1
|
Tao B, Li X, Hao M, Tian T, Li Y, Li X, Yang C, Li Q, Feng Q, Zhou H, Zhao Y, Wang D, Liu W. Organoid-Guided Precision Medicine: From Bench to Bedside. MedComm (Beijing) 2025; 6:e70195. [PMID: 40321594 PMCID: PMC12046123 DOI: 10.1002/mco2.70195] [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: 07/31/2024] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
Organoid technology, as an emerging field within biotechnology, has demonstrated transformative potential in advancing precision medicine. This review systematically outlines the translational trajectory of organoids from bench to bedside, emphasizing their construction methodologies, key regulatory factors, and multifaceted applications in personalized healthcare. By recapitulating physiological architectures and disease phenotypes through three-dimensional culture systems, organoids leverage natural and synthetic scaffolds, stem cell sources, and spatiotemporal cytokine regulation to model tissue-specific microenvironments. Diverse organoid types-including skin, intestinal, lung, and tumor organoids-have facilitated breakthroughs in modeling tissue development, drug efficacy and toxicity screening, disease pathogenesis studies, and patient-tailored diagnostics. For instance, patient-derived tumor organoids preserve tumor heterogeneity and genomic profiles, serving as predictive platforms for individualized chemotherapy responses. In precision medicine, organoid-guided multiomics analyses identify actionable biomarkers and resistance mechanisms, while clustered regularly interspaced short palindromic repeats-based functional screens optimize therapeutic targeting. Despite preclinical successes, challenges persist in standardization, vascularization, and ethical considerations. Future integration of artificial intelligence, microfluidics, and spatial transcriptomics will enhance organoid scalability, reproducibility, and clinical relevance. By bridging molecular insights with patient-specific therapies, organoids are poised to revolutionize precision medicine, offering dynamic platforms for drug development, regenerative strategies, and individualized treatment paradigms.
Collapse
Affiliation(s)
- Boqiang Tao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiaolan Li
- Laboratory of Allergy and Precision MedicineChengdu Institute of Respiratory Healththe Third People's Hospital of ChengduAffiliated Hospital of Southwest Jiaotong UniversityChengduChina
| | - Ming Hao
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Tian Tian
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yuyang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Xiang Li
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| | - Chun Yang
- College of Basic MedicineBeihua UniversityJilinChina
| | - Qirong Li
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Qiang Feng
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Hengzong Zhou
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Yicheng Zhao
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
| | - Dongxu Wang
- Laboratory Animal CenterCollege of Animal ScienceJilin UniversityChangchunChina
- Zhichuang Gene Editing Animal Model Research CenterWenzhou Institute of TechnologyWenzhouChina
| | - Weiwei Liu
- Department of Oral and Maxillofacial SurgeryHospital of StomatologyJilin UniversityChangchunChina
| |
Collapse
|
2
|
Fang C, He D, Qian Y, Shen X. BMP4-GPX4 can improve the ferroptosis phenotype of retinal ganglion cells and enhance their differentiation ability after retinal stem cell transplantation in glaucoma with high intraocular pressure. Hum Mol Genet 2025; 34:673-683. [PMID: 39877959 DOI: 10.1093/hmg/ddaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/07/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Activation of bone morphogenetic protein (BMP) 4 signaling promotes the survival of retinal ganglion cell (RGC) after acute injury. In this study, we investigated the role of the BMP4 signaling pathway in regulating the degeneration of retinal ganglion cells (RGCs) in a mouse glaucoma model and its potential application in retinal stem cell. Our results demonstrate that BMP4-GPX4 not only reduces oxidative stress and iron accumulation but also promotes neuroprotective factors that support the survival of transplanted RSCs into the host retina. These findings suggest a novel therapeutic approach for glaucoma involving the modulation of the BMP4-GPX4 pathway to protect RGCs and improve visual function through enhanced RSC differentiation.
Collapse
Affiliation(s)
- Chuankai Fang
- Ophthalmology Department, Tongxiang First People's hospital, No. 1918 Jiaochang East Road, Tongxiang, Zhejiang 314500, China
| | - Di He
- Otorhinolaryngology, Tongxiang First People's Hospital, No. 1918 Jiaochang East Road, Tongxiang, Zhejiang 314500, China
| | - Yafen Qian
- Department of Anesthesiology, Tongxiang First People's Hospital, No. 1918 Jiaochang East Road, Tongxiang, Zhejiang 314500, China
| | - Xiaomei Shen
- Ophthalmology Department, Tongxiang First People's hospital, No. 1918 Jiaochang East Road, Tongxiang, Zhejiang 314500, China
| |
Collapse
|
3
|
Ravlo E, Ianevski A, Schjølberg JO, Solvang V, Dumaru R, Lysvand H, Hankinson J, Vähä-Koskela M, Vainionpää S, Varhe A, Seppänen H, Smura T, Wang X, Wang Y, Li P, Pan Q, Dahl-Jorgensen K, Krogvold L, Kamyshnyi O, Nilsen HJS, Haugen IJ, Høyer E, Afset JE, Oksenych V, Galabov AS, Stoyanova A, Lam V, van Loon B, Lulla V, Bjørås M, Kainov DE. Synergistic combination of orally available safe-in-man pleconaril, AG7404, and mindeudesivir inhibits enterovirus infections in human cell and organoid cultures. Cell Mol Life Sci 2025; 82:57. [PMID: 39843710 PMCID: PMC11754576 DOI: 10.1007/s00018-025-05581-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/18/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025]
Abstract
Enteroviruses can infect various human organs, causing diseases such as meningitis, the common cold, hand-foot-and-mouth disease, myocarditis, pancreatitis, hepatitis, poliomyelitis, sepsis, and type 1 diabetes. Currently, there are no approved treatments for enterovirus infections. In this study, we identified a synergistic combination of orally available, safe-in-man pleconaril, AG7404, and mindeudesivir, that at non-toxic concentrations effectively inhibited enterovirus replication in human cell and organoid cultures. Importantly, the cocktail did not alter glucose and insulin levels in the culture medium of pancreatic β-cells and preserved the contraction rhythm of infected heart organoids. These findings highlight a promising drug cocktail for further preclinical studies and clinical trials targeting a broad range of enterovirus-mediated diseases.
Collapse
Affiliation(s)
- Erlend Ravlo
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | - Jørn-Ove Schjølberg
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
- Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo, 0372, Norway
| | - Vanessa Solvang
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | - Rabina Dumaru
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | - Hilde Lysvand
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | | | - Markus Vähä-Koskela
- Institute for Molecular Medicine FIMM, Helsinki Institute for Life Science, University of Helsinki, Helsinki, 00014, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, 00014, Finland
| | - Sanna Vainionpää
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, 00014, Finland
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Anni Varhe
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, 00014, Finland
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, 00014, Finland
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Teemu Smura
- Department of Virology, University of Helsinki, Helsinki, 00014, Finland
- Clinical Microbiology, Helsinki University Hospital, HUS Diagnostic Center, University of Helsinki, Helsinki, 00029, Finland
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, 3015, Netherlands
| | - Yining Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, 3015, Netherlands
| | - Pengfei Li
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, 3015, Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, 3015, Netherlands
| | | | - Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, 0424, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, 46001, Ukraine
| | | | - Inger Johanne Haugen
- Department of Medical Microbiology, Clinic for Laboratory Medicine, St. Olavs Hospital, Trondheim, 7028, Norway
| | - Erling Høyer
- Department of Medical Microbiology, Clinic for Laboratory Medicine, St. Olavs Hospital, Trondheim, 7028, Norway
| | - Jan Egil Afset
- Department of Medical Microbiology, Clinic for Laboratory Medicine, St. Olavs Hospital, Trondheim, 7028, Norway
| | - Valentyn Oksenych
- Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
| | - Angel S Galabov
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | - Adelina Stoyanova
- The Stephan Angeloff Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | | | - Barbara van Loon
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway
| | - Valeria Lulla
- Department of Pathology, University of Cambridge, Cambridge, CB21QP, UK
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway.
- Department of Microbiology, Oslo University Hospital and University of Oslo, Oslo, 0372, Norway.
- Centre for Embryology and Healthy Development (CRESCO), University of Oslo, Oslo, 0373, Norway.
| | - Denis E Kainov
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, 7028, Norway.
- Institute for Molecular Medicine FIMM, Helsinki Institute for Life Science, University of Helsinki, Helsinki, 00014, Finland.
| |
Collapse
|
4
|
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
|
5
|
Yan S, He Y, Zhu Y, Ye W, Chen Y, Zhu C, Zhan F, Ma Z. Human patient derived organoids: an emerging precision medicine model for gastrointestinal cancer research. Front Cell Dev Biol 2024; 12:1384450. [PMID: 38638528 PMCID: PMC11024315 DOI: 10.3389/fcell.2024.1384450] [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: 02/09/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Gastrointestinal cancers account for approximately one-third of the total global cancer incidence and mortality with a poor prognosis. It is one of the leading causes of cancer-related deaths worldwide. Most of these diseases lack effective treatment, occurring as a result of inappropriate models to develop safe and potent therapies. As a novel preclinical model, tumor patient-derived organoids (PDOs), can be established from patients' tumor tissue and cultured in the laboratory in 3D architectures. This 3D model can not only highly simulate and preserve key biological characteristics of the source tumor tissue in vitro but also reproduce the in vivo tumor microenvironment through co-culture. Our review provided an overview of the different in vitro models in current tumor research, the derivation of cells in PDO models, and the application of PDO model technology in gastrointestinal cancers, particularly the applications in combination with CRISPR/Cas9 gene editing technology, tumor microenvironment simulation, drug screening, drug development, and personalized medicine. It also elucidates the ethical status quo of organoid research and the current challenges encountered in clinical research, and offers a forward-looking assessment of the potential paths for clinical organoid research advancement.
Collapse
Affiliation(s)
- Sicheng Yan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxuan He
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuehong Zhu
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wangfang Ye
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Chen
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Cong Zhu
- Department of Colorectal Surgery, Huzhou Central Hospital, The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Fuyuan Zhan
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhihong Ma
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, The Affiliated Central Hospital of Huzhou University, Huzhou, China
- School of Basic Medicine College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
6
|
Starheim EJ, Ravlo E, Schjølberg JO, Solvang V, Wang W, Scrimgeour NR, Manaf A, Erlandsen SE, Aas PA, Hagen L, de Sousa MML, Bjørås M. NAxtra magnetic nanoparticles for low-cost, efficient isolation of mammalian DNA and RNA. Sci Rep 2023; 13:20836. [PMID: 38012172 PMCID: PMC10682382 DOI: 10.1038/s41598-023-46868-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023] Open
Abstract
A cost-effective, viral nucleic acid (NA) isolation kit based on NAxtra magnetic nanoparticles was developed at the Norwegian University of Science and Technology in response to the shortage of commercial kits for isolation of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA during the coronavirus disease 2019 (COVID-19) pandemic. This method showed comparable sensitivity to available kits at significantly reduced cost, making its application for other biological sources an intriguing prospect. Thus, based on this low-cost nucleic acid extraction technology, we developed a simple, low- and high-throughput, efficient method for isolation of high-integrity total NA, DNA and RNA from mammalian cell lines (monolayer) and organoids (3D-cultures). The extracted NA are compatible with downstream applications including (RT-)qPCR and next-generation sequencing. When automated, NA isolation can be performed in 14 min for up to 96 samples, yielding similar quantities to available kits.
Collapse
Affiliation(s)
- Eirin Johannessen Starheim
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Erlend Ravlo
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Jørn-Ove Schjølberg
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Vanessa Solvang
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Wei Wang
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Nathan Robert Scrimgeour
- Department of Circulation and Medical Imaging (ISB), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Adeel Manaf
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0372, Oslo, Norway
| | | | - Per Arne Aas
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
| | - Lars Hagen
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
- Proteomics and Modomics Experimental Core Facility (PROMEC) at NTNU, 7491, Trondheim, Norway
| | - Mirta Mittelstedt Leal de Sousa
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway
- Centre for Embryology and Healthy Development, University of Oslo, 0373, Oslo, Norway
| | - Magnar Bjørås
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology (NTNU), 7491, Trondheim, Norway.
- Department of Microbiology, Oslo University Hospital and University of Oslo, 0372, Oslo, Norway.
- Centre for Embryology and Healthy Development, University of Oslo, 0373, Oslo, Norway.
| |
Collapse
|
7
|
Riege D, Herschel S, Fenkl T, Schade D. Small-Molecule Probes as Pharmacological Tools for the Bone Morphogenetic Protein Signaling Pathway. ACS Pharmacol Transl Sci 2023; 6:1574-1599. [PMID: 37974621 PMCID: PMC10644459 DOI: 10.1021/acsptsci.3c00170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/21/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023]
Abstract
The bone morphogenetic protein (BMP) pathway is highly conserved and plays central roles in health and disease. The quality and quantity of its signaling outputs are regulated at multiple levels, offering pharmacological options for targeted modulation. Both target-centric and phenotypic drug discovery (PDD) approaches were applied to identify small-molecule BMP inhibitors and stimulators. In this Review, we accumulated and systematically classified the different reported chemotypes based on their targets as well as modes-of-action, and herein we illustrate the discovery history of selected candidates. A comprehensive summary of available biochemical, cellular, and in vivo activities is provided for the most relevant BMP modulators, along with recommendations on their preferred use as chemical probes to study BMP-related (patho)physiological processes. There are a number of high-quality probes used as BMP inhibitors that potently and selectively interrogate the kinase activities of distinct type I (16 chemotypes available) and type II receptors (3 chemotypes available). In contrast, only a few high-quality BMP stimulator modalities have been introduced to the field due to a lack of profound target knowledge. FK506-derived macrolides such as calcineurin-sparing FKBP12 inhibitors currently represent the best-characterized chemical tools for direct activation of BMP-SMAD signaling at the receptor level. However, several PDD campaigns succeeded in expanding the druggable space of BMP stimulators. Albeit the majority of them do not entirely fulfill the strict chemical probe criteria, many chemotypes exhibit unique and unrecognized mechanisms as pathway potentiators or synergizers, serving as valuable pharmacological tools for BMP perturbation.
Collapse
Affiliation(s)
- Daniel Riege
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Sven Herschel
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Teresa Fenkl
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
| | - Dennis Schade
- Department
of Pharmaceutical & Medicinal Chemistry, Christian-Albrechts-University of Kiel, Gutenbergstrasse 76, 24118 Kiel, Germany
- Partner
Site Kiel, DZHK, German Center for Cardiovascular
Research, 24105 Kiel, Germany
| |
Collapse
|
8
|
Carido M, Völkner M, Steinheuer LM, Wagner F, Kurth T, Dumler N, Ulusoy S, Wieneke S, Norniella AV, Golfieri C, Khattak S, Schönfelder B, Scamozzi M, Zoschke K, Canzler S, Hackermüller J, Ader M, Karl MO. Reliability of human retina organoid generation from hiPSC-derived neuroepithelial cysts. Front Cell Neurosci 2023; 17:1166641. [PMID: 37868194 PMCID: PMC10587494 DOI: 10.3389/fncel.2023.1166641] [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: 02/15/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
The possible applications for human retinal organoids (HROs) derived from human induced pluripotent stem cells (hiPSC) rely on the robustness and transferability of the methodology for their generation. Standardized strategies and parameters to effectively assess, compare, and optimize organoid protocols are starting to be established, but are not yet complete. To advance this, we explored the efficiency and reliability of a differentiation method, called CYST protocol, that facilitates retina generation by forming neuroepithelial cysts from hiPSC clusters. Here, we tested seven different hiPSC lines which reproducibly generated HROs. Histological and ultrastructural analyses indicate that HRO differentiation and maturation are regulated. The different hiPSC lines appeared to be a larger source of variance than experimental rounds. Although previous reports have shown that HROs in several other protocols contain a rather low number of cones, HROs from the CYST protocol are consistently richer in cones and with a comparable ratio of cones, rods, and Müller glia. To provide further insight into HRO cell composition, we studied single cell RNA sequencing data and applied CaSTLe, a transfer learning approach. Additionally, we devised a potential strategy to systematically evaluate different organoid protocols side-by-side through parallel differentiation from the same hiPSC batches: In an explorative study, the CYST protocol was compared to a conceptually different protocol based on the formation of cell aggregates from single hiPSCs. Comparing four hiPSC lines showed that both protocols reproduced key characteristics of retinal epithelial structure and cell composition, but the CYST protocol provided a higher HRO yield. So far, our data suggest that CYST-derived HROs remained stable up to at least day 200, while single hiPSC-derived HROs showed spontaneous pathologic changes by day 200. Overall, our data provide insights into the efficiency, reproducibility, and stability of the CYST protocol for generating HROs, which will be useful for further optimizing organoid systems, as well as for basic and translational research applications.
Collapse
Affiliation(s)
- Madalena Carido
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Manuela Völkner
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Lisa Maria Steinheuer
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Department of Computer Science, Leipzig University, Leipzig, Germany
| | - Felix Wagner
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Thomas Kurth
- Center for Molecular and Cellular Bioengineering (CMCB), Technology Platform, Core Facility Electron Microscopy and Histology, TU Dresden, Dresden, Germany
| | - Natalie Dumler
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Selen Ulusoy
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Stephanie Wieneke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | | | - Cristina Golfieri
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Shahryar Khattak
- Center for Molecular and Cellular Bioengineering (CMCB), Stem Cell Engineering Facility, TU Dresden, Dresden, Germany
| | - Bruno Schönfelder
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Maria Scamozzi
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Katja Zoschke
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| | - Sebastian Canzler
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
| | - Jörg Hackermüller
- Department Computational Biology, Helmholtz Centre for Environmental Research-UFZ, Leipzig, Germany
- Department of Computer Science, Leipzig University, Leipzig, Germany
| | - Marius Ader
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
| | - Mike O Karl
- Center for Regenerative Therapies Dresden (CRTD), TU Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Dresden, Dresden, Germany
| |
Collapse
|
9
|
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
|
10
|
Beaver D, Limnios IJ. A treatment within sight: challenges in the development of stem cell-derived photoreceptor therapies for retinal degenerative diseases. FRONTIERS IN TRANSPLANTATION 2023; 2:1130086. [PMID: 38993872 PMCID: PMC11235385 DOI: 10.3389/frtra.2023.1130086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 09/07/2023] [Indexed: 07/13/2024]
Abstract
Stem cell therapies can potentially treat various retinal degenerative diseases, including age-related macular degeneration (AMD) and inherited retinal diseases like retinitis pigmentosa. For these diseases, transplanted cells may include stem cell-derived retinal pigmented epithelial (RPE) cells, photoreceptors, or a combination of both. Although stem cell-derived RPE cells have progressed to human clinical trials, therapies using photoreceptors and other retinal cell types are lagging. In this review, we discuss the potential use of human pluripotent stem cell (hPSC)-derived photoreceptors for the treatment of retinal degeneration and highlight the progress and challenges for their efficient production and clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Davinia Beaver
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| | - Ioannis Jason Limnios
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| |
Collapse
|