1
|
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
|
2
|
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
|
3
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [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: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
4
|
Lee S, Chung WG, Jeong H, Cui G, Kim E, Lim JA, Seo H, Kwon YW, Byeon SH, Lee J, Park JU. Electrophysiological Analysis of Retinal Organoid Development Using 3D Microelectrodes of Liquid Metals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404428. [PMID: 38896876 DOI: 10.1002/adma.202404428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/17/2024] [Indexed: 06/21/2024]
Abstract
Despite of the substantial potential of human-derived retinal organoids, the degeneration of retinal ganglion cells (RGCs) during maturation limits their utility in assessing the functionality of later-born retinal cell subtypes. Additionally, conventional analyses primarily rely on fluorescent emissions, which limits the detection of actual cell functionality while risking damage to the 3D cytoarchitecture of organoids. Here, an electrophysiological analysis is presented to monitor RGC development in early to mid-stage retinal organoids, and compare distinct features with fully-mature mouse retina. This approach utilizes high-resolution 3D printing of liquid-metal microelectrodes, enabling precise targeting of specific inner retinal layers within organoids. The adaptable distribution and softness of these microelectrodes facilitate the spatiotemporal recording of inner retinal signals. This study not only demonstrates the functional properties of RGCs in retinal organoid development but also provides insights into their synaptic connectivity, reminiscent of fetal native retinas. Further comparison with fully-mature mouse retina in vivo verifies the organoid features, highlighting the potential of early-stage retinal organoids in biomedical research.
Collapse
Affiliation(s)
- Sanghoon Lee
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Won Gi Chung
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Han Jeong
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Gang Cui
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Enji Kim
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jeong Ah Lim
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Hunkyu Seo
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Won Kwon
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Suk Ho Byeon
- Institute of Vision Research, Department of Ophthalmology, Severance Eye Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Junwon Lee
- Institute of Vision Research, Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Jang-Ung Park
- Department of Materials Science & Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Neurosurgery, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| |
Collapse
|
5
|
Ashworth KE, Weisbrod J, Ballios BG. Inherited Retinal Diseases and Retinal Organoids as Preclinical Cell Models for Inherited Retinal Disease Research. Genes (Basel) 2024; 15:705. [PMID: 38927641 PMCID: PMC11203130 DOI: 10.3390/genes15060705] [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: 03/15/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Inherited retinal diseases (IRDs) are a large group of genetically and clinically diverse blinding eye conditions that result in progressive and irreversible photoreceptor degeneration and vision loss. To date, no cures have been found, although strides toward treatments for specific IRDs have been made in recent years. To accelerate treatment discovery, retinal organoids provide an ideal human IRD model. This review aims to give background on the development and importance of retinal organoids for the human-based in vitro study of the retina and human retinogenesis and retinal pathologies. From there, we explore retinal pathologies in the context of IRDs and the current landscape of IRD treatment discovery. We discuss the usefulness of retinal organoids in this context (as a patient-derived cell model for IRDs) to precisely understand the pathogenesis and potential mechanisms behind a specific IRD-causing variant of interest. Finally, we discuss the importance and promise of retinal organoids in treatment discovery for IRDs, now and in the future.
Collapse
Affiliation(s)
- Kristen E. Ashworth
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 3H2, Canada;
- Donald K. Johnson Eye Institute, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada;
| | - Jessica Weisbrod
- Donald K. Johnson Eye Institute, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada;
| | - Brian G. Ballios
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 3H2, Canada;
- Donald K. Johnson Eye Institute, Toronto Western Hospital, Toronto, ON M5T 2S8, Canada;
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON M5T 3A9, Canada
| |
Collapse
|
6
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
7
|
James E, Vielle A, Cusato K, Li H, Lee B, Parween S, Howell A, Johnson NR, Chial HJ, Potter H, Vergara MN. Human iPSC-derived retinal organoids develop robust Alzheimer's disease neuropathology. Front Cell Neurosci 2024; 18:1340448. [PMID: 38323188 PMCID: PMC10844524 DOI: 10.3389/fncel.2024.1340448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Alzheimer's disease (AD), characterized by memory loss and cognitive decline, affects nearly 50 million people worldwide. Amyloid beta (Aβ) plaques and intracellular neurofibrillary tangles (NFTs) of phosphorylated Tau protein (pTau) are key histopathological features of the disease in the brain, and recent advances have also identified AD histopathology in the retina. Thus, the retina represents a central nervous system (CNS) tissue highly amenable to non-invasive diagnostic imaging that shows promise as a biomarker for early AD. Given the devastating effects of AD on patients, their families, and society, new treatment modalities that can significantly alter the disease course are urgently needed. In this study, we have developed and characterized a novel human retinal organoid (RO) model derived from induced pluripotent stem cells (iPSCs) from patients with familial AD due to mutations in the amyloid precursor protein gene (APP). Using immunofluorescence and histological staining, we evaluated the cellular composition and AD histopathological features of AD-ROs compared to control ROs from healthy individuals. We found that AD-ROs largely resemble their healthy control counterparts in cellular composition but display increased levels of Aβ and pTau. We also present proof of principle of an assay to quantify amyloid levels in whole ROs. This in vitro model of the human AD retina constitutes a new tool for drug screening, biomarker discovery, and pathophysiological studies.
Collapse
Affiliation(s)
- Ethan James
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anne Vielle
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Karen Cusato
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Helen Li
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Byoungin Lee
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Shama Parween
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Anna Howell
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
| | - Noah R. Johnson
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Heidi J. Chial
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Huntington Potter
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - M. Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Research Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, CO, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- University of Colorado Alzheimer’s and Cognition Center, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
8
|
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
|
9
|
Keuthan CJ, Karma S, Zack DJ. Alternative RNA Splicing in the Retina: Insights and Perspectives. Cold Spring Harb Perspect Med 2023; 13:a041313. [PMID: 36690463 PMCID: PMC10547393 DOI: 10.1101/cshperspect.a041313] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alternative splicing is a fundamental and highly regulated post-transcriptional process that enhances transcriptome and proteome diversity. This process is particularly important in neuronal tissues, such as the retina, which exhibit some of the highest levels of differentially spliced genes in the body. Alternative splicing is regulated both temporally and spatially during neuronal development, can be cell-type-specific, and when altered can cause a number of pathologies, including retinal degeneration. Advancements in high-throughput sequencing technologies have facilitated investigations of the alternative splicing landscape of the retina in both healthy and disease states. Additionally, innovations in human stem cell engineering, specifically in the generation of 3D retinal organoids, which recapitulate many aspects of the in vivo retinal microenvironment, have aided studies of the role of alternative splicing in human retinal development and degeneration. Here we review these advances and discuss the ongoing development of strategies for the treatment of alternative splicing-related retinal disease.
Collapse
Affiliation(s)
- Casey J Keuthan
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Sadik Karma
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| | - Donald J Zack
- Departments of Ophthalmology, Wilmer Eye Institute, Neuroscience, Molecular Biology and Genetics, and Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, USA
| |
Collapse
|
10
|
Yee T, Wert KJ. Human retina-in-a-dish: Unlocking the potential to study mechanisms of inherited retinal disease. Mol Ther Methods Clin Dev 2023; 30:573-575. [PMID: 37693947 PMCID: PMC10485576 DOI: 10.1016/j.omtm.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Affiliation(s)
- Tiffany Yee
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katherine J. Wert
- Department of Ophthalmology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Peter O’Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Morales Pantoja IE, Smirnova L, Muotri AR, Wahlin KJ, Kahn J, Boyd JL, Gracias DH, Harris TD, Cohen-Karni T, Caffo BS, Szalay AS, Han F, Zack DJ, Etienne-Cummings R, Akwaboah A, Romero JC, Alam El Din DM, Plotkin JD, Paulhamus BL, Johnson EC, Gilbert F, Curley JL, Cappiello B, Schwamborn JC, Hill EJ, Roach P, Tornero D, Krall C, Parri R, Sillé F, Levchenko A, Jabbour RE, Kagan BJ, Berlinicke CA, Huang Q, Maertens A, Herrmann K, Tsaioun K, Dastgheyb R, Habela CW, Vogelstein JT, Hartung T. First Organoid Intelligence (OI) workshop to form an OI community. Front Artif Intell 2023; 6:1116870. [PMID: 36925616 PMCID: PMC10013972 DOI: 10.3389/frai.2023.1116870] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/08/2023] [Indexed: 03/08/2023] Open
Abstract
The brain is arguably the most powerful computation system known. It is extremely efficient in processing large amounts of information and can discern signals from noise, adapt, and filter faulty information all while running on only 20 watts of power. The human brain's processing efficiency, progressive learning, and plasticity are unmatched by any computer system. Recent advances in stem cell technology have elevated the field of cell culture to higher levels of complexity, such as the development of three-dimensional (3D) brain organoids that recapitulate human brain functionality better than traditional monolayer cell systems. Organoid Intelligence (OI) aims to harness the innate biological capabilities of brain organoids for biocomputing and synthetic intelligence by interfacing them with computer technology. With the latest strides in stem cell technology, bioengineering, and machine learning, we can explore the ability of brain organoids to compute, and store given information (input), execute a task (output), and study how this affects the structural and functional connections in the organoids themselves. Furthermore, understanding how learning generates and changes patterns of connectivity in organoids can shed light on the early stages of cognition in the human brain. Investigating and understanding these concepts is an enormous, multidisciplinary endeavor that necessitates the engagement of both the scientific community and the public. Thus, on Feb 22-24 of 2022, the Johns Hopkins University held the first Organoid Intelligence Workshop to form an OI Community and to lay out the groundwork for the establishment of OI as a new scientific discipline. The potential of OI to revolutionize computing, neurological research, and drug development was discussed, along with a vision and roadmap for its development over the coming decade.
Collapse
Affiliation(s)
- Itzy E. Morales Pantoja
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Lena Smirnova
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alysson R. Muotri
- Department of Pediatrics and Cellular and Molecular Medicine, School of Medicine, University of California, San Diego, San Diego, CA, United States
- Center for Academic Research and Training in Anthropogeny (CARTA), Archealization Center (ArchC), Kavli Institute for Brain and Mind, University of California, San Diego, San Diego, CA, United States
| | - Karl J. Wahlin
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, United States
| | - Jeffrey Kahn
- Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD, United States
| | - J. Lomax Boyd
- Berman Institute of Bioethics, Johns Hopkins University, Baltimore, MD, United States
| | - David H. Gracias
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Chemistry, Johns Hopkins University, Baltimore, MD, United States
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, United States
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD, United States
- Center for Microphysiological Systems (MPS), Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Timothy D. Harris
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Tzahi Cohen-Karni
- Departments of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Brian S. Caffo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Alexander S. Szalay
- Department of Computer Science, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Physics and Astronomy, Krieger School of Arts and Sciences, Johns Hopkins University, Baltimore, MD, United States
- Mark Foundation Center for Advanced Genomics and Imaging, Johns Hopkins University, Baltimore, MD, United States
| | - Fang Han
- Department of Statistics and Economics, University of Washington, Seattle, WA, United States
| | - Donald J. Zack
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ralph Etienne-Cummings
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Akwasi Akwaboah
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - July Carolina Romero
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Dowlette-Mary Alam El Din
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Jesse D. Plotkin
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Barton L. Paulhamus
- Department of Research and Exploratory Development, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States
| | - Erik C. Johnson
- Department of Research and Exploratory Development, Johns Hopkins University Applied Physics Laboratory, Laurel, MD, United States
| | - Frederic Gilbert
- Philosophy Program, School of Humanities, University of Tasmania, Hobart, TAS, Australia
| | | | | | - Jens C. Schwamborn
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Eric J. Hill
- School of Biosciences, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Paul Roach
- Department of Chemistry, School of Science, Loughborough University, Loughborough, Leicestershire, United Kingdom
| | - Daniel Tornero
- Department of Biomedical Sciences, Institute of Neuroscience, University of Barcelona, Barcelona, Spain
- Clinic Hospital August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Caroline Krall
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, United States
| | - Rheinallt Parri
- Aston Pharmacy School, College of Health and Life Sciences, Aston University, Birmingham, United Kingdom
| | - Fenna Sillé
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Andre Levchenko
- Department of Biomedical Engineering, Yale Systems Biology Institute, Yale University, New Haven, CT, United States
| | - Rabih E. Jabbour
- Department of Bioscience and Biotechnology, University of Maryland Global Campus, Rockville, MD, United States
| | | | - Cynthia A. Berlinicke
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Qi Huang
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Alexandra Maertens
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Kathrin Herrmann
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Katya Tsaioun
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Raha Dastgheyb
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Christa Whelan Habela
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Joshua T. Vogelstein
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Thomas Hartung
- Center for Alternatives to Animal Testing (CAAT), Department of Environmental Health and Engineering, Bloomberg School of Public Health and Whiting School of Engineering, Johns Hopkins University, Baltimore, MD, United States
- Center for Alternatives to Animal Testing (CAAT)-Europe, University of Konstanz, Konstanz, Germany
| |
Collapse
|
12
|
Agarwal D, Kuhns R, Dimitriou CN, Barlow E, Wahlin KJ, Enke RA. Bulk RNA sequencing analysis of developing human induced pluripotent cell-derived retinal organoids. Sci Data 2022; 9:759. [PMID: 36494376 PMCID: PMC9734101 DOI: 10.1038/s41597-022-01853-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Retinogenesis involves the transformation of the anterior developing brain into organized retinal lamellae coordinated by intricate gene signalling networks. This complex process has been investigated in several model organisms such as birds, fish, mammals and amphibians, yet many facets of retinal development are different in humans and remain unexplored. In this regard, human pluripotent stem cell (hPSC)-derived 3D retinal organoids and Next Generation Sequencing (NGS) have emerged as key technologies that have facilitated the discovery of previously unknown details about cell fate specification and gene regulation in the retina. Here we utilized hPSCs integrated with fluorescent reporter genes (SIX6-p2A-eGFP/CRX-p2A-h2b-mRuby3) to generate retinal organoids and carry out bulk RNA sequencing of samples encompassing the majority of retinogenesis (D0-D280). This data set will serve as a valuable reference for the vision research community to characterize differentially expressed genes in the developing human eye.
Collapse
Affiliation(s)
- Devansh Agarwal
- Viterbi Family Department of Ophthalmology at the Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Rian Kuhns
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA
| | | | - Emmalyn Barlow
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA
| | - Karl J Wahlin
- Viterbi Family Department of Ophthalmology at the Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Ray A Enke
- Department of Biology, James Madison University, Harrisonburg, VA, 22807, USA.
- The Center for Genome & Metagenome Studies, James Madison University Harrisonburg, Harrisonburg, VA, 22807, USA.
| |
Collapse
|
13
|
Onyak JR, Vergara MN, Renna JM. Retinal organoid light responsivity: current status and future opportunities. Transl Res 2022; 250:98-111. [PMID: 35690342 DOI: 10.1016/j.trsl.2022.06.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
The ability to generate human retinas in vitro from pluripotent stem cells opened unprecedented opportunities for basic science and for the development of therapeutic approaches for retinal degenerative diseases. Retinal organoid models not only mimic the histoarchitecture and cellular composition of the native retina, but they can achieve a remarkable level of maturation that allows them to respond to light stimulation. However, studies evaluating the nature, magnitude, and properties of light-evoked responsivity from each cell type, in each retinal organoid layer, have been sparse. In this review we discuss the current understanding of retinal organoid function, the technologies used for functional assessment in human retinal organoids, and the challenges and opportunities that lie ahead.
Collapse
Affiliation(s)
| | - M Natalia Vergara
- CellSight Ocular Stem Cell and Regeneration Program, Sue Anschutz-Rodgers Eye Center, University of Colorado School of Medicine, Aurora, Colorado.
| | - Jordan M Renna
- Department of Biology, The University of Akron, Akron, Ohio.
| |
Collapse
|
14
|
Landowski M, Bowes Rickman C. Targeting Lipid Metabolism for the Treatment of Age-Related Macular Degeneration: Insights from Preclinical Mouse Models. J Ocul Pharmacol Ther 2021; 38:3-32. [PMID: 34788573 PMCID: PMC8817708 DOI: 10.1089/jop.2021.0067] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related macular degeneration (AMD) is a major leading cause of irreversible visual impairment in the world with limited therapeutic interventions. Histological, biochemical, genetic, and epidemiological studies strongly implicate dysregulated lipid metabolism in the retinal pigmented epithelium (RPE) in AMD pathobiology. However, effective therapies targeting lipid metabolism still need to be identified and developed for this blinding disease. To test lipid metabolism-targeting therapies, preclinical AMD mouse models are needed to establish therapeutic efficacy and the role of lipid metabolism in the development of AMD-like pathology. In this review, we provide a comprehensive overview of current AMD mouse models available to researchers that could be used to provide preclinical evidence supporting therapies targeting lipid metabolism for AMD. Based on previous studies of AMD mouse models, we discuss strategies to modulate lipid metabolism as well as examples of studies evaluating lipid-targeting therapeutics to restore lipid processing in the RPE. The use of AMD mouse models may lead to worthy lipid-targeting candidate therapies for clinical trials to prevent the blindness caused by AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, Wisconsin, USA.,McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA.,Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Catherine Bowes Rickman
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|