1
|
Inui J, Ueyama-Toba Y, Imamura C, Nagai W, Asano R, Mizuguchi H. Two-dimensionally cultured functional hepatocytes generated from human induced pluripotent stem cell-derived hepatic organoids for pharmaceutical research. Biomaterials 2025; 318:123148. [PMID: 39904185 DOI: 10.1016/j.biomaterials.2025.123148] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Human induced pluripotent stem (iPS) cell-derived hepatocyte-like cells (HLCs) are expected to replace primary human hepatocytes (PHHs) as a new stable source of hepatocytes for pharmaceutical research. However, HLCs have lower hepatic functions than PHHs, require a long time for differentiation and cannot be prepared in large quantities because they do not proliferate after their terminal differentiation. To overcome these problems, we here established hepatic organoids (iHOs) from HLCs. We then showed that the iHOs could proliferate approximately 105-fold by more than 3 passages and expressed most hepatic genes more highly than HLCs. In addition, to enable their widespread use for in vitro drug discovery research, we developed a two-dimensional culture protocol for iHOs. Two-dimensionally cultured iHOs (iHO-Heps) expressed most of the major hepatocyte marker genes at much higher levels than HLCs, iHOs, and even PHHs. The iHO-Heps exhibited glycogen storage capacity, the capacity to uptake and release indocyanine green (ICG), albumin and urea secretion, and the capacity for bile canaliculi formation. Importantly, the iHO-Heps had the activity of major drug-metabolizing enzymes and responded to hepatotoxic drugs, much like PHHs. Thus, iHO-Heps overcome the limitations of the current models and promise to provide robust and reproducible pharmaceutical assays.
Collapse
Affiliation(s)
- Jumpei Inui
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Yukiko Ueyama-Toba
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Chiharu Imamura
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Wakana Nagai
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Rei Asano
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, 565-0871, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
2
|
Li N, Wei R, Yuan Y, Deng M, Hu Y, Cheng CW, Yang J, Ho WI, Au KW, Tse YL, Li F, Wu X, Lau YM, Liao S, Ma S, Liu P, Ng KM, Esteban MA, Tse HF. Enhancement of hepatic differentiation from induced pluripotent stem cells by suppressing epithelial-mesenchymal transition. Hepatol Commun 2025; 9:e0702. [PMID: 40377485 PMCID: PMC12088630 DOI: 10.1097/hc9.0000000000000702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/07/2024] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cells induced hepatocytes (iHeps) are widely used in modeling human liver diseases and as a potential cell source for replacement therapy. However, most iHeps are relatively immature and challenging to maintain for long-term in vitro culture. METHODS We optimized the differentiation protocol by addition of a combination of small molecules to inhibit epithelial-mesenchymal transition (EMT) in iHeps (iHeps EMTi), and further characterized their function both in vitro and in vivo analyses. RESULTS Inhibition of EMT extended the in vitro culture period of iHeps EMTi from day 24 to day 60. In vitro analysis revealed that, compared to control, iHeps EMTi exhibited significantly higher expression levels of hepatic functional markers and enhanced hepatocyte functions, including lipid accumulation, glycogen storage, albumin secretion, and urea acid metabolism. Moreover, the molecular profiles of iHeps EMTi are closer to those of primary human hepatocytes. In addition, the in vivo engraftment efficiency of iHeps EMTi in the chimeric mice model was also improved as compared to iHeps alone. CONCLUSIONS We established a robust protocol to generate human iHeps with improved function and capable of long-term in vitro culturing via the suppression of EMT. Moreover, those iHeps with EMT suppression have improved engraftment in human chimeric mice.
Collapse
Affiliation(s)
- Na Li
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Rui Wei
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yangyang Yuan
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Mingdan Deng
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yang Hu
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Chi-Wa Cheng
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Jiayin Yang
- Cell Inspire Therapeutics Co., Ltd and Cell Inspire Biotechnology Co., Ltd, Shenzhen, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wai-In Ho
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Ka-Wing Au
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Yiu-Lam Tse
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Fei Li
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Xinyi Wu
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Yee-Man Lau
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
| | - Songyan Liao
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
| | - Stephanie Ma
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Pentao Liu
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kwong-Man Ng
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
| | - Miguel A. Esteban
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
- 3DC STAR, Spatiotemporal Campus at BGI Shenzhen, Shenzhen, China
| | - Hung-Fat Tse
- Department of Medicine, The Cardiology Division, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Hong Kong-Guangdong Stem Cell and Regenerative Medicine Research Centre, The University of Hong Kong and Guangzhou Institutes of Biomedicine and Health, Hong Kong SAR, China
- Cardiac and Vascular Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Centre for Stem Cell Translational Biology, The University of Hong Kong, Hong Kong SAR, China
- Laboratory of Integrative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, New Territories, Hong Kong
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| |
Collapse
|
3
|
Xu ZY, Wang M, Shi JY, Liu Y, Yu C, Zhang XY, Zhang CW, He QF, Pan C, Zhou J, Xiao H, Cao HY, Ma Y. Engineering a dynamic extracellular matrix using thrombospondin-1 to propel hepatocyte organoids reprogramming and improve mouse liver regeneration post-transplantation. Mater Today Bio 2025; 32:101700. [PMID: 40225139 PMCID: PMC11986605 DOI: 10.1016/j.mtbio.2025.101700] [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: 01/14/2025] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/15/2025] Open
Abstract
Hepatocyte organoids (HOs) hold significant potential for constructing bioartificial liver construction, toxicology research, and liver failure therapies. However, challenges such as difficulties in induced pluripotent stem cells (iPSCs) harvest and differentiation, safety concerns of tumor-derived matrices, and limited primary cell regulation hinder clinical applications. In this study, we developed a non-tumor-derived decellularized extracellular matrix (dECM) system with tunable mechanical properties and viscoelasticity to enhance stem cell proliferation and organoid functionality using thrombospondin-1 (THBS1). Nanoindentation and transcriptomic analysis revealed that THBS1 mediates adaptation and remodeling between organoids and ECM proteins, exhibiting native tissue-like viscoelasticity and up-regulated reprogramming transcriptional factors KLF4 and SOX2 via the YAP/TAZ pathway. Transplanting HOs presenting reprogramming effects into a 70 % hepatectomy model demonstrated improved liver regeneration, underscoring the potential of the THBS1-based dynamic ECM system in organoids manipulation and liver regeneration.
Collapse
Affiliation(s)
- Zi-Yan Xu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Min Wang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jing-Yan Shi
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Ye Liu
- School of Medicine, Southeast University, Nanjing, China
| | - Chao Yu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-Yi Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chen-Wei Zhang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qi-Feng He
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chao Pan
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Zhou
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hua Xiao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hong-Yong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Ma
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Huang Y, Ye Q, Wang J, Zhu K, Yang H, Jiang X, Shen M. Recent progress in the identification and in vitro culture of skin organoids. Regen Ther 2025; 29:341-351. [PMID: 40242086 PMCID: PMC12000699 DOI: 10.1016/j.reth.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/13/2024] [Accepted: 01/04/2025] [Indexed: 04/18/2025] Open
Abstract
An organoid is a cell-based structure that shows organ-specific properties and shares a similar spatial organization as the corresponding organ. Organoids possess powerful capability to reproduce the key functions of the associated organ structures, and their similarity to the organs makes them physiologically relevant systems. The primary challenge associated with the development of skin organoids is the complexity of the human skin architecture, which encompasses the epidermis and the dermis as well as accessory structures, including hair follicles, sweat glands, and sebaceous glands, that perform various functions such as thermoregulation. The ultimate objectives of developing skin organoids are to regenerate the complete skin structure in vitro and reconstruct the skin in vivo. Consequently, safety, reliability, and the fidelity of the tissue interfaces are key considerations in this process. For this purpose, the present article reviews the most recent advances in this field, focusing on the cell sources, culture methods, culture conditions, and biomarkers for identifying the structure and function of skin organoids developed in vitro or in vivo. The subsequent sections summarize the recent applications of skin organoids in related disease diagnosis and treatments, and discuss the future prospects of these organoids in terms of clinical applications. This review of skin organoids can provide an important foundation for studies on human skin development, disease modeling, and reconstructive surgery, with broad utility for promising future opportunities in both biomedical research and clinical practice.
Collapse
Affiliation(s)
- Yanan Huang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Qing Ye
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | | | - Kaimin Zhu
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Haojie Yang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, China
| | - Xiaoping Jiang
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| | - Meihua Shen
- Shanghai Corps Hospital of Chinese People's Armed Police, China
| |
Collapse
|
5
|
Ansaryan S, Chiang YC, Liu YC, Tan J, Lorenzo-Martín LF, Lutolf MP, Tolstonog G, Altug H. Spatiotemporal Interrogation of Single Spheroids Using Multiplexed Nanoplasmonic-Fluorescence Imaging. SMALL METHODS 2025:e2500106. [PMID: 40434268 DOI: 10.1002/smtd.202500106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/14/2025] [Indexed: 05/29/2025]
Abstract
Advances in organoid models, as ex vivo mini-organs, and the development of screening imaging technologies have continuously driven each other forward. A complete understanding of organoids requires detailed insights into the intertwined intraorganoid and extraorganoid activities and how they change across time and space. This study introduces a multiplexed imaging platform that integrates label-free nanoplasmonic biosensing with fluorescence microscopy to offer simultaneous monitoring of dynamics occurring within and around arrays of single spheroids with spatiotemporal resolution. The label-free module employs nanoplasmonic biosensors with extraordinary optical transmission to track biomolecular secretions into the surroundings, while concurrent fluorescence imaging enables structural analysis and viability assessment. To perform multiparametric interrogation of the data from different channels over extended periods, a deep-learning-augmented image analysis is incorporated. The platform is applied to tumor spheroids to investigate vascular endothelial growth factor A secretion alongside morphometric changes and viability, showcasing its ability to capture variations in secretion and growth dynamics between untreated and drug-treated groups. This integrated approach advances comprehensive insights into organoid models and can complement existing technologies to accelerate discoveries in disease modeling and drug development.
Collapse
Affiliation(s)
- Saeid Ansaryan
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Yung-Cheng Chiang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Yen-Cheng Liu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Jiayi Tan
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | | | - Matthias P Lutolf
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, Basel, CH-4070, Switzerland
| | - Genrich Tolstonog
- Department of Otolaryngology-Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 21, Lausanne, 1011, Switzerland
- Agora Cancer Research Centre, Rue du Bugnon 25A, Lausanne, 1005, Switzerland
| | - Hatice Altug
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| |
Collapse
|
6
|
Chen L, Li W, Zai W, Zheng X, Meng X, Yao Q, Li W, Liang Y, Ye M, Zhou K, Liu M, Yang Z, Mao Z, Wei H, Yang S, Shi G, Yuan Z, Yu W. HBV sequence integrated to enhancer acting as oncogenic driver epigenetically promotes hepatocellular carcinoma development. J Exp Clin Cancer Res 2025; 44:155. [PMID: 40405227 PMCID: PMC12096768 DOI: 10.1186/s13046-025-03413-8] [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: 03/08/2025] [Accepted: 05/09/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND HBV integration is considered as the main contributor to hepatocellular carcinoma (HCC). However, whether HBV integrated sequences determine genotype pathogenicity and how to block their function during HCC progression remains unclear. METHODS An in vitro HBV-infected PHH model and liver cancer cell lines were established to confirm the pathogenic potential of HBV-SITEs. The roles of HBV-SITE-1 in HCC development were analyzed using cellular phenotypic assays and molecular biology techniques, including the combined analysis of RNA-seq and ChIP-seq. Animal models were also used to evaluate the therapeutic effect of HBV-miR-2 inhibitors. RESULTS We identified nine fragments of HBV Sequences Integrated To Enhancer, termed as "HBV-SITEs". Particularly, a single nucleotide variation (T > G) was embedded at seed sequence of HBV-miR-2 in the highest integrated HBV-SITE-1 between genotypes B and H. Unexpectedly, B-HBV-SITE-1, not H-HBV-SITE-1, could abnormally activate oncogenic genes including TERT and accelerate HCC cell proliferation and migration. Meanwhile, HBV-miR-2 was gradually increased in HBV-infected cells and patient plasma with different HCC stages. Importantly, 227 genes upregulated by HBV, were also activated by HBV-miR-2 through triggering HBV-SITE-1 enhancer. Conversely, enhancer activities were particularly decreased by HBV-miR-2 inhibitors, and further downregulated activated oncogenic genes. Finally, HCC growth was dramatically restrained and HBV-induced transcripts were systematically reduced via injection of HBV-miR-2 inhibitors in animal models. CONCLUSION HBV-SITEs were identified as novel oncogenic elements for HCC, which provides an insightful perspective for the other cancers caused by oncogenic DNA viruses. We demonstrated that the integrated HBV sequence itself acted as oncogenic enhancers and nucleotide variations of HBV genotypes account for particular pathogenic progression, supporting that the viral nucleotide sequences are vital pathogenic substances beyond viral proteins. And modulation of their enhancer activities could be clinically achievable strategy for blocking DNA viruses-related cancer progression in the future.
Collapse
Affiliation(s)
- Lu Chen
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenxuan Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Liver Cancer Institute, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangyi Zheng
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xianlong Meng
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qunyan Yao
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Liang
- Precision Pharmacy and Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Mu Ye
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kaicheng Zhou
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Mengxing Liu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhicong Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhanrui Mao
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyan Wei
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Yang
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
- Research and Development Department, Shanghai Epicurer Biotechnology Co., Ltd., Shanghai, China.
| | - Guoming Shi
- Department of Liver Surgery and Transplantation, Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wenqiang Yu
- Shanghai Public Health Clinical Center & Laboratory of RNA Epigenetics, Institutes of Biomedical Sciences & Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
7
|
Chen F, Wang Z, Yao H, Liu Q, Gan Y, Xu S, Bao H, Jin Y, Hu YP, Gao J, Song S, Wang MJ. Large-scale manufacturing of human gallbladder epithelial cell products and derived hepatocytes via a chemically defined approach. Trends Biotechnol 2025:S0167-7799(25)00136-2. [PMID: 40399214 DOI: 10.1016/j.tibtech.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Manufacturing sufficient quantities of high-quality hepatocytes holds significant promise for the treatment of liver diseases and drug screening. Here, we developed a chemically defined, animal-free method for the large-scale production of human gallbladder epithelial cells (hGBECs) under good manufacturing practice conditions, enabling their clinical application. The cell products were characterized for growth ability, phenotype, freeze-thaw viability, genetic stability, biological contamination, tumorigenicity, and acute toxicity to ensure quality control and biological safety. We also provide a protocol for generating functional hepatocytes from hGBECs. The derived hepatocytes demonstrated typical liver functions, including albumin secretion, urea production, and drug metabolism. In addition, these cells were used in drug toxicity testing. We conducted further functional experiments on Cu2+ transport and alcohol metabolism. Transplantation of these cells in vivo was able to rescue mice from liver failure. This large-scale, convenient strategy for manufacturing hGBECs serves as a biobank for clinical applications and provides a valuable model for studying liver diseases.
Collapse
Affiliation(s)
- Fei Chen
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Zijun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Hao Yao
- Department of Hematology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, China
| | - Qinggui Liu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yuwen Gan
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Shoujia Xu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Haili Bao
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiqiang Jin
- Shanghai Baixian Biotechnology Co., Ltd., Shanghai, China; Shanghai Angecon Biotechnology Co., Ltd., Shanghai, China
| | - Yi-Ping Hu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Junling Gao
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Shaohua Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Transplantation Center, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Min-Jun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
8
|
Li J, Yang Y, Yi Z, Zhu Y, Yang H, Chen B, Lobie PE, Ma S. Microdroplet-Engineered Skeletal Muscle Organoids from Primary Tissue Recapitulate Parental Physiology with High Reproducibility. RESEARCH (WASHINGTON, D.C.) 2025; 8:0699. [PMID: 40375923 PMCID: PMC12078942 DOI: 10.34133/research.0699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 05/18/2025]
Abstract
Achieving high maturity and functionality in in vitro skeletal muscle models is essential for advancing our understanding of muscle biology, disease mechanisms, and drug discovery. However, current models struggle to fully recapitulate key features such as sarcomere structure, muscle fiber composition, and contractile function while also ensuring consistency and rapid production. Adult stem cells residing in muscle tissue are known for their powerful regenerative potential, yet tissue-derived skeletal muscle organoids have not been established. In this study, we introduce droplet-engineered skeletal muscle organoids derived from primary tissue using cascade-tubing microfluidics. These droplet-engineered organoids (DEOs) exhibit high maturity, including well-developed striated sarcomeres, spontaneous and stimulated contractions, and recapitulation of parental muscle fiber types. Notably, DEOs are produced in just 8 d without the need for primary cell culture-substantially accelerating the 50- to 60-d process required by classical organoid models. Additionally, the cascade-tubing microfluidics platform enables high-throughput production of hundreds of uniform DEO replicates from a small tissue sample, providing a scalable and reproducible solution for skeletal muscle research and drug screening.
Collapse
Affiliation(s)
- Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Meatoid Biotechnology Limited, Shenzhen 518107, China
| | - Yiming Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Ziqi Yi
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Yu Zhu
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Haowei Yang
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
| | - Baiming Chen
- School of Medicine,
The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Peter E. Lobie
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS),
Tsinghua University, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education,
Tsinghua University, Beijing 100084, China
- Key Lab of Active Proteins and Peptides Green Biomanufacturing of Guangdong Higher Education Institutes,
Tsinghua Shenzhen International Graduate School, Shenzhen 518055, China
| |
Collapse
|
9
|
Shimizu T, Miyoshi M, Kakinuma S, Tsuchiya J, Yamane D, Watakabe K, Mochida T, Inada K, Yamada K, Shinozaki K, Sato A, Kaneko S, Kawai-Kitahata F, Murakawa M, Nitta S, Nakagawa M, Watanabe M, Asahina Y, Okamoto R. Bile acid-FXR signaling facilitates the long-term maintenance of hepatic characteristics in human iPSC-derived organoids. Cell Rep 2025:115675. [PMID: 40367952 DOI: 10.1016/j.celrep.2025.115675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 12/28/2024] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
Human induced pluripotent stem cells (iPSCs) can be differentiated into hepatocyte-like cells (iPS-Heps); however, maintaining the long-term proliferation and hepatic characteristics of iPS-Heps remains a challenge. In this study, we aimed to develop a human iPSC-derived hepatic organoid (iHO) culture system that effectively retains hepatic characteristics long term. Our original culture strategy, using bile acids and their receptor (farnesoid X receptor [FXR]) agonists, yielded human iHOs capable of long-term culture with a distinctive "grape-like" structure. Comprehensive analysis showed that these iHOs maintained hepatocyte-like phenotypes, even after multiple passages, whose gene expression profiles were consistent with those of fetal hepatocytes. In addition, the overexpression of small heterodimer partner (SHP), a downstream gene of FXR, in iHOs negatively regulated genes related to the intestine and cholangiocytes. Our data demonstrated that bile acid-FXR signaling promotes both the hepatic characteristics and proliferative potential of iHOs, offering promising potential for future applications in regenerative medicine and as a disease model.
Collapse
Affiliation(s)
- Taro Shimizu
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Sei Kakinuma
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan; Department of Clinical and Diagnostic Laboratory Science, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan.
| | - Jun Tsuchiya
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Daisuke Yamane
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo 1568506, Japan
| | - Keiya Watakabe
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Tomohiro Mochida
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Kento Inada
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Kaho Yamada
- Department of Clinical and Diagnostic Laboratory Science, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Kotomi Shinozaki
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo 1568506, Japan
| | - Ayako Sato
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| | - Mamoru Watanabe
- School of Medicine, Juntendo University, Tokyo 1138421, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan; Division of Hepatic Medical Science, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan.
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medical and Dental Science, Institute of Science Tokyo (Science Tokyo), Tokyo 1138519, Japan
| |
Collapse
|
10
|
Zhang J, Liang Z, Liu F, Wang Y, Huang W, Nie J. Research Progress on Hepatitis E Virus Culture. Pathogens 2025; 14:456. [PMID: 40430776 DOI: 10.3390/pathogens14050456] [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: 04/02/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatitis E virus (HEV) is a zoonotic pathogen and the main cause of acute viral hepatitis in China, resulting in a significant burden on public health. Developing a highly efficient in vitro culture system for HEV is crucial for understanding the determinants of HEV infection in humans and other animals, the pathogenic mechanisms, as well as the screening and evaluation of antiviral drugs. In this paper, the research progress on HEV in vitro culture systems is reviewed to provide a convenient reference for further research on HEV, aiding comprehensive efforts toward the widespread prevention and control of related diseases.
Collapse
Affiliation(s)
- Jie Zhang
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Ziteng Liang
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- National Institutes for Food and Drug Control, Chinese Academy of Medical Science & Peking Union Medical College, No. 9 Dongdan Santiao, Dongcheng District, Beijing 100730, China
| | - Fan Liu
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| | - Youchun Wang
- Institute of Medical Biology, Chinese Academy of Medical Science & Peking Union Medical College, Kunming 650118, China
| | - Weijin Huang
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- National Institutes for Food and Drug Control, Chinese Academy of Medical Science & Peking Union Medical College, No. 9 Dongdan Santiao, Dongcheng District, Beijing 100730, China
| | - Jianhui Nie
- Division of HIV/AIDS and Sexually-Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
- State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control (NIFDC), Beijing 102629, China
| |
Collapse
|
11
|
Ge Y, Yang S, Zhang T, Luo K, Zhang J, Wei Y, Pu Y, Chen Z, Liang G. Liver organoids uncover tire-derived 6-PPDQ-induced hepatotoxicity of: A preliminary application of environmental toxicology and safety assessment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118215. [PMID: 40249975 DOI: 10.1016/j.ecoenv.2025.118215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/23/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
The novel pollutant, N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) leaked out of the tire and has attracted extensive concerns due to its high lethal toxicity of salmon. However, the potential hepatotoxicity of 6-PPDQ exposure and its mechanisms are unknown. As a novel 3D cell culture, liver organoids (LOs) are more similar to real organ invitro in structure and function, which showed great potential for toxicity assessment. Herein, stable LOs were generated and their applicability on hepatotoxicity assessment was evaluated with four hepatotoxic compounds. The negative effect of 6-PPDQ was explored in LOs, live/dead staining visually demonstrated the damage to the liver, and the changes of ATP, LDH, ALT, and AST effectively reflected its hepatotoxicity. Meanwhile, machine learning-based quantitative assessments of LOs morphology changes provided objective data on area, circularity, and luminance changes, enabling sensitive detection of 6-PPDQ-induced hepatotoxicity. Furthermore, transcriptomic analysis revealed that the pathways related to DNA replication and repairment, cancers, and inflammation were significantly involved in the process of 6-PPDQ-induced liver injury; Disease enrichment analysis highlighted an increased risk of chronic liver diseases, and biliary atresia were validated by Cholyl-Lys-Fluorescein (CLF). Moreover, molecular docking analysis identified potential molecular targets of 6-PPDQ, including Slc6a9, Yes1, and Nos2. This study underscored the potential of LOs for toxicological studies and highlighted the toxic effects of 6-PPDQ on the liver, suggesting the need for further investigations to understand its long-term impact on human health.
Collapse
Affiliation(s)
- Yiling Ge
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China; Institute of Medical Devices (Suzhou), Southeast University, Suzhou, PR China.
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China; Institute of Medical Devices (Suzhou), Southeast University, Suzhou, PR China.
| | - Tianyi Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China; Institute of Medical Devices (Suzhou), Southeast University, Suzhou, PR China.
| | - Kai Luo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China.
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China.
| | - Yuan Wei
- Jiangsu Avatarget Biotechnology Co., Ltd. Suzhou, PR China.
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China.
| | - Zaozao Chen
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, PR China; Institute of Medical Devices (Suzhou), Southeast University, Suzhou, PR China.
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, PR China.
| |
Collapse
|
12
|
Goswami Y, Baghel A, Sharma G, Sharma PK, Biswas S, Yadav R, Garg PK, Shalimar, Tandon R. Liver Organoids From Hepatocytes of Healthy Humans and Non-alcoholic Fatty Liver Disease (NAFLD) Patients Display Multilineage Architecture and can be Used to Develop an In Vitro Model of Steatohepatitis. J Clin Exp Hepatol 2025; 15:102463. [PMID: 39872219 PMCID: PMC11761825 DOI: 10.1016/j.jceh.2024.102463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/15/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Aim Non-alcoholic fatty liver disease (NAFLD) is a global health concern with limited treatment options. The paucity of predictive i n v itro models in preclinical settings seems to be one of the limitations of identifying effective medicines. We therefore aimed to develop an i n v itro model that can display the key hallmarks of NAFLD, such as steatosis, inflammation, and fibrosis. Methods An in vitro model of steatohepatitis was developed using organoids prepared from hepatocytes of healthy individuals from a commercial source (HLOs) and the liver tissues collected from needle biopsies of NAFLD patients (HLONAFLD) using defined culture conditions. HLOs were treated with palmitic acid for 72 h to develop an i n v itro model of steatohepatitis, while HLONAFLD served as a natural model of steatohepatitis. Metformin and saroglitazar were used to validate the liver organoid model of steatohepatitis. Saroglitazar was also evaluated in the high-fat, high-fructose (HF-HF) diet-induced model of NAFLD using C57BL/6 mice to validate the findings from the i n v itro model. Results HLOs and HLONAFLD exhibited bipotent properties, showing the expression of markers of hepatocytes, ductal cells, and also stem cells. Furthermore, they demonstrated the expression of nonparenchymal cell markers such as stellate cells (CD166) and Kupffer-like cells (CD68 and EMR1). The steatohepatitis models developed using these organoids displayed markers associated with steatosis, inflammation and fibrosis, which were decreased by metformin and saroglitazar. Conclusion The in vitro models developed in our lab employing HLOs and HLONAFLD display all three key hallmarks of NAFLD: steatosis, inflammation, and fibrosis without the necessity for coculture with other nonparenchymal cells. The implementation of the HLONAFLD-based model is also expected to provide a more realistic assessment of test substances to develop therapeutics for NAFLD.
Collapse
Affiliation(s)
- Yamini Goswami
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Akash Baghel
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ghanshyam Sharma
- BRIC-Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Phulwanti K. Sharma
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sagnik Biswas
- All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- All India Institute of Medical Sciences, New Delhi, India
| | - Pramod K. Garg
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Shalimar
- All India Institute of Medical Sciences, New Delhi, India
| | - Ruchi Tandon
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
13
|
Ramandi A, Diehl AM, Sanyal AJ, de Jong YP. Experimental Models to Investigate PNPLA3 in Liver Steatosis. Liver Int 2025; 45:e70091. [PMID: 40231787 DOI: 10.1111/liv.70091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/16/2025]
Abstract
Patatin-like phospholipase domain-containing 3 (PNPLA3) was the first gene identified through genome-wide association studies to be linked to hepatic fat accumulation. A missense variant, encoding the PNPLA3-148M allele, has since been shown to increase the risk for the full spectrum of steatotic liver disease (SLD), from simple steatosis to steatohepatitis, cirrhosis, and hepatocellular carcinoma. Despite extensive validation of this association and ongoing research into its pathogenic role, the precise mechanisms by which PNPLA3-148M contributes to the progression of SLD remain poorly understood. In this review, we evaluate preclinical in vitro and in vivo models used to investigate PNPLA3 and its involvement in SLD, with particular emphasis on metabolic dysfunction-associated steatotic liver disease. We assess the strengths and limitations of these models, as well as the challenges arising from species differences in PNPLA3 expression and function between human and murine systems.
Collapse
Affiliation(s)
- Alireza Ramandi
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Anna-Mae Diehl
- Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
14
|
Ortuño-Costela MC, Pinzani M, Vallier L. Cell therapy for liver disorders: past, present and future. Nat Rev Gastroenterol Hepatol 2025; 22:329-342. [PMID: 40102584 DOI: 10.1038/s41575-025-01050-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
The liver fulfils a plethora of vital functions and, due to their importance, liver dysfunction has life-threatening consequences. Liver disorders currently account for more than two million deaths annually worldwide and can be classified broadly into three groups, considering their onset and aetiology, as acute liver diseases, inherited metabolic disorders and chronic liver diseases. In the most advanced and severe forms leading to liver failure, liver transplantation is the only treatment available, which has many associated drawbacks, including a shortage of organ donors. Cell therapy via fully mature cell transplantation is an advantageous alternative that may be able to restore a damaged organ's functionality or serve as a bridge until regeneration can occur. Pioneering work has shown that transplanting adult hepatocytes can support liver recovery. However, primary hepatocytes cannot be grown extensively in vitro as they rapidly lose their metabolic activity. Therefore, different cell sources are currently being tested as alternatives to primary cells. Human pluripotent stem cell-derived cells, chemically induced liver progenitors, or 'liver' organoids, hold great promise for developing new cell therapies for acute and chronic liver diseases. This Review focuses on the advantages and drawbacks of distinct cell sources and the relative strategies to address different therapeutic needs in distinct liver diseases.
Collapse
Affiliation(s)
- M Carmen Ortuño-Costela
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Massimo Pinzani
- University College London Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
- University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies (UPMC-ISMETT), Palermo, Italy
| | - Ludovic Vallier
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
15
|
Zhao Q, Shao M, Ma L, Zhou R. Insights into Modeling Inflammatory Bowel Disease from Stem Cell Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10887-8. [PMID: 40299197 DOI: 10.1007/s12015-025-10887-8] [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] [Accepted: 04/13/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), is a multifactorial, immune-mediated condition marked by chronic gastrointestinal inflammation. This condition significantly impairs patients' quality of life and represents a major public health challenge globally. Pathogenesis of IBD arises from complex interplay among genetic predisposition, environmental factors, immune dysregulation, and microbial dysbiosis. Although significant strides have been made in unraveling these mechanisms, existing therapeutic options remain inadequate in addressing the full spectrum of clinical needs, underscoring the urgent demand for innovative strategies. Regenerative medicine has emerged as a promising frontier, offering novel tools for therapeutic development. We briefly consolidated current knowledge on IBD pathogenesis and treatments, emphasized the pivotal potential of human intestinal organoids (including adult stem cell-derived organoids and pluripotent stem cell- derived organoids) as a robust platform for mechanistic studies and treatment exploration. Leveraging this technology, we aim to advance personalized and next-generation therapies for IBD.
Collapse
Affiliation(s)
- Qi Zhao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Miaoli Shao
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Lisha Ma
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China
| | - Renfang Zhou
- The Affiliated Wenling Hospital of Wenzhou Medical University (The First People's Hospital of Wenling), Taizhou, Zhejiang Province, China.
| |
Collapse
|
16
|
Lian J, An Y, Wei W, Lu Y, Zhang X, Sun G, Guo H, Xu L, Chen X, Hu H. Transcriptional landscape and chromatin accessibility reveal key regulators for liver regenerative initiation and organoid formation. Cell Rep 2025; 44:115633. [PMID: 40286271 DOI: 10.1016/j.celrep.2025.115633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 03/19/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Liver regeneration is a well-organized and phase-restricted process that involves chromatin remodeling and transcriptional alterations. However, the specific transcription factors (TFs) that act as key "switches" to initiate hepatocyte regeneration and organoid formation remain unclear. Comprehensive integration of RNA sequencing and ATAC sequencing reveals that ATF3 representing "Initiation_on" TF and ONECUT2 representing "Initiation_off" TF transiently modulate the occupancy of target promoters to license liver cells for regeneration. Knockdown of Atf3 or overexpression of Onecut2 not only reduces organoid formation but also delays tissue-damage repair after PHx or CCl4 treatment. Mechanistically, we demonstrate that ATF3 binds to the promoter of Slc7a5 to activate mTOR signals while the Hmgcs1 promoter loses ONECUT2 binding to facilitate regenerative initiation. The results identify the mechanism for initiating regeneration and reveal the remodeling of transcriptional landscapes and chromatin accessibility, thereby providing potential therapeutic targets for liver diseases with regenerative defects.
Collapse
Affiliation(s)
- Jiabei Lian
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yachun An
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Wenjing Wei
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Yao Lu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Xiyu Zhang
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Molecular Medicine and Genetics, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Gongping Sun
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Histology and Embryology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Haiyang Guo
- Department of Clinical Laboratory, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Longjin Xu
- Shandong Center for Disease Control and Prevention, Jinan, Shandong 250014, China
| | - Xuena Chen
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China
| | - Huili Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education, Department of Systems Biomedicine, School of Basic Medical Sciences, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
17
|
Sakaguchi K, Nakazono K, Tahara K, Hinata Y, Tobe Y, Homma J, Sekine H, Matsuura K, Iwasaki K, Tsuneda S, Shimizu T. Cardiomyocyte sheet stacking using fibrin enables high-speed construction of three-dimensional myocardial tissue and high transplantation efficiency. Biofabrication 2025; 17:035004. [PMID: 40209743 DOI: 10.1088/1758-5090/adcb6e] [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: 11/14/2024] [Accepted: 04/10/2025] [Indexed: 04/12/2025]
Abstract
Despite the development of three-dimensional (3D) tissues that promises remarkable advances in myocardial therapies and pharmaceutical research, vascularization is required for the repair of damaged hearts using cardiac tissue engineering. In this study, we developed a method for rapid generation of a 3D cardiac tissue, with extremely high engraftment efficiency, by stacking cardiomyocyte sheets using fibrin as an adhesive. Cell sheets were created by peeling off confluent cultured cells from a culture dish grafted with a polymer that induced surface hydrophilicity in response to low temperatures. The high engraftment rate was attributed to the retention of the adhesive protein. The multistacked vascularized cell sheets prepared using fibrin, when transplanted into the subcutaneous tissue and at myocardial infarction site in rats, yielded a transplanted 3D myocardial tissue. Furthermore, multilayered cardiomyocyte sheets were transplanted twice at 1 week intervals to create a 3D myocardial tissue. Our data suggest that fibrin-based rapidly layered cell sheets can advance tissue-engineered transplantation therapy and should aid the development of next-generation tissue-engineered products in the fields of regenerative medicine and drug screening.
Collapse
Affiliation(s)
- Katsuhisa Sakaguchi
- Department of Medical Engineering, Faculty of Science and Engineering, Tokyo City University, Setagaya-ku, Tokyo 158-8557, Japan
| | - Kazuki Nakazono
- Department of Integrative Bioscience and Biomedical Engineering, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Kodai Tahara
- Department of Integrative Bioscience and Biomedical Engineering, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yuto Hinata
- Department of Life Science and Medical Bioscience, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Yusuke Tobe
- Department of Modern Mechanical Engineering, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Jun Homma
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Hidekazu Sekine
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Shinjuku-ku, Tokyo 162-8666, Japan
| | - Kiyotaka Iwasaki
- Department of Integrative Bioscience and Biomedical Engineering, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
- Department of Modern Mechanical Engineering, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Satoshi Tsuneda
- Department of Life Science and Medical Bioscience, TWIns, Waseda University, Shinjuku-ku, Tokyo 162-8480, Japan
| | - Tatsuya Shimizu
- Institute of Advanced Biomedical Engineering and Science, TWIns, Tokyo Women's Medical University, Shinjuku-ku, Tokyo 162-8666, Japan
| |
Collapse
|
18
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025:10.1038/s41592-025-02647-w. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
19
|
Igarashi R, Oda M, Okada R, Yano T, Takahashi S, Pastuhov S, Matano M, Masuda N, Togasaki K, Ohta Y, Sato S, Hishiki T, Suematsu M, Itoh M, Fujii M, Sato T. Generation of human adult hepatocyte organoids with metabolic functions. Nature 2025:10.1038/s41586-025-08861-y. [PMID: 40240606 DOI: 10.1038/s41586-025-08861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Proliferating hepatocytes often undergo ductal metaplasia to balance the energy trade-off between cellular functions and replication, hindering the expansion of human adult hepatocytes with functional competency1. Here we demonstrate that the combined activation of Wnt and STAT3 signalling enables long-term self-renewal of human adult hepatocyte organoids. YAP activation facilitates hepatocyte proliferation but commits it towards the biliary duct lineage. By contrast, STAT3 activation by oncostatin M induces hepatocyte proliferation while counteracting ductal metaplasia and maintaining the hepatic identity. Xenotransplanted hepatocyte organoids repopulate the recipient mouse liver and reconstitute the metabolic zonation structure. Upon niche factor removal and hormone supplementation, hepatocyte organoids form cord-like structures with bile canalicular networks and exhibit major liver metabolic functions comparable to those of in vivo hepatocytes. Hepatocyte organoids are amenable to gene editing, prompting functional modelling of inherent metabolic liver diseases. The new culture system offers a promising avenue for developing therapeutic strategies against human liver diseases.
Collapse
Affiliation(s)
- Ryo Igarashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Okada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Tomoki Yano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Strahil Pastuhov
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Norio Masuda
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
20
|
Yang T, Zhu X, Long M, Hui L, Yuan X, Sun Z, Zhang L, Xia Q, Wan P. Phase I safety and tolerability dose escalation study of microencapsulated hepatocyte intraperitoneal transplantation therapy in adult patients with liver failure: a study protocol. BMJ Open 2025; 15:e087828. [PMID: 40233953 PMCID: PMC12004478 DOI: 10.1136/bmjopen-2024-087828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025] Open
Abstract
INTRODUCTION China has a high prevalence of liver diseases, with about 300 million patients suffering from various types of liver diseases, where the incidence of severe liver diseases is 1%-3%. More than half a million people die from end-stage liver disease every year in China. In situ liver transplantation is the most effective treatment for liver failure, but only a limited number of patients undergo liver transplantation due to the shortage of donors. Hepatocyte transplantation requires only a certain number of hepatocytes rather than the whole liver, regardless of complex issues such as in vitro reconstruction of the three-dimensional structure of the liver, blood vessels and biliary ducts, enabling it to be safer and easier to promote in clinical practice than liver transplantation. This study aims to evaluate the safety and tolerability of microencapsulated hepatocyte intraperitoneal transplantation therapy in adult patients with liver failure. METHODS AND ANALYSIS This study is a single-centre, unblinded, single-arm study comprising a dose escalation phase and a preliminary assessment of efficacy. Subjects who were diagnosed with liver failure (including chronic liver failure and acute-on-chronic liver failure), who received 3 days of regular treatment with no beneficial effect and who volunteered to participate in microencapsulated hepatocyte intraperitoneal transplantation therapy will be enrolled. To minimise the number of patients receiving an unbeneficial therapeutic dosage, the accelerated titration design and '3+3' design will be used jointly for the dosage escalation method. All patients with microencapsulated hepatocyte transplantation will be monitored on the 1st, 3rd, 7th, 14th, 28th, 60th and 90th days after the treatment for safety and primary efficacy analyses. ETHICS AND DISSEMINATION Ethical approval has been obtained from the Shanghai Jiao Tong University School of Medicine, Ren Ji Hospital Ethics Committee. Results will be disseminated through publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT05727722.
Collapse
Affiliation(s)
- Taihua Yang
- Department of liver surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Xinye Zhu
- Department of liver surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Mei Long
- Department of liver surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Lijian Hui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, Shanghai, China
| | - Xiang Yuan
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, Shanghai, China
| | - Zhen Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, Shanghai, China
| | - Ludi Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Shanghai, Shanghai, China
| | - Qiang Xia
- Department of liver surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Ping Wan
- Department of liver surgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| |
Collapse
|
21
|
Wu G, Wu D, Hu W, Lu Q, Zhou Y, Liu J, Du Q, Luo Z, Hu H, Jiang H, Hu B, Wang S. A novel microfluidic self-perfusion chip (MSPC) for pumpless 3D cell, microtissue and organoid culture. LAB ON A CHIP 2025. [PMID: 40206017 DOI: 10.1039/d5lc00030k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Microfluidic systems have revolutionized biological research by enabling precise control over cellular environments at microscale volumes. However, traditional pump-driven systems face challenges such as complexity, cost, cell-damaging shear stress, and limited portability. This study introduces a novel adjustable microfluidic self-perfusion chip (MSPC) that uses evaporation as a driving force, eliminating the need for external pumps. Our design offers improved metabolic waste management and simplified control over fluid dynamics. The chip features adjustable evaporation pore sizes, demonstrating a robust linear relationship (R2 = 0.95) between the pore size and fluid evaporation rate. This ensures consistent fluid flow and effective waste removal, shown by lower ammonia and lactate levels compared to conventional cultures. Its unidirectional flow system and integrated one-way valve maintain cell viability, even under complete evaporation conditions. This innovative platform facilitates the cultivation of complex tissue-like structures, providing a valuable tool for tissue and organ model development, as well as drug screening and toxicity testing. By addressing key limitations of traditional systems, our adjustable MSPC represents a significant advancement in microfluidic cell culture technology, offering improved accessibility and applicability in biological research.
Collapse
Affiliation(s)
- Guohua Wu
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Di Wu
- Frontiers Science Center Disease-related Molecular Network, Laboratory Pulmonary Immunology and Inflammation, Sichuan University, Chengdu, 610213, China
| | - Wenqi Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Qinrui Lu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Yusen Zhou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Jie Liu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Qijun Du
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
| | - Zhi Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Haijie Hu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongwei Jiang
- Luoyang Key Laboratory of Clinical Multiomics and Translational Medicine, Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang 471003, China.
| | - Bangchuan Hu
- Emergency and Critical Care Center, ICU, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, 310014, China.
| | - Shuqi Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China.
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Clinical Research Center for Respiratory Disease, West China Hospital, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| |
Collapse
|
22
|
Qin S, Bo X, Liu H, Zhang Z, Zhao Z, Xia Q. Cell therapies and liver organogenesis technologies: Promising strategies for end-stage liver disease. Hepatology 2025:01515467-990000000-01231. [PMID: 40178487 DOI: 10.1097/hep.0000000000001321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025]
Abstract
End-stage liver disease represents a critical hepatic condition with high mortality, for which liver transplantation remains the only effective treatment. However, the scarcity of suitable donors results in numerous patients dying while awaiting transplantation. Novel strategies, including cell therapies and technologies mimicking liver organogenesis, offer promising alternatives for treating end-stage liver disease by potentially providing new sources of liver grafts. Recently, significant progress has been made in this field, including stem cell transplantation, hepatocyte transplantation, in vitro liver tissue generation, and liver replacement technologies. Several clinical studies have demonstrated that stem cell transplantation and hepatocyte transplantation can prolong patient survival and serve as a bridge to liver transplantation. Furthermore, in vitro liver tissue generation technologies, such as liver organoids and three-dimensional bioprinting, can generate hepatic tissues with sophisticated structures and functions, making them promising transplantation materials. Notably, liver replacement technologies hold considerable potential for producing biologically functional and transplantable liver grafts. In this review, we discuss the fundamental principles and recent advancements in cell therapies and liver organogenesis technologies while also addressing the challenges and future prospects in this rapidly evolving field.
Collapse
Affiliation(s)
- Shaoyang Qin
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaochen Bo
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongyuan Liu
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhishuo Zhang
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhicong Zhao
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-German Gene and Cell Therapy Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sino-German Gene and Cell Therapy Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Engineering Research Centre of Transplantation and Immunology, Shanghai, China
- Shanghai Institute of Transplantation, Shanghai, China
| |
Collapse
|
23
|
Huang W, Jeong S, Kim W, Chen L. Biomedical applications of organoids in genetic diseases. MEDICAL REVIEW (2021) 2025; 5:152-163. [PMID: 40224362 PMCID: PMC11987506 DOI: 10.1515/mr-2024-0077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/04/2024] [Indexed: 04/15/2025]
Abstract
Organoid technology has significantly transformed biomedical research by providing exceptional prospects for modeling human tissues and disorders in a laboratory setting. It has significant potential for understanding the intricate relationship between genetic mutations, cellular phenotypes, and disease pathology, especially in the field of genetic diseases. The intersection of organoid technology and genetic research offers great promise for comprehending the pathophysiology of genetic diseases and creating innovative treatment approaches customized for specific patients. This review aimed to present a thorough analysis of the current advancements in organoid technology and its biomedical applications for genetic diseases. We examined techniques for modeling genetic disorders using organoid platforms, analyze the approaches for incorporating genetic disease organoids into clinical practice, and showcase current breakthroughs in preclinical application, individualized healthcare, and transplantation. Through the integration of knowledge from several disciplines, such as genetics, regenerative medicine, and biological engineering, our aim is to enhance our comprehension of the complex connection between genetic variations and organoid models in relation to human health and disease.
Collapse
Affiliation(s)
- Wenhua Huang
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, China
| | - Seogsong Jeong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, Korea
- Department of Biomedical Research Center, Korea University Guro Hospital, Seoul, Korea
| | - Won Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Divisions of Gastroenterology and Hepatology, Department of Internal Medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Lei Chen
- International Co-operation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital/National Center for Liver Cancer, Naval Medical University, Shanghai, China
| |
Collapse
|
24
|
Liu J, Wang Q, Le Y, Hu M, Li C, An N, Song Q, Yin W, Ma W, Pan M, Feng Y, Wang Y, Han L, Liu J. 3D-Bioprinting for Precision Microtissue Engineering: Advances, Applications, and Prospects. Adv Healthc Mater 2025; 14:e2403781. [PMID: 39648541 DOI: 10.1002/adhm.202403781] [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: 10/01/2024] [Revised: 11/18/2024] [Indexed: 12/10/2024]
Abstract
Microtissues, engineered to emulate the complexity of human organs, are revolutionizing the fields of regenerative medicine, disease modelling, and drug screening. Despite the promise of traditional microtissue engineering, it has yet to achieve the precision required to fully replicate organ-like structures. Enter 3D bioprinting, a transformative approach that offers unparalleled control over the microtissue's spatial arrangement and mechanical properties. This cutting-edge technology enables the detailed layering of bioinks, crafting microtissues with tissue-like 3D structures. It allows for the direct construction of organoids and the fine-tuning of the mechanical forces vital for tissue maturation. Moreover, 3D-printed devices provide microtissues with the necessary guidance and microenvironments, facilitating sophisticated tissue interactions. The applications of 3D-printed microtissues are expanding rapidly, with successful demonstrations of their functionality in vitro and in vivo. This technology excels at replicating the intricate processes of tissue development, offering a more ethical and controlled alternative to traditional animal models. By simulating in vivo conditions, 3D-printed microtissues are emerging as powerful tools for personalized drug screening, offering new avenues for pharmaceutical development and precision medicine.
Collapse
Affiliation(s)
- Jinrun Liu
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Qi Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yinpeng Le
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Min Hu
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Chen Li
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Ni An
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Qingru Song
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Wenzhen Yin
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Wenrui Ma
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Mingyue Pan
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yutian Feng
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yunfang Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Lu Han
- Beijing Institute of Graphic Communication, Beijing, 102600, China
| | - Juan Liu
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| |
Collapse
|
25
|
Li J, Isaakidou A, van Zanten LJ, Tas RP, Mirzaali MJ, Fratila-Apachitei LE, Zadpoor AA. Multi-scale additive manufacturing of 3D porous networks integrated with hydrogel for sustained in vitro tissue growth. Acta Biomater 2025; 196:198-212. [PMID: 40049309 DOI: 10.1016/j.actbio.2025.03.005] [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: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The development of high-fidelity three-dimensional (3D) tissue models can minimize the need for animal models in clinical medicine and drug development. However, physical limitations regarding the distances within which diffusion processes are effective impose limitations on the size of such constructs. That is because larger-size constructs experience necrosis, especially in their centers, due to the cells residing deep inside such constructs not receiving enough oxygen and nutrients. This hampers the sustained in vitro growth of the tissues which is required for achieving functional microtissues. To address this challenge, we used three types of 3D printing technologies to create perfusable networks at different length scales and integrate them into such constructs. Toward this aim, networks incorporating porous conduits with increasingly complex configurations were designed and fabricated using fused deposition modeling, stereolithography, and two-photon polymerization while optimizing the printing conditions for each of these technologies. Furthermore, following network embedding in hydrogels, contrast agent-enhanced micro-computed tomography and confocal fluorescence microscopy were employed to characterize one of the essential network functionalities, namely the diffusion function. The investigations revealed the effects of various design parameters on the diffusion behavior of the porous conduits over 24 h. We found that the number of pores exerts the most significant influence on the diffusion behavior of the contrast agent, followed by variations in the pore size and hydrogel concentration. The analytical approach and the findings of this study establish a solid base for a new technological platform to fabricate perfusable multiscale 3D porous networks with complex designs while enabling the customization of diffusion characteristics to meet specific requirements for sustained in vitro tissue growth. STATEMENT OF SIGNIFICANCE: This study addresses an essential limitation of current 3D tissue engineering, namely, sustaining tissue viability in larger constructs through optimized nutrient and oxygen delivery. By utilizing advanced 3D printing techniques this research proposes the fabrication of perfusable, multiscale and customizable networks that enhance diffusion and enable cell access to essential nutrients throughout the construct. The findings highlighted the role of network characteristics on the diffusion of a model compound within a hydrogel matrix. This work represents a promising technological platform for creating advanced in vitro 3D tissue models that can reduce the use of animal models in research involving tissue regeneration, disease models and drug development.
Collapse
Affiliation(s)
- J Li
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands.
| | - A Isaakidou
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - L J van Zanten
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - R P Tas
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - M J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - L E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands.
| | - A A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| |
Collapse
|
26
|
Alnasser SM. From gut to liver: organoids as platforms for next-generation toxicology assessment vehicles for xenobiotics. Stem Cell Res Ther 2025; 16:150. [PMID: 40140938 PMCID: PMC11948905 DOI: 10.1186/s13287-025-04264-y] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Traditional toxicological assessment relied heavily on 2D cell cultures and animal models of study, which were inadequate for the precise prediction of human response to chemicals. Researchers have now shifted focus on organoids for toxicological assessment. Organoids are 3D structures produced from stem cells that mimic the shape and functionality of human organs and have a number of advantages compared to traditional models of study. They have the capacity to replicate the intricate cellular microenvironment and in vivo interactions. They offer a physiologically pertinent platform that is useful for the researchers to monitor cellular responses in a more realistic manner and evaluate drug toxicity. Additionally, organoids can be created from cells unique to a patient, allowing for individualized toxicological research and providing understanding of the inter-individual heterogeneity in drug responses. Recent developments in the use of gut and liver organoids for assessment of the xenobiotics (environmental toxins and drugs) is reviewed in this article. Gut organoids can reveal potential damage to the digestive system and how xenobiotics affect nutrient absorption and barrier function. Liver is the primary site of detoxification and metabolism of xenobiotics, usually routed from the gut. Hence, these are linked and crucial for evaluating chemical or pollutant induced organ toxicity, forecasting their metabolism and pharmacokinetics. When incorporated into the drug development process, organoid models have the potential to improve the accuracy and efficiency of drug safety assessments, leading to safer and more effective treatments. We also discuss the limitations of using organoid-based toxicological assays, and future prospects, including the need for standardized protocols for overcoming reproducibility issues.
Collapse
Affiliation(s)
- Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, 51452, Buraydah, Qassim, Saudi Arabia.
| |
Collapse
|
27
|
Fukunaga I, Takebe T. In vitro liver models for toxicological research. Drug Metab Pharmacokinet 2025; 62:101478. [PMID: 40203632 DOI: 10.1016/j.dmpk.2025.101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Drug-induced liver injury (DILI) presents a major challenge not only in new drug development but also in post-marketing withdrawals and the safety of food, cosmetics, and chemicals. Experimental model organisms such as the rodents have been widely used for preclinical toxicological testing. However, the tension exists associated with the ethical and sustainable use of animals in part because animals do not necessarily inform the human-specific ADME (adsorption, dynamics, metabolism and elimination) profiling. To establish alternative models in humans, in vitro hepatic tissue models have been proposed, ranging from primary hepatocytes, immortal hepatocytes, to the development of new cell resources such as stem cell-derived hepatocytes. Given the evolving number of novel alternative methods, understanding possible combinations of cell sources and culture methods will be crucial to develop the context-of-use assays. This review primarily focuses on 3D liver organoid models for conducting. We will review the relevant cell sources, bioengineering methods, selection of training compounds, and biomarkers towards the rationale design of in vitro toxicology testing.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
28
|
Morrison MA, Artru F, Trovato FM, Triantafyllou E, McPhail MJ. Potential therapies for acute-on-chronic liver failure. Liver Int 2025; 45:e15545. [PMID: 36800487 PMCID: PMC11815631 DOI: 10.1111/liv.15545] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/16/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Acute-on-chronic liver failure (ACLF) is a syndrome that develops in approximately 30% of patients hospitalised with cirrhosis and is characterised by an acute decompensation of liver function associated with extra-hepatic organ failures and a high short-term mortality. At present, no specific therapies are available for ACLF, and current management is limited to treatment of the precipitating event and organ support. Given the high prevalence and high mortality of this severe liver disease, there is an urgent need for targeted treatments. There is increasing evidence of the important role played by systemic inflammation and immune dysfunction in the pathophysiology of ACLF and a better understanding of these immune processes is resulting in new therapeutic targets. The aim of this review is to present an overview of ongoing studies of potentially promising therapies and how they could be utilised in the management of ACLF.
Collapse
Affiliation(s)
- Maura A. Morrison
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Florent Artru
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Francesca M. Trovato
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| | - Evangelos Triantafyllou
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and ReproductionImperial College LondonLondonUK
| | - Mark J. McPhail
- Institute of Liver StudiesKing's College HospitalLondonUK
- Department of Inflammation Biology, School of Immunology and Microbial SciencesKing's College LondonLondonUK
| |
Collapse
|
29
|
Zhao J, Zhi Y, Ren H, Wang J, Zhao Y. Emerging biotechnologies for engineering liver organoids. Bioact Mater 2025; 45:1-18. [PMID: 39588483 PMCID: PMC11585797 DOI: 10.1016/j.bioactmat.2024.11.002] [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: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/27/2024] Open
Abstract
The engineering construction of the liver has attracted enormous attention. Organoids, as emerging miniature three-dimensional cultivation units, hold significant potential in the biomimetic simulation of liver structure and function. Despite notable successes, organoids still face limitations such as high variability and low maturity. To overcome these challenges, engineering strategies have been established to maintain organoid stability and enhance their efficacy, laying the groundwork for the development of advanced liver organoids. The present review comprehensively summarizes the construction of engineered liver organoids and their prospective applications in biomedicine. Initially, we briefly present the latest research progress on matrix materials that maintain the three-dimensional morphology of organoids. Next, we discuss the manipulative role of engineering technologies in organoid assembly. Additionally, we outline the impact of gene-level regulation on organoid growth and development. Further, we introduce the applications of liver organoids in disease modeling, drug screening and regenerative medicine. Lastly, we overview the current obstacles and forward-looking perspectives on the future of engineered liver organoids. We anticipate that ongoing innovations in engineered liver organoids will lead to significant advancements in medical applications.
Collapse
Affiliation(s)
- Junqi Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yue Zhi
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
30
|
Nagao M, Fukuda A, Kashima H, Matsuyama S, Iimori K, Nakayama S, Mizukoshi K, Kawai M, Yamakawa G, Omatsu M, Namikawa M, Masuda T, Hiramatsu Y, Muta Y, Maruno T, Nakanishi Y, Tsuruyama T, Seno H. Cholangiocyte organoids for disease, cancer, and regenerative medicine. Eur J Cell Biol 2025; 104:151472. [PMID: 39721346 DOI: 10.1016/j.ejcb.2024.151472] [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: 08/27/2024] [Revised: 11/19/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
The biliary tract is a ductal network comprising the intrahepatic (IHBDs) and extrahepatic bile duct (EHBDs). Biliary duct disorders include cholangitis, neoplasms, and injury. However, the underlying mechanisms are not fully understood. With advancements in 3D culture technology, cholangiocyte organoids (COs) derived from primary tissues or induced pluripotent stem cells (iPSCs) can accurately replicate the structural and functional properties of biliary tissues. These organoids have become powerful tools for studying the pathogenesis of biliary diseases, such as cystic fibrosis and primary sclerosing cholangitis, and for developing new therapeutic strategies for cholangiocarcinoma. Additionally, COs have the potential to repair bile duct injuries and facilitate transplantation therapies. This review also discusses the use of organoids in genetically engineered mouse models to provide mechanistic insights into tumorigenesis and cancer progression. Continued innovation and standardization of organoid technology are crucial for advancing precision medicine for biliary diseases and cancer.
Collapse
Affiliation(s)
- Munemasa Nagao
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, Osaka 589-8511, Japan
| | - Akihisa Fukuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan.
| | - Hirotaka Kashima
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Sho Matsuyama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kei Iimori
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shinnosuke Nakayama
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kenta Mizukoshi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Munenori Kawai
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Go Yamakawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mayuki Omatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mio Namikawa
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan; Department of Gastroenterology and Hepatology, The Japan Baptist Hospital, 47 Yamanomoto-cho, Kitashirakawa, Sakyo-ku, Kyoto 606-8273, Japan
| | - Tomonori Masuda
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yukiko Hiramatsu
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yu Muta
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Takahisa Maruno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tatsuaki Tsuruyama
- Department of Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, 54 Shogoin-Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| |
Collapse
|
31
|
Dwyer BJ, Tirnitz-Parker JEE. Patient-derived organoid models to decode liver pathophysiology. Trends Endocrinol Metab 2025; 36:235-248. [PMID: 39191607 DOI: 10.1016/j.tem.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Liver diseases represent a growing global health challenge, and the increasing prevalence of obesity and metabolic disorders is set to exacerbate this crisis. To meet evolving regulatory demands, patient-specific in vitro liver models are essential for understanding disease mechanisms and developing new therapeutic approaches. Organoid models, which faithfully recapitulate liver biology, can be established from both non-malignant and malignant liver tissues, offering insight into various liver conditions, from acute injuries to chronic diseases and cancer. Improved understanding of liver microenvironments, innovative biomaterials, and advanced imaging techniques now facilitate comprehensive and unbiased data analysis, paving the way for personalised medicine. In this review, we discuss state-of-the-art patient-derived liver organoid models, recent technological advancements, and strategies to enhance their clinical impact.
Collapse
Affiliation(s)
- Benjamin J Dwyer
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| | - Janina E E Tirnitz-Parker
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| |
Collapse
|
32
|
Barcena-Varela M, Monga SP, Lujambio A. Precision models in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2025; 22:191-205. [PMID: 39663463 DOI: 10.1038/s41575-024-01024-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) represents a global health challenge, and ranks among one of the most prevalent and deadliest cancers worldwide. Therapeutic advances have expanded the treatment armamentarium for patients with advanced HCC, but obstacles remain. Precision oncology, which aims to match specific therapies to patients who have tumours with particular features, holds great promise. However, its implementation has been hindered by the existence of numerous 'HCC influencers' that contribute to the high inter-patient heterogeneity. HCC influencers include tumour-related characteristics, such as genetic alterations, immune infiltration, stromal composition and aetiology, and patient-specific factors, such as sex, age, germline variants and the microbiome. This Review delves into the intricate world of HCC, describing the most innovative model systems that can be harnessed to identify precision and/or personalized therapies. We provide examples of how different models have been used to nominate candidate biomarkers, their limitations and strategies to optimize such models. We also highlight the importance of reproducing distinct HCC influencers in a flexible and modular way, with the aim of dissecting their relative contribution to therapy response. Next-generation HCC models will pave the way for faster discovery of precision therapies for patients with advanced HCC.
Collapse
Affiliation(s)
- Marina Barcena-Varela
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
33
|
Li T, Bo RQ, Yan J, Johnson NL, Liao MT, Li Y, Chen Y, Lin J, Li J, Chu FH, Ding X. Global landscape of hepatic organoid research: A bibliometric and visual study. World J Hepatol 2025; 17:95624. [PMID: 40027550 PMCID: PMC11866153 DOI: 10.4254/wjh.v17.i2.95624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/11/2024] [Accepted: 11/12/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Hepatic organoid-based modelling, through the elucidation of a range of in vivo biological processes and the recreation of the intricate liver microenvironment, is yielding groundbreaking insights into the pathophysiology and personalized medicine approaches for liver diseases. AIM This study was designed to analyse the global scientific output of hepatic organoid research and assess current achievements and future trends through bibliometric analysis. METHODS Articles were retrieved from the Web of Science Core Collection, and CiteSpace 6.3.R1 was employed to analyse the literature, including outputs, journals, and countries, among others. RESULTS Between 2010 and 2024, a total of 991 articles pertaining to hepatic organoid research were published. The journal Hepatology published the greatest number of papers, and journals with an impact factor greater than 10 constituted 60% of the top 10 journals. The United States and Utrecht University were identified as the most prolific country and institution, respectively. Clevers H emerged as the most prolific author, whereas Huch M had the highest number of cocitations, suggesting that both are ideal candidates for academic collaboration. Research on hepatic organoids has exhibited a progressive shift in focus, evolving from initial investigations into model building, differentiation research in stem cells, bile ducts, and progenitor cells, to a broader spectrum encompassing lipid metabolism, single-cell RNA sequencing, and therapeutic applications. The phrases exhibiting citation bursts from 2022 to 2024 include "drug resistance", "disease model", and "patient-derived tumor organoids". CONCLUSION Research on hepatic organoids has increased over the past decade and is expected to continue to grow. Key research areas include applications for liver diseases and drug development. Future trends likely to gain focus include patient-derived tumour organoids, disease modelling, and personalized medicine.
Collapse
Affiliation(s)
- Tao Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Rong-Qiang Bo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jun Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Nadia L Johnson
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Meng-Ting Liao
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yuan Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yan Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jie Lin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jian Li
- Department of Histology and Embryology, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Fu-Hao Chu
- Institute of Regulatory Science for Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100700, China
| |
Collapse
|
34
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [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: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
35
|
Yang J, Jiang Y, Li M, Wu K, Wei S, Zhao Y, Shen J, Du F, Chen Y, Deng S, Xiao Z, Yuan W, Wu X. Organoid, organ-on-a-chip and traditional Chinese medicine. Chin Med 2025; 20:22. [PMID: 39940016 PMCID: PMC11823035 DOI: 10.1186/s13020-025-01071-8] [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: 07/24/2024] [Accepted: 01/23/2025] [Indexed: 02/14/2025] Open
Abstract
In the past few years, the emergence of organoids and organ-on-a-chip (OOAC) technologies, which are complementary to animal models and two-dimensional cell culture methods and can better simulate the internal environment of the human body, provides a new platform for traditional Chinese medicine (TCM) studies. Organoids and OOAC techniques have been increasingly applied in the fields of drug screening, drug assessment and development, personalized therapies, and developmental biology, and there have been some application cases in the TCM studies. In this review, we summarized the current status of using organoid and OOAC technologies in TCM research and provide key insights for future study. It is believed that organoid and OOAC technologies will play more and more important roles in research and make greater contributions to the innovative development of TCM.
Collapse
Affiliation(s)
- Jiayue Yang
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yu Jiang
- Department of Gerontology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Mingxing Li
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Ke Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Shulin Wei
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Yueshui Zhao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Jing Shen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Fukuan Du
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Yu Chen
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Shuai Deng
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
| | - Zhangang Xiao
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou, 646000, China
- Gulin County Hospital of Traditional Chinese Medicine, Luzhou, 646500, China
- School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, 621000, Sichuan, China
| | - Wen Yuan
- School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Mianyang, 621000, Sichuan, China.
| | - Xu Wu
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Departments of Paediatrics & Paediatric Care, Luzhou People's Hospital, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
36
|
Zou RQ, Dai YS, Liu F, Yang SQ, Hu HJ, Li FY. Hepatobiliary organoid research: the progress and applications. Front Pharmacol 2025; 16:1473863. [PMID: 40008122 PMCID: PMC11850396 DOI: 10.3389/fphar.2025.1473863] [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: 07/31/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Organoid culture has emerged as a forefront technology in the life sciences field. As "in vitro micro-organs", organoids can faithfully recapitulate the organogenesis process, and conserve the key structure, physiological function and pathological state of the original tissue or organ. Consequently, it is widely used in basic and clinical studies, becoming important preclinical models for studying diseases and developing therapies. Here, we introduced the definition and advantages of organoids and described the development and advances in hepatobiliary organoids research. We focus on applying hepatobiliary organoids in benign and malignant diseases of the liver and biliary tract, drug research, and regenerative medicine to provide valuable reference information for the application of hepatobiliary organoids. Despite advances in research and treatment, hepatobiliary diseases including carcinoma, viral hepatitis, fatty liver and bile duct defects have still been conundrums of the hepatobiliary field. It is necessary and crucial to study disease mechanisms, establish efficient and accurate research models and find effective treatment strategies. The organoid culture technology shed new light on solving these issues. However, the technology is not yet mature, and many hurdles still exist that need to be overcome. The combination with new technologies such as CRISPR-HOT, organ-on-a-chip may inject new vitality into future development.
Collapse
Affiliation(s)
- Rui-Qi Zou
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Shi Dai
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fei Liu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Si-Qi Yang
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai-Jie Hu
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fu-Yu Li
- Division of Biliary Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Research Center for Biliary Diseases, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
37
|
Wu H, Wang J, Liu S, Wang Y, Tang X, Xie J, Wang N, Shan H, Chen S, Zhang X, Zeng W, Chen C, Fu Y, Lai L, Duan Y. Large-Scale Production of Expandable Hepatoblast Organoids and Polarised Hepatocyte Organoids From hESCs Under 3D Static and Dynamic Suspension Conditions. Cell Prolif 2025:e70001. [PMID: 39921573 DOI: 10.1111/cpr.70001] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
To date, generating viable and functional hepatocytes in large scale remains challenge. By employing 3D suspension condition with the support of low concentration Matrigel, a novel culture system was developed to generate expandable hepatoblast organoids (HB-orgs) and mature polarised hepatocyte organoids (P-hep-orgs) from human embryonic stem cells (hESCs) in both dishes and bioreactors. scRNA-seq and functional assays were used to characterise HB-orgs and P-hep-orgs. hESC-derived HB-orgs could proliferate at least for 15 passages, leading to 1012 in total cells in 4 weeks. P-hep-orgs differentiated from HB-orgs displayed characteristics of mature hepatocytes with polarisation. Moreover, single-cell RNA sequencing exhibited that over 40% of cells in P-hep-orgs were highly fidelity with human primary hepatocytes. Eventually, large-scale production of P-hep-orgs could be generated from massively expanded HB-orgs within 1 week with similar number in bioreactors, which were achieved by the enhancements in energy metabolism contribute to the expansion of HB-orgs and maturation of P-hep-orgs in bioreactors. By providing a cost-efficient and robust platform, our study represents a significant step toward manufacturing large-scale functioning hESC-derived hepatocytes for cell-based therapeutics, disease modelling, pharmacology and toxicology studies.
Collapse
Affiliation(s)
- Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yiyu Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xianglian Tang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanhuan Shan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Sen Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Weiping Zeng
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuxin Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yinjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
38
|
Zhou X, Liu WM, Sun HY, Peng Y, Huang RJ, Chen CY, Zhang HD, Zhou SA, Wu HP, Tang D, Huang WJ, Wu H, Li QG, Zhai B, Xia Q, Yu WF, Yan HX. Hepatocyte-derived liver progenitor-like cells attenuate liver cirrhosis via induction of apoptosis in hepatic stellate cells. Hepatol Commun 2025; 9:e0614. [PMID: 39878682 PMCID: PMC11781762 DOI: 10.1097/hc9.0000000000000614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/12/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Cell therapy demonstrates promising potential as a substitute therapeutic approach for liver cirrhosis. We have developed a strategy to effectively expand murine and human hepatocyte-derived liver progenitor-like cells (HepLPCs) in vitro. The primary objective of the present study was to apply HepLPCs to the treatment of liver cirrhosis and to elucidate the underlying mechanisms responsible for their therapeutic efficacy. METHODS The effects of allogeneic or xenogeneic HepLPC transplantation were investigated in rat model of liver cirrhosis. Liver tissues were collected and subjected to immunostaining to assess changes in histology. In vitro experiments used HSCs to explore the antifibrotic properties of HepLPC-secretomes and their underlying molecular mechanisms. Additionally, proteomic analysis was conducted to characterize the protein composition of HepLPC-secretomes. RESULTS Transplantation of HepLPCs resulted in decreased active fibrogenesis and net fibrosis in cirrhosis models. Apoptosis of HSCs was observed in vivo after HepLPC treatment. HepLPC-secretomes exhibited potent inhibition of TGF-β1-induced HSC activation and promoted apoptosis through signal transducer and activator of transcription (STAT)1-mediated pathways in vitro. Furthermore, synergistic effects between amphiregulin and FGF19 within HepLPC-secretomes were identified, contributing to HSC apoptosis and exerting antifibrotic effects via activation of the janus kinase-STAT1 pathway. CONCLUSIONS HepLPCs have the potential to ameliorate liver cirrhosis by inducing STAT1-dependent apoptosis in HSCs. Amphiregulin and FGF19 are key factors responsible for STAT1 activation, representing promising novel therapeutic targets for the treatment of liver cirrhosis.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Wen-Ming Liu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Han-Yong Sun
- Department of Hepatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuan Peng
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ren-Jie Huang
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
| | - Cai-Yang Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong-Dan Zhang
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
| | - Shen-Ao Zhou
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
| | - Hong-Ping Wu
- Molecular Epidemiology Laboratory, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Dan Tang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - Wei-Jian Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Han Wu
- Hubei Key Laboratory of Tumour Biological Behaviors, Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qi-Gen Li
- Department of Hepatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bo Zhai
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiang Xia
- Department of Hepatic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
| | - He-Xin Yan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Celliver Biotechnology Co. Ltd., Shanghai, China
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University), Ministry of Education, Shanghai, China
- Department of Interventional Oncology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Kim Y, Kang M, Mamo MG, Adisasmita M, Huch M, Choi D. Liver organoids: Current advances and future applications for hepatology. Clin Mol Hepatol 2025; 31:S327-S348. [PMID: 39722609 PMCID: PMC11925438 DOI: 10.3350/cmh.2024.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
The creation of self-organizing liver organoids represents a significant, although modest, step toward addressing the ongoing organ shortage crisis in allogeneic liver transplantation. However, researchers have recognized that achieving a fully functional whole liver remains a distant goal, and the original ambition of organoid-based liver generation has been temporarily put on hold. Instead, liver organoids have revolutionized the field of hepatology, extending their influence into various domains of precision and molecular medicine. These 3D cultures, capable of replicating key features of human liver function and pathology, have opened new avenues for human-relevant disease modeling, CRISPR gene editing, and high-throughput drug screening that animal models cannot accomplish. Moreover, advancements in creating more complex systems have led to the development of multicellular assembloids, dynamic organoid-on-chip systems, and 3D bioprinting technologies. These innovations enable detailed modeling of liver microenvironments and complex tissue interactions. Progress in regenerative medicine and transplantation applications continues to evolve and strives to overcome the obstacles of biocompatibility and tumorigenecity. In this review, we examine the current state of liver organoid research by offering insights into where the field currently stands, and the pivotal developments that are shaping its future.
Collapse
Affiliation(s)
- Yohan Kim
- Department of MetaBioHealth, Sungkyunkwan University, Suwon, Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Korea
| | - Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Michael Girma Mamo
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Michael Adisasmita
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
40
|
Shao W, Xu H, Zeng K, Ye M, Pei R, Wang K. Advances in liver organoids: replicating hepatic complexity for toxicity assessment and disease modeling. Stem Cell Res Ther 2025; 16:27. [PMID: 39865320 PMCID: PMC11771052 DOI: 10.1186/s13287-025-04139-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
The lack of in vivo accurate human liver models hinders the investigation of liver-related diseases, injuries, and drug-related toxicity, posing challenges for both basic research and clinical applications. Traditional cellular and animal models, while widely used, have significant limitations in replicating the liver's complex responses to various stressors. Liver organoids derived from human pluripotent stem cells, adult stem cells primary cells, or tissues can mimic diverse liver cell types, major physiological functions, and architectural features. Recent advancements in the field have shown that some liver organoids have sufficient accuracy to replicate specific aspects of the human liver's complexity. This review highlights recent progress in liver organoid research, with a particular emphasis on their potential for toxicity assessment and disease modeling. The intrinsic advantages of liver organoids include higher sensitivity and suitability for long-term studies, which enhance the predictive value in drug and nanomaterial toxicity testing. The integration of liver organoids with microfluidic devices enables the simulation of the liver microenvironment and facilitates high-throughput drug screening. The liver organoids also serve as ideal platforms for studying liver diseases such as hepatitis, liver fibrosis, viral liver diseases, and monogenic diseases. Additionally, this review discusses the advantages and limitations of liver organoids along with potential avenues for future research.
Collapse
Affiliation(s)
- Weidong Shao
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
- China Pharmaceutical University, 639 Longmian Rd, Nanjing, Jiangsu, 210009, China
| | - Hui Xu
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Kanghua Zeng
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Mingzhou Ye
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Renjun Pei
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| | - Kai Wang
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
41
|
Gong D, Mo J, Zhai M, Zhou F, Wang G, Ma S, Dai X, Deng X. Advances, challenges and future applications of liver organoids in experimental regenerative medicine. Front Med (Lausanne) 2025; 11:1521851. [PMID: 39927267 PMCID: PMC11804114 DOI: 10.3389/fmed.2024.1521851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/20/2024] [Indexed: 02/11/2025] Open
Abstract
The liver is a vital organ responsible for numerous metabolic processes in the human body, including the metabolism of drugs and nutrients. After liver damage, the organ can rapidly return to its original size if the causative factor is promptly eliminated. However, when the harmful stimulus persists, the liver's regenerative capacity becomes compromised. Substantial theoretical feasibility has been demonstrated at the levels of gene expression, molecular interactions, and intercellular dynamics, complemented by numerous successful animal studies. However, a robust model and carrier that closely resemble human physiology are still lacking for translating these theories into practice. The potential for liver regeneration has been a central focus of ongoing research. Over the past decade, the advent of organoid technology has provided improved models and materials for advancing research efforts. Liver organoid technology represents a novel in vitro culture system. After several years of refinement, human liver organoids can now accurately replicate the liver's morphological structure, nutrient and drug metabolism, gene expression, and secretory functions, providing a robust model for liver disease research. Regenerative medicine aims to replicate human organ or tissue functions to repair or replace damaged tissues, restore their structure or function, or stimulate the regeneration of tissues or organs within the body. Liver organoids possess the same structure and function as liver tissue, offering the potential to serve as a viable replacement for the liver, aligning with the goals of regenerative medicine. This review examines the role of liver organoids in regenerative medicine.
Collapse
Affiliation(s)
- Da Gong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Jiaye Mo
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
- Guangxi University of Chinese Medicine, Nanning, China
| | - Mei Zhai
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| | - Fulin Zhou
- Department of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Guocai Wang
- Department of Physiology, School of Medicine and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, Guangdong, China
| | - Xiaoyong Dai
- Department of Physiology, School of Medicine and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
- Institute of Biopharmaceutical and Health Engineering, Shenzhen Key Laboratory of Gene and Antibody Therapy, State Key Laboratory of Chemical Oncogenomics, Tsinghua University Shenzhen International Graduate School, Guangdong, China
| | - Xuesong Deng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Shenzhen University, Health Science Center, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
42
|
Shrestha S, Vanga MG, Jonnadula C, Acharya P, Lee M, Lee MY. Reproducible, Scale-Up Production of Human Liver Organoids (HLOs) on a Pillar Plate Platform via Microarray 3D Bioprinting. Methods Mol Biol 2025. [PMID: 39821806 DOI: 10.1007/7651_2024_603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Human liver organoids (HLOs) derived from pluripotent stem cells hold potential for disease modeling and high-throughput compound screening due to their architectural and functional resemblance to human liver tissues. However, reproducible, scale-up production of HLOs for high-throughput screening (HTS) presents challenges. These include the high costs of additives and growth factors required for cell differentiation, variability in organoid size and function from batch to batch, suboptimal maturity of HLOs compared to primary hepatocytes, and low assay throughput due to excessive manual processes and the absence of assay-ready plates with HLOs. To address some of these issues, here we present standard operating procedures (SOPs) for the scale-up production of HLOs using a pillar plate through microarray 3D bioprinting. This technology facilitates the rapid, uniform seeding of foregut cells onto the pillar plate, maintaining cell viability and enabling the scale-up generation of HLOs. The assay-ready pillar plate with HLOs is suitable for compound testing, as well as in situ organoid staining and analysis.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | | | - Charishma Jonnadula
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Minseong Lee
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, TX, USA.
- Bioprinting Laboratories Inc., Dallas, TX, USA.
| |
Collapse
|
43
|
Kaur I, Vasudevan A, Sanchez-Romero N, Sanyal A, Sharma A, Hemati H, Juneja P, Sharma A, Pla Palacin I, Rastogi A, Vijayaragavan P, Ghosh S, Ramakrishna S, Sarin SK, Baptista PM, Tripathi DM, Kaur S. In vivo transplantation of intrahepatic cholangiocyte organoids with decellularized liver-derived hydrogels supports hepatic cellular proliferation and differentiation in chronic liver injury. J Mater Chem B 2025; 13:918-928. [PMID: 39656267 DOI: 10.1039/d4tb01503g] [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: 01/16/2025]
Abstract
The limited replicative potential of primary hepatocytes (Hep) is a major hurdle for obtaining sufficient quantity and quality hepatocytes during cell therapy in patients with liver failure. Intrahepatic cholangiocyte organoids (ICOs) derived from intrahepatic bile ducts differentiate into both hepatocytes and cholangiocytes in vitro. Here, we studied in vivo effects of transplanting ICOs and Hep in chronic liver injury mice models. Well characterized primary mouse ICOs and Hep were mixed in decellularized liver (DCL) matrix hydrogels and injected into the subcapsular left lateral liver lobe of CCl4-induced liver injury models whereas mice given DCL alone were in the sham group. Two weeks post-transplantation, transplanted liver lobes were collected and studied by histology and RNA sequencing. Transplanted animals did not exhibit any tumors, mortality or morbidity. Mice livers transplanted with ICOs had increased cellular proliferation and vascularization as compared to Hep transplanted mice or sham. Collagen deposition in the liver was significantly reduced and serum albumin levels were significantly increased in transplanted groups compared to the sham group. Expression of genes associated with hepatocyte differentiation was highest in Hep transplanted livers among the three groups, but they were also upregulated in ICO transplanted livers compared to sham. Our study demonstrates that ICOs encapsulated in DCL hydrogels when transplanted in chronically injured mice livers engraft well and show hepatocyte differentiation and reduction of fibrosis, indicating that hydrogel transplanted cholangiocyte organoids may serve as an efficient cell source and therapy for renewal of hepatocytes, restoration of hepatocyte functions and resolution of liver injury.
Collapse
Affiliation(s)
- Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida 201301, Uttar Pradesh, India
| | - Natalia Sanchez-Romero
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Be Cytes Biotechnologies, Barcelona, Spain
- Facultad de Ciencias de la Salud, Universidad San Jorge, Campus Universitario, Autov A23 km 299, 50830, Villanueva de Gallego, Zaragoza, Spain
| | - Arka Sanyal
- Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, India
| | - Aarushi Sharma
- Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, India
| | - Hamed Hemati
- Department of Toxicology and Cancer Biology, University of Kentucky, KY, USA
| | - Pinky Juneja
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Aarti Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Iris Pla Palacin
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
| | | | - Pooja Vijayaragavan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Sector-125, Noida 201301, Uttar Pradesh, India
| | - Sourabh Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, India
| | | | - Shiv K Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Pedro M Baptista
- Instituto de Investigación Sanitária de Aragón (IIS Aragón), Zaragoza, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas (CIBERehd), Madrid, Spain
- Fundación ARAID, Zaragoza, Spain
- Department of Biomedical and Aerospace Engineering, Universidad Carlos III de Madrid, Madrid, Spain
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
44
|
Hu Y, Zhang Z, Adiham A, Li H, Gu J, Gong P. In Vivo and In Vitro Models of Hepatic Fibrosis for Pharmacodynamic Evaluation and Pathology Exploration. Int J Mol Sci 2025; 26:696. [PMID: 39859410 PMCID: PMC11766297 DOI: 10.3390/ijms26020696] [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: 10/22/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Hepatic fibrosis (HF) is an important pathological state in the progression of chronic liver disease to end-stage liver disease and is usually triggered by alcohol, nonalcoholic fatty liver, chronic hepatitis viruses, autoimmune hepatitis (AIH), or cholestatic liver disease. Research on novel therapies has become a hot topic due to the reversibility of HF. Research into the molecular mechanisms of the pathology of HF and potential drug screening relies on reliable and rational biological models, mainly including animals and cells. Hence, a number of modeling approaches have been attempted based on human dietary, pathological, and physiological factors in the development of HF. In this review, classical and novel methods of modeling HF in the last 10 years were collected from electronic databases, including Web of Science, PubMed, ScienceDirect, ResearchGate, Baidu Scholar, and CNKI. Animal models of HF are usually induced by chemical toxicants, special diets, pathogenic microorganisms, surgical operations, and gene editing. The advantages and limitations of hepatic stellate cells (HSCs), organoids, and 3D coculture-based HF modeling methods established in vitro were also proposed and summarized. This information provides a scientific basis for the discovery of the pathological mechanism and treatment of HF.
Collapse
Affiliation(s)
| | | | | | | | - Jian Gu
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| | - Puyang Gong
- College of Pharmacy and Food, Southwest Minzu University, Chengdu 610093, China; (Y.H.); (Z.Z.); (A.A.); (H.L.)
| |
Collapse
|
45
|
Durazzo M, Ferro A, Navarro-Tableros VM, Gaido A, Fornengo P, Altruda F, Romagnoli R, Moestrup SK, Calvo PL, Fagoonee S. Current Treatment Regimens and Promising Molecular Therapies for Chronic Hepatobiliary Diseases. Biomolecules 2025; 15:121. [PMID: 39858515 PMCID: PMC11763965 DOI: 10.3390/biom15010121] [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: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
Chronic hepatobiliary damage progressively leads to fibrosis, which may evolve into cirrhosis and/or hepatocellular carcinoma. The fight against the increasing incidence of liver-related morbidity and mortality is challenged by a lack of clinically validated early-stage biomarkers and the limited availability of effective anti-fibrotic therapies. Current research is focused on uncovering the pathogenetic mechanisms that drive liver fibrosis. Drugs targeting molecular pathways involved in chronic hepatobiliary diseases, such as inflammation, hepatic stellate cell activation and proliferation, and extracellular matrix production, are being developed. Etiology-specific treatments, such as those for hepatitis B and C viruses, are already in clinical use, and efforts to develop new, targeted therapies for other chronic hepatobiliary diseases are ongoing. In this review, we highlight the major molecular changes occurring in patients affected by metabolic dysfunction-associated steatotic liver disease, viral hepatitis (Delta virus), and autoimmune chronic liver diseases (autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis). Further, we describe how this knowledge is linked to current molecular therapies as well as ongoing preclinical and clinical research on novel targeting strategies, including nucleic acid-, mesenchymal stromal/stem cell-, and extracellular vesicle-based options. Much clinical development is obviously still missing, but the plethora of promising potential treatment strategies in chronic hepatobiliary diseases holds promise for a future reversal of the current increase in morbidity and mortality in this group of patients.
Collapse
Affiliation(s)
- Marilena Durazzo
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Arianna Ferro
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Victor Manuel Navarro-Tableros
- 2i3T, Società per la Gestione dell’Incubatore di Imprese e per il Trasferimento Tecnologico, University of Turin, 10126 Turin, Italy;
| | - Andrea Gaido
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Paolo Fornengo
- Department of Medical Sciences, University of Turin, C.so A.M. Dogliotti 14, 10126 Turin, Italy; (M.D.); (A.F.); (A.G.); (P.F.)
| | - Fiorella Altruda
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre “Guido Tarone”, University of Turin, 10126 Turin, Italy;
| | - Renato Romagnoli
- General Surgery 2U-Liver Transplant Unit, Department of Surgical Sciences, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, University of Turin, Corso Bramante 88-90, 10126 Turin, Italy;
| | - Søren K. Moestrup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark;
- Department of Clinical Biochemistry, Aarhus University Hospital, 8000 Aarhus, Denmark
| | - Pier Luigi Calvo
- Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, Città della Salute e della Scienza, 10126 Turin, Italy;
| | - Sharmila Fagoonee
- Institute for Biostructure and Bioimaging, National Research Council, Molecular Biotechnology Centre “Guido Tarone”, 10126 Turin, Italy
| |
Collapse
|
46
|
Ravichandran A, Mahajan V, van de Kemp T, Taubenberger A, Bray LJ. Phenotypic analysis of complex bioengineered 3D models. Trends Cell Biol 2025:S0962-8924(24)00257-5. [PMID: 39794253 DOI: 10.1016/j.tcb.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
With advances in underlying technologies such as complex multicellular systems, synthetic materials, and bioengineering techniques, we can now generate in vitro miniaturized human tissues that recapitulate the organotypic features of normal or diseased tissues. Importantly, these 3D culture models have increasingly provided experimental access to diverse and complex tissues architectures and their morphogenic assembly in vitro. This review presents an analytical toolbox for biological researchers using 3D modeling technologies through which they can find a collation of currently available methods to phenotypically assess their 3D models in their normal state as well as their response to therapeutic or pathological agents.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Vaibhav Mahajan
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Tom van de Kemp
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Anna Taubenberger
- Biotechnology Center, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, 01307 Dresden, Germany
| | - Laura J Bray
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; School of Mechanical, Medical, and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia; Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia.
| |
Collapse
|
47
|
Yoneyama Y, Wu Y, Mori K, Takebe T. In toto biological framework: Modeling interconnectedness during human development. Dev Cell 2025; 60:8-20. [PMID: 39765224 DOI: 10.1016/j.devcel.2024.09.027] [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: 08/30/2023] [Revised: 04/21/2024] [Accepted: 09/20/2024] [Indexed: 05/24/2025]
Abstract
Recent advancements in pluripotent stem cell and synthetic tissue technology have brought significant breakthroughs in studying early embryonic development, particularly within the first trimester of development in humans. However, during fetal stage development, investigating further biological events represents a major challenge, partly due to the evolving complexity and continued interaction across multiple organ systems. To bridge this gap, we propose an "in toto" biological framework that leverages a triad of technologies: synthetic tissues, intravital microscopy, and computer vision to capture in vivo cellular morphodynamics, conceptualized as single-cell choreography. This perspective will discuss the inherent challenges in capturing such complexities and explore engineering technologies to delve into the less-explored phase of human development. We also propose reframing the organ-centric to a system-centric paradigm, as such a framework broadens the value of the in vivo-embedded synthetic-tissue-based approach for interrogating the multifaceted interplay of human developmental processes during this crucial stage.
Collapse
Affiliation(s)
- Yosuke Yoneyama
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Graduate School of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan; Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yunheng Wu
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | - Kensaku Mori
- Graduate School of Informatics, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Information Technology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan; Research Center for Medical Bigdata, National Institute of Informatics, Tokyo 100-0003, Japan
| | - Takanori Takebe
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Graduate School of Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita-shi, Osaka 565-0871, Japan; Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, and Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229-3039, USA.
| |
Collapse
|
48
|
Yang L, Wang X, Zhou X, Chen H, Song S, Deng L, Yao Y, Yin X. A tunable human intestinal organoid system achieves controlled balance between self-renewal and differentiation. Nat Commun 2025; 16:315. [PMID: 39747097 PMCID: PMC11697020 DOI: 10.1038/s41467-024-55567-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
A balance between stem cell self-renewal and differentiation is required to maintain concurrent proliferation and cellular diversification in organoids; however, this has proven difficult in homogeneous cultures devoid of in vivo spatial niche gradients for adult stem cell-derived organoids. In this study, we leverage a combination of small molecule pathway modulators to enhance the stemness of organoid stem cells, thereby amplifying their differentiation potential and subsequently increasing cellular diversity within human intestinal organoids without the need for artificial spatial or temporal signaling gradients. Moreover, we demonstrate that this balance between self-renewal and differentiation can be effectively and reversibly shifted from secretory cell differentiation to the enterocyte lineage with enhanced proliferation using BET inhibitors, or unidirectional differentiation towards specific intestinal cell types by manipulating in vivo niche signals such as Wnt, Notch, and BMP. As a result, we establish an optimized human small intestinal organoid (hSIO) system characterized by high proliferative capacity and increased cell diversity under a single culture condition. This optimization facilitates the scalability and utility of the organoid system in high-throughput applications.
Collapse
Affiliation(s)
- Li Yang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xulei Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xingyu Zhou
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hongyu Chen
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Sentao Song
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Liling Deng
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yao Yao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiaolei Yin
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
49
|
Zhu Z, Cheng Y, Liu X, Ding W, Liu J, Ling Z, Wu L. Advances in the Development and Application of Human Organoids: Techniques, Applications, and Future Perspectives. Cell Transplant 2025; 34:9636897241303271. [PMID: 39874083 PMCID: PMC11775963 DOI: 10.1177/09636897241303271] [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] [Revised: 10/10/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025] Open
Abstract
Organoids are three-dimensional (3D) cell cultures derived from human pluripotent stem cells or adult stem cells that recapitulate the cellular heterogeneity, structure, and function of human organs. These microstructures are invaluable for biomedical research due to their ability to closely mimic the complexity of native tissues while retaining human genetic material. This fidelity to native organ systems positions organoids as a powerful tool for advancing our understanding of human biology and for enhancing preclinical drug testing. Recent advancements have led to the successful development of a variety of organoid types, reflecting a broad range of human organs and tissues. This progress has expanded their application across several domains, including regenerative medicine, where organoids offer potential for tissue replacement and repair; disease modeling, which allows for the study of disease mechanisms and progression in a controlled environment; drug discovery and evaluation, where organoids provide a more accurate platform for testing drug efficacy and safety; and microecological research, where they contribute to understanding the interactions between microbes and host tissues. This review provides a comprehensive overview of the historical development of organoid technology, highlights the key achievements and ongoing challenges in the field, and discusses the current and emerging applications of organoids in both laboratory research and clinical practice.
Collapse
Affiliation(s)
- Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| |
Collapse
|
50
|
Peng L, Xiang S, Wang T, Yang M, Duan Y, Ma X, Li S, Yu C, Zhang X, Hu H, Liu Z, Sun J, Sun C, Wang C, Liu B, Wang Z, Qian M. The hepatic clock synergizes with HIF-1α to regulate nucleotide availability during liver damage repair. Nat Metab 2025; 7:148-165. [PMID: 39775529 DOI: 10.1038/s42255-024-01184-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Nucleotide availability is crucial for DNA replication and repair; however, the coordinating mechanisms in vivo remain unclear. Here, we show that the circadian clock in the liver controls the activity of the pentose phosphate pathway (PPP) to support de novo nucleotide biosynthesis for DNA synthesis demands. We demonstrate that disrupting the hepatic clock by genetic manipulation or mistimed feeding impairs PPP activity in male mice, leading to nucleotide imbalance. Such defects not only elicit DNA replication stress to limit liver regeneration after resection but also allow genotoxin-induced hepatocyte senescence and STING signalling-dependent inflammation. Mechanistically, the molecular clock activator BMAL1 synergizes with hypoxia-inducible factor-1α (HIF-1α) to regulate the transcription of the PPP rate-limiting enzyme glucose-6-phosphate dehydrogenase (G6PD), which is enhanced during liver regeneration. Overexpressing G6PD restores the compromised regenerative capacity of the BMAL1- or HIF-1α-deficient liver. Moreover, boosting G6PD expression genetically or through preoperative intermittent fasting potently facilitates liver repair in normal mice. Hence, our findings highlight the physiological importance of the hepatic clock and suggest a promising pro-regenerative strategy.
Collapse
Affiliation(s)
- Linyuan Peng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Siliang Xiang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Tianzhi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mei Yang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yajun Duan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xiaoyu Ma
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Su Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Cong Yu
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xin Zhang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, China
| | - Jie Sun
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Chunmeng Sun
- State Key Laboratory of Natural Medicines, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Center for Anti-aging and Regenerative Medicine, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
| | - Zhongyuan Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| | - Minxian Qian
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|