1
|
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] [MESH Headings] [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
|
2
|
Ajekiigbe VO, Agbo CE, Ogieuhi IJ, Anthony CS, Adewole OA, Ahmed B, Akingbola A, Nwankwo CK, Kayode AT, Chima UE, Adaobi OM. Innovative approaches to treatment of eye diseases: advances in stem cell therapy use in ophthalmology. Int Ophthalmol 2025; 45:113. [PMID: 40120030 DOI: 10.1007/s10792-025-03493-7] [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: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION The human eye, a photo-sensory organ with an array of neuronal and tissue networks, remains susceptible to damage from various diseases and disorders despite its being a flawless masterpiece. It is estimated that over 2 billion people suffer from vision loss with common causative factors such as; age-related macular degeneration (AMD), glaucoma, cataracts, diabetic retinopathy, and infections amongst others. The use of Orthodox procedures has only helped mitigate the pathology; however, it doesn't serve any substantial curative purpose. More recently, the incorporation of new therapies via ocular delivery of nanomaterials and stem cell intervention has helped to change tides in the treatment of various ophthalmic pathologies. MAIN TEXT This review provides an overview of the current trends and breakthroughs in ophthalmology via stem cell therapy, with emphasis on its types, mechanisms, applications, and benefits. Mesenchymal stem cells which can arise from embryonic or adult origin possess some immunomodulatory effects that contribute to the therapeutic relevance of the MSCs and the ability to evade rejection from the host. However, the major drawback has been uncontrolled growth which can result in unintended side effects. Moreso, religious and ethical issues concerning the employment of MSCs from embryonic origin have also hindered clinical progression with its use. The use of stem cell therapy in the treatment of eye pathologies which is still undergoing clinical trials has shown to be a more viable treatment approach in ophthalmology as it targets retinal degenerative diseases thereby offering novel pathways for vision restoration. And also serves as a revolutionary alternative for treating severe ocular diseases. Stem cell delivery techniques might be quite cumbersome as the eye is a very delicate organ. The therapeutic interventional technique employed is aimed to ensure the reduction or absence of undesired effects in the deposition of the active pharmaceutical ingredient (API) being the stem cells. Techniques such as hydrogel-based injectables, which offer delivery of the APIs to the desired site of action without change in the physicochemical properties of the drug molecule, the scaffold delivery techniques, and the use of 3D bio-printing which can be used to develop scaffolds for retinal degeneration. The employment of artificial intelligence and machine learning in stem cell therapy has shown to be very fast and efficient in stem cell delivery and preventing likely human errors. CONCLUSIONS Unlike conventional treatments that often focus on managing symptoms, stem cells have the unique ability to repair and regenerate damaged tissues, addressing the root causes of the diseases. However, limitations due to economic, regulatory, and ethical challenges have posed barriers to advancing stem cell therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Bisharat Ahmed
- Allama Iqbal Medical College, University of Health Sciences, Lahore, Pakistan
| | - Adewunmi Akingbola
- Department of Public Health, University of Cambridge Cambridgeshire Old Trinity Schools, Cambridge, CB2 1TN, England, UK
| | | | | | | | | |
Collapse
|
3
|
Yang C, Du Z, Mei L, Chen X, Liao Y, Ge L, Kang J, Gu Z, Fan X, Xu H. Influences of lead-based perovskite nanoparticles exposure on early development of human retina. J Nanobiotechnology 2025; 23:144. [PMID: 40001141 PMCID: PMC11863764 DOI: 10.1186/s12951-025-03245-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Lead-based perovskite nanoparticles (Pb-PNPs) are widely utilized in the photovoltaic industry. However, due to their poor stability and high water solubility, lead often gets released into the environment, which can negatively impact the development of the central nervous system (CNS). As an extension of the CNS, the effects and mechanisms of Pb-PNPs on human retinal development have remained elusive. OBJECTIVES We aimed to investigate the effects of Pb-PNPs on human retinal development. METHODS Human embryonic stem cell-derived three-dimensional floating retinal organoids (hEROs) were established to simulate early retinal development. Using immunofluorescence staining, biological-transmission electron microscopy analysis, inductively coupled plasma-mass spectrometry, two-dimensional element distribution detection, and RNA sequencing, we evaluated and compared the toxicity of CsPbBr3 nanoparticles (a representative substance of Pb-PNPs) and Pb(AC)2 and investigated the toxicity-reducing effects of SiO2 encapsulation. RESULTS Our findings revealed that CsPbBr3 nanoparticles exposure resulted in a concentration-dependent decrease in the area and thickness of the neural retina in hEROs. Additionally, CsPbBr3 nanoparticles exposure hindered cell proliferation and promoted cell apoptosis while suppressing the retinal ganglion cell differentiation, an alteration that further led to the disruption of retinal structure. By contrast, CsPbBr3 nanoparticles exposure to hEROs was slightly less toxic than Pb(AC)2. Mechanistically, CsPbBr3 nanoparticles exposure activated endoplasmic reticulum stress, which promoted apoptosis by up-regulating Caspase-3 and inhibited retinal ganglion cell development by down-regulating Pax6. Interestingly, after coating CsPbBr3 nanoparticles with silica, it exhibited lower toxicities to hEROs by alleviating endoplasmic reticulum stress. CONCLUSION Overall, our study provides evidence of Pb-PNPs-induced developmental toxicity in the human retina, explores the potential mechanisms of CsPbBr3 nanoparticles' developmental toxicity, and suggests a feasible strategy to reduce toxicity.
Collapse
Affiliation(s)
- Cao Yang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing, 400038, China
| | - Zhulin Du
- Key Laboratory of Extreme Environmental Medicine Ministry of Education, Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Linqiang Mei
- Institute of High Energy Physics and National Center for Nanoscience and Technology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xia Chen
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing, 400038, China
| | - You Liao
- Institute of High Energy Physics and National Center for Nanoscience and Technology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, Beijing, 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingling Ge
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing, 400038, China
| | - Jiahui Kang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing, 400038, China
| | - Zhanjun Gu
- Institute of High Energy Physics and National Center for Nanoscience and Technology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, Chinese Academy of Sciences, Beijing, 100049, China.
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xiaotang Fan
- Key Laboratory of Extreme Environmental Medicine Ministry of Education, Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Southwest Eye Hospital, Southwest Hospital, Chongqing, 400038, China.
| |
Collapse
|
4
|
Watson A, Queen R, Ferrández-Peral L, Dorgau B, Collin J, Nelson A, Hussain R, Coxhead J, McCorkindale M, Atkinson R, Zerti D, Chichagova V, Conesa A, Armstrong L, Cremers FPM, Lako M. Unravelling genotype-phenotype correlations in Stargardt disease using patient-derived retinal organoids. Cell Death Dis 2025; 16:108. [PMID: 39971915 PMCID: PMC11840025 DOI: 10.1038/s41419-025-07420-7] [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: 09/09/2024] [Revised: 12/18/2024] [Accepted: 02/03/2025] [Indexed: 02/21/2025]
Abstract
Stargardt disease is an inherited retinopathy affecting approximately 1:8000 individuals. It is characterised by biallelic variants in ABCA4 which encodes a vital protein for the recycling of retinaldehydes in the retina. Despite its prevalence and impact, there are currently no treatments available for this condition. Furthermore, 35% of STGD1 cases remain genetically unsolved. To investigate the cellular and molecular characteristics associated with STGD1, we generated iPSCs from two monoallelic unresolved (PT1 & PT2), late-onset STGD1 cases with the heterozygous complex allele - c.[5461-10 T > C;5603 A > T]. Both patient iPSCs and those from a biallelic affected control (AC) carrying -c.4892 T > C and c.4539+2001G > A, were differentiated to retinal organoids, which developed all key retinal neurons and photoreceptors with outer segments positive for ABCA4 expression. We observed patient-specific disruption to lamination with OPN1MW/LW+ cone photoreceptor retention in the retinal organoid centre during differentiation. Photoreceptor retention was more severe in the AC case affecting both cones and rods, suggesting a genotype/phenotype correlation. scRNA-Seq suggests retention may be due to the induction of stress-related pathways in photoreceptors. Whole genome sequencing successfully identified the missing alleles in both cases; PT1 reported c.-5603A > T in homozygous state and PT2 uncovered a rare hypomorph - c.-4685T > C. Furthermore, retinal organoids were able to recapitulate the retina-specific splicing defect in PT1 as shown by long-read RNA-seq data. Collectively, these results highlight the suitability of retinal organoids in STGD1 modelling. Their ability to display genotype-phenotype correlations enhances their utility as a platform for therapeutic development.
Collapse
Affiliation(s)
- Avril Watson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcells Biotech Ltd., Newcastle upon Tyne, UK
| | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Luis Ferrández-Peral
- Institute for Integrative Systems Biology, University of Valencia, Valencia, Spain
| | - Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew Nelson
- NU-OMICs, Northumbria University, Newcastle Upon Tyne, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Robert Atkinson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Darin Zerti
- Department of Biotechnological and Applied Clinical Sciences, Università degli Studi dell'Aquila, L'Aquila, Italy
| | | | - Ana Conesa
- Institute for Integrative Systems Biology, University of Valencia, Valencia, Spain
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Newcells Biotech Ltd., Newcastle upon Tyne, UK
| | - Frans P M Cremers
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Duncan JL, Bowman A, Laster A, Gelfman C, Birch DG, Boye SE, Daiger SP, Del Priore L, Zack DJ, Handa JT. Inherited Retinal Degenerations and Non-Neovascular Age-Related Macular Degeneration: Progress and Unmet Needs. Transl Vis Sci Technol 2024; 13:28. [PMID: 39688851 DOI: 10.1167/tvst.13.12.28] [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: 12/18/2024] Open
Abstract
Inherited retinal degeneration (IRD) disease and age-related macular degeneration (AMD) are leading causes of irreversible vision loss and blindness. Although significant progress has advanced the field in the past 5 years, significant challenges remain. The current article reviews the accomplishments and research advances that have fueled the development of treatments for patients with IRD and AMD, including the first approved gene-augmentation treatment for RPE65-related retinal degeneration and complement inhibition therapies to slow progression of geographic atrophy (GA) in AMD. The article outlines opportunities to address gaps and unmet needs that should lead to additional progress toward the development of treatments for patients with IRDs and non-neovascular AMD in the future.
Collapse
Affiliation(s)
- Jacque L Duncan
- Wayne and Gladys Valley Center for Vision, Department of Ophthalmology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Amy Laster
- Foundation Fighting Blindness, Columbia, MD, USA
| | | | - David G Birch
- Rose-Silverthorne Retinal Degenerations Laboratory, Retina Foundation of the Southwest, Dallas, TX, USA
| | - Shannon E Boye
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Stephen P Daiger
- Human Genetics Center, Epidemiology Dept., School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Lucian Del Priore
- Department of Ophthalmology and Visual Science, Yale School of Medicine, New Haven, CT, USA
| | - Donald J Zack
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James T Handa
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
6
|
Cui X, Li X, Zheng H, Su Y, Zhang S, Li M, Hao X, Zhang S, Hu Z, Xia Z, Shi C, Xu Y, Mao C. Human midbrain organoids: a powerful tool for advanced Parkinson's disease modeling and therapy exploration. NPJ Parkinsons Dis 2024; 10:189. [PMID: 39428415 PMCID: PMC11491477 DOI: 10.1038/s41531-024-00799-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder marked by the loss of dopaminergic neurons in the substantia nigra. Despite progress, the pathogenesis remains unclear. Human midbrain organoids (hMLOs) have emerged as a promising model for studying PD, drug screening, and potential treatments. This review discusses the development of hMLOs, their application in PD research, and current challenges in organoid construction, highlighting possible optimization strategies.
Collapse
Affiliation(s)
- Xin Cui
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuyu Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Neuro-Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengjie Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xiaoyan Hao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shuo Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zhengwei Hu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Academy of Medical Sciences of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Zongping Xia
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Clinical Systems Biology Laboratories, Zhengzhou University, Zhengzhou, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China.
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
7
|
Ma SC, Xie YL, Wang Q, Fu SG, Wu HZ. Application of eye organoids in the study of eye diseases. Exp Eye Res 2024; 247:110068. [PMID: 39233304 DOI: 10.1016/j.exer.2024.110068] [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: 04/16/2024] [Revised: 08/22/2024] [Accepted: 09/01/2024] [Indexed: 09/06/2024]
Abstract
The eyes are one of the most important sensory organs in the human body. Currently, diseases such as limbal stem cell deficiency, cataract, retinitis pigmentosa and dry eye seriously threaten the quality of people's lives, and the treatment of advanced blinding eye disease and dry eye is ineffective and costly. Thus, new treatment modalities are urgently needed to improve patients' symptoms and suffering. In recent years, stem cell-derived three-dimensional structural organoids have been shown to mimic specific structures and functions similar to those of organs in the human body. Currently, 3D culture systems are used to construct organoids for different ocular growth and development models and ocular disease models to explore their physiological and pathological mechanisms. Eye organoids can also be used as a platform for drug screening. This paper reviews the latest research progress in regard to eye organoids (the cornea, lens, retina, lacrimal gland, and conjunctiva).
Collapse
Affiliation(s)
- Shi-Chao Ma
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-Lin Xie
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qian Wang
- School of Ophthalmology and Optometry, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shan-Gui Fu
- The Second School of Clinical Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hong-Ze Wu
- Department of Traditional Chinese Medicine, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, 332007, Jiangxi, China.
| |
Collapse
|
8
|
Kang S, Chen EC, Cifuentes H, Co JY, Cole G, Graham J, Hsia R, Kiyota T, Klein JA, Kroll KT, Nieves Lopez LM, Norona LM, Peiris H, Potla R, Romero-Lopez M, Roth JG, Tseng M, Fullerton AM, Homan KA. Complex in vitromodels positioned for impact to drug testing in pharma: a review. Biofabrication 2024; 16:042006. [PMID: 39189069 DOI: 10.1088/1758-5090/ad6933] [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: 12/22/2023] [Accepted: 07/30/2024] [Indexed: 08/28/2024]
Abstract
Recent years have seen the creation and popularization of various complexin vitromodels (CIVMs), such as organoids and organs-on-chip, as a technology with the potential to reduce animal usage in pharma while also enhancing our ability to create safe and efficacious drugs for patients. Public awareness of CIVMs has increased, in part, due to the recent passage of the FDA Modernization Act 2.0. This visibility is expected to spur deeper investment in and adoption of such models. Thus, end-users and model developers alike require a framework to both understand the readiness of current models to enter the drug development process, and to assess upcoming models for the same. This review presents such a framework for model selection based on comparative -omics data (which we term model-omics), and metrics for qualification of specific test assays that a model may support that we term context-of-use (COU) assays. We surveyed existing healthy tissue models and assays for ten drug development-critical organs of the body, and provide evaluations of readiness and suggestions for improving model-omics and COU assays for each. In whole, this review comes from a pharma perspective, and seeks to provide an evaluation of where CIVMs are poised for maximum impact in the drug development process, and a roadmap for realizing that potential.
Collapse
Affiliation(s)
- Serah Kang
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Eugene C Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Helen Cifuentes
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julia Y Co
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Gabrielle Cole
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica Graham
- Product Quality & Occupational Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of Americaica
| | - Rebecca Hsia
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Tomomi Kiyota
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Jessica A Klein
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Katharina T Kroll
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Lenitza M Nieves Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Leah M Norona
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Heshan Peiris
- Human Genetics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Ratnakar Potla
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Monica Romero-Lopez
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Julien G Roth
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Min Tseng
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Aaron M Fullerton
- Investigative Toxicology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| | - Kimberly A Homan
- Complex in vitro Systems Group, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States of America
| |
Collapse
|
9
|
Tsikandelova R, Galo E, Cerniauskas E, Hallam D, Georgiou M, Cerna-Chavez R, Atkinson R, Palmowski P, Burté F, Davies T, Steel DH, McKibbin M, Bond J, Haggarty J, Whitfield P, Korolchuk V, Armstrong L, Yang C, Dorgau B, Kurzawa-Akanbi M, Lako M. Retinal cells derived from patients with DRAM2-dependent CORD21 dystrophy exhibit key lysosomal enzyme deficiency and lysosomal content accumulation. Stem Cell Reports 2024; 19:1107-1121. [PMID: 38964324 PMCID: PMC11368688 DOI: 10.1016/j.stemcr.2024.06.002] [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: 04/04/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
Biallelic mutations in DRAM2 lead to an autosomal recessive cone-rod dystrophy known as CORD21, which typically presents between the third and sixth decades of life. Although DRAM2 localizes to the lysosomes of photoreceptor and retinal pigment epithelium (RPE) cells, its specific role in retinal degeneration has not been fully elucidated. In this study, we generated and characterized retinal organoids (ROs) and RPE cells from induced pluripotent stem cells (iPSCs) derived from two CORD21 patients. Our investigation revealed that CORD21-ROs and RPE cells exhibit abnormalities in lipid metabolism, defects in autophagic flux, accumulation of aberrant lysosomal content, and reduced lysosomal enzyme activity. We identified potential interactions of DRAM2 with vesicular trafficking proteins, suggesting its involvement in this cellular process. These findings collectively suggest that DRAM2 plays a crucial role in maintaining the integrity of photoreceptors and RPE cells by regulating lysosomal function, autophagy, and potentially vesicular trafficking.
Collapse
Affiliation(s)
| | - Eldo Galo
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | - Dean Hallam
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Maria Georgiou
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | | | | | - Florence Burté
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Tracey Davies
- Electron Microscopy Research Services, Newcastle University, Newcastle, UK
| | - David H Steel
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Martin McKibbin
- Leeds Teaching Hospitals NHS Trust, Leeds UK and Leeds Institute for Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Jacquelyn Bond
- Leeds Teaching Hospitals NHS Trust, Leeds UK and Leeds Institute for Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Jennifer Haggarty
- Shared Research Facilities, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Phil Whitfield
- Glasgow Polyomics and Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Chunbo Yang
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle, UK.
| |
Collapse
|
10
|
Matsushita T, Onishi A, Matsuyama T, Masuda T, Ogino Y, Kageyama M, Takahashi M, Uchiumi F. Rapid and efficient generation of mature retinal organoids derived from human pluripotent stem cells via optimized pharmacological modulation of Sonic hedgehog, activin A, and retinoic acid signal transduction. PLoS One 2024; 19:e0308743. [PMID: 39121095 PMCID: PMC11315325 DOI: 10.1371/journal.pone.0308743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/29/2024] [Indexed: 08/11/2024] Open
Abstract
Human retinal organoids have become indispensable tools for retinal disease modeling and drug screening. Despite its versatile applications, the long timeframe for their differentiation and maturation limits the throughput of such research. Here, we successfully shortened this timeframe by accelerating human retinal organoid development using unique pharmacological approaches. Our method comprised three key steps: 1) a modified self-formed ectodermal autonomous multizone (SEAM) method, including dual SMAD inhibition and bone morphogenetic protein 4 treatment, for initial neural retinal induction; 2) the concurrent use of a Sonic hedgehog agonist SAG, activin A, and all-trans retinoic acid for rapid retinal cell specification; and 3) switching to SAG treatment alone for robust retinal maturation and lamination. The generated retinal organoids preserved typical morphological features of mature retinal organoids, including hair-like surface structures and well-organized outer layers. These features were substantiated by the spatial immunostaining patterns of several retinal cell markers, including rhodopsin and L/M opsin expression in the outermost layer, which was accompanied by reduced ectopic cone photoreceptor generation. Importantly, our method required only 90 days for retinal organoid maturation, which is approximately two-thirds the time necessary for other conventional methods. These results indicate that thoroughly optimized pharmacological interventions play a pivotal role in rapid and precise photoreceptor development during human retinal organoid differentiation and maturation. Thus, our present method may expedite human retinal organoid research, eventually contributing to the development of better treatment options for various degenerative retinal diseases.
Collapse
Affiliation(s)
- Tokiyoshi Matsushita
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Akishi Onishi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Takahiro Matsuyama
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Tomohiro Masuda
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Yoko Ogino
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
| | - Masaaki Kageyama
- Product Discovery, Ophthalmology Innovation Center, Santen Pharmaceutical Co., Ltd., Ikoma, Nara, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Cell and Gene Therapy in Ophthalmology Laboratory, Baton Zone Program, RIKEN, Wako, Saitama, Japan
| | - Fumiaki Uchiumi
- Faculty of Pharmaceutical Sciences, Department of Gene Regulation, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
11
|
Harkin J, Peña KH, Gomes C, Hernandez M, Lavekar SS, So K, Lentsch K, Feder EM, Morrow S, Huang KC, Tutrow KD, Morris A, Zhang C, Meyer JS. A highly reproducible and efficient method for retinal organoid differentiation from human pluripotent stem cells. Proc Natl Acad Sci U S A 2024; 121:e2317285121. [PMID: 38870053 PMCID: PMC11194494 DOI: 10.1073/pnas.2317285121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 05/15/2024] [Indexed: 06/15/2024] Open
Abstract
Human pluripotent stem cell (hPSC)-derived retinal organoids are three-dimensional cellular aggregates that differentiate and self-organize to closely mimic the spatial and temporal patterning of the developing human retina. Retinal organoid models serve as reliable tools for studying human retinogenesis, yet limitations in the efficiency and reproducibility of current retinal organoid differentiation protocols have reduced the use of these models for more high-throughput applications such as disease modeling and drug screening. To address these shortcomings, the current study aimed to standardize prior differentiation protocols to yield a highly reproducible and efficient method for generating retinal organoids. Results demonstrated that through regulation of organoid size and shape using quick reaggregation methods, retinal organoids were highly reproducible compared to more traditional methods. Additionally, the timed activation of BMP signaling within developing cells generated pure populations of retinal organoids at 100% efficiency from multiple widely used cell lines, with the default forebrain fate resulting from the inhibition of BMP signaling. Furthermore, given the ability to direct retinal or forebrain fates at complete purity, mRNA-seq analyses were then utilized to identify some of the earliest transcriptional changes that occur during the specification of these two lineages from a common progenitor. These improved methods also yielded retinal organoids with expedited differentiation timelines when compared to traditional methods. Taken together, the results of this study demonstrate the development of a highly reproducible and minimally variable method for generating retinal organoids suitable for analyzing the earliest stages of human retinal cell fate specification.
Collapse
Affiliation(s)
- Jade Harkin
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kiersten H. Peña
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Cátia Gomes
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Melody Hernandez
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Sailee S. Lavekar
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Kaman So
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kelly Lentsch
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Elyse M. Feder
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN46202
| | - Sarah Morrow
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
| | - Kang-Chieh Huang
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN46202
| | - Kaylee D. Tutrow
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Ann Morris
- Department of Biology, University of Kentucky, Lexington, KY40506
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
| | - Jason S. Meyer
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN46202
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN46202
| |
Collapse
|
12
|
Eintracht J, Owen N, Harding P, Moosajee M. Disruption of common ocular developmental pathways in patient-derived optic vesicle models of microphthalmia. Stem Cell Reports 2024; 19:839-858. [PMID: 38821055 PMCID: PMC11390689 DOI: 10.1016/j.stemcr.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 06/02/2024] Open
Abstract
Genetic perturbations influencing early eye development can result in microphthalmia, anophthalmia, and coloboma (MAC). Over 100 genes are associated with MAC, but little is known about common disease mechanisms. In this study, we generated induced pluripotent stem cell (iPSC)-derived optic vesicles (OVs) from two unrelated microphthalmia patients and healthy controls. At day 20, 35, and 50, microphthalmia patient OV diameters were significantly smaller, recapitulating the "small eye" phenotype. RNA sequencing (RNA-seq) analysis revealed upregulation of apoptosis-initiating and extracellular matrix (ECM) genes at day 20 and 35. Western blot and immunohistochemistry revealed increased expression of lumican, nidogen, and collagen type IV, suggesting ECM overproduction. Increased apoptosis was observed in microphthalmia OVs with reduced phospho-histone 3 (pH3+) cells confirming decreased cell proliferation at day 35. Pharmacological inhibition of caspase-8 activity with Z-IETD-FMK decreased apoptosis in one patient model, highlighting a potential therapeutic approach. These data reveal shared pathophysiological mechanisms contributing to a microphthalmia phenotype.
Collapse
Affiliation(s)
| | | | | | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; Moorfields Eye Hospital NHS Foundation Trust, London EC1V 9EL, UK; Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
13
|
Kawai K, Ho MT, Ueno Y, Abdo D, Xue C, Nonaka H, Nishida H, Honma Y, Wallace VA, Shoichet MS. Hyaluronan improves photoreceptor differentiation and maturation in human retinal organoids. Acta Biomater 2024; 181:117-132. [PMID: 38705224 DOI: 10.1016/j.actbio.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
Human stem cell-derived organoids enable both disease modeling and serve as a source of cells for transplantation. Human retinal organoids are particularly important as a source of human photoreceptors; however, the long differentiation period required and lack of vascularization in the organoid often results in a necrotic core and death of inner retinal cells before photoreceptors are fully mature. Manipulating the in vitro environment of differentiating retinal organoids through the incorporation of extracellular matrix components could influence retinal development. We investigated the addition of hyaluronan (HA), a component of the interphotoreceptor matrix, as an additive to promote long-term organoid survival and enhance retinal maturation. HA treatment had a significant reduction in the proportion of proliferating (Ki67+) cells and increase in the proportion of photoreceptors (CRX+), suggesting that HA accelerated photoreceptor commitment in vitro. HA significantly upregulated genes specific to photoreceptor maturation and outer segment development. Interestingly, prolonged HA-treatment significantly decreased the length of the brush border layer compared to those in control retinal organoids, where the photoreceptor outer segments reside; however, HA-treated organoids also had more mature outer segments with organized discs structures, as revealed by transmission electron microscopy. The brush border layer length was inversely proportional to the molar mass and viscosity of the hyaluronan added. This is the first study to investigate the role of exogenous HA, viscosity, and polymer molar mass on photoreceptor maturation, emphasizing the importance of material properties on organoid culture. STATEMENT OF SIGNIFICANCE: Retinal organoids are a powerful tool to study retinal development in vitro, though like many other organoid systems, can be highly variable. In this work, Shoichet and colleagues investigated the use of hyaluronan (HA), a native component of the interphotoreceptor matrix, to improve photoreceptor maturation in developing human retinal organoids. HA promoted human photoreceptor differentiation leading to mature outer segments with disc formation and more uniform and healthy retinal organoids. These findings highlight the importance of adding components native to the developing retina to generate more physiologically relevant photoreceptors for cell therapy and in vitro models to drive drug discovery and uncover novel disease mechanisms.
Collapse
Affiliation(s)
- Kotoe Kawai
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Margaret T Ho
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Yui Ueno
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada
| | - Dhana Abdo
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Chang Xue
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada
| | - Hidenori Nonaka
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Hiroyuki Nishida
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Yoichi Honma
- Regenerative Medicine Research and Planning Division, Rohto Pharmaceutical Co., Ltd., 6-5-4 Kunimidai, Kizugawa, Kyoto 619-0216, Japan
| | - Valerie A Wallace
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Canada; Department of Ophthalmology and Vision Sciences, University of Toronto, Canada
| | - Molly S Shoichet
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada; Institute of Biomedical Engineering, University of Toronto, Canada; Department of Chemical Engineering and Applied Chemistry, University of Toronto, Canada; Department of Chemistry, University of Toronto, Canada.
| |
Collapse
|
14
|
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
|
15
|
Dorgau B, Collin J, Rozanska A, Boczonadi V, Moya-Molina M, Unsworth A, Hussain R, Coxhead J, Dhanaseelan T, Armstrong L, Queen R, Lako M. Deciphering the spatiotemporal transcriptional and chromatin accessibility of human retinal organoid development at the single-cell level. iScience 2024; 27:109397. [PMID: 38510120 PMCID: PMC10952046 DOI: 10.1016/j.isci.2024.109397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/28/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Molecular information on the early stages of human retinal development remains scarce due to limitations in obtaining early human eye samples. Pluripotent stem cell-derived retinal organoids (ROs) provide an unprecedented opportunity for studying early retinogenesis. Using a combination of single cell RNA-seq and spatial transcriptomics we present for the first-time a single cell spatiotemporal transcriptome of RO development. Our data demonstrate that ROs recapitulate key events of retinogenesis including optic vesicle/cup formation, presence of a putative ciliary margin zone, emergence of retinal progenitor cells and their orderly differentiation to retinal neurons. Combining the scRNA- with scATAC-seq data, we were able to reveal cell-type specific transcription factor binding motifs on accessible chromatin at each stage of organoid development, and to show that chromatin accessibility is highly correlated to the developing human retina, but with some differences in the temporal emergence and abundance of some of the retinal neurons.
Collapse
Affiliation(s)
- Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Joseph Collin
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Agata Rozanska
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Veronika Boczonadi
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Marina Moya-Molina
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
- Newcells Biotech, Newcastle upon Tyne NE4 5BX, UK
| | - Adrienne Unsworth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rafiqul Hussain
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Jonathan Coxhead
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Tamil Dhanaseelan
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Rachel Queen
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Zhao H, Yan F. Retinal Organoids: A Next-Generation Platform for High-Throughput Drug Discovery. Stem Cell Rev Rep 2024; 20:495-508. [PMID: 38079086 PMCID: PMC10837228 DOI: 10.1007/s12015-023-10661-8] [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: 11/28/2023] [Indexed: 02/03/2024]
Abstract
Retinal diseases are leading causes of blindness globally. Developing new drugs is of great significance for preventing vision loss. Current drug discovery relies mainly on two-dimensional in vitro models and animal models, but translation to human efficacy and safety is biased. In recent years, the emergence of retinal organoid technology platforms, utilizing three-dimensional microenvironments to better mimic retinal structure and function, has provided new platforms for exploring pathogenic mechanisms and drug screening. This review summarizes the latest advances in retinal organoid technology, emphasizing its application advantages in high-throughput drug screening, efficacy and toxicity evaluation, and translational medicine research. The review also prospects the combination of emerging technologies such as organ-on-a-chip, 3D bioprinting, single cell sequencing, gene editing with retinal organoid technology, which is expected to further optimize retinal organoid models and advance the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Hongkun Zhao
- Key Laboratory of Yunnan Province, Yunnan Eye Institute, Affiliated Hospital of Yunnan University, Yunnan University, Kunming, Yunnan, China
| | - Fei Yan
- Department of Pathology and Pathophysiology, Faculty of Basic Medicine School, Kunming Medical University, 1168 Yuhua Street, Chunrong West Road, Chenggong District, Kunming, Yunnan, 650500, China.
| |
Collapse
|
19
|
Keuthan CJ, Zack DJ. RNA Isolation from Human Stem Cell-Derived Retinal Organoids. Methods Mol Biol 2024; 2822:3-11. [PMID: 38907907 DOI: 10.1007/978-1-0716-3918-4_1] [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: 06/24/2024]
Abstract
RNA isolation is an essential first step for many types of molecular analyses, including reverse transcription PCR (RT-PCR)/quantitative RT-PCR (qRT-PCR), Northern blotting, microarrays, and RNA-sequencing. While many RNA purification methods have been reported, it can be challenging to extract sufficient quantity, and suitable quality, of RNA from very small amounts of tissue and/or samples containing low numbers of cells. Here we outline a total RNA isolation method that reproducibly yields high-quality RNA from human stem cell-derived retinal organoids for downstream transcriptomic analysis.
Collapse
Affiliation(s)
- Casey J Keuthan
- Department of Ophthalmology | Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Donald J Zack
- Department of Ophthalmology | Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Gong Y, Ge L, Li Q, Gong J, Chen M, Gao H, Kang J, Yu T, Li J, Xu H. Ethanol Causes Cell Death and Neuronal Differentiation Defect During Initial Neurogenesis of the Neural Retina by Disrupting Calcium Signaling in Human Retinal Organoids. Stem Cell Rev Rep 2023; 19:2790-2806. [PMID: 37603136 DOI: 10.1007/s12015-023-10604-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
Fetal Alcohol Syndrome (FAS) affects a significant proportion, exceeding 90%, of afflicted children, leading to severe ocular aberrations such as microphthalmia and optic nerve hypoplasia. During the early stages of pregnancy, the commencement of neural retina neurogenesis represents a critical period for human eye development, concurrently exposing the developing retinal structures to the highest risk of prenatal ethanol exposure due to a lack of awareness. Despite the paramount importance of this period, the precise influence and underlying mechanisms of short-term ethanol exposure on the developmental process of the human neural retina have remained largely elusive. In this study, we utilize the human embryonic stem cells derived retinal organoids (hROs) to recapitulate the initial retinal neurogenesis and find that 1% (v/v) ethanol slows the growth of hROs by inducing robust cell death and retinal ganglion cell differentiation defect. Bulk RNA-seq analysis and two-photon microscope live calcium imaging reveal altered calcium signaling dynamics derived from ethanol-induced down-regulation of RYR1 and CACNA1S. Moreover, the calcium-binding protein RET, one of the downstream effector genes of the calcium signaling pathway, synergistically integrates ethanol and calcium signals to abort neuron differentiation and cause cell death. To sum up, our study illustrates the effect and molecular mechanism of ethanol on the initial neurogenesis of the human embryonic neural retina, providing a novel interpretation of the ocular phenotype of FAS and potentially informing preventative measures for susceptible populations.
Collapse
Affiliation(s)
- Yu Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Lingling Ge
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Qiyou Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jing Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Min Chen
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Hui Gao
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jiahui Kang
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Ting Yu
- Department of Clinical Laboratory, The 89th Hospital of The People's Liberation Army, Weifang, People's Republic of China
| | - Jiawen Li
- Department of Ophthalmology, 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, People's Republic of China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China.
| |
Collapse
|
21
|
Chakrabarty K, Nayak D, Debnath J, Das D, Shetty R, Ghosh A. Retinal organoids in disease modeling and drug discovery: Opportunities and challenges. Surv Ophthalmol 2023:S0039-6257(23)00127-3. [PMID: 37778668 DOI: 10.1016/j.survophthal.2023.09.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Diseases leading to retinal cell loss can cause severe visual impairment and blindness. The lack of effective therapies to address retinal cell loss and the absence of intrinsic regeneration in the human retina leads to an irreversible pathological condition. Progress in recent years in the generation of human three-dimensional retinal organoids from pluripotent stem cells makes it possible to recreate the cytoarchitecture and associated cell-cell interactions of the human retina in remarkable detail. These human three-dimensional retinal organoid systems made of distinct retinal cell types and possessing contextual physiological responses allow the study of human retina development and retinal disease pathology in a way animal model and two-dimensional cell cultures were unable to achieve. We describe the derivation of retinal organoids from human pluripotent stem cells and their application for modeling retinal disease pathologies, while outlining the opportunities and challenges for its application in academia and industry.
Collapse
Affiliation(s)
- Koushik Chakrabarty
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India.
| | - Divyani Nayak
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Jayasree Debnath
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| | - Debashish Das
- Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bangalore, Karnataka, India
| |
Collapse
|
22
|
Móvio MI, de Lima-Vasconcellos TH, Dos Santos GB, Echeverry MB, Colombo E, Mattos LS, Resende RR, Kihara AH. Retinal organoids from human-induced pluripotent stem cells: From studying retinal dystrophies to early diagnosis of Alzheimer's and Parkinson's disease. Semin Cell Dev Biol 2023; 144:77-86. [PMID: 36210260 DOI: 10.1016/j.semcdb.2022.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 09/15/2022] [Accepted: 09/17/2022] [Indexed: 11/18/2022]
Abstract
Human-induced pluripotent stem cells (hiPSCs) have provided new methods to study neurodegenerative diseases. In addition to their wide application in neuronal disorders, hiPSCs technology can also encompass specific conditions, such as inherited retinal dystrophies. The possibility of evaluating alterations related to retinal disorders in 3D organoids increases the truthfulness of in vitro models. Moreover, both Alzheimer's (AD) and Parkinson's disease (PD) have been described as causing early retinal alterations, generating beta-amyloid protein accumulation, or affecting dopaminergic amacrine cells. This review addresses recent advances and future perspectives obtained from in vitro modeling of retinal diseases, focusing on retinitis pigmentosa (RP). Additionally, we depicted the possibility of evaluating changes related to AD and PD in retinal organoids obtained from potential patients long before the onset of the disease, constituting a valuable tool in early diagnosis. With this, we pointed out prospects in the study of retinal dystrophies and early diagnosis of AD and PD.
Collapse
Affiliation(s)
- Marília Inês Móvio
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | | | | | - Marcela Bermudez Echeverry
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Elisabetta Colombo
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Leonardo S Mattos
- Biomedical Robotics Laboratory, Department of Advanced Robotics, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Rodrigo Ribeiro Resende
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexandre Hiroaki Kihara
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil; Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil.
| |
Collapse
|
23
|
Smirnov A, Melino G, Candi E. Gene expression in organoids: an expanding horizon. Biol Direct 2023; 18:11. [PMID: 36964575 PMCID: PMC10038780 DOI: 10.1186/s13062-023-00360-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/20/2023] [Indexed: 03/26/2023] Open
Abstract
Recent development of human three-dimensional organoid cultures has opened new doors and opportunities ranging from modelling human development in vitro to personalised cancer therapies. These new in vitro systems are opening new horizons to the classic understanding of human development and disease. However, the complexity and heterogeneity of these models requires cutting-edge techniques to capture and trace global changes in gene expression to enable identification of key players and uncover the underlying molecular mechanisms. Rapid development of sequencing approaches made possible global transcriptome analyses and epigenetic profiling. Despite challenges in organoid culture and handling, these techniques are now being adapted to embrace organoids derived from a wide range of human tissues. Here, we review current state-of-the-art multi-omics technologies, such as single-cell transcriptomics and chromatin accessibility assays, employed to study organoids as a model for development and a platform for precision medicine.
Collapse
Affiliation(s)
- Artem Smirnov
- Department of Experimental Medicine, Torvergata Oncoscience Research, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, Torvergata Oncoscience Research, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy
| | - Eleonora Candi
- Department of Experimental Medicine, Torvergata Oncoscience Research, University of Rome "Tor Vergata", Via Montpellier 1, 00133, Rome, Italy.
- Biochemistry Laboratory, Istituto Dermopatico Immacolata (IDI-IRCCS), 00166, Rome, Italy.
| |
Collapse
|
24
|
Kim AH, Kolesnikova M, Ngo WK, Tsang SH. Effects of medications on hypoxia-inducible factor in the retina: A review. Clin Exp Ophthalmol 2023; 51:205-216. [PMID: 36594241 DOI: 10.1111/ceo.14161] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 01/04/2023]
Abstract
Hypoxia-inducible factor (HIF) plays a critical role in the mechanisms that allow cells to adapt to various oxygen levels in the environment. Specifically, HIF-1⍺ has shown to be widely involved in cellular repair, survival, and energy metabolism. HIF-1⍺ has also been found in increased levels in cancer cells, highlighting the importance of balance in the hypoxic response. Promoting HIF-1⍺ activity as a potential therapy for degenerative diseases and inhibiting HIF-1⍺ as a therapy for pathologies with overactive cell proliferation are actively being explored. Digoxin and metformin, HIF-1⍺ inhibitors, and deferoxamine and ⍺-ketoglutarate analogues, HIF-1⍺ activators, are being studied for application in age-related macular degeneration, diabetic retinopathy, and retinitis pigmentosa. However, these same medications have retinal toxicities that must be assessed before implementation of therapeutic care. Herein, we highlight the duality of therapeutic and toxic potential of HIF-1⍺ that must be carefully assessed prior to its clinical application in retinal disorders.
Collapse
Affiliation(s)
- Angela H Kim
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,SUNY Downstate Medical School, Brooklyn, New York, USA
| | - Masha Kolesnikova
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,SUNY Downstate Medical School, Brooklyn, New York, USA
| | - Wei Kiong Ngo
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore, Singapore
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York-Presbyterian Hospital, New York, New York, USA.,Edward S. Harkness Eye Institute, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, New York, USA.,Departments of Pathology & Cell Biology, Columbia Stem Cell Initiative, New York, New York, USA.,Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
25
|
Li M, Gong J, Ge L, Gao H, Yang J, Yang C, Kang J, Fang Y, Xu H. Development of human retinal organoid models for bisphenol toxicity assessment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 245:114094. [PMID: 36126549 DOI: 10.1016/j.ecoenv.2022.114094] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/30/2022] [Accepted: 09/13/2022] [Indexed: 06/15/2023]
Abstract
Bisphenols, including Bisphenol A (BPA), Tetrabromobisphenol A (TBBPA), and Tetrabromobisphenol S (TBBPS), have been widely applied in the production of polycarbonate plastics and epoxy resins and have been detected in the environment worldwide. The frequent detection of bisphenols in maternal and fetal samples has raised concerns about their toxic effects on human embryonic development, especially on the development of the central nervous system. However, the effect of bisphenols on human retinal development is still unknown. In this study, to evaluate the toxicity of bisphenols on early retinal development, human embryonic stem cells were induced to differentiate into retinal organoids that responded to BPA, TBBPA, and TBBPS, at human exposure relevant concentrations. The global gene expression of retinal organoids was analyzed by RNA sequencing (RNA-seq). A set of retinal development-related biological processes, including neuron differentiation, phototransduction, axon guidance, and retina layer formation, were identified in retinal organoids corresponding to different developmental stages. The RNA-seq data also showed that BPA, TBBPA, and TBBPS influenced retinal development by interfering with the Cytokine-cytokine receptor interaction pathway. HSPA6, HIF1A-AS3, CDC20B, IL19, OAS1, HSPA7, and RN7SK were dysregulated by these chemicals. Additionally, BPA, TBBPA, and TBBPS exhibited different toxic effects on neural retina development, with TBBPA appearing to exert more toxicity than BPA and TBBPS. Furthermore, three bisphenols exhibited different effects at different stages of neural retina development. The sensitivity of retinal development to bisphenols depends on their developmental stage. This study provides new insights into the deep dissection of retinotoxicity after prenatal bisphenol exposure.
Collapse
Affiliation(s)
- Minghui Li
- 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
| | - Lingling Ge
- 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
| | - Hui Gao
- 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
| | - Junling 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
| | - 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
| | - 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
| | - Yajie Fang
- 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
| | - 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
|
26
|
Retinal Toxicity Induced by Chemical Agents. Int J Mol Sci 2022; 23:ijms23158182. [PMID: 35897758 PMCID: PMC9331776 DOI: 10.3390/ijms23158182] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Vision is an important sense for humans, and visual impairment/blindness has a huge impact in daily life. The retina is a nervous tissue that is essential for visual processing since it possesses light sensors (photoreceptors) and performs a pre-processing of visual information. Thus, retinal cell dysfunction or degeneration affects visual ability and several general aspects of the day-to-day of a person's lives. The retina has a blood-retinal barrier, which protects the tissue from a wide range of molecules or microorganisms. However, several agents, coming from systemic pathways, reach the retina and influence its function and survival. Pesticides are still used worldwide for agriculture, contaminating food with substances that could reach the retina. Natural products have also been used for therapeutic purposes and are another group of substances that can get to the retina. Finally, a wide number of medicines administered for different diseases can also affect the retina. The present review aimed to gather recent information about the hazard of these products to the retina, which could be used to encourage the search for more healthy, suitable, or less risky agents.
Collapse
|