1
|
Geng Y, Chen Z, Luo T, Liu Y, Kong S, Yan X, Bai H, Wang Y. Innovative construction and application of bile duct organoids: Unraveling the complexity of bile duct diseases and potential therapeutic strategies. Cancer Lett 2025; 618:217619. [PMID: 40074068 DOI: 10.1016/j.canlet.2025.217619] [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: 01/13/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
The biliary system is crucial for liver function, regulating bile production, secretion, and transport. Dysfunctions within this system can lead to various diseases, such as cholangiopathies and biliary fibrosis, which may progress from benign to malignant states like cholangiocarcinoma. While liver organoid research is well-established and technologically advanced, bile duct organoids (BDOs) offer significant potential. BDOs can accurately simulate the physiological structure and function of bile ducts, making them valuable tools for in-vitro biliary disease research. Here, we review the development of BDO models, focusing on stem cell-derived organoids and tissue-derived organoids. We also illustrate the role of cultivation strategies and extracellular scaffolds in supporting organoid growth and stability, including the influence of cellular components of the microenvironment and physicochemical factors. Furthermore, we discuss the applications of BDOs in biliary development, disease modeling, regenerative medicine, and drug screening. Additionally, we emphasize the transformative potential in BDO biobanks and personalized medicine, which helps to pave the way for innovative therapeutic strategies and personalized medicine. Finally, we summarize the current and prospective advancements in BDO technologies, highlighting the integration of emerging technologies such as artificial intelligence, 3D bioprinting, and organoid-on-chip systems. These technologies hold great promise for significantly enhancing both clinical and research applications in the field of biliary diseases.
Collapse
Affiliation(s)
- Yadi Geng
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China; School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Ziye Chen
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Tianzi Luo
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Yakun Liu
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Siming Kong
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Hui Bai
- Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China.
| | - Yunfang Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, 102218, China; Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
2
|
Kiyuna LA, Horcas‐Nieto JM, Odendaal C, Langelaar‐Makkinje M, Gerding A, Broekhuis MJC, Bonanini F, Singh M, Kurek D, Harms AC, Hankemeier T, Foijer F, Derks TGJ, Bakker BM. iPSC-Derived Liver Organoids as a Tool to Study Medium Chain Acyl-CoA Dehydrogenase Deficiency. J Inherit Metab Dis 2025; 48:e70028. [PMID: 40199742 PMCID: PMC11978564 DOI: 10.1002/jimd.70028] [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: 03/06/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/10/2025]
Abstract
Medium chain acyl-CoA dehydrogenase deficiency (MCADD) is an inherited metabolic disease, characterized by biallelic variants in the ACADM gene. Interestingly, even with the same genotype, patients often present with very heterogeneous symptoms, ranging from fully asymptomatic to life-threatening hypoketotic hypoglycemia. The mechanisms underlying this heterogeneity remain unclear. Therefore, there is a need for in vitro models of MCADD that recapitulate the clinical phenotype as a tool to study the pathophysiology of the disease. Fibroblasts of control and symptomatic MCADD patients with the c.985A>G (p.K329E) were reprogrammed into induced pluripotent stem cells (iPSCs). iPSCs were then differentiated into hepatic expandable organoids (EHOs), further matured to Mat-EHOs, and functionally characterized. EHOs and Mat-EHOs performed typical hepatic metabolic functions, such as albumin and urea production. The organoids metabolized fatty acids, as confirmed by acyl-carnitine profiling and high-resolution respirometry. MCAD protein was fully ablated in MCADD organoids, in agreement with the instability of the mutated MCAD protein. MCADD organoids accumulated medium-chain acyl-carnitines, with a strongly elevated C8/C10 ratio, characteristic of the biochemical phenotype of the disease. Notably, C2 and C14 acyl-carnitines were found decreased in MCADD Mat-EHOs. Finally, MCADD organoids exhibited differential expression of genes involved in ω-oxidation, mitochondrial β-oxidation, TCA cycle, and peroxisomal coenzyme A metabolism, particularly upregulation of NUDT7. iPSC-derived organoids of MCADD patients recapitulated the major biochemical phenotype of the disease. Mat-EHOs expressed relevant pathways involved in putative compensatory mechanisms, notably CoA metabolism and the TCA cycle. The upregulation of NUDT7 expression may play a role in preventing excessive accumulation of dicarboxylic acids in MCADD. This patient-specific hepatic organoid system is a promising platform to study the phenotypic heterogeneity between MCADD patients.
Collapse
Affiliation(s)
- Ligia A. Kiyuna
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - José M. Horcas‐Nieto
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Christoff Odendaal
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Miriam Langelaar‐Makkinje
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Albert Gerding
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
- Department of Laboratory MedicineUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Mathilde J. C. Broekhuis
- European Research Institute for the Biology of AgeingUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | | | - Madhulika Singh
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | | | - Amy C. Harms
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | - Thomas Hankemeier
- Metabolomics and Analytics CentreLeiden Academic Centre for Drug Research, Leiden UniversityLeidenthe Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of AgeingUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| | - Terry G. J. Derks
- Section of Metabolic Diseases, Beatrix Children's HospitalUniversity Medical Centre Groningen, University of GroningenGroningenthe Netherlands
| | - Barbara M. Bakker
- Laboratory of PediatricsUniversity Medical Center Groningen, University of GroningenGroningenthe Netherlands
| |
Collapse
|
3
|
Zhou L, Huang J, Li C, Gu Q, Li G, Li ZA, Xu J, Zhou J, Tuan RS. Organoids and organs-on-chips: Recent advances, applications in drug development, and regulatory challenges. MED 2025; 6:100667. [PMID: 40220744 DOI: 10.1016/j.medj.2025.100667] [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: 03/21/2024] [Revised: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025]
Abstract
Organoids and organs-on-chips (OoCs) are rapidly evolving technologies for creating miniature human tissue models. They can mimic complex physiological functions and pathological conditions, offering more realistic platforms for disease modeling, drug screening, precision medicine, and regenerative therapies. The passing of the FDA Modernization Act 2.0 has reduced animal testing requirements for drug trials, marking a significant milestone in using advanced in vitro models such as organoids and OoCs for therapeutic discovery. Apart from technical and ethical challenges, regulatory issues persist in ensuring the reliability, scientificity, and applicability of these models in drug development. This perspective explores the concept, advancements, pros and cons, and applications of organoids and OoCs, particularly in drug research and development. It also examines global regulatory agencies' policies and actions on using these models in drug evaluation, aiming to guide industry standard setting and advance regulatory science.
Collapse
Affiliation(s)
- Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingjing Huang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cun Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qi Gu
- Key Laboratory of Organ Regeneration and Reconstruction, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gang Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital of the Southern Medical University, Guangzhou, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jie Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Papamichail L, Koch LS, Veerman D, Broersen K, van der Meer AD. Organoids-on-a-chip: microfluidic technology enables culture of organoids with enhanced tissue function and potential for disease modeling. Front Bioeng Biotechnol 2025; 13:1515340. [PMID: 40134772 PMCID: PMC11933005 DOI: 10.3389/fbioe.2025.1515340] [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: 10/22/2024] [Accepted: 02/12/2025] [Indexed: 03/27/2025] Open
Abstract
Organoids are stem-cell derived tissue structures mimicking specific structural and functional characteristics of human organs. Despite significant advancements in the field over the last decade, challenges like limited long-term functional culture and lack of maturation are hampering the implementation of organoids in biomedical research. Culture of organoids in microfluidic chips is being used to tackle these challenges through dynamic and precise control over the organoid microenvironment. This review highlights the significant breakthroughs that have been made in the innovative field of "organoids-on-chip," demonstrating how these have contributed to advancing organoid models. We focus on the incorporation of organoids representative for various tissues into chips and discuss the latest findings in multi-organoids-on-chip approaches. Additionally, we examine current limitations and challenges of the field towards the development of reproducible organoids-on-chip systems. Finally, we discuss the potential of organoids-on-chip technology for both in vitro and in vivo applications.
Collapse
Affiliation(s)
- Lito Papamichail
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lena S. Koch
- Applied Stem Cell Technologies, Department of Bioengineering Technologies, University of Twente, Enschede, Netherlands
| | - Devin Veerman
- Applied Stem Cell Technologies, Department of Bioengineering Technologies, University of Twente, Enschede, Netherlands
- BIOS Lab on a Chip Group, MESA+ Institute for Nanotechnology, University of Twente, Enschede, Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Department of Bioengineering Technologies, University of Twente, Enschede, Netherlands
| | - Andries D. van der Meer
- Applied Stem Cell Technologies, Department of Bioengineering Technologies, University of Twente, Enschede, Netherlands
| |
Collapse
|
5
|
Maity A, Maidantchik VD, Weidenfeld K, Larisch S, Barkan D, Haick H. Chemical Tomography of Cancer Organoids and Cyto-Proteo-Genomic Development Stages Through Chemical Communication Signals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413017. [PMID: 39935131 PMCID: PMC11938034 DOI: 10.1002/adma.202413017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Organoids mimic human organ function, offering insights into development and disease. However, non-destructive, real-time monitoring is lacking, as traditional methods are often costly, destructive, and low-throughput. In this article, a non-destructive chemical tomographic strategy is presented for decoding cyto-proteo-genomics of organoid using volatile signaling molecules, hereby, Volatile Organic Compounds (VOCs), to indicate metabolic activity and development of organoids. Combining a hierarchical design of graphene-based sensor arrays with AI-driven analysis, this method maps VOC spatiotemporal distribution and generate detailed digital profiles of organoid morphology and proteo-genomic features. Lens- and label-free, it avoids phototoxicity, distortion, and environmental disruption. Results from testing organoids with the reported chemical tomography approach demonstrate effective differentiation between cyto-proteo-genomic profiles of normal and diseased states, particularly during dynamic transitions such as epithelial-mesenchymal transition (EMT). Additionally, the reported approach identifies key VOC-related biochemical pathways, metabolic markers, and pathways associated with cancerous transformations such as aromatic acid degradation and lipid metabolism. This real-time, non-destructive approach captures subtle genetic and structural variations with high sensitivity and specificity, providing a robust platform for multi-omics integration and advancing cancer biomarker discovery.
Collapse
Affiliation(s)
- Arnab Maity
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
| | - Vivian Darsa Maidantchik
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
| | - Keren Weidenfeld
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Sarit Larisch
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Dalit Barkan
- Department of Human Biology and Medical SciencesUniversity of HaifaHaifa3498838Israel
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology InstituteTechnion – Israel Institute of TechnologyHaifa3200003Israel
- Life Science Technology (LiST) GroupDanube Private UniversityFakultät Medizin/Zahnmedizin, Steiner Landstraße 124Krems‐Stein3500Austria
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Fan H, Shang J, Li J, Yang B, Zhou D, Jiang S, Fan Y, Zhou Y, Wang Y, Liu P, Li C, Chen Z, Chen P. High-Throughput Formation of Pre-Vascularized hiPSC-Derived Hepatobiliary Organoids on a Chip via Nonparenchymal Cell Grafting. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407945. [PMID: 39755926 PMCID: PMC11848576 DOI: 10.1002/advs.202407945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/17/2024] [Indexed: 01/06/2025]
Abstract
Liver organoids have been increasingly adopted as a critical in vitro model to study liver development and diseases. However, the pre-vascularization of liver organoids without affecting liver parenchymal specification remains a long-lasting challenge, which is essential for their application in regenerative medicine. Here, the large-scale formation of pre-vascularized human hepatobiliary organoids (vhHBOs) is presented without affecting liver epithelial specification via a novel strategy, namely nonparenchymal cell grafting (NCG). Endothelial and mesenchymal cells are grafted to human hepatobiliary organoids (hHBOs) at the different liver epithelial differentiation stages without supplementing with nonparenchymal culture medium and growth factors. Endothelial grafting at the stage of hepatic maturation offers an optimal integration efficiency compared to the stage of hepatic specification. Additionally, grafting with mesenchymal proves crucial in endothelial invading and sprouting into the liver epithelial cells during the establishment of vhHBOs. Ectopic liver implants into mice further displayed integration of vhHBOs into mice vascular networks. Notably, transplanted vhHBOs self-organized into native liver tissue like hepatic zone and bile ducts, indicating their potential to regenerate damaged hepatic and bile duct tissues. It is believed that nonparenchymal cell grafting will offer a novel technical route to form a high-fidelity complex in vitro model for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Han Fan
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Jia Shang
- Department of Biological RepositoriesZhongnan Hospital of Wuhan UniversityWuhan430071China
| | - Junbo Li
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Bo Yang
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Ding Zhou
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Shanqing Jiang
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Yuhang Fan
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Ying Zhou
- Research Center for Medicine and Structural Biology of Wuhan UniversityWuhan UniversityWuhanHubei430071China
| | - Yuwen Wang
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Peidi Liu
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
| | - Changyong Li
- Department of PhysiologyWuhan University TaiKang Medical School (School of Basic Medical Sciences)WuhanHubei430071China
| | - Zhishui Chen
- Key Laboratory of Organ TransplantationInstitute of Organ TransplantationTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Pu Chen
- Tissue Engineering and Organ Manufacturing (TEOM) LabDepartment of Biomedical EngineeringWuhan University TaiKang Medical School (School of Basic Medical Sciences)Wuhan430071China
- TaiKang Center for Life and Medical SciencesWuhan UniversityWuhan430071China
| |
Collapse
|
9
|
Dutta A, Chowdhury N, Chandra S, Guha P, Saha V, GuhaSarkar D. Gallbladder cholangiocyte organoids. Biol Cell 2025; 117:e2400132. [PMID: 39945546 PMCID: PMC11823593 DOI: 10.1111/boc.202400132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/23/2024] [Accepted: 01/30/2025] [Indexed: 02/16/2025]
Abstract
Organoids are miniature three-dimensional (3D) organ-like structures developed from primary cells that closely mimic the key histological, functional, and molecular characteristics of their parent organs. These structures self-organize through cell-cell and cell-matrix interaction in culture. In the last decade, organoids and allied 3D culture technologies have catalyzed studies involving developmental biology, disease biology, high-throughput drug screening, personalized medicine, biomarker discovery, tissue engineering, and regenerative medicine. Many organoid systems have been generated from the gastrointestinal system, for example, intestine, stomach, liver, pancreas, or colon. Gallbladder cancer (GBC) is the most common and highly aggressive form of biliary tract cancer. GBC is rare in the west but has a high incidence in South America and India. Prolonged chronic inflammation is implicated in the pathogenesis of GBC but the driving molecular pathways leading to neoplasia are not well understood. Gallbladder cholangiocyte organoids (GCO) will facilitate the understanding of the evolution of the disease and novel therapeutic strategies. In this review, we have discussed alternative methodologies and culture conditions developed to generate GCO models, applications that these models have been subjected to and the current limitations for the use of GCOs in addressing the challenges in GBC research.
Collapse
Affiliation(s)
- Ankita Dutta
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
- School of Medical Science and TechnologyIndian Institute of Technology KharagpurKharagpurIndia
| | - Nandita Chowdhury
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Shinjini Chandra
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Payel Guha
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Vaskar Saha
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
- Department of Paediatric Haematology and Oncology Tata Medical CenterKolkataIndia
- Division of Cancer SciencesFaculty of BiologyMedicine and HealthSchool of Medical SciencesUniversity of ManchesterManchesterUK
| | - Dwijit GuhaSarkar
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Shah DD, Chorawala MR, Pandya AJ, Kothari N, Prajapati BG, Parekh PS. Advancing the Battle against Cystic Fibrosis: Stem Cell and Gene Therapy Insights. Curr Med Sci 2024; 44:1155-1174. [PMID: 39676146 DOI: 10.1007/s11596-024-2936-5] [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: 03/01/2024] [Accepted: 09/03/2024] [Indexed: 12/17/2024]
Abstract
Cystic fibrosis (CF) is a hereditary disorder characterized by mutations in the CFTR gene, leading to impaired chloride ion transport and subsequent thickening of mucus in various organs, particularly the lungs. Despite significant progress in CF management, current treatments focus mainly on symptom relief and do not address the underlying genetic defects. Stem cell and gene therapies present promising avenues for tackling CF at its root cause. Stem cells, including embryonic, induced pluripotent, mesenchymal, hematopoietic, and lung progenitor cells, offer regenerative potential by differentiating into specialized cells and modulating immune responses. Similarly, gene therapy aims to correct CFTR gene mutations by delivering functional copies of the gene into affected cells. Various approaches, such as viral and nonviral vectors, gene editing with CRISPR-Cas9, small interfering RNA (siRNA) therapy, and mRNA therapy, are being explored to achieve gene correction. Despite their potential, challenges such as safety concerns, ethical considerations, delivery system optimization, and long-term efficacy remain. This review provides a comprehensive overview of the current understanding of CF pathophysiology, the rationale for exploring stem cell and gene therapies, the types of therapies available, their mechanisms of action, and the challenges and future directions in the field. By addressing these challenges, stem cell and gene therapies hold promise for transforming CF management and improving the quality of life of affected individuals.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Aanshi J Pandya
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Nirjari Kothari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, India
| | - Bhupendra G Prajapati
- Department of Pharmaceutics and Pharmaceutical Technology, Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat University, Mehsana, 384012, India.
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| | | |
Collapse
|
13
|
Frau C, Vallier L. Exploiting the plasticity of cholangiocytes to repair the biliary tree. Curr Opin Genet Dev 2024; 89:102257. [PMID: 39255689 DOI: 10.1016/j.gde.2024.102257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/13/2024] [Accepted: 08/17/2024] [Indexed: 09/12/2024]
Abstract
Cholangiocytes are the main cell type lining the epithelium of the biliary tree of the liver. This cell type has been implicated not only in diseases affecting the biliary tree but also in chronic liver diseases targeting other hepatic cells such as hepatocytes. However, the isolation and culture of cholangiocytes have been particularly arduous, thereby limiting the development of new therapies. The emergence of organoids has the potential to address in part this challenge. Indeed, cholangiocyte organoids can be established from both the intra- and extrahepatic regions of the biliary tree, providing an advantageous platform for disease modeling and mechanism investigations. Accordingly, recent studies on cholangiocyte organoids, together with the advent of single-cell -omics, have opened the field to exciting discoveries concerning the plastic nature of these cells and their capability to adapt to different environments and stimuli. This review will focus on describing how these plasticity properties could be exploited in regenerative medicine and cell-based therapy, opening new frontiers for treating disorders affecting the biliary tree and beyond.
Collapse
Affiliation(s)
- Carla Frau
- Berlin Institute of Health Centre for Regenerative Therapies, Berlin, Germany; Max Planck Institute for Molecular Genetics, Berlin, Germany; Berlin Institute of Health @Charite, Berlin, Germany.
| | - Ludovic Vallier
- Berlin Institute of Health Centre for Regenerative Therapies, Berlin, Germany; Max Planck Institute for Molecular Genetics, Berlin, Germany; Berlin Institute of Health @Charite, Berlin, Germany.
| |
Collapse
|
14
|
Shrestha S, Acharya P, Kang SY, Vanga MG, Lekkala VKR, Liu J, Yang Y, Joshi P, Lee MY. Regenerative human liver organoids (HLOs) in a pillar/perfusion plate for hepatotoxicity assays. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.25.586638. [PMID: 38586058 PMCID: PMC10996672 DOI: 10.1101/2024.03.25.586638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Human liver organoids (HLOs) differentiated from embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and adult stem cells (ASCs) can recapitulate the structure and function of human fetal liver tissues, thus being considered as a promising tissue model for liver diseases and predictive compound screening. However, the adoption of HLOs in drug discovery faces several technical challenges, which include the lengthy differentiation process with multiple culture media leading to batch-to-batch variation, short-term maintenance of hepatic functions post-maturation, low assay throughput due to Matrigel dissociation and HLO transfer to a microtiter well plate, and insufficient maturity levels compared to primary hepatocytes. To address these issues, expandable HLOs (Exp-HLOs) derived from human iPSCs were generated by optimizing differentiation protocols, which were rapidly printed on a 144-pillar plate with sidewalls and slits (144PillarPlate) and dynamically cultured for up to 20 days into differentiated HLOs (Diff-HLOs) in a 144-perfusion plate with perfusion wells and reservoirs (144PerfusionPlate) for in situ organoid culture and analysis. The dynamically cultured Diff-HLOs exhibited greater maturity and reproducibility than those cultured statically, especially after a 10-day differentiation period. In addition, Diff-HLOs in the pillar/perfusion plate were tested with acetaminophen and troglitazone for 3 days to assess drug-induced liver injury (DILI) and then incubated in an expansion medium for 10 days to evaluate liver recovery from DILI. The assessment of liver regeneration post-injury is critical to understanding the mechanism of recovery and determining the threshold drug concentration beyond which there will be a sharp decrease in the liver's regenerative capacity. We envision that bioprinted Diff-HLOs in the pillar/perfusion plate could be used for high-throughput screening (HTS) of hepatotoxic compounds due to the short-term differentiation of passage-able Exp-HLOs, stable hepatic function post-maturation, high reproducibility, and high throughput with capability of in situ organoid culture, testing, staining, imaging, and analysis.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | | | | | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
| | - Pranav Joshi
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, Texas, USA
- Bioprinting Laboratories Inc., Dallas, Texas, USA
| |
Collapse
|
15
|
Nijiati N, Wubuli D, Li X, Zhou Z, Julaiti M, Huang P, Hu B. The Construction of Stem Cell-Induced Hepatocyte Model and Its Application in Evaluation of Developmental Hepatotoxicity of Environmental Pollutants. Stem Cells Dev 2024; 33:575-585. [PMID: 39109950 DOI: 10.1089/scd.2024.0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Stem cells, with their ability to self-renew and differentiate into various cell types, are a unique and valuable resource for medical research and toxicological studies. The liver is the most crucial metabolic organ in the human body and serves as the primary site for the accumulation of environmental pollutants. Enrichment with environmental pollutants can disrupt the early developmental processes of the liver and have a significant impact on liver function. The liver comprises a complex array of cell types, and different environmental pollutants have varying effects on these cells. Currently, there is a lack of well-established research models that can effectively demonstrate the mechanisms by which environmental pollutants affect human liver development. The emergence of liver cells and organoids derived from stem cells offers a promising tool for investigating the impact of environmental pollutants on human health. Therefore, this study systematically reviewed the developmental processes of different types of liver cells and provided an overview of studies on the developmental toxicity of various environmental pollutants using stem cell models.
Collapse
Affiliation(s)
- Nadire Nijiati
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Dilixiati Wubuli
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Xiaobing Li
- The Third Clinical Medicine College of Xinjiang Medical University, Urumqi, China
| | - Zidong Zhou
- The Third Clinical Medicine College of Xinjiang Medical University, Urumqi, China
| | - Mulati Julaiti
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Pengfei Huang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Bowen Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
16
|
Panchuk I, Smirnikhina S. Toolbox for creating three-dimensional liver models. Biochem Biophys Res Commun 2024; 731:150375. [PMID: 39018971 DOI: 10.1016/j.bbrc.2024.150375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Research within the hepato-biliary system and hepatic function is currently experiencing heightened interest, this is due to the high frequency of relapse rates observed in chronic conditions, as well as the imperative for the development of innovative therapeutic strategies to address both inherited and acquired diseases within this domain. The most commonly used sources for studying hepatocytes include primary human hepatocytes, human hepatic cancer cell lines, and hepatic-like cells derived from induced pluripotent stem cells. However, a significant challenge in primary hepatic cell culture is the rapid decline in their phenotypic characteristics, dedifferentiation and short cultivation time. This limitation creates various problems, including the inability to maintain long-term cell cultures, which can lead to failed experiments in drug development and the creation of relevant disease models for researchers' purposes. To address these issues, the creation of a powerful 3D cell model could play a pivotal role as a personalized disease model and help reduce the use of animal models during certain stages of research. Such a cell model could be used for disease modelling, genome editing, and drug discovery purposes. This review provides an overview of the main methods of 3D-culturing liver cells, including a discussion of their characteristics, advantages, and disadvantages.
Collapse
Affiliation(s)
- Irina Panchuk
- Research Centre for Medical Genetics, Moscow, Russian Federation.
| | | |
Collapse
|
17
|
Wang L, Koui Y, Kanegae K, Kido T, Tamura-Nakano M, Yabe S, Tai K, Nakajima Y, Kusuhara H, Sakai Y, Miyajima A, Okochi H, Tanaka M. Establishment of human induced pluripotent stem cell-derived hepatobiliary organoid with bile duct for pharmaceutical research use. Biomaterials 2024; 310:122621. [PMID: 38815455 DOI: 10.1016/j.biomaterials.2024.122621] [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: 01/05/2024] [Revised: 04/26/2024] [Accepted: 05/19/2024] [Indexed: 06/01/2024]
Abstract
In vitro models of the human liver are promising alternatives to animal tests for drug development. Currently, primary human hepatocytes (PHHs) are preferred for pharmacokinetic and cytotoxicity tests. However, they are unable to recapitulate the flow of bile in hepatobiliary clearance owing to the lack of bile ducts, leading to the limitation of bile analysis. To address the issue, a liver organoid culture system that has a functional bile duct network is desired. In this study, we aimed to generate human iPSC-derived hepatobiliary organoids (hHBOs) consisting of hepatocytes and bile ducts. The two-step differentiation process under 2D and semi-3D culture conditions promoted the maturation of hHBOs on culture plates, in which hepatocyte clusters were covered with monolayered biliary tubes. We demonstrated that the hHBOs reproduced the flow of bile containing a fluorescent bile acid analog or medicinal drugs from hepatocytes into bile ducts via bile canaliculi. Furthermore, the hHBOs exhibited pathophysiological responses to troglitazone, such as cholestasis and cytotoxicity. Because the hHBOs can recapitulate the function of bile ducts in hepatobiliary clearance, they are suitable as a liver disease model and would be a novel in vitro platform system for pharmaceutical research use.
Collapse
Affiliation(s)
- Luyao Wang
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan; Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Yuta Koui
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Kazuko Kanegae
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Taketomo Kido
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Miwa Tamura-Nakano
- Communal Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shigeharu Yabe
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kenpei Tai
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiko Nakajima
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kusuhara
- Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Atsushi Miyajima
- Laboratory of Cell Growth and Differentiation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan
| | - Hitoshi Okochi
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Minoru Tanaka
- Department of Regenerative Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan; Laboratory of Stem Cell Regulation, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
18
|
Luo X, Gong Y, Gong Z, Fan K, Suo T, Liu H, Ni X, Ni X, Abudureyimu M, Liu H. Liver and bile duct organoids and tumoroids. Biomed Pharmacother 2024; 178:117104. [PMID: 39024834 DOI: 10.1016/j.biopha.2024.117104] [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: 03/28/2024] [Revised: 06/23/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024] Open
Abstract
Organoids refer to 3D cultures established to recapitulate histology, pathology, architecture, and genetic traits of various organs and tissues in the body, thereby replacing 2D cell cultures, xenograft, and animal models. Organoids form a 3D in vitro mimic of original tissues like the liver and are derived from embryonic or adult tissue stem cells. Liver and bile duct tumor organoids, also called, tumoroids capture genetic diversity, cellular, and pathophysiological properties of original tumors. Moreover, co-culture techniques along with genetic modulation of organoids allow for using tumoroids in liver and bile duct cancer research and drug screening/testing. Therefore, tumoroids are promising platforms for studying liver and bile duct cancer, which paves the way for the new era of personalized therapies. In the current review, we aimed to discuss liver and bile duct organoids with special emphasis on tumoroids and their applications, advantages, and shortcomings.
Collapse
Affiliation(s)
- Xuanming Luo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Yuda Gong
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Zijun Gong
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Kun Fan
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China
| | - Tao Suo
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Han Liu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Xiaoling Ni
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Xiaojian Ni
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China
| | - Miyesaier Abudureyimu
- Cardiovascular Department, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China.
| | - Houbao Liu
- Department of Biliary Surgery, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Center of Zhongshan Hospital, Fudan University, China; Cancer Center, Zhongshan Hospital, Fudan University, China; Biliary Tract Disease Institute, Fudan University, China; Shanghai Engineering Research Center of Biliary Tract Minimal Invasive Surgery and Materials, China; Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Kasper VL, Assis DN. Pathophysiology of Cystic Fibrosis Liver Disease. Pediatr Pulmonol 2024; 59 Suppl 1:S98-S106. [PMID: 39105342 DOI: 10.1002/ppul.26869] [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: 08/05/2023] [Revised: 12/14/2023] [Accepted: 01/10/2024] [Indexed: 08/07/2024]
Abstract
Hepatobiliary complications of Cystic Fibrosis (CF) constitute a significant burden for persons with CF of all ages, with advanced CF liver disease in particular representing a leading cause of mortality. The causes of the heterogeneity of clinical manifestations, ranging from steatosis to focal biliary cholestasis and biliary strictures, are poorly understood and likely reflect a variety of environmental and disease-modifying factors in the setting of underlying CFTR mutations. This review summarizes the current understanding of the pathophysiology of hepatobiliary manifestations of CF, and discusses emerging disease models and therapeutic approaches that hold promise to impact this important yet incompletely addressed aspect of CF care.
Collapse
Affiliation(s)
- Vania L Kasper
- The Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Hasbro Children's Hospital, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - David N Assis
- Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
20
|
Carolina E, Kuse Y, Okumura A, Aoshima K, Tadokoro T, Matsumoto S, Kanai E, Okumura T, Kasai T, Yamabe S, Nishikawa Y, Yamaguchi K, Furukawa Y, Tanimizu N, Taniguchi H. Generation of human iPSC-derived 3D bile duct within liver organoid by incorporating human iPSC-derived blood vessel. Nat Commun 2024; 15:7424. [PMID: 39198465 PMCID: PMC11358266 DOI: 10.1038/s41467-024-51487-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
In fetal development, tissue interaction such as the interplay between blood vessel (BV) and epithelial tissue is crucial for organogenesis. Here we recapitulate the spatial arrangement between liver epithelial tissue and the portal vein to observe the formation of intrahepatic bile ducts (BDs) from human induced pluripotent stem cells (hiPSC). We co-culture hiPSC-liver progenitors on the artificial BV consisting of immature smooth muscle cells and endothelial cells, both derived from hiPSCs. After 3 weeks, liver progenitors within hiPSC-BV-incorporated liver organoids (BVLO) differentiate to cholangiocytes and acquire epithelial characteristics, including intercellular junctions, microvilli on the apical membrane, and secretory functions. Furthermore, liver surface transplanted-BVLO temporarily attenuates cholestatic injury symptoms. Single cell RNA sequence analysis suggests that BD interact with the BV in BVLO through TGFβ and Notch pathways. Knocking out JAG1 in hiPSC-BV significantly attenuates bile duct formation, highlighting BVLO potential as a model for Alagille syndrome, a congenital biliary disease. Overall, we develop a novel 3D co-culture method that successfully establishes functional human BDs by emulating liver epithelial-BV interaction.
Collapse
Affiliation(s)
- Erica Carolina
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yoshiki Kuse
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ayumu Okumura
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Kenji Aoshima
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Tomomi Tadokoro
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Shinya Matsumoto
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Eriko Kanai
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Okumura
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshiharu Kasai
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Souichiro Yamabe
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yuji Nishikawa
- Division of Tumor Pathology, Department of Pathology, Asahikawa Medical University, Asahikawa, Hokkaido, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hideki Taniguchi
- Division of Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Department of Regenerative Medicine, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.
| |
Collapse
|
21
|
Karnawat K, Parthasarathy R, Sakhrie M, Karthik H, Krishna KV, Balachander GM. Building in vitro models for mechanistic understanding of liver regeneration in chronic liver diseases. J Mater Chem B 2024; 12:7669-7691. [PMID: 38973693 DOI: 10.1039/d4tb00738g] [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: 07/09/2024]
Abstract
The liver has excellent regeneration potential and attains complete functional recovery from partial hepatectomy. The regenerative mechanisms malfunction in chronic liver diseases (CLDs), which fuels disease progression. CLDs account for 2 million deaths per year worldwide. Pathophysiological studies with clinical correlation have shown evidence of deviation of normal regenerative mechanisms and its contribution to fueling fibrosis and disease progression. However, we lack realistic in vitro models that can allow experimental manipulation for mechanistic understanding of liver regeneration in CLDs and testing of candidate drugs. In this review, we aim to provide the framework for building appropriate organotypic models for dissecting regenerative responses in CLDs, with the focus on non-alcoholic steatohepatitis (NASH). By drawing parallels with development and hepatectomy, we explain the selection of critical components such as cells, signaling, and, substrate-driven biophysical cues to build an appropriate CLD model. We highlight the organoid-based organotypic models available for NASH disease modeling, including organ-on-a-chip and 3D bioprinted models. With the focus on bioprinting as a fabrication method, we prescribe building in vitro CLD models and testing schemes for exploring the regenerative responses in the bioprinted model.
Collapse
Affiliation(s)
- Khushi Karnawat
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Rithika Parthasarathy
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Mesevilhou Sakhrie
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Harikeshav Karthik
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Konatala Vibhuvan Krishna
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Gowri Manohari Balachander
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| |
Collapse
|
22
|
Liu S, Cheng C, Zhu L, Zhao T, Wang Z, Yi X, Yan F, Wang X, Li C, Cui T, Yang B. Liver organoids: updates on generation strategies and biomedical applications. Stem Cell Res Ther 2024; 15:244. [PMID: 39113154 PMCID: PMC11304926 DOI: 10.1186/s13287-024-03865-3] [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: 04/25/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
The liver is the most important metabolic organ in the body. While mouse models and cell lines have further deepened our understanding of liver biology and related diseases, they are flawed in replicating key aspects of human liver tissue, particularly its complex structure and metabolic functions. The organoid model represents a major breakthrough in cell biology that revolutionized biomedical research. Organoids are in vitro three-dimensional (3D) physiological structures that recapitulate the morphological and functional characteristics of tissues in vivo, and have significant advantages over traditional cell culture methods. In this review, we discuss the generation strategies and current advances in the field focusing on their application in regenerative medicine, drug discovery and modeling diseases.
Collapse
Affiliation(s)
- Sen Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
| | | | - Liuyang Zhu
- First Central Clinical College of Tianjin Medical University, Tianjin, 300192, China
| | - Tianyu Zhao
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
| | - Ze Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
- Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiulin Yi
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
- Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Fengying Yan
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
- Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xiaoliang Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China
| | - Chunli Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Tao Cui
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, 300301, China.
- Research Unit for Drug Metabolism, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Baofeng Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- School of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
23
|
Wang J, Wu X, Zhao J, Ren H, Zhao Y. Developing Liver Microphysiological Systems for Biomedical Applications. Adv Healthc Mater 2024; 13:e2302217. [PMID: 37983733 DOI: 10.1002/adhm.202302217] [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: 07/13/2023] [Revised: 11/15/2023] [Indexed: 11/22/2023]
Abstract
Microphysiological systems (MPSs), also known as organ chips, are micro-units that integrate cells with diverse physical and biochemical environmental cues. In the field of liver MPSs, cellular components have advanced from simple planar cell cultures to more sophisticated 3D formations such as spheroids and organoids. Additionally, progress in microfluidic devices, bioprinting, engineering of matrix materials, and interdisciplinary technologies have significant promise for producing MPSs with biomimetic structures and functions. This review provides a comprehensive summary of biomimetic liver MPSs including their clinical applications and future developmental potential. First, the key components of liver MPSs, including the principal cell types and engineered structures utilized for cell cultivation, are briefly introduced. Subsequently, the biomedical applications of liver MPSs, including the creation of disease models, drug absorption, distribution, metabolism, excretion, and toxicity, are discussed. Finally, the challenges encountered by MPSs are summarized, and future research directions for their development are proposed.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Xiangyi Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Junqi Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Southeast University Shenzhen Research Institute, Shenzhen, 518071, China
| |
Collapse
|
24
|
Rejas C, Junger H. Cholangiocyte Organoids in Liver Transplantation; a Comprehensive Review. Transpl Int 2024; 37:12708. [PMID: 39100755 PMCID: PMC11294148 DOI: 10.3389/ti.2024.12708] [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: 01/19/2024] [Accepted: 06/17/2024] [Indexed: 08/06/2024]
Abstract
Liver transplantation is the only curative option for many liver diseases that end up in liver failure, and cholangiopathy remains a challenging complication post-liver transplant, associated with significant morbidity and potential graft loss. The low availability of organs and high demand for transplantation motivate scientists to find novel interventions. Organoids, as three-dimensional cell cultures derived from adult cells or induced pluripotent cells, may help to address this problem. Different types of organoids have been described, from which cholangiocyte organoids offer a high level of versatility and plasticity for a deeper study of liver disease mechanisms. Cholangiocytes can be obtained from different segments of the biliary tree and have shown a remarkable capacity to adapt to new environments, presenting an effective system for studying cholangiopathies. Studies using cholangiocyte organoids show promising results for disease modeling, where organoids offer fundamental features to recapitulate the complexities of tissues in vitro and uncover fundamental pathological pathways to potentially reveal therapeutic strategies for personalized medicine. Organoids could hold the potential for regeneration of injured livers, representing tools of clinical impact in regenerative medicine when tissue damage is already present.
Collapse
Affiliation(s)
| | - H. Junger
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
25
|
Budi NYP, Lai WY, Huang YH, Ho HN. 3D organoid cultivation improves the maturation and functional differentiation of cholangiocytes from human pluripotent stem cells. Front Cell Dev Biol 2024; 12:1361084. [PMID: 39040044 PMCID: PMC11260683 DOI: 10.3389/fcell.2024.1361084] [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: 12/24/2023] [Accepted: 05/29/2024] [Indexed: 07/24/2024] Open
Abstract
Idiopathic cholangiopathies are diseases that affect cholangiocytes, and they have unknown etiologies. Currently, orthotopic liver transplantation is the only treatment available for end-stage liver disease. Limited access to the bile duct makes it difficult to model cholangiocyte diseases. In this study, by mimicking the embryonic development of cholangiocytes and using a robust, feeder- and serum-free protocol, we first demonstrate the generation of unique functional 3D organoids consisting of small and large cholangiocytes derived from human pluripotent stem cells (PSCs), as opposed to traditional 2D culture systems. At day 28 of differentiation, the human PSC-derived cholangiocytes expressed markers of mature cholangiocytes, such as CK7, CK19, and cystic fibrosis transmembrane conductance regulator (CFTR). Compared with the 2D culture system-generated cholangiocytes, the 3D cholangiocyte organoids (COs) showed higher expression of the region-specific markers of intrahepatic cholangiocytes YAP1 and JAG1 and extrahepatic cholangiocytes AQP1 and MUC1. Furthermore, the COs had small-large tube-like structures and functional assays revealed that they exhibited characteristics of mature cholangiocytes, such as multidrug resistance protein 1 transporter function and CFTR channel activity. In addition to the extracellular matrix supports, the epidermal growth factor receptor (EGFR)-mediated signaling regulation might be involved in this cholangiocyte maturation and differentiation. These results indicated the successful generation of intrahepatic and extrahepatic cholangiocytes by using our 3D organoid protocol. The results highlight the advantages of our 3D culture system over the 2D culture system in promoting the functional differentiation and maturation of cholangiocytes. In summary, in advance of the previous works, our study provides a possible concept of small-large cholangiocyte transdifferentiation of human PSCs under cost-effective 3D culture conditions. The study findings have implications for the development of effective cell-based therapy using COs for patients with cholangiopathies.
Collapse
Affiliation(s)
- Nova Yuli Prasetyo Budi
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Pediatric Surgery Division, Department of Surgery/Genetics Working Group, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada/Dr. Sardjito Hospital, Yogyakarta, Indonesia
| | - Wei-Yu Lai
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Hua Huang
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hong-Nerng Ho
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Taipei Municipal Wanfang Hospital, Taipei Medical University, Taipei, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
26
|
Okumura A, Aoshima K, Tanimizu N. Generation of in vivo-like multicellular liver organoids by mimicking developmental processes: A review. Regen Ther 2024; 26:219-234. [PMID: 38903867 PMCID: PMC11186971 DOI: 10.1016/j.reth.2024.05.020] [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: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/22/2024] Open
Abstract
Liver is involved in metabolic reactions, ammonia detoxification, and immunity. Multicellular liver tissue cultures are more desirable for drug screening, disease modeling, and researching transplantation therapy, than hepatocytes monocultures. Hepatocytes monocultures are not stable for long. Further, hepatocyte-like cells induced from pluripotent stem cells and in vivo hepatocytes are functionally dissimilar. Organoid technology circumvents these issues by generating functional ex vivo liver tissue from intrinsic liver progenitor cells and extrinsic stem cells, including pluripotent stem cells. To function as in vivo liver tissue, the liver organoid cells must be arranged precisely in the 3-dimensional space, closely mimicking in vivo liver tissue. Moreover, for long term functioning, liver organoids must be appropriately vascularized and in contact with neighboring epithelial tissues (e.g., bile canaliculi and intrahepatic bile duct, or intrahepatic and extrahepatic bile ducts). Recent discoveries in liver developmental biology allows one to successfully induce liver component cells and generate organoids. Thus, here, in this review, we summarize the current state of knowledge on liver development with a focus on its application in generating different liver organoids. We also cover the future prospects in creating (functionally and structurally) in vivo-like liver organoids using the current knowledge on liver development.
Collapse
Affiliation(s)
- Ayumu Okumura
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Kenji Aoshima
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| | - Naoki Tanimizu
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-0071, Japan
| |
Collapse
|
27
|
Afonso MB, Marques V, van Mil SW, Rodrigues CM. Human liver organoids: From generation to applications. Hepatology 2024; 79:1432-1451. [PMID: 36815360 PMCID: PMC11095893 DOI: 10.1097/hep.0000000000000343] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023]
Abstract
In the last decade, research into human hepatology has been revolutionized by the development of mini human livers in a dish. These liver organoids are formed by self-organizing stem cells and resemble their native counterparts in cellular content, multicellular architecture, and functional features. Liver organoids can be derived from the liver tissue or pluripotent stem cells generated from a skin biopsy, blood cells, or renal epithelial cells present in urine. With the development of liver organoids, a large part of previous hurdles in modeling the human liver is likely to be solved, enabling possibilities to better model liver disease, improve (personalized) drug testing, and advance bioengineering options. In this review, we address strategies to generate and use organoids in human liver disease modeling, followed by a discussion of their potential application in drug development and therapeutics, as well as their strengths and limitations.
Collapse
Affiliation(s)
- Marta B. Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Saskia W.C. van Mil
- Center for Molecular Medicine, University Medical Center Utrecht and Utrecht University, The Netherlands
| | - Cecilia M.P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| |
Collapse
|
28
|
Zhang W, Wu H, Luo S, Lu X, Tan X, Wen L, Ma X, Efferth T. Molecular insights into experimental models and therapeutics for cholestasis. Biomed Pharmacother 2024; 174:116594. [PMID: 38615607 DOI: 10.1016/j.biopha.2024.116594] [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: 02/02/2024] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
Cholestatic liver disease (CLD) is a range of conditions caused by the accumulation of bile acids (BAs) or disruptions in bile flow, which can harm the liver and bile ducts. To investigate its pathogenesis and treatment, it is essential to establish and assess experimental models of cholestasis, which have significant clinical value. However, owing to the complex pathogenesis of cholestasis, a single modelling method can merely reflect one or a few pathological mechanisms, and each method has its adaptability and limitations. We summarize the existing experimental models of cholestasis, including animal models, gene-knockout models, cell models, and organoid models. We also describe the main types of cholestatic disease simulated clinically. This review provides an overview of targeted therapy used for treating cholestasis based on the current research status of cholestasis models. In addition, we discuss the respective advantages and disadvantages of different models of cholestasis to help establish experimental models that resemble clinical disease conditions. In sum, this review not only outlines the current research with cholestasis models but also projects prospects for clinical treatment, thereby bridging basic research and practical therapeutic applications.
Collapse
Affiliation(s)
- Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hefei Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiman Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
29
|
Kim HY, Charton C, Shim JH, Lim SY, Kim J, Lee S, Ohn JH, Kim BK, Heo CY. Patient-Derived Organoids Recapitulate Pathological Intrinsic and Phenotypic Features of Fibrous Dysplasia. Cells 2024; 13:729. [PMID: 38727265 PMCID: PMC11083396 DOI: 10.3390/cells13090729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Fibrous dysplasia (FD) is a rare bone disorder characterized by the replacement of normal bone with benign fibro-osseous tissue. Developments in our understanding of the pathophysiology and treatment options are impeded by the lack of suitable research models. In this study, we developed an in vitro organotypic model capable of recapitulating key intrinsic and phenotypic properties of FD. Initially, transcriptomic profiling of individual cells isolated from patient lesional tissues unveiled intralesional molecular and cellular heterogeneity. Leveraging these insights, we established patient-derived organoids (PDOs) using primary cells obtained from patient FD lesions. Evaluation of PDOs demonstrated preservation of fibrosis-associated constituent cell types and transcriptional signatures observed in FD lesions. Additionally, PDOs retained distinct constellations of genomic and metabolic alterations characteristic of FD. Histological evaluation further corroborated the fidelity of PDOs in recapitulating important phenotypic features of FD that underscore their pathophysiological relevance. Our findings represent meaningful progress in the field, as they open up the possibility for in vitro modeling of rare bone lesions in a three-dimensional context and may signify the first step towards creating a personalized platform for research and therapeutic studies.
Collapse
Affiliation(s)
- Ha-Young Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Clémentine Charton
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
| | - Jung Hee Shim
- Department of Research Administration Team, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - So Young Lim
- Department of Plastic and Reconstructive Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Republic of Korea
| | - Sejoon Lee
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam 13605, Republic of Korea; (C.C.); (J.K.); (S.L.)
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Jung Hun Ohn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Baek Kyu Kim
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| | - Chan Yeong Heo
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea;
- Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
- Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea;
| |
Collapse
|
30
|
Wang Z, Xing C, van der Laan LJW, Verstegen MMA, Spee B, Masereeuw R. Cholangiocyte organoids to study drug-induced injury. Stem Cell Res Ther 2024; 15:78. [PMID: 38475870 DOI: 10.1186/s13287-024-03692-6] [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: 10/05/2023] [Accepted: 03/07/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Drug induced bile duct injury is a frequently observed clinical problem leading to a wide range of pathological features. During the past decades, several agents have been identified with various postulated mechanisms of bile duct damage, however, mostly still poorly understood. METHODS Here, we investigated the mechanisms of chlorpromazine (CPZ) induced bile duct injury using advanced in vitro cholangiocyte cultures. Intrahepatic cholangiocyte organoids (ICOs) were driven into mature cholangiocyte like cells (CLCs), which were exposed to CPZ under cholestatic or non-cholestatic conditions through the addition of a bile acid cocktail. RESULTS CPZ caused loss of monolayer integrity by reducing expression levels of tight junction protein 1 (TJP1), E-cadherin 1 (CDH1) and lysyl oxidase homolog 2 (LOXL2). Loss of zonula occuludens-1 (ZO-1) and E-cadherin was confirmed by immunostaining after exposure to CPZ and rhodamine-123 leakage further confirmed disruption of the cholangiocyte barrier function. Furthermore, oxidative stress seemed to play a major role in the early damage response by CPZ. The drug also decreased expression of three main basolateral bile acid transporters, ABCC3 (ATP binding cassette subfamily C member 3), SLC51A/B (solute carrier family 51 subunit alpha/beta) and multidrug resistance transporter ABCB1 (ATP binding cassette subfamily B member 1), thereby contributing to bile acid accumulation. CPZ did not induce an inflammatory response by itself, but addition of TNFα revealed a synergistic effect. CONCLUSION These results show that ICOs present a model to identify toxic drugs affecting the bile ducts while providing mechanistic insights into hepatotoxicity.
Collapse
Affiliation(s)
- Zhenguo Wang
- Division of Pharmacology, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Chen Xing
- Division of Pharmacology, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, The Netherlands
| | - Bart Spee
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Faculty of Sciences, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
31
|
Zhang Y, Liu K, He H, Xiao H, Fang Z, Chen X, Li H. Innovative explorations: unveiling the potential of organoids for investigating environmental pollutant exposure. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:16256-16273. [PMID: 38342830 DOI: 10.1007/s11356-024-32256-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/25/2024] [Indexed: 02/13/2024]
Abstract
As the economy rapidly develops, chemicals are widely produced and used. This has exacerbated the problems associated with environmental pollution, raising the need for efficient toxicological evaluation techniques to investigate the toxic effects and mechanisms of toxicity of environmental pollutants. The progress in the techniques of cell culture in three dimensions has resulted in the creation of models that are more relevant in terms of biology and physiology. This enables researchers to study organ development, toxicology, and drug screening. Adult stem cells (ASCs) and induced pluripotent stem cells (iPSCs) can be obtained from various mammalian tissues, including cancerous and healthy tissues. Such stem cells exhibit a significant level of tissue memory and ability to self-assemble. When cultivated in 3D in vitro environments, the resulting organoids demonstrate a remarkable capacity to recapitulate the cellular composition and function of organs in vivo. Recently, many tumors' tissue-derived organoids have been widely used in research on tumor pathogenesis, drug development, precision medicine, and other fields, including those derived from colon cancer, cholangiocarcinoma, liver cancer, and gastric cancer. However, the application of organoid models for evaluating the toxicity of environmental pollutants is still in its infancy. This review introduces the characteristics of the toxicity responses of organoid models upon exposure to pollutants from the perspectives of organoid characteristics, tissue types, and their applications in toxicology; discusses the feasibility of using organoid models in evaluating the toxicity of pollutants; and provides a reference for future toxicological studies on environmental pollutants based on organoid models.
Collapse
Affiliation(s)
- Yuanhang Zhang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Kai Liu
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huan He
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China
| | - Hui Xiao
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Zhihong Fang
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Xianxian Chen
- School of Environment, Nanjing Normal University, Nanjing, 210023, China
| | - Huiming Li
- School of Environment, Nanjing Normal University, Nanjing, 210023, China.
- Jiangsu Province Engineering Research Center of Environmental Risk Prevention and Emergency Response Technology, Nanjing, 210023, China.
| |
Collapse
|
32
|
Deng S, Zhao X, Kou Z, Zhu Y, Zhang X, Chan HF. Effect of Valproic Acid on Promoting the Differentiation of Human Embryonic Stem Cells Into Cholangiocyte-Like Cells. Stem Cells Transl Med 2024; 13:166-176. [PMID: 37995322 PMCID: PMC10872666 DOI: 10.1093/stcltm/szad079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/06/2023] [Indexed: 11/25/2023] Open
Abstract
Cholangiocytes form a complex 3D network of bile ducts in the liver and contribute to liver function. The damage or destruction of cholangiocytes can lead to biliary diseases, and the shortage of cholangiocytes remains an obstacle for drug development targeting biliary diseases. Valproic acid (VPA) is a potent activator of Notch signaling pathway that is essential for cholangiocyte differentiation. Here, we report a VPA-based approach for cholangiocyte differentiation of human pluripotent stem cells. VPA activated Notch2 expression and upregulated HES-1, HEY-1, and Sox9 gene expression in hESC-derived hepatoblast. After 7 days treatment, VPA promoted successful differentiation of hepatoblast into cholangiocytes expressing cholangiocyte marker genes (AE2, AQP1, CFTR) and proteins (CK19 and CK7). In addition, the differentiated cholangiocytes formed bile duct-like structures after implantation into the spleen of NOD/SCID mice. Our results suggested that VPA can promote hESC differentiation to cholangiocyte-like cells. The induced cholangiocytes may serve as a potential cell source for both in vitro modeling and regenerative therapy of cholangiopathies. The findings can also support further development of small-molecule based differentiation protocols for cholangiocyte production.
Collapse
Affiliation(s)
- Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, People’s Republic of China
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Xiaoyu Zhao
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Ziyan Kou
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Yanlun Zhu
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, People’s Republic of China
| | - Xuerao Zhang
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Hon Fai Chan
- Key Laboratory for Regenerative Medicine of the Ministry of Education of China, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong SAR, People’s Republic of China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, Hong Kong SAR, People’s Republic of China
| |
Collapse
|
33
|
Rizwan M. Shaping Tomorrow's Liver Organoids: A Journey Toward Integrating Bile Ducts. Adv Biol (Weinh) 2024; 8:e2300450. [PMID: 37845008 DOI: 10.1002/adbi.202300450] [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: 08/25/2023] [Revised: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Liver tissue engineering has undergone remarkable developments since the late 20th century, transitioning from simple two-dimensional cultures to sophisticated three-dimensional organoid models for drug toxicity assessments. Stem cell innovations have enabled the creation of liver organoids for disease modelling and tissue engineering. However, a key limitation is the absence of functional bile ducts in these organoids, crucial for replicating bile-duct related diseases. Bile, synthesized by hepatocytes, plays a vital role in digesting fats and expelling lipid-soluble wastes, including drug byproducts. Diseases impeding bile flow are responsible for many liver transplants and can cause severe conditions such as liver cirrhosis, causing over 50,000 annual deaths in the US. Current liver organoids, while bile-producing, are devoid of bile ducts, limiting their efficacy in mimicking diseases related to bile flow. This article underscores the pressing need to incorporate bile ducts in engineered liver tissues, delves into the challenges faced in this effort, and highlights potential solutions through biomaterial and bioengineering techniques. Such advancements will offer researchers enhanced insights into bile duct disorders and pave the way for exploring innovative therapeutic strategies.
Collapse
Affiliation(s)
- Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Dr, 305 MM. Bldg., Houghton, MI, 49931, USA
| |
Collapse
|
34
|
Coulet F, Coton M, Iperi C, Belinger Podevin M, Coton E, Hymery N. Cytotoxic Effects of Major and Emerging Mycotoxins on HepaRG Cells and Transcriptomic Response after Exposure of Spheroids to Enniatins B and B1. Toxins (Basel) 2024; 16:54. [PMID: 38251270 PMCID: PMC10819306 DOI: 10.3390/toxins16010054] [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: 12/01/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Mycotoxins, produced by fungi, frequently occur at different stages in the food supply chain between pre- and postharvest. Globally produced cereal crops are known to be highly susceptible to contamination, thus constituting a major public health concern. Among the encountered mycotoxigenic fungi in cereals, Fusarium spp. are the most frequent and produce both regulated (i.e., T-2 toxin, deoxynivalenol -DON-, zearalenone -ZEA-) and emerging (i.e., enniatins -ENNs-, beauvericin -BEA-) mycotoxins. In this study, we investigated the in vitro cytotoxic effects of regulated and emerging fusariotoxins on HepaRG cells in 2D and 3D models using undifferentiated and differentiated cells. We also studied the impact of ENN B1 and ENN B exposure on gene expression of HepaRG spheroids. Gene expression profiling pinpointed the differentially expressed genes (DEGs) and overall similar pathways were involved in responses to mycotoxin exposure. Complement cascades, metabolism, steroid hormones, bile secretion, and cholesterol pathways were all negatively impacted by both ENNs. For cholesterol biosynthesis, 23/27 genes were significantly down-regulated and could be correlated to a 30% reduction in cholesterol levels. Our results show the impact of ENNs on the cholesterol biosynthesis pathway for the first time. This finding suggests a potential negative effect on human health due to the essential role this pathway plays.
Collapse
Affiliation(s)
- France Coulet
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, F-29280 Plouzané, France; (F.C.); (M.C.); (M.B.P.); (E.C.)
| | - Monika Coton
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, F-29280 Plouzané, France; (F.C.); (M.C.); (M.B.P.); (E.C.)
| | - Cristian Iperi
- Autoimmunité et Immunothérapies UMR 51227, Inserm, University Brest, Lymphocytes B, F-29200 Brest, France;
| | - Marine Belinger Podevin
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, F-29280 Plouzané, France; (F.C.); (M.C.); (M.B.P.); (E.C.)
| | - Emmanuel Coton
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, F-29280 Plouzané, France; (F.C.); (M.C.); (M.B.P.); (E.C.)
| | - Nolwenn Hymery
- Univ Brest, INRAE, Laboratoire Universitaire de Biodiversité et Ecologie Microbienne, F-29280 Plouzané, France; (F.C.); (M.C.); (M.B.P.); (E.C.)
| |
Collapse
|
35
|
Cui M, Wu W, Li Q, Qi G, Liu X, Bai J, Chen M, Li P, Sun X(S. Unlocking the Potential of Human-Induced Pluripotent Stem Cells: Cellular Responses and Secretome Profiles in Peptide Hydrogel 3D Culture. Cells 2024; 13:143. [PMID: 38247835 PMCID: PMC10814310 DOI: 10.3390/cells13020143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) have shown great potential for human health, but their growth and properties have been significantly limited by the traditional monolayer (2D) cell culture method for more than 15 years. Three-dimensional (3D) culture technology has demonstrated tremendous advantages over 2D. In particular, the 3D PGmatrix hiPSC derived from a peptide hydrogel offers a breakthrough pathway for the maintenance and expansion of physiologically relevant hiPSC 3D colonies (spheroids). In this study, the impact of 3D culture conditions in PGmatrix hiPSC on cell performance, integrity, and secretome profiles was determined across two commonly used hiPSC cell lines derived from fibroblast cells (hiPSC-F) and peripheral blood mononuclear cells (hiPSC-P) in the two most popular hiPSC culture media (mTeSR1 and essential eight (E8)). The 3D culture conditions varied in hydrogel strength, 3D embedded matrix, and 3D suspension matrix. The results showed that hiPSCs cultured in 3D PGmatrix hiPSC demonstrated the ability to maintain a consistently high cell viability that was above 95% across all the 3D conditions with cell expansion rates of 10-20-fold, depending on the 3D conditions and cell lines. The RT-qPCR analysis suggested that pluripotent gene markers are stable and not significantly affected by the cell lines or 3D PGmatrix conditions tested in this study. Mass spectrometry-based analysis of secretome from hiPSCs cultured in 3D PGmatrix hiPSC revealed a significantly higher quantity of unique proteins, including extracellular vesicle (EV)-related proteins and growth factors, compared to those in the 2D culture. Moreover, this is the first evidence to identify that hiPSCs in a medium with a rich supplement (i.e., mTeSR1) released more growth-regulating factors, while in a medium with fewer supplements (i.e., E8) hiPSCs secreted more survival growth factors and extracellular proteins. These findings offer insights into how these differences may impact hiPSC behavior, and they deepen our understanding of how hiPSCs respond to 3D culture conditions, aiding the optimization of hiPSC properties in translational biomedical research toward clinical applications.
Collapse
Affiliation(s)
- Muyun Cui
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA; (M.C.); (G.Q.)
| | - Wei Wu
- Department of Chemistry, Kansas State University, Manhattan, KS 66506, USA; (W.W.); (P.L.)
| | - Quan Li
- Carl and Melinda Helwig Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA;
| | - Guangyan Qi
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA; (M.C.); (G.Q.)
| | - Xuming Liu
- USDA-ARS and Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (X.L.); (M.C.)
| | - Jianfa Bai
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA;
| | - Mingshun Chen
- USDA-ARS and Department of Entomology, Kansas State University, Manhattan, KS 66506, USA; (X.L.); (M.C.)
| | - Ping Li
- Department of Chemistry, Kansas State University, Manhattan, KS 66506, USA; (W.W.); (P.L.)
| | - Xiuzhi (Susan) Sun
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA; (M.C.); (G.Q.)
- Carl and Melinda Helwig Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA;
| |
Collapse
|
36
|
Mun SJ, Hong YH, Shin Y, Lee J, Cho HS, Kim DS, Chung KS, Son MJ. Efficient and reproducible generation of human induced pluripotent stem cell-derived expandable liver organoids for disease modeling. Sci Rep 2023; 13:22935. [PMID: 38129682 PMCID: PMC10739970 DOI: 10.1038/s41598-023-50250-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/17/2023] [Indexed: 12/23/2023] Open
Abstract
Genetic liver disease modeling is difficult because it is challenging to access patient tissue samples and to develop practical and relevant model systems. Previously, we developed novel proliferative and functional liver organoids from pluripotent stem cells; however, the protocol requires improvement for standardization and reproducible mass production. Here, we improved the method such that it is suitable for scalable expansion and relatively homogenous production, resulting in an efficient and reproducible process. Moreover, three medium components critical for long-term expansion were defined. Detailed transcriptome analysis revealed that fibroblast growth factor signaling, the essential pathway for hepatocyte proliferation during liver regeneration, was mainly enriched in proliferative liver organoids. Short hairpin RNA-mediated knockdown of FGFR4 impaired the generation and proliferation of organoids. Finally, glycogen storage disease type Ia (GSD1a) patient-specific liver organoids were efficiently and reproducibly generated using the new protocol. They well maintained disease-specific phenotypes such as higher lipid and glycogen accumulation in the liver organoids and lactate secretion into the medium consistent with the main pathologic characteristics of patients with GSD1a. Therefore, our newly established liver organoid platform can provide scalable and practical personalized disease models and help to find new therapies for incurable liver diseases including genetic liver diseases.
Collapse
Affiliation(s)
- Seon Ju Mun
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Yeon-Hwa Hong
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Yongbo Shin
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jaeseo Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Hyun-Soo Cho
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
- Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Dae-Soo Kim
- Department of Bioinformatics, UST, 217 Gajungro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- Department of Digital Biotech Innovation Center, KRIBB, 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea
| | - Kyung-Sook Chung
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
- Biomedical Translational Research Center, KRIBB, 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-Ro, Yuseong-Gu, Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, Korea University of Science & Technology (UST), 217 Gajungro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
37
|
Salas-Silva S, Kim Y, Kim TH, Kim M, Seo D, Choi J, Factor VM, Seo HR, Song Y, Choi GS, Jung YK, Kim K, Lee KG, Jeong J, Shin JH, Choi D. Human chemically-derived hepatic progenitors (hCdHs) as a source of liver organoid generation: Application in regenerative medicine, disease modeling, and toxicology testing. Biomaterials 2023; 303:122360. [PMID: 38465578 DOI: 10.1016/j.biomaterials.2023.122360] [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: 05/09/2023] [Revised: 09/24/2023] [Accepted: 10/19/2023] [Indexed: 03/12/2024]
Abstract
BACKGROUND & AIMS Several types of human stem cells from embryonic (ESCs) and induced pluripotent (iPSCs) to adult tissue-specific stem cells are commonly used to generate 3D liver organoids for modeling tissue physiology and disease. We have recently established a protocol for direct conversion of primary human hepatocytes (hPHs) from healthy donor livers into bipotent progenitor cells (hCdHs). Here we extended this culture system to generate hCdH-derived liver organoids for diverse biomedical applications. METHODS To obtain hCdHs, hPHs were cultured in reprogramming medium containing A83-01 and CHIR99021 for 7 days. Liver organoids were established from hCdHs (hCdHOs) and human liver cells (hLOs) using the same donor livers for direct comparison, as well as from hiPSCs. Organoid properties were analyzed by standard in vitro assays. Molecular changes were determined by RT-qPCR and RNA-seq. Clinical relevance was evaluated by transplantation into FRG mice, modeling of alcohol-related liver disease (ARLD), and in vitro drug-toxicity tests. RESULTS hCdHs were clonally expanded as organoid cultures with low variability between starting hCdH lines. Similar to the hLOs, hCdHOs stably maintained stem cell phenotype based on accepted criteria. However, hCdHOs had an advantage over hLOs in terms of EpCAM expression, efficiency of organoid generation and capacity for directed hepatic differentiation as judged by molecular profiling, albumin secretion, glycogen accumulation, and CYP450 activities. Accordingly, FRG mice transplanted with hCdHOs survived longer than mice injected with hLOs. When exposed to ethanol, hCdHOs developed stronger ARLD phenotype than hLOs as evidenced by transcriptional profiling, lipid accumulation and mitochondrial dysfunction. In drug-induced injury assays in vitro, hCdHOs showed a similar or higher sensitivity response than hPHs. CONCLUSION hCdHOs provide a novel patient-specific stem cell-based platform for regenerative medicine, toxicology testing and modeling liver diseases.
Collapse
Affiliation(s)
- Soraya Salas-Silva
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Yohan Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea; Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany; Department of MetaBioHealth, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Tae Hun Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea
| | - Myounghoi Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea
| | - Daekwan Seo
- Office of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Researcj, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Jeonghoon Choi
- Office of Cellular, Tissue and Gene Therapies, Center for Biologics Evaluation and Researcj, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Valentina M Factor
- Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Haeng Ran Seo
- Advanced Biomedical Research Laboratory, Institute Pasteur Korea, 16, Daewangpangyo-ro 712-beon gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Yeonhwa Song
- Advanced Biomedical Research Laboratory, Institute Pasteur Korea, 16, Daewangpangyo-ro 712-beon gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Gyu Sung Choi
- Department of General Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yun Kyung Jung
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Kungsik Kim
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Kyeong Geun Lee
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea
| | - Jaemin Jeong
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, KIRAMS, Republic of Korea
| | - Ji Hyun Shin
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea.
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, 04763, Republic of Korea; Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, 04763, Republic of Korea; Department of HY-KIST Bio-convergence, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
38
|
Hu Y, Hu X, Luo J, Huang J, Sun Y, Li H, Qiao Y, Wu H, Li J, Zhou L, Zheng S. Liver organoid culture methods. Cell Biosci 2023; 13:197. [PMID: 37915043 PMCID: PMC10619312 DOI: 10.1186/s13578-023-01136-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/20/2023] [Indexed: 11/03/2023] Open
Abstract
Organoids, three-dimensional structures cultured in vitro, can recapitulate the microenvironment, complex architecture, and cellular functions of in vivo organs or tissues. In recent decades, liver organoids have been developed rapidly, and their applications in biomedicine, such as drug screening, disease modeling, and regenerative medicine, have been widely recognized. However, the lack of repeatability and consistency, including the lack of standardized culture conditions, has been a major obstacle to the development and clinical application of liver organoids. It is time-consuming for researchers to identify an appropriate medium component scheme, and the usage of some ingredients remains controversial. In this review, we summarized and compared different methods for liver organoid cultivation that have been published in recent years, focusing on controversial medium components and discussing their advantages and drawbacks. We aimed to provide an effective reference for the development and standardization of liver organoid cultivation.
Collapse
Affiliation(s)
- Yiqing Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Xiaoyi Hu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jia Luo
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jiacheng Huang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yaohan Sun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Haoyu Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Yinbiao Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
| | - Jianhui Li
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China
- The Organ Repair and Regeneration Medicine Institute of Hangzhou, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Zhejiang Shuren University School of Medicine, Hangzhou, 310015, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, 250117, China.
| |
Collapse
|
39
|
Ock SA, Kim SY, Ju WS, Kim YI, Wi HY, Lee P. Adipose Tissue-Derived Mesenchymal Stem Cells Extend the Lifespan and Enhance Liver Function in Hepatocyte Organoids. Int J Mol Sci 2023; 24:15429. [PMID: 37895114 PMCID: PMC10607770 DOI: 10.3390/ijms242015429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/29/2023] Open
Abstract
In this study, we generated hepatocyte organoids (HOs) using frozen-thawed primary hepatocytes (PHs) within a three-dimensional (3D) Matrigel dome culture in a porcine model. Previously studied hepatocyte organoid analogs, spheroids, or hepatocyte aggregates created using PHs in 3D culture systems have limitations in their in vitro lifespans. By co-culturing adipose tissue-derived mesenchymal stem cells (A-MSCs) with HOs within a 3D Matrigel dome culture, we achieved a 3.5-fold increase in the in vitro lifespan and enhanced liver function compared to a conventional two-dimensional (2D) monolayer culture, i.e., more than twice that of the HO group cultured alone, reaching up to 126 d. Although PHs were used to generate HOs, we identified markers associated with cholangiocyte organoids such as cytokeratin 19 and epithelial cellular adhesion molecule (EPCAM). Co-culturing A-MSCs with HOs increased the secretion of albumin and urea and glucose consumption compared to HOs cultured alone. After more than 100 d, we observed the upregulation of tumor protein P53 (TP53)-P21 and downregulation of EPCAM, albumin (ALB), and cytochrome P450 family 3 subfamily A member 29 (CYP3A29). Therefore, HOs with function and longevity improved through co-culturing with A-MSCs can be used to create large-scale human hepatotoxicity testing models and precise livestock nutrition assessment tools.
Collapse
Affiliation(s)
- Sun A Ock
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, 1500 Kongjwipatjwi-ro, Iseo-myeon, Wanju-gun 55365, Republic of Korea
| | | | | | | | | | | |
Collapse
|
40
|
Brun C, Allain C, Ferron PJ, Younoussa H, Colicchio B, Jeandidier E, M’Kacher R, Guguen-Guillouzo C, Bertile F. Extended lifespan and improved genome stability in HepaRG-derived cell lines through reprogramming by high-density stress. Proc Natl Acad Sci U S A 2023; 120:e2219298120. [PMID: 37639591 PMCID: PMC10483629 DOI: 10.1073/pnas.2219298120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 07/26/2023] [Indexed: 08/31/2023] Open
Abstract
The characteristics and fate of cancer cells partly depend on their environmental stiffness, i.e., the local mechanical cues they face. HepaRG progenitors are liver carcinoma cells exhibiting transdifferentiation properties; however, the underlying mechanisms remain unknown. To evaluate the impact of external physical forces mimicking the tumor microenvironment, we seeded them at very high density for 20 h, keeping the cells round and unanchored to the substrate. Applied without corticoids, spatial confinement due to very high density induced reprogramming of HepaRG cells into stable replicative stem-like cells after replating at normal density. Redifferentiation of these stem-like cells into cells very similar to the original HepaRG cells was then achieved using the same stress but in the presence of corticoids. This demonstrates that the cells retained the memory required to run the complete hepatic differentiation program, after bypassing the Hayflick limit twice. We show that physical stress improved chromosome quality and genomic stability, through greater efficiency of DNA repair and restoration of telomerase activity, thus enabling cells to escape progression to a more aggressive cancer state. We also show the primary importance of high-density seeding, possibly triggering compressive stress, in these processes, rather than that of cell roundness or intracellular tensional signals. The HepaRG-derived lines established here considerably extend the lifespan and availability of this surrogate cell system for mature human hepatocytes. External physical stress is a promising way to create a variety of cell lines, and it paves the way for the development of strategies to improve cancer prognosis.
Collapse
Affiliation(s)
- Charlotte Brun
- Université de Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien UMR 7178, StrasbourgF-67000, France
- Proteomics French Infrastructure, FR2048, ProFI, StrasbourgF-67000, France
| | - Coralie Allain
- Université de Rennes 1, INSERM U1241, Nutrition, Métabolismes et Cancer, RennesF-35033, France
| | - Pierre-Jean Ferron
- Université de Rennes 1, INSERM U1241, Nutrition, Métabolismes et Cancer, RennesF-35033, France
| | | | - Bruno Colicchio
- Université de Haute-Alsace, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, MulhouseF-68093, France
| | - Eric Jeandidier
- Groupe Hospitalier de la Région de Mulhouse et Sud Alsace Mulhouse, Service de génétique, MulhouseF-68070, France
| | - Radhia M’Kacher
- Cell Environment DNA Damage R&D, Genopole, EvryF-91058, France
| | | | - Fabrice Bertile
- Université de Strasbourg, CNRS, Institut Pluridisciplinaire Hubert Curien UMR 7178, StrasbourgF-67000, France
- Proteomics French Infrastructure, FR2048, ProFI, StrasbourgF-67000, France
| |
Collapse
|
41
|
Odendaal C, Jager EA, Martines ACMF, Vieira-Lara MA, Huijkman NCA, Kiyuna LA, Gerding A, Wolters JC, Heiner-Fokkema R, van Eunen K, Derks TGJ, Bakker BM. Personalised modelling of clinical heterogeneity between medium-chain acyl-CoA dehydrogenase patients. BMC Biol 2023; 21:184. [PMID: 37667308 PMCID: PMC10478272 DOI: 10.1186/s12915-023-01652-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 06/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Monogenetic inborn errors of metabolism cause a wide phenotypic heterogeneity that may even differ between family members carrying the same genetic variant. Computational modelling of metabolic networks may identify putative sources of this inter-patient heterogeneity. Here, we mainly focus on medium-chain acyl-CoA dehydrogenase deficiency (MCADD), the most common inborn error of the mitochondrial fatty acid oxidation (mFAO). It is an enigma why some MCADD patients-if untreated-are at risk to develop severe metabolic decompensations, whereas others remain asymptomatic throughout life. We hypothesised that an ability to maintain an increased free mitochondrial CoA (CoASH) and pathway flux might distinguish asymptomatic from symptomatic patients. RESULTS We built and experimentally validated, for the first time, a kinetic model of the human liver mFAO. Metabolites were partitioned according to their water solubility between the bulk aqueous matrix and the inner membrane. Enzymes are also either membrane-bound or in the matrix. This metabolite partitioning is a novel model attribute and improved predictions. MCADD substantially reduced pathway flux and CoASH, the latter due to the sequestration of CoA as medium-chain acyl-CoA esters. Analysis of urine from MCADD patients obtained during a metabolic decompensation showed an accumulation of medium- and short-chain acylcarnitines, just like the acyl-CoA pool in the MCADD model. The model suggested some rescues that increased flux and CoASH, notably increasing short-chain acyl-CoA dehydrogenase (SCAD) levels. Proteome analysis of MCADD patient-derived fibroblasts indeed revealed elevated levels of SCAD in a patient with a clinically asymptomatic state. This is a rescue for MCADD that has not been explored before. Personalised models based on these proteomics data confirmed an increased pathway flux and CoASH in the model of an asymptomatic patient compared to those of symptomatic MCADD patients. CONCLUSIONS We present a detailed, validated kinetic model of mFAO in human liver, with solubility-dependent metabolite partitioning. Personalised modelling of individual patients provides a novel explanation for phenotypic heterogeneity among MCADD patients. Further development of personalised metabolic models is a promising direction to improve individualised risk assessment, management and monitoring for inborn errors of metabolism.
Collapse
Affiliation(s)
- Christoff Odendaal
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Emmalie A Jager
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Anne-Claire M F Martines
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Marcel A Vieira-Lara
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Nicolette C A Huijkman
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Ligia A Kiyuna
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Rebecca Heiner-Fokkema
- Department of Laboratory Medicine, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Karen van Eunen
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Terry G J Derks
- Section of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.
| | - Barbara M Bakker
- Laboratory of Paediatrics, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands.
| |
Collapse
|
42
|
Zhang W, Kyritsi K, Isidan A, Park Y, Li P, Cross-Najafi AA, Lopez K, Kennedy L, Sato K, Glaser S, Francis H, Alpini G, Ekser B. Development of Scaffold-Free Three-Dimensional Cholangiocyte Organoids to Study the Progression of Primary Sclerosing Cholangitis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1156-1169. [PMID: 37263345 DOI: 10.1016/j.ajpath.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 06/03/2023]
Abstract
Organoids are novel in vitro models to study intercellular cross talk between the different types of cells in disease pathophysiology. To better understand the underlying mechanisms driving the progression of primary sclerosing cholangitis (PSC), scaffold-free multicellular three-dimensional cholangiocyte organoids (3D-CHOs) were developed using primary liver cells derived from normal subjects and patients with PSC. Human liver samples from healthy donors and patients with PSC were used to isolate primary cholangiocytes [epithelial cell adhesion molecule (EpCam)+/ cytokeratin-19+], liver endothelial cells (CD31+), and hepatic stellate cells (HSCs; CD31-/CD68-/desmin+/vitamin A+). 3D-CHOs were formed using cholangiocytes, HSCs, and liver endothelial cells, and kept viable for up to 1 month. Isolated primary cell lines and 3D-CHOs were further characterized by immunofluorescence, quantitative RT-PCR, and transmission electron microscopy. Transcription profiles for cholangiocytes (SOX9, CFTR, EpCAM, AE, SCT, and SCTR), fibrosis (ACTA2, COL1A1, DESMIN, and TGFβ1), angiogenesis (PECAM, VEGF, CDH5, and vWF), and inflammation (IL-6 and TNF-α) confirmed PSC phenotypes of 3D-CHOs. Because cholangiocytes develop a neuroendocrine phenotype and express neuromodulators, confocal immunofluorescence was used to demonstrate localization of the neurokinin-1 receptor within cytokeratin-19+ cholangiocytes and desmin+ HSCs. Moreover, 3D-CHOs from patients with PSC confirmed PSC phenotypes with up-regulated neurokinin-1 receptor, tachykinin precursor 1, and down-regulated membrane metalloendopeptidase. Scaffold-free multicellular 3D-CHOs showed superiority as an in vitro model in mimicking PSC in vivo phenotypes compared with two-dimensional cell culture, which can be used in PSC disease-related research.
Collapse
Affiliation(s)
- Wenjun Zhang
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana
| | - Konstantina Kyritsi
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Abdulkadir Isidan
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana
| | - Yujin Park
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana
| | - Ping Li
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana
| | | | - Kevin Lopez
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana
| | - Lindsey Kennedy
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Keisaku Sato
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, Texas
| | - Heather Francis
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Gianfranco Alpini
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Division of Research, Richard L. Roudebush VA Medical Center, Indianapolis, Indiana
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indianapolis, Indiana.
| |
Collapse
|
43
|
Zhang XS, Xie G, Ma H, Ding S, Wu YX, Fei Y, Cheng Q, Huang Y, Wang Y. Highly reproducible and cost-effective one-pot organoid differentiation using a novel platform based on PF-127 triggered spheroid assembly. Biofabrication 2023; 15:045014. [PMID: 37552975 DOI: 10.1088/1758-5090/acee21] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
Organoid technology offers sophisticatedin vitrohuman models for basic research and drug development. However, low batch-to-batch reproducibility and high cost due to laborious procedures and materials prevent organoid culture standardization for automation and high-throughput applications. Here, using a novel platform based on the findings that Pluronic F-127 (PF-127) could trigger highly uniform spheroid assembly through a mechanism different from plate coating, we develop a one-pot organoid differentiation strategy. Using our strategy, we successfully generate cortical, nephron, hepatic, and lung organoids with improved reproducibility compared to previous methods while reducing the original costs by 80%-95%. In addition, we adapt our platform to microfluidic chips allowing automated culture. We showcase that our platform can be applied to tissue-specific screening, such as drug toxicity and transfection reagents testing. Finally, we generateNEAT1knockout tissue-specific organoids and showNEAT1modulates multiple signaling pathways fine-tuning the differentiation of nephron and hepatic organoids and suppresses immune responses in cortical organoids. In summary, our strategy provides a powerful platform for advancing organoid research and studying human development and diseases.
Collapse
Affiliation(s)
- Xiao-Shan Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, People's Republic of China
| | - Gang Xie
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, People's Republic of China
| | - Honghao Ma
- Peking-Tsinghua Center for Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing, People's Republic of China
| | - Shuangjin Ding
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, People's Republic of China
| | - Yi-Xia Wu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, People's Republic of China
| | - Yuan Fei
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, People's Republic of China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, People's Republic of China
| | - Yanyi Huang
- Peking-Tsinghua Center for Life Sciences, Biomedical Pioneering Innovation Center, Peking University, Beijing, People's Republic of China
- College of Chemistry and Molecular Engineering, Beijing National Laboratory for Molecular Sciences, Peking University, Beijing, People's Republic of China
- Institute for Cell Analysis, Shenzhen Bay Laboratory, Shenzhen, People's Republic of China
| | - Yangming Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, People's Republic of China
| |
Collapse
|
44
|
Cardinale V, Lanthier N, Baptista PM, Carpino G, Carnevale G, Orlando G, Angelico R, Manzia TM, Schuppan D, Pinzani M, Alvaro D, Ciccocioppo R, Uygun BE. Cell transplantation-based regenerative medicine in liver diseases. Stem Cell Reports 2023; 18:1555-1572. [PMID: 37557073 PMCID: PMC10444572 DOI: 10.1016/j.stemcr.2023.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 08/11/2023] Open
Abstract
This review aims to evaluate the current preclinical state of liver bioengineering, the clinical context for liver cell therapies, the cell sources, the delivery routes, and the results of clinical trials for end-stage liver disease. Different clinical settings, such as inborn errors of metabolism, acute liver failure, chronic liver disease, liver cirrhosis, and acute-on-chronic liver failure, as well as multiple cellular sources were analyzed; namely, hepatocytes, hepatic progenitor cells, biliary tree stem/progenitor cells, mesenchymal stromal cells, and macrophages. The highly heterogeneous clinical scenario of liver disease and the availability of multiple cellular sources endowed with different biological properties make this a multidisciplinary translational research challenge. Data on each individual liver disease and more accurate endpoints are urgently needed, together with a characterization of the regenerative pathways leading to potential therapeutic benefit. Here, we critically review these topics and identify related research needs and perspectives in preclinical and clinical settings.
Collapse
Affiliation(s)
- Vincenzo Cardinale
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy.
| | - Nicolas Lanthier
- Service d'Hépato-gastroentérologie, Cliniques Universitaires Saint-Luc, Laboratory of Hepatogastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria 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
| | - Guido Carpino
- Department of Anatomical, Histological, Forensic Medicine and Orthopedic Sciences, Sapienza University of Rome, Italy
| | - Gianluca Carnevale
- Department of Surgery, Medicine, Dentistry, and Morphological Sciences with Interest in Transplant, Oncology, and Regenerative Medicine, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giuseppe Orlando
- Section of Transplantation, Department of Surgery, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Roberta Angelico
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Tommaso Maria Manzia
- Hepatobiliary Surgery and Transplant Unit, Department of Surgical Sciences, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Detlef Schuppan
- Institute of Translational Immunology, Research Center for Immune Therapy, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
| | - Domenico Alvaro
- Department of Translation and Precision Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy.
| | - Basak E Uygun
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Hospitals for Children, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
45
|
Szabó L, Seubert AC, Kretzschmar K. Modelling adult stem cells and their niche in health and disease with epithelial organoids. Semin Cell Dev Biol 2023; 144:20-30. [PMID: 36127261 DOI: 10.1016/j.semcdb.2022.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 10/14/2022]
Abstract
Adult stem cells are responsible for homoeostasis and regeneration of epithelial tissues. Stem cell function is regulated by both cell autonomous mechanisms as well as the niche. Deregulated stem cell function contributes to diseases such as cancer. Epithelial organoid cultures generated from tissue-resident adult stem cells have allowed unprecedented insights into the biology of epithelial tissues. The subsequent adaptation of organoid technology enabled the modelling of the communication of stem cells with their cellular and non-cellular niche as well as diseases. Starting from its first model described in 2009, the murine small intestinal organoid, we discuss here how epithelial organoid cultures have been become a prime in vitro research tool for cell and developmental biology, bioengineering, and biomedicine in the last decade.
Collapse
Affiliation(s)
- Lili Szabó
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany
| | - Anna C Seubert
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany
| | - Kai Kretzschmar
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, IZKF/MSNZ, Würzburg, Germany.
| |
Collapse
|
46
|
Han DW, Xu K, Jin ZL, Xu YN, Li YH, Wang L, Cao Q, Kim KP, Ryu D, Hong K, Kim NH. Customized liver organoids as an advanced in vitro modeling and drug discovery platform for non-alcoholic fatty liver diseases. Int J Biol Sci 2023; 19:3595-3613. [PMID: 37497008 PMCID: PMC10367556 DOI: 10.7150/ijbs.85145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its progressive form non-alcoholic steatohepatitis (NASH) have presented a major and common health concern worldwide due to their increasing prevalence and progressive development of severe pathological conditions such as cirrhosis and liver cancer. Although a large number of drug candidates for the treatment of NASH have entered clinical trial testing, all have not been released to market due to their limited efficacy, and there remains no approved treatment for NASH available to this day. Recently, organoid technology that produces 3D multicellular aggregates with a liver tissue-like cytoarchitecture and improved functionality has been suggested as a novel platform for modeling the human-specific complex pathophysiology of NAFLD and NASH. In this review, we describe the cellular crosstalk between each cellular compartment in the liver during the pathogenesis of NAFLD and NASH. We also summarize the current state of liver organoid technology, describing the cellular diversity that could be recapitulated in liver organoids and proposing a future direction for liver organoid technology as an in vitro platform for disease modeling and drug discovery for NAFLD and NASH.
Collapse
Affiliation(s)
- Dong Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - KangHe Xu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Zhe-Long Jin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
| | - Lin Wang
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Qilong Cao
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
| | - Kee-Pyo Kim
- Department of Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - DongHee Ryu
- Department of Surgery, College of Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, The institute of advanced regenerative science, Konkuk University, Seoul, Republic of Korea
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
- International Healthcare Innovation Institute (Jiangmen), Jianghai, Jiangmen, Guangdong Province, China
- Research and Development, Qingdao Haier Biotech Co. Ltd, Qingdao, China
- Guangdong ORGANOID Biotechnology Co. Ltd, Jiangmen, China
| |
Collapse
|
47
|
Cuevas-Ocaña S, Yang JY, Aushev M, Schlossmacher G, Bear CE, Hannan NRF, Perkins ND, Rossant J, Wong AP, Gray MA. A Cell-Based Optimised Approach for Rapid and Efficient Gene Editing of Human Pluripotent Stem Cells. Int J Mol Sci 2023; 24:10266. [PMID: 37373413 PMCID: PMC10299534 DOI: 10.3390/ijms241210266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Introducing or correcting disease-causing mutations through genome editing in human pluripotent stem cells (hPSCs) followed by tissue-specific differentiation provide sustainable models of multiorgan diseases, such as cystic fibrosis (CF). However, low editing efficiency resulting in extended cell culture periods and the use of specialised equipment for fluorescence activated cell sorting (FACS) make hPSC genome editing still challenging. We aimed to investigate whether a combination of cell cycle synchronisation, single-stranded oligodeoxyribonucleotides, transient selection, manual clonal isolation, and rapid screening can improve the generation of correctly modified hPSCs. Here, we introduced the most common CF mutation, ΔF508, into the CFTR gene, using TALENs into hPSCs, and corrected the W1282X mutation using CRISPR-Cas9, in human-induced PSCs. This relatively simple method achieved up to 10% efficiency without the need for FACS, generating heterozygous and homozygous gene edited hPSCs within 3-6 weeks in order to understand genetic determinants of disease and precision medicine.
Collapse
Affiliation(s)
- Sara Cuevas-Ocaña
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Jin Ye Yang
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Magomet Aushev
- Wellcome Trust Centre for Mitochondrial Research, Institute of Genetic Medicine, Biomedicine West Wing, Centre for Life, Times Square, Newcastle upon Tyne NE1 3BZ, UK;
| | - George Schlossmacher
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| | - Christine E. Bear
- Programme in Molecular Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada;
| | - Nicholas R. F. Hannan
- Biodiscovery Institute, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Neil D. Perkins
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| | - Janet Rossant
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Amy P. Wong
- Programme in Developmental & Stem Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; (J.Y.Y.); (J.R.); (A.P.W.)
| | - Michael A. Gray
- Biosciences Institute, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK; (G.S.); (N.D.P.); (M.A.G.)
| |
Collapse
|
48
|
Yang S, Hu H, Kung H, Zou R, Dai Y, Hu Y, Wang T, Lv T, Yu J, Li F. Organoids: The current status and biomedical applications. MedComm (Beijing) 2023; 4:e274. [PMID: 37215622 PMCID: PMC10192887 DOI: 10.1002/mco2.274] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Organoids are three-dimensional (3D) miniaturized versions of organs or tissues that are derived from cells with stem potential and can self-organize and differentiate into 3D cell masses, recapitulating the morphology and functions of their in vivo counterparts. Organoid culture is an emerging 3D culture technology, and organoids derived from various organs and tissues, such as the brain, lung, heart, liver, and kidney, have been generated. Compared with traditional bidimensional culture, organoid culture systems have the unique advantage of conserving parental gene expression and mutation characteristics, as well as long-term maintenance of the function and biological characteristics of the parental cells in vitro. All these features of organoids open up new opportunities for drug discovery, large-scale drug screening, and precision medicine. Another major application of organoids is disease modeling, and especially various hereditary diseases that are difficult to model in vitro have been modeled with organoids by combining genome editing technologies. Herein, we introduce the development and current advances in the organoid technology field. We focus on the applications of organoids in basic biology and clinical research, and also highlight their limitations and future perspectives. We hope that this review can provide a valuable reference for the developments and applications of organoids.
Collapse
Affiliation(s)
- Siqi Yang
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Haijie Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Hengchung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Ruiqi Zou
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yushi Dai
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Yafei Hu
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Tiantian Wang
- Key Laboratory of Rehabilitation Medicine in Sichuan ProvinceWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tianrun Lv
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| | - Jun Yu
- Departments of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Departments of OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Fuyu Li
- Division of Biliary Tract SurgeryDepartment of General SurgeryWest China HospitalSichuan UniversityChengduSichuan ProvinceChina
| |
Collapse
|
49
|
Xu Z, Yang J, Xin X, Liu C, Li L, Mei X, Li M. Merits and challenges of iPSC-derived organoids for clinical applications. Front Cell Dev Biol 2023; 11:1188905. [PMID: 37305682 PMCID: PMC10250752 DOI: 10.3389/fcell.2023.1188905] [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: 03/18/2023] [Accepted: 04/18/2023] [Indexed: 06/13/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have entered an unprecedented state of development since they were first generated. They have played a critical role in disease modeling, drug discovery, and cell replacement therapy, and have contributed to the evolution of disciplines such as cell biology, pathophysiology of diseases, and regenerative medicine. Organoids, the stem cell-derived 3D culture systems that mimic the structure and function of organs in vitro, have been widely used in developmental research, disease modeling, and drug screening. Recent advances in combining iPSCs with 3D organoids are facilitating further applications of iPSCs in disease research. Organoids derived from embryonic stem cells, iPSCs, and multi-tissue stem/progenitor cells can replicate the processes of developmental differentiation, homeostatic self-renewal, and regeneration due to tissue damage, offering the potential to unravel the regulatory mechanisms of development and regeneration, and elucidate the pathophysiological processes involved in disease mechanisms. Herein, we have summarized the latest research on the production scheme of organ-specific iPSC-derived organoids, the contribution of these organoids in the treatment of various organ-related diseases, in particular their contribution to COVID-19 treatment, and have discussed the unresolved challenges and shortcomings of these models.
Collapse
Affiliation(s)
- Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
- Department of Clinical Laboratory, Lequn Branch, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiaxu Yang
- Department of Neonatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xianyi Xin
- Department of Pediatric Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Chengrun Liu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| | - Xianglin Mei
- Department of pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, Jilin, China
| |
Collapse
|
50
|
Giuli L, Santopaolo F, Pallozzi M, Pellegrino A, Coppola G, Gasbarrini A, Ponziani FR. Cellular therapies in liver and pancreatic diseases. Dig Liver Dis 2023; 55:563-579. [PMID: 36543708 DOI: 10.1016/j.dld.2022.11.013] [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: 08/11/2022] [Revised: 10/21/2022] [Accepted: 11/22/2022] [Indexed: 04/29/2023]
Abstract
Over the past two decades, developments in regenerative medicine in gastroenterology have been greatly enhanced by the application of stem cells, which can self-replicate and differentiate into any somatic cell. The discovery of induced pluripotent stem cells has opened remarkable perspectives on tissue regeneration, including their use as a bridge to transplantation or as supportive therapy in patients with organ failure. The improvements in DNA manipulation and gene editing strategies have also allowed to clarify the physiopathology and to correct the phenotype of several monogenic diseases, both in vivo and in vitro. Further progress has been made with the development of three-dimensional cultures, known as organoids, which have demonstrated morphological and functional complexity comparable to that of a miniature organ. Hence, owing to its protean applications and potential benefits, cell and organoid transplantation has become a hot topic for the management of gastrointestinal diseases. In this review, we describe current knowledge on cell therapies in hepatology and pancreatology, providing insight into their future applications in regenerative medicine.
Collapse
Affiliation(s)
- Lucia Giuli
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Pallozzi
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Pellegrino
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gaetano Coppola
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology, Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|