem cells and retinal organoids in LCA treatment research. Furthermore, we explore preventive strategies and future treatment directions for LCA, including the development of novel gene therapy vectors, the optimization of combinatorial treatment strategies, and the formulation of personalized treatment approaches. These advancements hold significant potential to offer improved treatment options and enhance the quality of life for LCA patients.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
6
|
Song HB, Campello L, Mondal A, Chen HY, English MA, Glen M, Vanlandingham P, Farjo R, Swaroop A. Sex-specific attenuation of photoreceptor degeneration by reserpine in a rhodopsin P23H rat model of autosomal dominant retinitis pigmentosa. eLife 2025; 14:RP103888. [PMID: 40231721 PMCID: PMC11999695 DOI: 10.7554/elife.103888] [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: 04/16/2025] Open
Abstract
Inherited retinal degenerations (IRDs) constitute a group of clinically and genetically diverse vision-impairing disorders. Retinitis pigmentosa (RP), the most common form of IRD, is characterized by gradual dysfunction and degeneration of rod photoreceptors, followed by the loss of cone photoreceptors. Recently, we identified reserpine as a lead molecule for maintaining rod survival in mouse and human retinal organoids as well as in the rd16 mouse, which phenocopy Leber congenital amaurosis caused by mutations in the cilia-centrosomal gene CEP290 (Chen et al., 2023). Here, we show the therapeutic potential of reserpine in a rhodopsin P23H rat model of autosomal dominant RP. At postnatal day (P) 68, when males and females are analyzed together, the reserpine-treated rats exhibit higher rod-derived scotopic b-wave amplitudes compared to the controls with little or no change in scotopic a-wave or cone-derived photopic b-wave. Interestingly, the reserpine-treated female rats display enhanced scotopic a- and b-waves and photopic b-wave responses at P68, along with a better contrast threshold and increased outer nuclear layer thickness. The female rats demonstrate better preservation of both rod and cone photoreceptors following reserpine treatment. Retinal transcriptome analysis reveals sex-specific responses to reserpine, with significant upregulation of phototransduction genes and proteostasis-related pathways, and notably, genes associated with stress response. This study builds upon our previously reported results reaffirming the potential of reserpine for gene-agnostic treatment of IRDs and emphasizes the importance of biological sex in retinal disease research and therapy development.
Collapse
Affiliation(s)
- Hyun Beom Song
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Anupam Mondal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Holly Y Chen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Milton A English
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Michael Glen
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | | | | | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
7
|
Galindo-Cabello N, Caballano-Infantes E, Benites G, Pastor-Idoate S, Diaz-Corrales FJ, Usategui-Martín R. Retinal Organoids: Innovative Tools for Understanding Retinal Degeneration. Int J Mol Sci 2025; 26:3263. [PMID: 40244125 PMCID: PMC11990004 DOI: 10.3390/ijms26073263] [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: 03/01/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Retinal degenerative diseases (RDDs) comprise diverse genetic and phenotypic conditions that cause progressive retinal dysfunction and cell loss, leading to vision impairment or blindness. Most RDDs lack appropriate animal models for their study, which affects understanding their disease mechanisms and delays the progress of new treatment development. Recent advances in stem cell engineering, omics, and organoid technology are facilitating research into diseases for which there are no previously existing models. The development of retinal organoids produced from human stem cells has impacted the study of retinal development as well as the development of in vitro models of diseases, opening possibilities for applications in regenerative medicine, drug discovery, and precision medicine. In this review, we recapitulate research in the retinal organoid models for RDD, mentioning some of the main pathways underlying retinal neurodegeneration that can be studied in these new models, as well as their limitations and future challenges in this rapidly advancing field.
Collapse
Affiliation(s)
- Nadia Galindo-Cabello
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain; (G.B.); (S.P.-I.)
| | - Estefanía Caballano-Infantes
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Spain;
| | - Gregorio Benites
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain; (G.B.); (S.P.-I.)
| | - Salvador Pastor-Idoate
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain; (G.B.); (S.P.-I.)
- Department of Ophthalmology, University Clinical Hospital of Valladolid, 47003 Valladolid, Spain
| | - Francisco J. Diaz-Corrales
- Department of Integrative Pathophysiology and Therapies, Andalusian Molecular Biology and Regenerative Medicine Centre (CABIMER), Junta de Andalucía, CSIC, Universidad de Sevilla, Universidad Pablo de Olavide, Avda. Américo Vespucio 24, 41092 Seville, Spain;
| | - Ricardo Usategui-Martín
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47003 Valladolid, Spain;
- Institute of Applied Ophthalmobiology (IOBA), University of Valladolid, 47011 Valladolid, Spain; (G.B.); (S.P.-I.)
| |
Collapse
|
8
|
Liu X, Zhou Z, Zhang Y, Zhong H, Cai X, Guan R. Recent progress on the organoids: Techniques, advantages and applications. Biomed Pharmacother 2025; 185:117942. [PMID: 40043462 DOI: 10.1016/j.biopha.2025.117942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/30/2025] [Accepted: 02/24/2025] [Indexed: 03/23/2025] Open
Abstract
Organoids are a cutting-edge technology in the life sciences field, with applications in precision medicine, bionic organs, and toxicological evaluations of chemicals. Their 3D structure closely resembles that of real organs, allowing more accurate functional mimicry. The 3D organoid culture system can simulate the growth state of cells in vivo and establish a suspension culture system for organoid 3D culture by using scaffold-less or scaffold technology to avoid direct contact between cells and plastic culture vessels. Furthermore, organoids can simulate the pathophysiological state of tissues and organs in vitro. This paper primarily discusses the construction methodologies, as well as the advantages and disadvantages of 3D culture systems for both scaffold-free organoids and scaffolded organoids. This review also summarizes the application of organoid models in chemical toxicology evaluation, drug screening and functional evaluation, establishment of in vitro disease models, and research on disease occurrence and potential mechanisms. The aim is to provide a reference for the research and practical applications of organoid-related scientific fields.
Collapse
Affiliation(s)
- Xiaofeng Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Zhiyuan Zhou
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yao Zhang
- Zhejiang Provincial Key Lab for Chem and Bio Processing Technology of Farm Produces, School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, Zhejiang 310023, China
| | - Hao Zhong
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Xiulei Cai
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Rongfa Guan
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China; Moganshan Institute ZJUT, Kangqian District, Deqing 313200, China.
| |
Collapse
|
9
|
Lin S, Arno G, Robson AG, Schiff ER, Mohamed MD, Michaelides M, Webster AR, Mahroo OA. Bifocal retinal degeneration observed on ultra-widefield autofluorescence in some cases of CRX-associated retinopathy. Eye (Lond) 2025; 39:951-957. [PMID: 39632990 PMCID: PMC11933272 DOI: 10.1038/s41433-024-03522-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/12/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Variants in CRX are associated with dominantly inherited retinopathy with considerable phenotypic variability. Many patients have central retinal degeneration; in some patients, we have observed an additional focus of degeneration in the nasal retina. This study explores this phenotypic association amongst patients with CRX-associated disease. METHODS A retrospective review was conducted for all patients with dominant CRX-associated retinopathy at two UK centres. Analysis focused on patients with available ultra-widefield autofluorescence imaging and aimed to identify those with a specific bifocal degeneration pattern involving the nasal retina in both eyes. RESULTS Sixty patients were identified, with ultra-widefield fundus imaging available for 50 patients. Of these, six male patients aged 26-74 years displayed a distinct pattern characterised by central retinal degeneration and an additional discrete area of altered autofluorescence in the nasal periphery. Pattern and full-field ERGs indicated macular dysfunction in all 6 cases, with generalised cone (n = 2) or cone and rod (n = 4) system involvement, with a locus that appeared to be post-phototransduction. The CRX variants found in these patients included missense variants (n = 2), frameshifting variants (n = 3), and a CRX whole gene deletion (n = 1), with no clear genotype-phenotype correlation identified. CONCLUSIONS We report a distinct pattern of bifocal retinal degeneration in some cases of CRX-associated retinopathy (12% in our cohort), not typically seen in other forms of inherited retinal disease. Recognising such phenotypes can guide genetic investigations or their interpretation, facilitating molecular diagnoses for effective family counselling, given the autosomal dominant inheritance and phenotypic variability of CRX-associated retinopathy.
Collapse
Affiliation(s)
- Siying Lin
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Gavin Arno
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
- Greenwood Genetic Center, Greenwood, SC, USA
| | - Anthony G Robson
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Elena R Schiff
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Moin D Mohamed
- Department of Ophthalmology, St Thomas' Hospital, London, UK
| | - Michel Michaelides
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Andrew R Webster
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK
- UCL Institute of Ophthalmology, University College London, London, UK
| | - Omar A Mahroo
- National Institute of Health Research Biomedical Research Centre at Moorfields Eye Hospital and The UCL Institute of Ophthalmology, London, UK.
- UCL Institute of Ophthalmology, University College London, London, UK.
- Department of Ophthalmology, St Thomas' Hospital, London, UK.
| |
Collapse
|
10
|
Kaiser VM, Gonzalez-Cordero A. Organoids - the future of pre-clinical development of AAV gene therapy for CNS disorders. Gene Ther 2025:10.1038/s41434-025-00527-8. [PMID: 40148593 DOI: 10.1038/s41434-025-00527-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Advancements in our understanding of genetic disease and adeno-associated virus has prompted great excitement into the field of AAV-mediated gene therapy, particularly for genetic diseases of the central nervous system, including retinal disorders. Despite significant progress, exemplified by the approval of therapies such as Luxturna® and Zolgensma®, a substantial number of therapies remain in pre-clinical or early clinical stages, with many failing to advance to later phases. Whilst the use of animal models to test safety and delivery route efficacy of AAV treatments is imperative, differences in tissue structure and physiology between humans and animal models has restricted precise disease modelling and gene therapy development for many CNS disorders. Alongside the FDA push for non-animal alternative models, researchers are increasingly turning to human-based models, including stem cell-derived organoids, which can offer a more accurate representation of human cellular microenvironments and niches. As such, this review explores the advantages and limitations of brain and retinal organoids as pre-clinical models of disease, with a primary focus on their utility in identifying novel AAV capsids, cell-specific promoters, and their role in recent pre-clinical AAV gene therapy studies.
Collapse
Affiliation(s)
- Vivienne M Kaiser
- Stem Cell Medicine Unit, Children's Medical Research Institute, Westmead, NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Unit, Children's Medical Research Institute, Westmead, NSW, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
11
|
Leigh A, Swaroop A, Kruczek K, Ullah E, Brooks BP. Cone Rod Homeobox ( CRX): literature review and new insights. Ophthalmic Genet 2025:1-9. [PMID: 40074530 DOI: 10.1080/13816810.2025.2458086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 03/14/2025]
Abstract
The development of the neural retina requires a complex, spatiotemporally regulated network of gene expression. Here we review the role of the cone rod homeobox (CRX) transcription factor in specification and differentiation of retinal photoreceptors and its function in inherited retinal diseases such as cone-rod dystrophy (CoRD), dominant retinitis pigmentosa (RP), and Leber's congenital amaurosis (LCA). We delineate the findings of animal models and, more recently, human retinal organoids in elucidating molecular mechanisms of CRX activity and the pathogenesis of inherited photoreceptor degenerations. Lastly, we discuss implications of these findings in the development of therapies for inherited retinal diseases.
Collapse
Affiliation(s)
- Arnold Leigh
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, Bethesda, Virginia, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kamil Kruczek
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ehsan Ullah
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, Bethesda, Virginia, USA
| | - Brian P Brooks
- Ophthalmic Genetics & Visual Function Branch, National Eye Institute, Bethesda, Virginia, USA
| |
Collapse
|
12
|
Soundara Pandi SP, Winter H, Smith MR, Harkin K, Bojdo J. Preclinical Retinal Disease Models: Applications in Drug Development and Translational Research. Pharmaceuticals (Basel) 2025; 18:293. [PMID: 40143072 PMCID: PMC11944893 DOI: 10.3390/ph18030293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/10/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Retinal models play a pivotal role in translational drug development, bridging preclinical research and therapeutic applications for both ocular and systemic diseases. This review highlights the retina as an ideal organ for studying advanced therapies, thanks to its immune privilege, vascular and neuronal networks, accessibility, and advanced imaging capabilities. Preclinical retinal disease models offer unparalleled insights into inflammation, angiogenesis, fibrosis, and hypoxia, utilizing clinically translatable bioimaging tools like fundoscopy, optical coherence tomography, confocal scanning laser ophthalmoscopy, fluorescein angiography, optokinetic tracking, and electroretinography. These models have driven innovations in anti-inflammatory, anti-angiogenic, and neuroprotective strategies, with broader implications for systemic diseases such as rheumatoid arthritis, Alzheimer's, and fibrosis-related conditions. By emphasizing the integration of the 3Rs principles and novel imaging modalities, this review highlights how retinal research not only enhances therapeutic precision but also minimizes ethical concerns, paving the way for more predictive and human-relevant approaches in drug development.
Collapse
Affiliation(s)
| | - Hanagh Winter
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| | - Madeleine R. Smith
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| | - Kevin Harkin
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - James Bojdo
- Medinect Bioservices Ltd., Belfast BT7 1NF, UK; (S.P.S.P.); (H.W.); (M.R.S.); (K.H.)
| |
Collapse
|
13
|
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
|
14
|
Hahn LC, van der Veen I, Georgiou M, van Schooneveld MJ, Ten Brink JB, Florijn RJ, Mahroo OA, de Carvalho ER, Webster AR, Bergen AA, Michaelides M, Boon CJF. Clinical, Genetic, and Histopathological Characteristics of CRX-associated Retinal Dystrophies. Ophthalmol Retina 2025; 9:78-88. [PMID: 39128788 DOI: 10.1016/j.oret.2024.08.003] [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: 02/25/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE To describe phenotypic, genotypic, and histopathological features of inherited retinal dystrophies associated with the CRX gene (CRX-RDs). DESIGN Retrospective multicenter cohort study including histopathology. SUBJECTS Thirty-nine patients from 31 families with pathogenic variants in the CRX gene. METHODS Clinical data of 152 visits were collected from medical records. The median follow-up time was 9.1 years (interquartile range (IQR), 3.3-15.3 years; range, 0.0-48.8 years). Histopathologic examination of the eye of a 17-year-old patient with advanced early-onset CRX-RD was performed. MAIN OUTCOME MEASURES Visual acuity, retinal imaging, electroretinography, genotype-phenotype correlation, and histopathological examination were evaluated. RESULTS The age at onset ranged from birth to the eighth decade of life. Median visual acuity was 1.00 logarithm of the minimum angle of resolution (logMAR) (IQR, 0.69-1.48 logMAR; range, 0.06-3.00 logMAR) at a mean age of 52.0 ± 19.9 years (range, 4.6-81.9 years). Sufficient imaging was available for 36 out of 39 patients (92.3%), and all showed degeneration of at least the macula. Of these 36 patients, 22 (61.1%) had only macular dystrophy. Another 10 patients (27.8%) had additional degeneration beyond the vascular arcades, and 4 patients (11.1%) panretinal degeneration. Two patients (5.1%) had Leber congenital amaurosis. In total, 21 different disease-associated heterozygous CRX variants were identified (10 frameshift, 7 missense, 2 deletion, 1 nonsense, 1 deletion-insertion variants). Missense variants in the CRX homeodomain and 2 variants deleting all functional domains, thus causing haploinsufficiency, generally tended to cause milder late-onset phenotypes. Histopathologic examination of the eye of a 17-year-old patient with advanced early-onset retinal dystrophy due to a heterozygous deletion of exons 3 and 4 of the CRX gene revealed loss of laminar integrity and widespread photoreceptor degeneration especially in the central retina, with extensive loss of photoreceptor nuclei and outer segments. CONCLUSIONS This study illustrates the large clinical and genetic heterogenic spectrum of CRX-RDs, ranging from Leber congenital amaurosis to mild late-onset maculopathy resembling occult macular dystrophy. Haploinsufficiency and missense variants tended to be associated with milder phenotypes. Patients showed degeneration predominantly affecting the central retina on imaging. The histopathological findings also mirror these clinical findings and features similar to previously reported animal models of CRX-RDs. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Leo C Hahn
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Isa van der Veen
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Michalis Georgiou
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Department of Ophthalmology, Jones Eye Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Mary J van Schooneveld
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Department of Ophthalmology, St Thomas' Hospital, London, United Kingdom
| | - Jacoline B Ten Brink
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Ralph J Florijn
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Omar A Mahroo
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom; Department of Ophthalmology, St Thomas' Hospital, London, United Kingdom; Section of Ophthalmology, King's College London, London, United Kingdom; Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | | | - Andrew R Webster
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Arthur A Bergen
- Department of Human Genetics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; The Netherlands Institute for Neuroscience (NIN-KNAW), Amsterdam, The Netherlands
| | - Michel Michaelides
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom; UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Camiel J F Boon
- Department of Ophthalmology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands; Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
15
|
Huang W, Xu Z, Li S, Zhou J, Zhao B. Living Biobanks of Organoids: Valuable Resource for Translational Research. Biopreserv Biobank 2024; 22:543-549. [PMID: 38959173 PMCID: PMC11656124 DOI: 10.1089/bio.2023.0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024] Open
Abstract
The emergence of organoids is considered a revolutionary model, changing the landscape of traditional translational research. These three-dimensional miniatures of human organs or tissues, cultivated from stem cells or biospecimens obtained from patients, faithfully replicate the structural and functional characteristics of specific target organs or tissues. In this extensive review, we explore the profound impact of organoids and assess the current state of living organoid biobanks, which are essential repositories for cryopreserving organoids derived from a variety of diseases. These resources hold significant value for translational research. We delve into the diverse origins of organoids, the underlying technologies, and their roles in recapitulating human development, disease modeling, as well as their potential applications in the pharmaceutical field. With a particular emphasis on biobanking organoids for prospective applications, we discuss how these advancements expedite the transition from bench to bedside translational research, thereby fostering personalized medicine and enriching our comprehension of human health.
Collapse
Affiliation(s)
- Wenqing Huang
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Zhaoting Xu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, Republic of China
| | - Shuang Li
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Junmei Zhou
- Department of Central Laboratory, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, Republic of China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, Republic of China
| |
Collapse
|
16
|
Zhao N, Li YY, Xu JM, Yang MY, Li YZ, Lam TC, Zhou L, Tong QH, Zhang JT, Wang SZ, Hu XX, Wu YF, Lu QK, Lang TY. Cone-rod homeobox transcriptionally activates TCF7 to promote the proliferation of retinal pigment epithelial and retinoblastoma cells in vitro. Int J Ophthalmol 2024; 17:1995-2006. [PMID: 39559312 PMCID: PMC11528266 DOI: 10.18240/ijo.2024.11.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 07/19/2024] [Indexed: 11/20/2024] Open
Abstract
AIM To investigate the proliferation regulatory effect of cone-rod homeobox (CRX) in retinal pigment epithelium (RPE) and retinoblastoma (RB) cells to explore the potential application and side effect (oncogenic potential) of CRX-based gene therapy in RPE-based retinopathies. METHODS Adult human retinal pigment epithelial (ARPE)-19 and human retinal pigment epithelial (RPE)-1 cells and Y79 RB cell were used in the study. Genetic manipulation was performed by lentivirus-based technology. The cell proliferation was determined by a CellTiter-Glo Reagent. The mRNA and protein levels were determined by quantitative real-time polymerase chain reaction (qPCR) and Western blot assay. The transcriptional activity of the promoter was determined by luciferase reporter gene assay. The bindings between CRX and transcription factor 7 (TCF7) promoter as well as TCF7 and the promoters of TCF7 target genes were examined by chromatin immunoprecipitation (ChIP) assay. The transcription of the TCF7 was determined by a modified nuclear run-on assay. RESULTS CRX overexpression and knockdown significantly increased (n=3, P<0.05 in all the cells) and decreased (n=3, P<0.01 in all the cells) the proliferation of RPE and RB cells. CRX overexpression and knockdown significantly increased and deceased the mRNA levels of Wnt signaling target genes [including MYC proto-oncogene (MYC), JUN, FOS like 1 (FOSL1), CCND1, cyclin D2 (CCND2), cyclin D3 (CCND3), cellular communication network factor 4 (CCN4), peroxisome proliferator activated receptor delta (PPARD), and matrix metallopeptidase 7 (MMP7)] and the luciferase activity driven by the Wnt signaling transcription factor (TCF7). TCF7 overexpression and knockdown significantly increased and decreased the proliferation of RPE and RB cells and depletion of TCF7 significantly abolished the stimulatory effect of CRX on the proliferation of RPE and RB cells. CRX overexpression and knockdown significantly increased and decreased the mRNA level of TCF7 and the promoter of TCF7 was significantly immunoprecipitated by CRX antibody. CONCLUSION CRX transcriptionally activates TCF7 to promote the proliferation of RPE and RB cells in vitro. CRX is a potential target for RPE-based regenerative medicine. The potential risk of this strategy, tumorigenic potential, should be considered.
Collapse
Affiliation(s)
- Na Zhao
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Ying-Ying Li
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Jia-Man Xu
- Reproductive Medicine Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Mu-Yao Yang
- College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yun-Zhe Li
- Department of Gynaecology and Obstetrics, the First Affiliated Hospital of Chongqing Medical University; Department of Gynecology, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042; State Key Laboratory of Maternal and Fetal Medicine of Chongqing, Chongqing Medical University, Chongqing 400016, China
| | - Thomas Chuen Lam
- Centre for Myopia Research, School of Optometry, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong 999077, China
| | - Lei Zhou
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong 999077, China
- School of Optometry; Department of Applied Biology and Chemical Technology; Research Centre for SHARP Vision (RCSV); The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Qi-Hu Tong
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Jun-Tao Zhang
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Sheng-Zhan Wang
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Xin-Xin Hu
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yu-Fei Wu
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Qin-Kang Lu
- Ophthalmology Center, the Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
- Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Ting-Yuan Lang
- Reproductive Medicine Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
17
|
Howard L, Ishikawa Y, Katayama T, Park SJ, Hill MJ, Blake DJ, Nishida K, Hayashi R, Quantock AJ. Single-cell transcriptomics reveals the molecular basis of human iPS cell differentiation into ectodermal ocular lineages. Commun Biol 2024; 7:1495. [PMID: 39532995 PMCID: PMC11557866 DOI: 10.1038/s42003-024-07130-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
The generation of a self-formed, ectodermal, autonomous multi-zone (SEAM) from human induced pluripotent stem cells (hiPSCs) offers a unique perspective to study the dynamics of ocular cell differentiation over time. Here, by utilising single-cell transcriptomics, we have (i) identified, (ii) molecularly characterised and (iii) ascertained the developmental trajectories of ectodermally-derived ocular cell populations which emerge within SEAMs as they form. Our analysis reveals interdependency between tissues of the early eye and delineates the sequential formation and maturation of distinct cell types over a 12-week period. We demonstrate a progression from pluripotency through to tissue specification and differentiation which encompasses both surface ectodermal and neuroectodermal ocular lineages and the generation of iPSC-derived components of the developing cornea, conjunctiva, lens, and retina. Our findings not only advance the understanding of ocular development in a stem cell-based system of human origin, but also establish a robust methodological paradigm for exploring cellular and molecular dynamics during SEAM formation at single-cell resolution and highlight the potential of hiPSC-derived systems as powerful platforms for modelling human eye development and disease.
Collapse
Affiliation(s)
- Laura Howard
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Yuki Ishikawa
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohiko Katayama
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sung-Joon Park
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Matthew J Hill
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Derek J Blake
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | - Ryuhei Hayashi
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Andrew J Quantock
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
18
|
Alsalloum A, Gornostal E, Mingaleva N, Pavlov R, Kuznetsova E, Antonova E, Nadzhafova A, Kolotova D, Kadyshev V, Mityaeva O, Volchkov P. A Comparative Analysis of Models for AAV-Mediated Gene Therapy for Inherited Retinal Diseases. Cells 2024; 13:1706. [PMID: 39451224 PMCID: PMC11506034 DOI: 10.3390/cells13201706] [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: 09/12/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Inherited retinal diseases (IRDs) represent a diverse group of genetic disorders leading to progressive degeneration of the retina due to mutations in over 280 genes. This review focuses on the various methodologies for the preclinical characterization and evaluation of adeno-associated virus (AAV)-mediated gene therapy as a potential treatment option for IRDs, particularly focusing on gene therapies targeting mutations, such as those in the RPE65 and FAM161A genes. AAV vectors, such as AAV2 and AAV5, have been utilized to deliver therapeutic genes, showing promise in preserving vision and enhancing photoreceptor function in animal models. Despite their advantages-including high production efficiency, low pathogenicity, and minimal immunogenicity-AAV-mediated therapies face limitations such as immune responses beyond the retina, vector size constraints, and challenges in large-scale manufacturing. This review systematically compares different experimental models used to investigate AAV-mediated therapies, such as mouse models, human retinal explants (HREs), and induced pluripotent stem cell (iPSC)-derived retinal organoids. Mouse models are advantageous for genetic manipulation and detailed investigations of disease mechanisms; however, anatomical differences between mice and humans may limit the translational applicability of results. HREs offer valuable insights into human retinal pathophysiology but face challenges such as tissue degradation and lack of systemic physiological effects. Retinal organoids, on the other hand, provide a robust platform that closely mimics human retinal development, thereby enabling more comprehensive studies on disease mechanisms and therapeutic strategies, including AAV-based interventions. Specific outcomes targeted in these studies include vision preservation and functional improvements of retinas damaged by genetic mutations. This review highlights the strengths and weaknesses of each experimental model and advocates for their combined use in developing targeted gene therapies for IRDs. As research advances, optimizing AAV vector design and delivery methods will be critical for enhancing therapeutic efficacy and improving clinical outcomes for patients with IRDs.
Collapse
Affiliation(s)
- Almaqdad Alsalloum
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
| | | | - Natalia Mingaleva
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Roman Pavlov
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | | | - Ekaterina Antonova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Aygun Nadzhafova
- Moscow Center for Advanced Studies, Kulakova Str. 20, 123592 Moscow, Russia
| | - Daria Kolotova
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, 117485 Moscow, Russia
| | | | - Olga Mityaeva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Pavel Volchkov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, 125315 Moscow, Russia (P.V.)
- Department of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
- Moscow Clinical Scientific Center N.A. A.S. Loginov, 111123 Moscow, Russia
| |
Collapse
|
19
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [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] [Received: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|
20
|
Liu Y, Zong X, Cao W, Zhang W, Zhang N, Yang N. Gene Therapy for Retinitis Pigmentosa: Current Challenges and New Progress. Biomolecules 2024; 14:903. [PMID: 39199291 PMCID: PMC11352491 DOI: 10.3390/biom14080903] [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: 06/21/2024] [Revised: 07/14/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Retinitis pigmentosa (RP) poses a significant threat to eye health worldwide, with prevalence rates of 1 in 5000 worldwide. This genetically diverse retinopathy is characterized by the loss of photoreceptor cells and atrophy of the retinal pigment epithelium. Despite the involvement of more than 3000 mutations across approximately 90 genes in its onset, finding an effective treatment has been challenging for a considerable time. However, advancements in scientific research, especially in gene therapy, are significantly expanding treatment options for this most prevalent inherited eye disease, with the discovery of new compounds, gene-editing techniques, and gene loci offering hope for more effective treatments. Gene therapy, a promising technology, utilizes viral or non-viral vectors to correct genetic defects by either replacing or silencing disease-causing genes, potentially leading to complete recovery. In this review, we primarily focus on the latest applications of gene editing research in RP. We delve into the most prevalent genes associated with RP and discuss advancements in genome-editing strategies currently employed to correct various disease-causing mutations.
Collapse
Affiliation(s)
| | | | | | | | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan 430060, China; (Y.L.); (X.Z.); (W.C.); (W.Z.)
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road #238, Wuhan 430060, China; (Y.L.); (X.Z.); (W.C.); (W.Z.)
| |
Collapse
|
21
|
Kurzawa-Akanbi M, Tzoumas N, Corral-Serrano JC, Guarascio R, Steel DH, Cheetham ME, Armstrong L, Lako M. Pluripotent stem cell-derived models of retinal disease: Elucidating pathogenesis, evaluating novel treatments, and estimating toxicity. Prog Retin Eye Res 2024; 100:101248. [PMID: 38369182 DOI: 10.1016/j.preteyeres.2024.101248] [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: 12/07/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Blindness poses a growing global challenge, with approximately 26% of cases attributed to degenerative retinal diseases. While gene therapy, optogenetic tools, photosensitive switches, and retinal prostheses offer hope for vision restoration, these high-cost therapies will benefit few patients. Understanding retinal diseases is therefore key to advance effective treatments, requiring in vitro models replicating pathology and allowing quantitative assessments for drug discovery. Pluripotent stem cells (PSCs) provide a unique solution given their limitless supply and ability to differentiate into light-responsive retinal tissues encompassing all cell types. This review focuses on the history and current state of photoreceptor and retinal pigment epithelium (RPE) cell generation from PSCs. We explore the applications of this technology in disease modelling, experimental therapy testing, biomarker identification, and toxicity studies. We consider challenges in scalability, standardisation, and reproducibility, and stress the importance of incorporating vasculature and immune cells into retinal organoids. We advocate for high-throughput automation in data acquisition and analyses and underscore the value of advanced micro-physiological systems that fully capture the interactions between the neural retina, RPE, and choriocapillaris.
Collapse
|
22
|
Kandoi S, Martinez C, Chen KX, Mehine M, Reddy LVK, Mansfield BC, Duncan JL, Lamba DA. Disease modeling and pharmacological rescue of autosomal dominant retinitis pigmentosa associated with RHO copy number variation. eLife 2024; 12:RP90575. [PMID: 38661530 PMCID: PMC11045220 DOI: 10.7554/elife.90575] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Retinitis pigmentosa (RP), a heterogenous group of inherited retinal disorder, causes slow progressive vision loss with no effective treatments available. Mutations in the rhodopsin gene (RHO) account for ~25% cases of autosomal dominant RP (adRP). In this study, we describe the disease characteristics of the first-ever reported mono-allelic copy number variation (CNV) in RHO as a novel cause of adRP. We (a) show advanced retinal degeneration in a male patient (68 years of age) harboring four transcriptionally active intact copies of rhodopsin, (b) recapitulated the clinical phenotypes using retinal organoids, and (c) assessed the utilization of a small molecule, Photoregulin3 (PR3), as a clinically viable strategy to target and modify disease progression in RP patients associated with RHO-CNV. Patient retinal organoids showed photoreceptors dysgenesis, with rod photoreceptors displaying stunted outer segments with occasional elongated cilia-like projections (microscopy); increased RHO mRNA expression (quantitative real-time PCR [qRT-PCR] and bulk RNA sequencing); and elevated levels and mislocalization of rhodopsin protein (RHO) within the cell body of rod photoreceptors (western blotting and immunohistochemistry) over the extended (300 days) culture time period when compared against control organoids. Lastly, we utilized PR3 to target NR2E3, an upstream regulator of RHO, to alter RHO expression and observed a partial rescue of RHO protein localization from the cell body to the inner/outer segments of rod photoreceptors in patient organoids. These results provide a proof-of-principle for personalized medicine and suggest that RHO expression requires precise control. Taken together, this study supports the clinical data indicating that RHO-CNV associated adRPdevelops as a result of protein overexpression, thereby overloading the photoreceptor post-translational modification machinery.
Collapse
Affiliation(s)
- Sangeetha Kandoi
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research University of California, San FranciscoSan FranciscoUnited States
| | - Cassandra Martinez
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research University of California, San FranciscoSan FranciscoUnited States
| | - Kevin Xu Chen
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research University of California, San FranciscoSan FranciscoUnited States
| | | | - L Vinod K Reddy
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research University of California, San FranciscoSan FranciscoUnited States
| | - Brian C Mansfield
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of HealthBethesdaUnited States
| | - Jacque L Duncan
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
| | - Deepak A Lamba
- Department of Ophthalmology, University of California, San FranciscoSan FranciscoUnited States
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research University of California, San FranciscoSan FranciscoUnited States
- Immunology and Regenerative Medicine, GenentechSouth San FranciscoUnited States
| |
Collapse
|
23
|
Zheng Y, Stormo GD, Chen S. Aberrant homeodomain-DNA cooperative dimerization underlies distinct developmental defects in two dominant CRX retinopathy models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584677. [PMID: 38559186 PMCID: PMC10979960 DOI: 10.1101/2024.03.12.584677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Paired-class homeodomain transcription factors (HD TFs) play essential roles in vertebrate development, and their mutations are linked to human diseases. One unique feature of paired-class HD is cooperative dimerization on specific palindrome DNA sequences. Yet, the functional significance of HD cooperative dimerization in animal development and its dysregulation in diseases remain elusive. Using the retinal TF Cone-rod Homeobox (CRX) as a model, we have studied how blindness-causing mutations in the paired HD, p.E80A and p.K88N, alter CRX's cooperative dimerization, lead to gene misexpression and photoreceptor developmental deficits in dominant manners. CRXE80A maintains binding at monomeric WT CRX motifs but is deficient in cooperative binding at dimeric motifs. CRXE80A's cooperativity defect impacts the exponential increase of photoreceptor gene expression in terminal differentiation and produces immature, non-functional photoreceptors in the CrxE80A retinas. CRXK88N is highly cooperative and localizes to ectopic genomic sites with strong enrichment of dimeric HD motifs. CRXK88N's altered biochemical properties disrupt CRX's ability to direct dynamic chromatin remodeling during development to activate photoreceptor differentiation programs and silence progenitor programs. Our study here provides in vitro and in vivo molecular evidence that paired-class HD cooperative dimerization regulates neuronal development and dysregulation of cooperative binding contributes to severe dominant blinding retinopathies.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Gary D. Stormo
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| |
Collapse
|
24
|
Spirig SE, Renner M. Toward Retinal Organoids in High-Throughput. Cold Spring Harb Perspect Med 2024; 14:a041275. [PMID: 37217280 PMCID: PMC10910359 DOI: 10.1101/cshperspect.a041275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Human retinal organoids recapitulate the cellular diversity, arrangement, gene expression, and functional aspects of the human retina. Protocols to generate human retinal organoids from pluripotent stem cells are typically labor intensive, include many manual handling steps, and the organoids need to be maintained for several months until they mature. To generate large numbers of human retinal organoids for therapy development and screening purposes, scaling up retinal organoid production, maintenance, and analysis is of utmost importance. In this review, we discuss strategies to increase the number of high-quality retinal organoids while reducing manual handling steps. We further review different approaches to analyze thousands of retinal organoids with currently available technologies and point to challenges that still await to be overcome both in culture and analysis of retinal organoids.
Collapse
Affiliation(s)
- Stefan Erich Spirig
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Magdalena Renner
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| |
Collapse
|
25
|
McDonald A, Wijnholds J. Retinal Ciliopathies and Potential Gene Therapies: A Focus on Human iPSC-Derived Organoid Models. Int J Mol Sci 2024; 25:2887. [PMID: 38474133 PMCID: PMC10932180 DOI: 10.3390/ijms25052887] [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: 01/26/2024] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
The human photoreceptor function is dependent on a highly specialised cilium. Perturbation of cilial function can often lead to death of the photoreceptor and loss of vision. Retinal ciliopathies are a genetically diverse range of inherited retinal disorders affecting aspects of the photoreceptor cilium. Despite advances in the understanding of retinal ciliopathies utilising animal disease models, they can often lack the ability to accurately mimic the observed patient phenotype, possibly due to structural and functional deviations from the human retina. Human-induced pluripotent stem cells (hiPSCs) can be utilised to generate an alternative disease model, the 3D retinal organoid, which contains all major retinal cell types including photoreceptors complete with cilial structures. These retinal organoids facilitate the study of disease mechanisms and potential therapies in a human-derived system. Three-dimensional retinal organoids are still a developing technology, and despite impressive progress, several limitations remain. This review will discuss the state of hiPSC-derived retinal organoid technology for accurately modelling prominent retinal ciliopathies related to genes, including RPGR, CEP290, MYO7A, and USH2A. Additionally, we will discuss the development of novel gene therapy approaches targeting retinal ciliopathies, including the delivery of large genes and gene-editing techniques.
Collapse
Affiliation(s)
- Andrew McDonald
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZC Leiden, The Netherlands;
- Netherlands Institute of Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
26
|
Zheng Y, Chen S. Transcriptional precision in photoreceptor development and diseases - Lessons from 25 years of CRX research. Front Cell Neurosci 2024; 18:1347436. [PMID: 38414750 PMCID: PMC10896975 DOI: 10.3389/fncel.2024.1347436] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 01/19/2024] [Indexed: 02/29/2024] Open
Abstract
The vertebrate retina is made up of six specialized neuronal cell types and one glia that are generated from a common retinal progenitor. The development of these distinct cell types is programmed by transcription factors that regulate the expression of specific genes essential for cell fate specification and differentiation. Because of the complex nature of transcriptional regulation, understanding transcription factor functions in development and disease is challenging. Research on the Cone-rod homeobox transcription factor CRX provides an excellent model to address these challenges. In this review, we reflect on 25 years of mammalian CRX research and discuss recent progress in elucidating the distinct pathogenic mechanisms of four CRX coding variant classes. We highlight how in vitro biochemical studies of CRX protein functions facilitate understanding CRX regulatory principles in animal models. We conclude with a brief discussion of the emerging systems biology approaches that could accelerate precision medicine for CRX-linked diseases and beyond.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Saint Louis, MO, United States
- Department of Ophthalmology and Visual Sciences, Saint Louis, MO, United States
| | - Shiming Chen
- Molecular Genetics and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Saint Louis, MO, United States
- Department of Ophthalmology and Visual Sciences, Saint Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
27
|
de Lemos L, Antas P, Ferreira IS, Santos IP, Felgueiras B, Gomes CM, Brito C, Seabra MC, Tenreiro S. Modelling neurodegeneration and inflammation in early diabetic retinopathy using 3D human retinal organoids. IN VITRO MODELS 2024; 3:33-48. [PMID: 39872068 PMCID: PMC11756505 DOI: 10.1007/s44164-024-00068-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 01/29/2025]
Abstract
Purpose Diabetic retinopathy (DR) is a complication of diabetes and a primary cause of visual impairment amongst working-age individuals. DR is a degenerative condition in which hyperglycaemia results in morphological and functional changes in certain retinal cells. Existing treatments mainly address the advanced stages of the disease, which involve vascular defects or neovascularization. However, it is now known that retinal neurodegeneration and inflammation precede these vascular changes as early events of DR. Therefore, there is a pressing need to develop a reliable human in vitro model that mimics the early stage of DR to identify new therapeutic approaches to prevent and delay its progression. Methods Here, we used human-induced pluripotent stem cells (hiPSCs) differentiated into three-dimensional (3D) retinal organoids, which resemble the complexity of the retinal tissue. Retinal organoids were subjected to high-glucose conditions to generate a model of early DR. Results Our model showed well-established molecular and cellular features of early DR, such as (i) loss of retinal ganglion and amacrine cells; (ii) glial reactivity and inflammation, with increased expression of the vascular endothelial-derived growth factor (VEGF) and interleukin-1β (IL-1β), and monocyte chemoattractant protein-1 (MCP-1) secretion; and (iii) increased levels of reactive oxygen species accompanied by activation of key enzymes involved in antioxidative stress response. Conclusion The data provided highlight the utility of retinal organoid technology in modelling early-stage DR. This offers new avenues for the development of targeted therapeutic interventions on neurodegeneration and inflammation in the initial phase of DR, potentially slowing the disease's progression. Supplementary Information The online version contains supplementary material available at 10.1007/s44164-024-00068-1.
Collapse
Affiliation(s)
- Luisa de Lemos
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| | - Pedro Antas
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| | - Inês S. Ferreira
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| | - Inês Paz Santos
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| | - Beatriz Felgueiras
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| | - Catarina M. Gomes
- iBET, Instituto de Biologia Experimental E Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química E Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Catarina Brito
- iBET, Instituto de Biologia Experimental E Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
- Instituto de Tecnologia Química E Biológica António Xavier, Universidade Nova de Lisboa, Avenida da República, 2780-157 Oeiras, Portugal
| | - Miguel C. Seabra
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
- UCL Institute of Ophthalmology, London, UK
| | - Sandra Tenreiro
- iNOVA4Health, NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Rua Camara Pestana, 6, Lisbon, Portugal
| |
Collapse
|
28
|
Qu Z, Batz Z, Singh N, Marchal C, Swaroop A. Stage-specific dynamic reorganization of genome topology shapes transcriptional neighborhoods in developing human retinal organoids. Cell Rep 2023; 42:113543. [PMID: 38048222 PMCID: PMC10790351 DOI: 10.1016/j.celrep.2023.113543] [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: 05/12/2023] [Revised: 09/21/2023] [Accepted: 11/17/2023] [Indexed: 12/06/2023] Open
Abstract
We have generated a high-resolution Hi-C map of developing human retinal organoids to elucidate spatiotemporal dynamics of genomic architecture and its relationship with gene expression patterns. We demonstrate progressive stage-specific alterations in DNA topology and correlate these changes with transcription of cell-type-restricted gene markers during retinal differentiation. Temporal Hi-C reveals a shift toward A compartment for protein-coding genes and B compartment for non-coding RNAs, displaying high and low expression, respectively. Notably, retina-enriched genes are clustered near lost boundaries of topologically associated domains (TADs), and higher-order assemblages (i.e., TAD cliques) localize in active chromatin regions with binding sites for eye-field transcription factors. These genes gain chromatin contacts at their transcription start site as organoid differentiation proceeds. Our study provides a global view of chromatin architecture dynamics associated with diversification of cell types during retinal development and serves as a foundational resource for in-depth functional investigations of retinal developmental traits.
Collapse
Affiliation(s)
- Zepeng Qu
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Zachary Batz
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Nivedita Singh
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Claire Marchal
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA; In silichrom Ltd, 15 Digby Road, Newbury RG14 1TS, UK
| | - Anand Swaroop
- Neurobiology, Neurodegeneration, and Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
29
|
Pan D, Zhang X, Jin K, Jin ZB. CRX haploinsufficiency compromises photoreceptor precursor translocation and differentiation in human retinal organoids. Stem Cell Res Ther 2023; 14:346. [PMID: 38049871 PMCID: PMC10696917 DOI: 10.1186/s13287-023-03590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND The CRX-associated autosomal dominant retinopathies suggest a possible pathogenic mechanism of gene haploinsufficiency. However, based on reported human patient cases and studies with mouse models, it is hard to confirm the specific weight of haploinsufficiency in pathogenesis due to the interspecies gaps between gene expression and function. METHODS We created monoallelic CRX by replacing one allele with tdTomato in human embryonic stem cells (hESCs) and subsequently dissect pathogenesis in hESCs-derived retinal organoids. We used transcriptome and immunofluorescence analyses to dissect phenotypic differences between CRX-monoallelic knockout and control wildtype organoids. For location analysis of CRX+ cells, a CRX-expression-tracing system was constructed in control hESCs. We implemented long-term live-cell imaging to describe the translocation of CRX+ cells between two groups in early organoid differentiation. The expression pattern of these dynamic differences was validated using RNA-seq and immunofluorescence assays. RESULTS We identified delayed differentiation of outer nuclear layer (ONL) stratification along with thinner ONL, serious loss of photoreceptor outer segments, as well as downregulated expression of gene for phototransduction and inner/outer segment formation. By live-cell imaging and immunostaining, we observed the overtension of actomyosin network and the arrested translocation of monoallelic CRX+ cells in the early stage of retinal differentiation. CONCLUSIONS We confirmed that gene haploinsufficiency is the mechanism for the dominant pathogenicity of CRX and discovered that CRX regulated postmitotic photoreceptor precursor translocation in addition to its specification of photoreceptor cell fates during human retinal development. These findings revealed a new underlying mechanism of CRX dominant pathogenesis and provided a new clue for the treatment of CRX-associated human retinopathies.
Collapse
Affiliation(s)
- Deng Pan
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Xiao Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Kangxin Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zi-Bing Jin
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
30
|
Santiago CP, Gimmen MY, Lu Y, McNally MM, Duncan LH, Creamer TJ, Orzolek LD, Blackshaw S, Singh MS. Comparative Analysis of Single-cell and Single-nucleus RNA-sequencing in a Rabbit Model of Retinal Detachment-related Proliferative Vitreoretinopathy. OPHTHALMOLOGY SCIENCE 2023; 3:100335. [PMID: 37496518 PMCID: PMC10365955 DOI: 10.1016/j.xops.2023.100335] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 07/28/2023]
Abstract
Purpose Proliferative vitreoretinopathy (PVR) is the most common cause of failure of retinal reattachment surgery, and the molecular changes leading to this aberrant wound healing process are currently unknown. Our ultimate goal is to study PVR pathogenesis by employing single-cell transcriptomics to dissect cellular heterogeneity. Design Here we aimed to compare single-cell RNA sequencing (scRNA-seq) and single-nucleus RNA-sequencing (snRNA-seq) of retinal PVR samples in the rabbit model. Participants Unilateral induction of PVR lesions in rabbit eyes with contralateral eyes serving as controls. Methods Proliferative vitreoretinopathy was induced unilaterally in Dutch Belted rabbits. At different timepoints after PVR induction, retinas were dissociated into either cells or nuclei suspension and processed for scRNA-seq or snRNA-seq. Main Outcome Measures Single cell and nuclei transcriptomic profiles of retinas after PVR induction. Results Single-cell RNA sequencing and snRNA-seq were conducted on retinas at 4 hours and 14 days after disease induction. Although the capture rate of unique molecular identifiers and genes were greater in scRNA-seq samples, overall gene expression profiles of individual cell types were highly correlated between scRNA-seq and snRNA-seq. A major disparity between the 2 sequencing modalities was the cell type capture rate, however, with glial cell types overrepresented in scRNA-seq, and inner retinal neurons were enriched by snRNA-seq. Furthermore, fibrotic Müller glia were overrepresented in snRNA-seq samples, whereas reactive Müller glia were overrepresented in scRNA-seq samples. Trajectory analyses were similar between the 2 methods, allowing for the combined analysis of the scRNA-seq and snRNA-seq data sets. Conclusions These findings highlight limitations of both scRNA-seq and snRNA-seq analysis and imply that use of both techniques together can more accurately identify transcriptional networks critical for aberrant fibrogenesis in PVR than using either in isolation. Financial Disclosures Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Clayton P. Santiago
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
| | - Megan Y. Gimmen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
| | - Yuchen Lu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Minda M. McNally
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leighton H. Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
| | - Tyler J. Creamer
- Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Linda D. Orzolek
- Institute for Basic Biomedical Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, Maryland
| | - Mandeep S. Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
31
|
Wahle P, Brancati G, Harmel C, He Z, Gut G, Del Castillo JS, Xavier da Silveira Dos Santos A, Yu Q, Noser P, Fleck JS, Gjeta B, Pavlinić D, Picelli S, Hess M, Schmidt GW, Lummen TTA, Hou Y, Galliker P, Goldblum D, Balogh M, Cowan CS, Scholl HPN, Roska B, Renner M, Pelkmans L, Treutlein B, Camp JG. Multimodal spatiotemporal phenotyping of human retinal organoid development. Nat Biotechnol 2023; 41:1765-1775. [PMID: 37156914 PMCID: PMC10713453 DOI: 10.1038/s41587-023-01747-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/13/2023] [Indexed: 05/10/2023]
Abstract
Organoids generated from human pluripotent stem cells provide experimental systems to study development and disease, but quantitative measurements across different spatial scales and molecular modalities are lacking. In this study, we generated multiplexed protein maps over a retinal organoid time course and primary adult human retinal tissue. We developed a toolkit to visualize progenitor and neuron location, the spatial arrangements of extracellular and subcellular components and global patterning in each organoid and primary tissue. In addition, we generated a single-cell transcriptome and chromatin accessibility timecourse dataset and inferred a gene regulatory network underlying organoid development. We integrated genomic data with spatially segmented nuclei into a multimodal atlas to explore organoid patterning and retinal ganglion cell (RGC) spatial neighborhoods, highlighting pathways involved in RGC cell death and showing that mosaic genetic perturbations in retinal organoids provide insight into cell fate regulation.
Collapse
Affiliation(s)
- Philipp Wahle
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Giovanna Brancati
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Christoph Harmel
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Zhisong He
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Gabriele Gut
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | - Aline Xavier da Silveira Dos Santos
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Qianhui Yu
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Pascal Noser
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Jonas Simon Fleck
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Bruno Gjeta
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Dinko Pavlinić
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Simone Picelli
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Max Hess
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Gregor W Schmidt
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Tom T A Lummen
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Yanyan Hou
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Patricia Galliker
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - David Goldblum
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Marton Balogh
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Cameron S Cowan
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | - Hendrik P N Scholl
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Botond Roska
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Magdalena Renner
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
- Department of Ophthalmology, University of Basel, Basel, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.
| | - Barbara Treutlein
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland.
| | - J Gray Camp
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland.
- Department of Ophthalmology, University of Basel, Basel, Switzerland.
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
| |
Collapse
|
32
|
Liang Y, Sun X, Duan C, Tang S, Chen J. Application of patient-derived induced pluripotent stem cells and organoids in inherited retinal diseases. Stem Cell Res Ther 2023; 14:340. [PMID: 38012786 PMCID: PMC10683306 DOI: 10.1186/s13287-023-03564-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
Inherited retinal diseases (IRDs) can induce severe sight-threatening retinal degeneration and impose a considerable economic burden on patients and society, making efforts to cure blindness imperative. Transgenic animals mimicking human genetic diseases have long been used as a primary research tool to decipher the underlying pathogenesis, but there are still some obvious limitations. As an alternative strategy, patient-derived induced pluripotent stem cells (iPSCs), particularly three-dimensional (3D) organoid technology, are considered a promising platform for modeling different forms of IRDs, including retinitis pigmentosa, Leber congenital amaurosis, X-linked recessive retinoschisis, Batten disease, achromatopsia, and best vitelliform macular dystrophy. Here, this paper focuses on the status of patient-derived iPSCs and organoids in IRDs in recent years concerning disease modeling and therapeutic exploration, along with potential challenges for translating laboratory research to clinical application. Finally, the importance of human iPSCs and organoids in combination with emerging technologies such as multi-omics integration analysis, 3D bioprinting, or microfluidic chip platform are highlighted. Patient-derived retinal organoids may be a preferred choice for more accurately uncovering the mechanisms of human retinal diseases and will contribute to clinical practice.
Collapse
Affiliation(s)
- Yuqin Liang
- Aier Eye Institute, Changsha, 410015, China
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xihao Sun
- Aier Eye Institute, Changsha, 410015, China
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Chunwen Duan
- Aier Eye Institute, Changsha, 410015, China
- Changsha Aier Eye Hospital, Changsha, 410015, China
| | - Shibo Tang
- Aier Eye Institute, Changsha, 410015, China.
- Changsha Aier Eye Hospital, Changsha, 410015, China.
| | - Jiansu Chen
- Aier Eye Institute, Changsha, 410015, China.
- Changsha Aier Eye Hospital, Changsha, 410015, China.
- Key Laboratory for Regenerative Medicine, Ministry of Education, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
33
|
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
|
34
|
Kandoi S, Martinez C, Chen KX, Reddy LVK, Mehine M, Mansfield BC, Duncan JL, Lamba DA. Disease modeling and pharmacological rescue of autosomal dominant Retinitis Pigmentosa associated with RHO copy number variation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.27.23286248. [PMID: 36909455 PMCID: PMC10002783 DOI: 10.1101/2023.02.27.23286248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Retinitis pigmentosa (RP), a heterogenous group of inherited retinal disorder causes slow progressive vision loss with no effective treatments available. Mutations in the rhodopsin gene (RHO), account for ~25% cases of autosomal dominant RP (adRP). In this study, we describe the disease characteristics of the first ever reported mono-allelic copy number variation (CNV) in RHO as a novel cause of adRP. We (1) show advanced retinal degeneration in a male patient (60-70 year old) harboring four transcriptionally active intact copies of rhodopsin, (2) recapitulated the clinical phenotypes using retinal organoids, and (3) assessed the utilization of a small molecule, Photoregulin3 (PR3), as a clinically viable strategy to target and modify disease progression in RP patients associated with RHO-CNV. Patient retinal organoids showed photoreceptors dysgenesis, with rod photoreceptors displaying stunted outer segments with occasional elongated cilia-like projections (microscopy); increased RHO mRNA expression (qRT-PCR and bulk RNA-sequencing); and elevated levels and mislocalization of rhodopsin protein (RHO) within the cell body of rod photoreceptors (western blotting and immunohistochemistry) over the extended (300-days) culture time period when compared against control organoids. Lastly, we utilized PR3 to target NR2E3, an upstream regulator of RHO, to alter RHO expression and observed a partial rescue of RHO protein localization from the cell body to the inner/outer segments of rod photoreceptors in patient organoids. These results provide a proof-of-principle for personalized medicine and suggest that RHO expression requires precise control. Taken together, this study supports the clinical data indicating that adRP due to RHO-CNV develops due protein overexpression overloading the photoreceptor post-translational modification machinery.
Collapse
Affiliation(s)
- Sangeetha Kandoi
- Department of Ophthalmology, University of California San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California San Francisco, CA, USA
| | - Cassandra Martinez
- Department of Ophthalmology, University of California San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California San Francisco, CA, USA
| | - Kevin Xu Chen
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California San Francisco, CA, USA
| | - L Vinod K. Reddy
- Department of Ophthalmology, University of California San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California San Francisco, CA, USA
| | | | - Brian C. Mansfield
- Section on Cellular Differentiation, Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD USA
| | - Jacque L. Duncan
- Department of Ophthalmology, University of California San Francisco, CA, USA
| | - Deepak A. Lamba
- Department of Ophthalmology, University of California San Francisco, CA, USA
- Eli and Edythe Broad Center of Regeneration Medicine & Stem Cell Research, University of California San Francisco, CA, USA
| |
Collapse
|
35
|
Kruczek K, Swaroop A. Patient stem cell-derived in vitro disease models for developing novel therapies of retinal ciliopathies. Curr Top Dev Biol 2023; 155:127-163. [PMID: 38043950 PMCID: PMC12050124 DOI: 10.1016/bs.ctdb.2023.09.003] [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: 12/05/2023]
Abstract
Primary cilia are specialized organelles on the surface of almost all cells in vertebrate tissues and are primarily involved in the detection of extracellular stimuli. In retinal photoreceptors, cilia are uniquely modified to form outer segments containing components required for the detection of light in stacks of membrane discs. Not surprisingly, vision impairment is a frequent phenotype associated with ciliopathies, a heterogeneous class of conditions caused by mutations in proteins required for formation, maintenance and/or function of primary cilia. Traditionally, immortalized cell lines and model organisms have been used to provide insights into the biology of ciliopathies. The advent of methods for reprogramming human somatic cells into pluripotent stem cells has enabled the generation of in vitro disease models directly from patients suffering from ciliopathies. Such models help us in investigating pathological mechanisms specific to human physiology and in developing novel therapeutic approaches. In this article, we review current protocols to differentiate human pluripotent stem cells into retinal cell types, and discuss how these cellular and/or organoid models can be utilized to interrogate pathobiology of ciliopathies affecting the retina and for testing prospective treatments.
Collapse
Affiliation(s)
- Kamil Kruczek
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MD, United States.
| |
Collapse
|
36
|
Sanjurjo-Soriano C, Jimenez-Medina C, Erkilic N, Cappellino L, Lefevre A, Nagel-Wolfrum K, Wolfrum U, Van Wijk E, Roux AF, Meunier I, Kalatzis V. USH2A variants causing retinitis pigmentosa or Usher syndrome provoke differential retinal phenotypes in disease-specific organoids. HGG ADVANCES 2023; 4:100229. [PMID: 37654703 PMCID: PMC10465966 DOI: 10.1016/j.xhgg.2023.100229] [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: 06/05/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
There is an emblematic clinical and genetic heterogeneity associated with inherited retinal diseases (IRDs). The most common form is retinitis pigmentosa (RP), a rod-cone dystrophy caused by pathogenic variants in over 80 different genes. Further complexifying diagnosis, different variants in individual RP genes can also alter the clinical phenotype. USH2A is the most prevalent gene for autosomal-recessive RP and one of the most challenging because of its large size and, hence, large number of variants. Moreover, USH2A variants give rise to non-syndromic and syndromic RP, known as Usher syndrome (USH) type 2, which is associated with vision and hearing loss. The lack of a clear genotype-phenotype correlation or prognostic models renders diagnosis highly challenging. We report here a long-awaited differential non-syndromic RP and USH phenotype in three human disease-specific models: fibroblasts, induced pluripotent stem cells (iPSCs), and mature iPSC-derived retinal organoids. Moreover, we identified distinct retinal phenotypes in organoids from multiple RP and USH individuals, which were validated by isogenic-corrected controls. Non-syndromic RP organoids showed compromised photoreceptor differentiation, whereas USH organoids showed a striking and unexpected cone phenotype. Furthermore, complementary clinical investigations identified macular atrophy in a high proportion of USH compared with RP individuals, further validating our observations that USH2A variants differentially affect cones. Overall, identification of distinct non-syndromic RP and USH phenotypes in multiple models provides valuable and robust readouts for testing the pathogenicity of USH2A variants as well as the efficacy of therapeutic approaches in complementary cell types.
Collapse
Affiliation(s)
- Carla Sanjurjo-Soriano
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Carla Jimenez-Medina
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Nejla Erkilic
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Luisina Cappellino
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| | - Arnaud Lefevre
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Kerstin Nagel-Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Uwe Wolfrum
- Institute of Molecular Physiology, Molecular Cell Biology, and Photoreceptor Cell Biology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Erwin Van Wijk
- Department of Otorhinolaryngology, Hearing, & Genes, Radboud University Medical Center, Nijmegen, the Netherlands
- Donders Institute for Brain, Cognition, and Behavior, Nijmegen, the Netherlands
| | - Anne-Françoise Roux
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- Molecular Genetics Laboratory, University of Montpellier, CHU, Montpellier, France
| | - Isabelle Meunier
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
- National Reference Centre for Inherited Sensory Diseases, University of Montpellier, CHU, Montpellier, France
| | - Vasiliki Kalatzis
- Institute for Neurosciences of Montpellier (INM), University of Montpellier, INSERM, Montpellier, France
| |
Collapse
|
37
|
Kelley RA, Wu Z. Utilization of the retinal organoid model to evaluate the feasibility of genetic strategies to ameliorate retinal disease(s). Vision Res 2023; 210:108269. [PMID: 37295270 DOI: 10.1016/j.visres.2023.108269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Organoid models have quickly become a popular research tool to evaluate novel therapeutics on 3-D recapitulated tissue. This has enabled researchers to use physiologically relevant human tissue in vitro to augment the standard use of immortalized cells and animal models. Organoids can also provide a model when an engineered animal cannot recreate a specific disease phenotype. In particular, the retinal research field has taken advantage of this burgeoning technology to provide insight into inherited retinal disease(s) mechanisms and therapeutic intervention to ameliorate their effects. In this review we will discuss the use of both wild-type and patient-specific retinal organoids to further gene therapy research that could potentially prevent retinal disease(s) progression. Furthermore, we will discuss the pitfalls of current retinal organoid technology and present potential solutions that could overcome these hurdles in the near future.
Collapse
Affiliation(s)
- Ryan A Kelley
- PTC Therapeutics, 100 Corporate Ct #2400, South Plainfield, NJ 07080, USA.
| | - Zhijian Wu
- PTC Therapeutics, 100 Corporate Ct #2400, South Plainfield, NJ 07080, USA
| |
Collapse
|
38
|
Jones MK, Orozco LD, Qin H, Truong T, Caplazi P, Elstrott J, Modrusan Z, Chaney SY, Jeanne M. Integration of human stem cell-derived in vitro systems and mouse preclinical models identifies complex pathophysiologic mechanisms in retinal dystrophy. Front Cell Dev Biol 2023; 11:1252547. [PMID: 37691820 PMCID: PMC10483287 DOI: 10.3389/fcell.2023.1252547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Rare DRAM2 coding variants cause retinal dystrophy with early macular involvement via unknown mechanisms. We found that DRAM2 is ubiquitously expressed in the human eye and expression changes were observed in eyes with more common maculopathy such as Age-related Macular Degeneration (AMD). To gain insights into pathogenicity of DRAM2-related retinopathy, we used a combination of in vitro and in vivo models. We found that DRAM2 loss in human pluripotent stem cell (hPSC)-derived retinal organoids caused the presence of additional mesenchymal cells. Interestingly, Dram2 loss in mice also caused increased proliferation of cells from the choroid in vitro and exacerbated choroidal neovascular lesions in vivo. Furthermore, we observed that DRAM2 loss in human retinal pigment epithelial (RPE) cells resulted in increased susceptibility to stress-induced cell death in vitro and that Dram2 loss in mice caused age-related photoreceptor degeneration. This highlights the complexity of DRAM2 function, as its loss in choroidal cells provided a proliferative advantage, whereas its loss in post-mitotic cells, such as photoreceptor and RPE cells, increased degeneration susceptibility. Different models such as human pluripotent stem cell-derived systems and mice can be leveraged to study and model human retinal dystrophies; however, cell type and species-specific expression must be taken into account when selecting relevant systems.
Collapse
Affiliation(s)
- Melissa K. Jones
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
- Product Development Clinical Science Ophthalmology, Genentech Inc., South San Francisco, CA, United States
| | - Luz D. Orozco
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, United States
| | - Han Qin
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
| | - Tom Truong
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA, United States
| | - Patrick Caplazi
- Department of Research Pathology, Genentech Inc., South San Francisco, CA, United States
| | - Justin Elstrott
- Department of Translational Imaging, Genentech Inc., South San Francisco, CA, United States
| | - Zora Modrusan
- Department of Microchemistry, Proteomics, Lipidomics and Next-Generation Sequencing, Genentech Inc., South San Francisco, CA, United States
| | - Shawnta Y. Chaney
- Department of Translational Immunology, Genentech Inc., South San Francisco, CA, United States
| | - Marion Jeanne
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, United States
| |
Collapse
|
39
|
Xia J, Gu L, Pan Q. The landscape of basic gene therapy approaches in inherited retinal dystrophies. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1193595. [PMID: 38983091 PMCID: PMC11182181 DOI: 10.3389/fopht.2023.1193595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 06/09/2023] [Indexed: 07/11/2024]
Abstract
The study of gene therapies has been of particular interest in recent decades due to their promising potential to slow or even rescue the degeneration of the retina in inherited retinal dystrophies (IRDs). Here, we review the current approaches to gene therapy trials on IRDs, including the selection of animal models, therapeutic window, vectors and dosages. Mice are typically the first choice of animal models and recombinant adeno-associated virus (rAAV) of serotype 8 is the most common vector for loss-of-function IRDs. Furthermore, the therapeutic window should be considered to ensure efficacy before retinal degeneration occurs if possible, and dosages must be tailored to each approach.
Collapse
Affiliation(s)
| | | | - Qing Pan
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
40
|
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
|
41
|
Yang S, Hu H, Kung H, Zou R, Dai Y, Hu Y, Wang T, Lv T, Yu J, Li F. Organoids: The current status and biomedical applications. MedComm (Beijing) 2023; 4:e274. [PMID: 37215622 PMCID: PMC10192887 DOI: 10.1002/mco2.274] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Organoids are three-dimensional (3D) miniaturized versions of organs or tissues that are derived from cells with stem potential and can self-organize and differentiate into 3D cell masses, recapitulating the morphology and functions of their in vivo counterparts. Organoid culture is an emerging 3D culture technology, and organoids derived from various organs and tissues, such as the brain, lung, heart, liver, and kidney, have been generated. Compared with traditional bidimensional culture, organoid culture systems have the unique advantage of conserving parental gene expression and mutation characteristics, as well as long-term maintenance of the function and biological characteristics of the parental cells in vitro. All these features of organoids open up new opportunities for drug discovery, large-scale drug screening, and precision medicine. Another major application of organoids is disease modeling, and especially various hereditary diseases that are difficult to model in vitro have been modeled with organoids by combining genome editing technologies. Herein, we introduce the development and current advances in the organoid technology field. We focus on the applications of organoids in basic biology and clinical research, and also highlight their limitations and future perspectives. We hope that this review can provide a valuable reference for the developments and applications of organoids.
Collapse
Affiliation(s)
- Siqi Yang
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Haijie Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Hengchung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ruiqi Zou
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yushi Dai
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yafei Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tianrun Lv
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Jun Yu
- Departments of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fuyu Li
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
42
|
Boon N, Lu X, Andriessen CA, Moustakas I, Buck TM, Freund C, Arendzen CH, Böhringer S, Mei H, Wijnholds J. AAV-mediated gene augmentation therapy of CRB1 patient-derived retinal organoids restores the histological and transcriptional retinal phenotype. Stem Cell Reports 2023; 18:1123-1137. [PMID: 37084726 DOI: 10.1016/j.stemcr.2023.03.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/23/2023] Open
Abstract
Retinitis pigmentosa and Leber congenital amaurosis are inherited retinal dystrophies that can be caused by mutations in the Crumbs homolog 1 (CRB1) gene. CRB1 is required for organizing apical-basal polarity and adhesion between photoreceptors and Müller glial cells. CRB1 patient-derived induced pluripotent stem cells were differentiated into CRB1 retinal organoids that showed diminished expression of variant CRB1 protein observed by immunohistochemical analysis. Single-cell RNA sequencing revealed impact on, among others, the endosomal pathway and cell adhesion and migration in CRB1 patient-derived retinal organoids compared with isogenic controls. Adeno-associated viral (AAV) vector-mediated hCRB2 or hCRB1 gene augmentation in Müller glial and photoreceptor cells partially restored the histological phenotype and transcriptomic profile of CRB1 patient-derived retinal organoids. Altogether, we show proof-of-concept that AAV.hCRB1 or AAV.hCRB2 treatment improved the phenotype of CRB1 patient-derived retinal organoids, providing essential information for future gene therapy approaches for patients with mutations in the CRB1 gene.
Collapse
Affiliation(s)
- Nanda Boon
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Xuefei Lu
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Charlotte A Andriessen
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Ioannis Moustakas
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Thilo M Buck
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Christian Freund
- hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Christiaan H Arendzen
- hiPSC Hotel, Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Stefan Böhringer
- Department of Biomedical Data Sciences, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Jan Wijnholds
- Department of Ophthalmology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA Amsterdam, the Netherlands.
| |
Collapse
|
43
|
Völkner M, Wagner F, Kurth T, Sykes AM, Del Toro Runzer C, Ebner LJA, Kavak C, Alexaki VI, Cimalla P, Mehner M, Koch E, Karl MO. Modeling inducible neuropathologies of the retina with differential phenotypes in organoids. Front Cell Neurosci 2023; 17:1106287. [PMID: 37213216 PMCID: PMC10196395 DOI: 10.3389/fncel.2023.1106287] [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] [Received: 11/23/2022] [Accepted: 04/06/2023] [Indexed: 05/23/2023] Open
Abstract
Neurodegenerative diseases remain incompletely understood and therapies are needed. Stem cell-derived organoid models facilitate fundamental and translational medicine research. However, to which extent differential neuronal and glial pathologic processes can be reproduced in current systems is still unclear. Here, we tested 16 different chemical, physical, and cell functional manipulations in mouse retina organoids to further explore this. Some of the treatments induce differential phenotypes, indicating that organoids are competent to reproduce distinct pathologic processes. Notably, mouse retina organoids even reproduce a complex pathology phenotype with combined photoreceptor neurodegeneration and glial pathologies upon combined (not single) application of HBEGF and TNF, two factors previously associated with neurodegenerative diseases. Pharmacological inhibitors for MAPK signaling completely prevent photoreceptor and glial pathologies, while inhibitors for Rho/ROCK, NFkB, and CDK4 differentially affect them. In conclusion, mouse retina organoids facilitate reproduction of distinct and complex pathologies, mechanistic access, insights for further organoid optimization, and modeling of differential phenotypes for future applications in fundamental and translational medicine research.
Collapse
Affiliation(s)
- Manuela Völkner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Felix Wagner
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Thomas Kurth
- Technische Universität Dresden, Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform Core Facility Electron Microscopy and Histology, Dresden, Germany
| | - Alex M. Sykes
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Lynn J. A. Ebner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Cagri Kavak
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Vasileia Ismini Alexaki
- Technische Universität Dresden, Institute of Clinical Chemistry and Laboratory Medicine, University Clinic Carl Gustav Carus, Dresden, Germany
| | - Peter Cimalla
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mirko Mehner
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Edmund Koch
- Technische Universität Dresden, Carl Gustav Carus Faculty of Medicine, Department of Anesthesiology and Intensive Care Medicine, Clinical Sensoring and Monitoring, Dresden, Germany
| | - Mike O. Karl
- Technische Universität Dresden, Center for Regenerative Therapies Dresden (CRTD), Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
- *Correspondence: Mike O. Karl, ,
| |
Collapse
|
44
|
Cheng L, Kuehn MH. Human Retinal Organoids in Therapeutic Discovery: A Review of Applications. Handb Exp Pharmacol 2023; 281:157-187. [PMID: 37608005 PMCID: PMC11631198 DOI: 10.1007/164_2023_691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Human embryonic stem cells (hESCs)- and induced pluripotent stem cells (hiPSCs)-derived retinal organoids (ROs) are three-dimensional laminar structures that recapitulate the developmental trajectory of the human retina. The ROs provide a fascinating tool for basic science research, eye disease modeling, treatment development, and biobanking for tissue/cell replacement. Here we review the previous studies that paved the way for RO technology, the two most widely accepted, standardized protocols to generate ROs, and the utilization of ROs in medical discovery. This review is conducted from the perspective of basic science research, transplantation for regenerative medicine, disease modeling, and therapeutic development for drug screening and gene therapy. ROs have opened avenues for new technologies such as assembloids, coculture with other organoids, vasculature or immune cells, microfluidic devices (organ-on-chip), extracellular vesicles for drug delivery, biomaterial engineering, advanced imaging techniques, and artificial intelligence (AI). Nevertheless, some shortcomings of ROs currently limit their translation for medical applications and pose a challenge for future research. Despite these limitations, ROs are a powerful tool for functional studies and therapeutic strategies for retinal diseases.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
- Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, IA, USA.
| | - Markus H Kuehn
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Center for the Prevention and Treatment of Visual Loss, Veterans Affairs Medical Center, Iowa City, IA, USA
- Institute for Vision Research, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
45
|
Shen W, Shao A, Zhou W, Lou L, Grzybowski A, Jin K, Ye J. Retinogenesis in a Dish: Bibliometric Analysis and Visualization of Retinal Organoids From 2011 to 2022. Cell Transplant 2023; 32:9636897231214321. [PMID: 38044501 PMCID: PMC10695087 DOI: 10.1177/09636897231214321] [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: 08/20/2023] [Revised: 10/12/2023] [Accepted: 10/31/2023] [Indexed: 12/05/2023] Open
Abstract
Retinal organoid (RO) is the three-dimensional (3D) retinal culture derived from pluripotent or embryonic stem cells which recapitulates organ functions, which was a revolutionary milestone in stem cell technology. The purpose of this study is to explore the hotspots and future directions on ROs, as well as to better understand the fields of greatest research opportunities. Eligible publications related to RO from 2011 to 2022 were acquired from the Web of Science (WoS) Core Collection database. Bibliometric analysis was performed by using software including VOSviewer, CiteSpace, and ArcGIS. A total of 520 articles were included, and the number of annual publications showed a rapid increase with an average rate of 40.86%. The United States published the most articles (241/520, 46.35%) with highest total citation frequencies (5,344). University College London (UK) contributed the largest publication output (40/520, 7.69%) and received highest total citation frequencies. Investigative Ophthalmology & Visual Science was the most productive journal with 129 articles. Majlinda Lako contributed the most research with 32 articles, while Olivier Goureau has the strongest collaboration work. Research could be subdivided into four keyword clusters: "culture and differentiation," "morphogenesis and modeling," "gene therapy," and "transplantation and visual restoration," and evolution of keywords was identified. Last decade has witnessed the huge progress in the field of RO, which is a young and promising research area with extensive and in-depth studies. More attention should be paid to RO-related models and therapies based on specific retinal diseases, especially inherited retinopathies.
Collapse
Affiliation(s)
- Wenyue Shen
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - An Shao
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Wuyuan Zhou
- Zhejiang Academy of Science and Technology Information, Hangzhou, China
| | - Lixia Lou
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Andrzej Grzybowski
- Institute for Research in Ophthalmology, Foundation for Ophthalmology Development, Poznan, Poland
| | - Kai Jin
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| | - Juan Ye
- Eye Center, The Second Affiliated Hospital School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, China
| |
Collapse
|
46
|
Kandoi S, Lamba DA. Retinal Organoids: A Human Model System for Development, Diseases, and Therapies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:549-554. [PMID: 37440085 DOI: 10.1007/978-3-031-27681-1_80] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Inherited retinal degenerations (IRD) encompasses a group of heterogeneous disorders causing debilitating visual diseases and blindness, affecting more than two million people worldwide, in all age groups. The inheritance patterns vary from autosomal dominant, autosomal recessive, X-linked, and sporadic with mutations in over 260 genes identified to date. Despite the significant advances in clinical diagnosis, there is no effective treatment available. Human-induced pluripotent stem cells (hiPSC) derived in vitro 3D retinal organoids offer a powerful preclinical tool to investigate the molecular mechanism(s) of inherited diseases. Organoids have the potential for the development of personalized therapies by modeling the disease-specific and patient-specific IRD. This mini-review will elaborate on the utility of the advanced culture model system by focusing on staging the in vitro human retinogenesis, modeling retinal diseases, and as a tool for testing potential therapeutic approaches to restore or prevent vision loss in affected individuals.
Collapse
Affiliation(s)
- Sangeetha Kandoi
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA.
- Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA.
| | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
- Eli & Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
47
|
Sun C, Chen S. Disease-causing mutations in genes encoding transcription factors critical for photoreceptor development. Front Mol Neurosci 2023; 16:1134839. [PMID: 37181651 PMCID: PMC10172487 DOI: 10.3389/fnmol.2023.1134839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Photoreceptor development of the vertebrate visual system is controlled by a complex transcription regulatory network. OTX2 is expressed in the mitotic retinal progenitor cells (RPCs) and controls photoreceptor genesis. CRX that is activated by OTX2 is expressed in photoreceptor precursors after cell cycle exit. NEUROD1 is also present in photoreceptor precursors that are ready to specify into rod and cone photoreceptor subtypes. NRL is required for the rod fate and regulates downstream rod-specific genes including the orphan nuclear receptor NR2E3 which further activates rod-specific genes and simultaneously represses cone-specific genes. Cone subtype specification is also regulated by the interplay of several transcription factors such as THRB and RXRG. Mutations in these key transcription factors are responsible for ocular defects at birth such as microphthalmia and inherited photoreceptor diseases such as Leber congenital amaurosis (LCA), retinitis pigmentosa (RP) and allied dystrophies. In particular, many mutations are inherited in an autosomal dominant fashion, including the majority of missense mutations in CRX and NRL. In this review, we describe the spectrum of photoreceptor defects that are associated with mutations in the above-mentioned transcription factors, and summarize the current knowledge of molecular mechanisms underlying the pathogenic mutations. At last, we deliberate the outstanding gaps in our understanding of the genotype-phenotype correlations and outline avenues for future research of the treatment strategies.
Collapse
Affiliation(s)
- Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Chi Sun,
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
48
|
Ng L, Liu H, Liu Y, Forrest D. Biphasic expression of thyroid hormone receptor TRβ1 in mammalian retina and anterior ocular tissues. Front Endocrinol (Lausanne) 2023; 14:1174600. [PMID: 37033230 PMCID: PMC10076699 DOI: 10.3389/fendo.2023.1174600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 03/08/2023] [Indexed: 04/11/2023] Open
Abstract
The retina is increasingly recognized as a target of thyroid hormone. We previously reported critical functions for thyroid hormone receptor TRβ2, encoded by Thrb, in cones, the photoreceptors that mediate color vision. TRβ1, another Thrb receptor isoform, is widely expressed in other tissues but little studied in the retina. Here, we investigate these N-terminal isoforms by RNA-sequencing analysis and reveal a striking biphasic profile for TRβ1 in mouse and human retina. In contrast to the early TRβ2 peak, TRβ1 peaks later during retinal maturation or later differentiation of human retinal organoids. This switch in receptor expression profiles was confirmed using lacZ reporter mice. TRβ1 localized in cones, amacrine cells and ganglion cells in contrast to the restricted expression of TRβ2 in cones. Intriguingly, TRβ1 was also detected in the retinal pigmented epithelium and in anterior structures in the ciliary margin zone, ciliary body and iris, suggesting novel functions in non-retinal eye tissues. Although TRβ1 was detected in cones, TRβ1-knockout mice displayed only minor changes in opsin photopigment expression and normal electroretinogram responses. Our results suggest that strikingly different temporal and cell-specific controls over TRβ1 and TRβ2 expression may underlie thyroid hormone actions in a range of ocular cell types. The TRβ1 expression pattern suggests novel functions in retinal and non-neural ocular tissues.
Collapse
Affiliation(s)
- Lily Ng
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, MD, United States
| | - Hong Liu
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, MD, United States
| | - Ye Liu
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, MD, United States
| | - Douglas Forrest
- National Institute of Diabetes and Digestive and Kidney Diseases, Laboratory of Endocrinology and Receptor Biology, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
49
|
Connolly K, Gonzalez-Cordero A. Modelling inner ear development and disease using pluripotent stem cells - a pathway to new therapeutic strategies. Dis Model Mech 2022; 15:dmm049593. [PMID: 36331565 PMCID: PMC10621662 DOI: 10.1242/dmm.049593] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023] Open
Abstract
The sensory epithelia of the mammalian inner ear enable sound and movement to be perceived. Damage to these epithelia can cause irreversible sensorineural hearing loss and vestibular dysfunction because they lack regenerative capacity. The human inner ear cannot be biopsied without causing permanent damage, significantly limiting the tissue samples available for research. Investigating disease pathology and therapeutic developments have therefore traditionally relied on animal models, which often cannot completely recapitulate the human otic systems. These challenges are now being partly addressed using induced pluripotent stem cell-derived cultures, which generate the sensory epithelial-like tissues of the inner ear. Here, we review how pluripotent stem cells have been used to produce two-dimensional and three-dimensional otic cultures, the strengths and limitations of these new approaches, and how they have been employed to investigate genetic and acquired forms of audiovestibular dysfunction. This Review provides an overview of the progress in pluripotent stem cell-derived otic cultures thus far, focusing on their applications in disease modelling and therapeutic trials. We survey their current limitations and future directions, highlighting their prospective utility for high-throughput drug screening and developing personalised medicine approaches.
Collapse
Affiliation(s)
- Keeva Connolly
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children's Medical Research Institute, Westmead, 2145 NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, 2145 NSW, Australia
| |
Collapse
|
50
|
Kruczek K, Qu Z, Welby E, Shimada H, Hiriyanna S, English MA, Zein WM, Brooks BP, Swaroop A. In vitro modeling and rescue of ciliopathy associated with IQCB1/NPHP5 mutations using patient-derived cells. Stem Cell Reports 2022; 17:2172-2186. [PMID: 36084637 PMCID: PMC9561628 DOI: 10.1016/j.stemcr.2022.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/12/2022] [Accepted: 08/12/2022] [Indexed: 11/26/2022] Open
Abstract
Mutations in the IQ calmodulin-binding motif containing B1 (IQCB1)/NPHP5 gene encoding the ciliary protein nephrocystin 5 cause early-onset blinding disease Leber congenital amaurosis (LCA), together with kidney dysfunction in Senior-Løken syndrome. For in vitro disease modeling, we obtained dermal fibroblasts from patients with NPHP5-LCA that were reprogrammed into induced pluripotent stem cells (iPSCs) and differentiated into retinal pigment epithelium (RPE) and retinal organoids. Patient fibroblasts and RPE demonstrated aberrantly elongated ciliary axonemes. Organoids revealed impaired development of outer segment structures, which are modified primary cilia, and mislocalization of visual pigments to photoreceptor cell soma. All patient-derived cells showed reduced levels of CEP290 protein, a critical cilia transition zone component interacting with NPHP5, providing a plausible mechanism for aberrant ciliary gating and cargo transport. Disease phenotype in NPHP5-LCA retinal organoids could be rescued by adeno-associated virus (AAV)-mediated IQCB1/NPHP5 gene augmentation therapy. Our studies thus establish a human disease model and a path for treatment of NPHP5-LCA.
Collapse
Affiliation(s)
- Kamil Kruczek
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Zepeng Qu
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Emily Welby
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Hiroko Shimada
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Suja Hiriyanna
- Ocular Gene Therapy Core, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Milton A English
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA
| | - Wadih M Zein
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brian P Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, MSC0610, 6 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|