1
|
Zhang B, Fan Z, Liu X, Wu Y, Cheng L, Wang L, Liu H. Bisphenol AF induces lipid metabolism disorders, oxidative stress and upregulation of heat shock protein 70 in zebrafish. Comp Biochem Physiol C Toxicol Pharmacol 2025; 293:110164. [PMID: 40020955 DOI: 10.1016/j.cbpc.2025.110164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Bisphenol AF (BPAF) is a widespread endocrine disruptor in the environment, and the use of BPAF has been strongly associated with the development of several diseases. In this study, we investigated the effects of BPAF on growth, development, oxidative stress and lipid metabolism in zebrafish. We chose the concentrations based on the measured LC50 at 96 h post-fertilization (96 hpf), and the zebrafish embryos were exposed to three different concentrations (0.125, 0.5 and 2 μmol/L). The findings indicated that BPAF exposure in zebrafish leaded to alterations in heart rate, body length and hatching rate, as well as an accumulation of red blood cells in the heart. Additionally, BPAF exposure resulted in increased levels of neutrophils, reactive oxygen species (ROS) and malondialdehyde (MDA), and decreased activity of antioxidant enzymes (superoxide dismutase (SOD) and catalase (CAT)), thus disturbing the balance between oxidative and antioxidative systems. BPAF promoted fatty acid catabolism and inhibited fatty acid synthesis, ultimately leading to a reduction in fatty acid content. Mechanistically, RNA-seq analysis and RT-qPCR revealed a significant upregulation of heat shock protein 70 (hsp70) after BPAF exposure. Inhibition of hsp70 with VER-155008 ameliorated BPAF-induced oxidative stress. These data provided a novel approach to investigate BPAF-induced oxidative stress and suggested that regulation of hsp70 is a crucial target for alleviating this process.
Collapse
Affiliation(s)
- Bingya Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Zhonghua Fan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Xuan Liu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Yuanyuan Wu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Lin Cheng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Li Wang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China.
| | - Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China.
| |
Collapse
|
2
|
Paranthaman S, Uthaiah CA, Md S, Alkreathy HM. Comprehensive strategies for constructing efficient CRISPR/Cas based cancer therapy: Target gene selection, sgRNA optimization, delivery methods and evaluation. Adv Colloid Interface Sci 2025; 341:103497. [PMID: 40157335 DOI: 10.1016/j.cis.2025.103497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/17/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Cancer is a complicated disease that results from the interplay between specific changes in cellular genetics and diverse microenvironments. The application of high-performance and customizable clustered regularly interspaced palindromic repeats/associated protein (CRISPR/Cas) nuclease systems has significantly enhanced genome editing for accurate cancer modeling and facilitated simultaneous genetic modification for cancer therapy and mutation identification. Achieving an effective CRISPR/Cas platform for cancer treatment depends on the identification, selection, and optimization of specific mutated genes in targeted cancer tissues. However, overcoming the off-target effects, specificity, and immunogenicity are additional challenges that must be addressed while developing a gene editing system for cancer therapy. From this perspective, we briefly covered the pipeline of CRISPR/Cas cancer therapy, identified target genes to optimize gRNAs and sgRNAs, and explored alternative delivery modalities, including viral, non-viral, and extracellular vesicles. In addition, the list of patents and current clinical trials related to this unique cancer therapy method is discussed. In summary, we have discussed comprehensive start-to-end pipeline strategies for CRISPR/Cas development to advance the precision, effectiveness, and safety of clinical applications for cancer therapy.
Collapse
Affiliation(s)
- Sathishbabu Paranthaman
- Department of Cell Biology and Molecular Genetics, Sri Devaraj Urs Medical College, Sri Devaraj Urs Academy of Higher Education and Research, Tamaka, Kolar 563103, Karnataka, India.
| | - Chinnappa A Uthaiah
- Genetics Laboratory, Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raipur, Chhattisgarh 492099, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Mohammed Alkreathy
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Chao L, Liang Y, Hu X. Recent advances in field effect transistor biosensors for drug screening applications. Analyst 2025. [PMID: 40566960 DOI: 10.1039/d5an00206k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Clinical drug screening is a critical component in disease treatment and in elucidating the impact of drug monitoring on cellular-level biochemical reactions. Field effect transistor (FET) based biosensors represent a novel technology in biomedical research, playing an essential role in clinical drug screening due to their high specificity, sensitivity, label-free detection, real-time monitoring, cost-effectiveness, and integration capabilities. This review provides a detailed description of the structural mechanisms of various types of FET biosensors. Furthermore, it introduces the classification, structural mechanisms, and immobilization methods of biological probes. Additionally, the methodologies and effects of utilizing the FET biosensor platform for screening cancer drugs, anti-infective drugs, inhibitor drugs, anti-histamine drugs, psychotropic drugs, cardiovascular drugs, and other drug categories are highlighted. Finally, the challenges encountered by FET biosensors in drug screening applications are discussed, and advanced strategies for future clinical drug screening are proposed.
Collapse
Affiliation(s)
- Lemeng Chao
- Department of Medical Engineering, Inner Mongolia People's Hospital, Hohhot 010017, China.
| | - Ying Liang
- School of Computer Information, Inner Mongolia Medical University, Hohhot 010110, China
| | - Xiao Hu
- School of Computer Information, Inner Mongolia Medical University, Hohhot 010110, China
| |
Collapse
|
4
|
Chen Y, Yu K, Jiang Z, Yang G. CRISPR-based genetically modified scaffold-free biomaterials for tissue engineering and regenerative medicine. Biomater Sci 2025; 13:3149-3175. [PMID: 40326747 DOI: 10.1039/d5bm00194c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
CRISPR-based genetically modified scaffold-free biomaterials, including extracellular vehicles, cell sheets, cell aggregates, organoids and organs, have attracted significant attention in the fields of regenerative medicine and tissue engineering in recent years. With a wide range of applications in gene therapy, modeling disease, tissue regeneration, organ xenotransplantation, modeling organogenesis as well as gene and drug screening, they are at a critical juncture from clinical trials to therapeutic applications. Xenografts have already been tested on non-human primates and humans. However, we have to admit that a series of obstacles still need to be addressed, such as immune response, viral infection, off-target effects, difficulty in mass production, and ethical issues. Therefore, future research should pay more attention to improving their safety, accuracy of gene editing, flexibility of production, and ethical rationality. This review summarizes various types of CRISPR-based genetically modified scaffold-free biomaterials, including their preparation procedures, applications, and possible improvements.
Collapse
Affiliation(s)
- Yunxuan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Ke Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
5
|
Liu Y, Gilchrist AE, Johansson PK, Guan Y, Deras JD, Liu Y, Ceva S, Huang MS, Navarro RS, Enejder A, Peltz G, Heilshorn SC. Engineered Hydrogels for Organoid Models of Human Nonalcoholic Fatty Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e17332. [PMID: 40364726 PMCID: PMC12165117 DOI: 10.1002/advs.202417332] [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] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/22/2025] [Indexed: 05/15/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by increased lipid accumulation and excessive deposition of extracellular matrix (ECM) that results in tissue stiffening. The potential interplay between matrix stiffness and hepatocyte lipid accumulation during NAFLD has not been established. Here, an in vitro NAFLD model is developed using chemically defined, engineered hydrogels and human induced pluripotent stem cell-derived hepatic organoids (HOs). Specifically, dynamic covalent chemistry crosslinking, along with transient small molecule competitors, are used to create dynamic stiffening hydrogels that enable the reproducible culture of HOs. Within matrices that mimic the stiffness of healthy to diseased tissue (≈1-6 kPa), lipid droplet accumulation in HOs is triggered by exposure to an NAFLD-associated free fatty acid. These NAFLD model suggests that higher stiffness microenvironments result in increased hepatic lipid droplet accumulation, increased expression of fibrosis markers, and increased metabolic dysregulation. By targeting the ROCK mechanosignaling pathway, the synergy between matrix stiffness and lipid droplet accumulation is disrupted. The in vitro model of NAFLD has the potential to understand the role of mechanosignaling in disease progression and identify new pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Aidan E. Gilchrist
- Department of Biomedical EngineeringUniversity of CaliforniaDavisCA95616USA
| | - Patrik K. Johansson
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Yuan Guan
- Department of AnesthesiologyPain and Perioperative MedicineStanford University School of MedicineStanfordCA94305USA
| | - Jaydon D. Deras
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Yu‐Chung Liu
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | - Sofia Ceva
- Department of BiologyStanford UniversityStanfordCA94305USA
| | - Michelle S. Huang
- Department of Chemical EngineeringStanford UniversityStanfordCA94305USA
| | - Renato S. Navarro
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Annika Enejder
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Gary Peltz
- Department of AnesthesiologyPain and Perioperative MedicineStanford University School of MedicineStanfordCA94305USA
| | - Sarah C. Heilshorn
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| |
Collapse
|
6
|
Zhuang C, Cui F, Chen J, He D, Sun T, Wang P. Rbm39 ameliorates metabolic dysfunction-associated steatotic liver disease by regulating Apob and Fabp4. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167815. [PMID: 40147697 DOI: 10.1016/j.bbadis.2025.167815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/12/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
Excessive hepatic lipid accumulation is the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD), yet its underlying mechanisms still not fully understood. In this study, we identified RNA binding motif protein 39 (Rbm39) as a key modulator of hepatic lipid homeostasis during MASLD progression. To establish in vivo MASLD model, mice were fed either a high-fat diet (HFD) or a Gubra-Amylin NASH (GAN) diet. We employed adeno-associated virus to manipulate Rbm39 expression levels to assess its role in MASLD. Transcriptome analysis was conducted to pinpoint the genes targeted by Rbm39. Western blot, RT-PCR, dual-luciferase reporter gene assays, and alternative splicing analysis were utilized to delve into the molecular mechanisms. Our results showed that Rbm39 expression was notably decreased in the livers of MASLD mice. Knockdown of hepatic Rbm39 aggravated HFD-induced hepatic steatosis and GAN diet-induced MASH, along with a notable decrease in serum lipid levels. Conversely, overexpression of Rbm39 attenuated MASLD development and progression. RNA sequencing data analysis indicated that Rbm39 regulated the expression of apolipoprotein B (Apob) and fatty acid-binding protein 4 (Fabp4), both of which are crucial for lipid transport. Mechanistically, Rbm39 enhanced the transcription of Apob by upregulating hepatocyte nuclear factor 4α (Hnf4α), while it suppressed Fabp4 transcription by regulating alternative splicing of hypoxia inducible factor-1α (Hif-1α). These findings highlight the pivotal role of Rbm39 in maintaining hepatic lipid homeostasis and suggest its potential as a therapeutic target for MASLD.
Collapse
Affiliation(s)
- Chunbo Zhuang
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Fangfang Cui
- Department of Gastroenterology, Kaifeng People's Hospital, Kaifeng, Henan 475000, PR China
| | - Jin Chen
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Dezhi He
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Ting Sun
- Department of Clinical Laboratory, Key Clinical Laboratory of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Pei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China.
| |
Collapse
|
7
|
Schmidt-Arras D. Should I Stay or Should I Go - ERK as Control Lever in Hepatocyte Expansion. Cell Mol Gastroenterol Hepatol 2025:101532. [PMID: 40456274 DOI: 10.1016/j.jcmgh.2025.101532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 06/22/2025]
Affiliation(s)
- Dirk Schmidt-Arras
- Department of Biosciences and Medical Biology, Paris-Lodron-University, Salzburg, Austria; Center for Tumor Biology and Immunology, Paris-Lodron-University, Salzburg, Austria; Cancer Cluster Salzburg, Salzburg, Austria.
| |
Collapse
|
8
|
Panchuk I, Kovalskaia V, Balinova N, Ryzhkova O, Smirnikhina S. The Optimization of a Protocol for the Directed Differentiation of Induced Pluripotent Stem Cells into Liver Progenitor Cells and the Delivery of Transgenes. BIOLOGY 2025; 14:586. [PMID: 40563838 DOI: 10.3390/biology14060586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/23/2025] [Accepted: 05/12/2025] [Indexed: 06/28/2025]
Abstract
The liver plays a pivotal role in metabolism, detoxification, and protein synthesis and comprises several cell types, including hepatocytes and cholangiocytes. Primary human hepatocytes in 2D cultures rapidly dedifferentiate and lose their function, making their use as a reliable cell model challenging. Therefore, developing robust three-dimensional cell culture models is crucial, especially for diseases lacking reliable animal models. The aim of this study was to optimize a protocol for the directed differentiation of induced pluripotent stem cells into liver progenitor cells, achieving the high-level expression of specific markers. As a result, we established a 2D culture of liver progenitor cells capable of differentiating into three cell types: a 3D organoid culture containing hepatocyte- and cholangiocyte-like cells and a 2D cell culture comprising stellate-like cells. To evaluate gene delivery efficiency, liver progenitor cells were transduced with various rAAV serotypes carrying an eGFP reporter cassette at different multiplicities of infection (MOIs). Our results revealed that rAAV serotype 2/2 at MOI of 100,000 achieved the highest transduction efficiency of 93.6%, while electroporation demonstrated a plasmid delivery efficiency of 54.3%. These findings suggest that liver progenitor cells are a promising tissue-like cell model for regenerative medicine and demonstrate high amenability to genetic manipulation, underscoring their potential in gene therapy and genome editing studies.
Collapse
Affiliation(s)
- Irina Panchuk
- Research Centre for Medical Genetics, Moskvorechye Str., 1, 115478 Moscow, Russia
| | - Valeriia Kovalskaia
- Research Centre for Medical Genetics, Moskvorechye Str., 1, 115478 Moscow, Russia
| | - Natalia Balinova
- Research Centre for Medical Genetics, Moskvorechye Str., 1, 115478 Moscow, Russia
| | - Oxana Ryzhkova
- Research Centre for Medical Genetics, Moskvorechye Str., 1, 115478 Moscow, Russia
| | - Svetlana Smirnikhina
- Research Centre for Medical Genetics, Moskvorechye Str., 1, 115478 Moscow, Russia
| |
Collapse
|
9
|
Chen F, Wang Z, Yao H, Liu Q, Gan Y, Xu S, Bao H, Jin Y, Hu YP, Gao J, Song S, Wang MJ. Large-scale manufacturing of human gallbladder epithelial cell products and derived hepatocytes via a chemically defined approach. Trends Biotechnol 2025:S0167-7799(25)00136-2. [PMID: 40399214 DOI: 10.1016/j.tibtech.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/26/2025] [Accepted: 04/08/2025] [Indexed: 05/23/2025]
Abstract
Manufacturing sufficient quantities of high-quality hepatocytes holds significant promise for the treatment of liver diseases and drug screening. Here, we developed a chemically defined, animal-free method for the large-scale production of human gallbladder epithelial cells (hGBECs) under good manufacturing practice conditions, enabling their clinical application. The cell products were characterized for growth ability, phenotype, freeze-thaw viability, genetic stability, biological contamination, tumorigenicity, and acute toxicity to ensure quality control and biological safety. We also provide a protocol for generating functional hepatocytes from hGBECs. The derived hepatocytes demonstrated typical liver functions, including albumin secretion, urea production, and drug metabolism. In addition, these cells were used in drug toxicity testing. We conducted further functional experiments on Cu2+ transport and alcohol metabolism. Transplantation of these cells in vivo was able to rescue mice from liver failure. This large-scale, convenient strategy for manufacturing hGBECs serves as a biobank for clinical applications and provides a valuable model for studying liver diseases.
Collapse
Affiliation(s)
- Fei Chen
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Zijun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Hao Yao
- Department of Hematology, People's Liberation Army The General Hospital of Western Theater Command, Chengdu, China
| | - Qinggui Liu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yuwen Gan
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Shoujia Xu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Haili Bao
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yiqiang Jin
- Shanghai Baixian Biotechnology Co., Ltd., Shanghai, China; Shanghai Angecon Biotechnology Co., Ltd., Shanghai, China
| | - Yi-Ping Hu
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Junling Gao
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| | - Shaohua Song
- Department of General Surgery, Ruijin Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China; Transplantation Center, Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Min-Jun Wang
- Department of Cell Biology, Second Military Medical University (Naval Medical University), Shanghai, China.
| |
Collapse
|
10
|
Zhu L, Liu S, Wang Z, Yang Y, Han P, Tong W, Zhao T, Wang L, Cui T, Yang L, Zhang Y. Modeling hepatic steatosis with human adult stem cell-derived liver organoids. iScience 2025; 28:112344. [PMID: 40276762 PMCID: PMC12019286 DOI: 10.1016/j.isci.2025.112344] [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: 05/18/2024] [Revised: 11/05/2024] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) remains the most common chronic liver disease worldwide, and appropriate in vitro models are of great significance for investigating pathogenesis and drug screening of MASLD. In this study, human expandable cholangiocyte organoids were derived from adult stem cells of normal liver tissue. After differentiation, liver organoids (LOs) exhibited the functional characteristics and genomic features of mature hepatocytes. To induce steatosis, LOs were incubated with a gradient concentration oleic acid, and it was found that the model could recapitulate the development of lipid accumulation and inflammation. In addition, the drug sensitivity of the hepatic steatosis model was further verified through anti-steatosis drug testing. In summary, LOs have great potential for disease modeling, and the results indicate that the hepatic steatosis model may serve as a useful tool for exploring the molecular mechanisms and drug screening of MASLD.
Collapse
Affiliation(s)
- Liuyang Zhu
- First Central Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Sen Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ze Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Yueyue Yang
- College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Pinsheng Han
- Nankai University of Medicine College, Tianjin 300071, China
| | - Wen Tong
- First Central Clinical College of Tianjin Medical University, Tianjin 300070, China
| | - Tianyu Zhao
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Libo Wang
- State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin 300000, China
| | - Tao Cui
- Tianhui Biotechnology Co., Ltd., Hefei 230000, China
| | - Long Yang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, Tianjin 300192, China
| |
Collapse
|
11
|
Zhang X, Xiong W, Gao F, Yu Z, Ren F, Lei XG. Impacts and mechanism of liver-specific knockout of selenoprotein I on hepatic phospholipid metabolism, selenogenome expression, redox status, and resistance to CCl 4 toxicity. Free Radic Biol Med 2025; 235:426-442. [PMID: 40345504 DOI: 10.1016/j.freeradbiomed.2025.05.387] [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: 03/13/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/11/2025]
Abstract
Selenoprotein I (SELENOI) was known initially as ethanolamine phosphotransferase 1 (EPT1) and later as a selenoprotein. Because global knockout of Selenoi in mice is embryonically lethal, we generated liver-specific Selenoi knockout (cKO) mice to reveal functions and mechanism of SELENOI in the liver. Compared with control mice, cKO mice (8 weeks old) had no differences in body weight, glucose metabolism, energy expenditure, overall health status, or liver histology. However, these mice had lower (P < 0.05) mRNA levels of 13 selenoprotein genes, contents of Se, GSH, and T-AOC (12-40%), and activities of antioxidant enzymes (17-51%), but higher (P < 0.05) mRNA levels of oxidative stress-related genes (34%-46%) in the liver than the control mice. They had a higher (P < 0.05) ratio of phosphatidylcholine (PC) to phosphatidylethanolamine (PE) due to increases of the former and decreases of the latter, altered PE and PC constituents such as n-6/n-3 PUFA ratios, and elevated mRNA levels (95%-2-fold, P < 0.05) of lipolysis genes, compared with the control mice. The knockout attenuated hepatic injury and fibrosis induced by 14 intraperitoneal injections of CCl4 (0.5 mL/kg). The protection was associated with adaptive cytoprotective mechanisms induced by the overall decline of redox status mediated by SELENOI as a selenoprotein and activations of PPAR signaling, fatty acid desaturase 2 (FADS2), glutathione S-transferase, and lipid peroxide hydrolysis through modulating biosynthesis and(or) constituents of PC, PE, and n-6/n-3 PUFAs mediated by SELENOI as EPT1. Inhibition of FADS2 in CCl4-treated cKO hepatocytes partially removed the protection by the knockout. In conclusion, hepatic SELENOI expression was not essential for survival, but served as a multifunctional regulator of hepatic selenogenome expression, Se metabolism, redox status, biosyntheses and profiles of PC and PE, and resistance to CCI4.
Collapse
Affiliation(s)
- Xu Zhang
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Wei Xiong
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China; Food Laboratory of Zhongyuan, Luohe, Henan, 462300, China.
| | - Fei Gao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhengquan Yu
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China; Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Fazheng Ren
- Department of Nutrition and Health, China Agricultural University, Beijing, 100193, China
| | - Xin Gen Lei
- Department of Animal Science, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
12
|
He J, Wu X, Qiao J, Xie X, Wang Y, Zhang H, Zhang W. Tubular specific glutathione peroxidase 3 deletion exacerbates kidney damage in IRI-AKI mice. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167895. [PMID: 40345457 DOI: 10.1016/j.bbadis.2025.167895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Ischemia-reperfusion injury stands as a primary instigator of acute kidney injury (AKI), prominently driven by oxidative stress. Among the critical antioxidant defenses is glutathione peroxidase 3 (GPX3), an enzyme generated by renal tubular epithelial cells. Our prior investigations have unveiled a substantial downregulation of GPX3 in renal tissues gleaned from AKI patients and murine models. This study aims to investigate the role of tubular cell-specific Gpx3 deletion on ischemia-reperfusion injury-induced AKI (IRI-AKI) in a murine model and delineate the potential underlying mechanisms. By generating renal tubular epithelial cell-specific Gpx3 knockout mice and inducing IRI-AKI, we assessed a spectrum of kidney injury indices including renal function, oxidative stress, apoptosis and mitochondrial dynamics. Additionally, we conducted transcriptome sequencing and bioinformatics analyses. The outcomes underscore that the deficiency of GPX3 in tubular cells exacerbates tubular injury, renal dysfunction, oxidative stress, apoptosis, and mitochondrial dynamic disturbances in the context of IRI-AKI. Sequencing and bioinformatics analysis suggest that the Gpx3 deletion predominantly impacts pathways associated with metabolism and inflammation. In conclusion, the tubular cell-specific deficiency of GPX3 exacerbates renal injury by intensifying oxidative stress, fostering mitochondrial impairment, perturbing metabolic processes and fueling inflammation. The targeted restoration of GPX3 in the renal tubular emerges as a potential therapeutic avenue for mitigating IRI-AKI.
Collapse
Affiliation(s)
- Jinrong He
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China
| | - Xueqin Wu
- Department of Nephrology, The Central Hospital of Shaoyang, Hunan Province 422000, China
| | - Jie Qiao
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China
| | - Xian Xie
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China
| | - Yu Wang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China.
| | - Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China; Clinical Research Center for Critical Kidney Disease in Hunan Province, Changsha, Hunan 410013, China.
| |
Collapse
|
13
|
Goswami Y, Baghel A, Sharma G, Sharma PK, Biswas S, Yadav R, Garg PK, Shalimar, Tandon R. Liver Organoids From Hepatocytes of Healthy Humans and Non-alcoholic Fatty Liver Disease (NAFLD) Patients Display Multilineage Architecture and can be Used to Develop an In Vitro Model of Steatohepatitis. J Clin Exp Hepatol 2025; 15:102463. [PMID: 39872219 PMCID: PMC11761825 DOI: 10.1016/j.jceh.2024.102463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/15/2024] [Indexed: 01/30/2025] Open
Abstract
Background/Aim Non-alcoholic fatty liver disease (NAFLD) is a global health concern with limited treatment options. The paucity of predictive i n v itro models in preclinical settings seems to be one of the limitations of identifying effective medicines. We therefore aimed to develop an i n v itro model that can display the key hallmarks of NAFLD, such as steatosis, inflammation, and fibrosis. Methods An in vitro model of steatohepatitis was developed using organoids prepared from hepatocytes of healthy individuals from a commercial source (HLOs) and the liver tissues collected from needle biopsies of NAFLD patients (HLONAFLD) using defined culture conditions. HLOs were treated with palmitic acid for 72 h to develop an i n v itro model of steatohepatitis, while HLONAFLD served as a natural model of steatohepatitis. Metformin and saroglitazar were used to validate the liver organoid model of steatohepatitis. Saroglitazar was also evaluated in the high-fat, high-fructose (HF-HF) diet-induced model of NAFLD using C57BL/6 mice to validate the findings from the i n v itro model. Results HLOs and HLONAFLD exhibited bipotent properties, showing the expression of markers of hepatocytes, ductal cells, and also stem cells. Furthermore, they demonstrated the expression of nonparenchymal cell markers such as stellate cells (CD166) and Kupffer-like cells (CD68 and EMR1). The steatohepatitis models developed using these organoids displayed markers associated with steatosis, inflammation and fibrosis, which were decreased by metformin and saroglitazar. Conclusion The in vitro models developed in our lab employing HLOs and HLONAFLD display all three key hallmarks of NAFLD: steatosis, inflammation, and fibrosis without the necessity for coculture with other nonparenchymal cells. The implementation of the HLONAFLD-based model is also expected to provide a more realistic assessment of test substances to develop therapeutics for NAFLD.
Collapse
Affiliation(s)
- Yamini Goswami
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Akash Baghel
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ghanshyam Sharma
- BRIC-Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Phulwanti K. Sharma
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sagnik Biswas
- All India Institute of Medical Sciences, New Delhi, India
| | - Rajni Yadav
- All India Institute of Medical Sciences, New Delhi, India
| | - Pramod K. Garg
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Shalimar
- All India Institute of Medical Sciences, New Delhi, India
| | - Ruchi Tandon
- BRIC-Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
14
|
Ortuño-Costela MC, Pinzani M, Vallier L. Cell therapy for liver disorders: past, present and future. Nat Rev Gastroenterol Hepatol 2025; 22:329-342. [PMID: 40102584 DOI: 10.1038/s41575-025-01050-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/20/2025]
Abstract
The liver fulfils a plethora of vital functions and, due to their importance, liver dysfunction has life-threatening consequences. Liver disorders currently account for more than two million deaths annually worldwide and can be classified broadly into three groups, considering their onset and aetiology, as acute liver diseases, inherited metabolic disorders and chronic liver diseases. In the most advanced and severe forms leading to liver failure, liver transplantation is the only treatment available, which has many associated drawbacks, including a shortage of organ donors. Cell therapy via fully mature cell transplantation is an advantageous alternative that may be able to restore a damaged organ's functionality or serve as a bridge until regeneration can occur. Pioneering work has shown that transplanting adult hepatocytes can support liver recovery. However, primary hepatocytes cannot be grown extensively in vitro as they rapidly lose their metabolic activity. Therefore, different cell sources are currently being tested as alternatives to primary cells. Human pluripotent stem cell-derived cells, chemically induced liver progenitors, or 'liver' organoids, hold great promise for developing new cell therapies for acute and chronic liver diseases. This Review focuses on the advantages and drawbacks of distinct cell sources and the relative strategies to address different therapeutic needs in distinct liver diseases.
Collapse
Affiliation(s)
- M Carmen Ortuño-Costela
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Massimo Pinzani
- University College London Institute for Liver and Digestive Health, Division of Medicine, Royal Free Hospital, London, UK
- University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies (UPMC-ISMETT), Palermo, Italy
| | - Ludovic Vallier
- Berlin Institute of Health, BIH Centre for Regenerative Therapies, Charité-Universitätsmedizin, Berlin, Germany.
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
| |
Collapse
|
15
|
Andreatta F, Hendriks D, Artegiani B. Human Organoids as an Emerging Tool for Genome Screenings. Annu Rev Biomed Eng 2025; 27:157-183. [PMID: 40310889 DOI: 10.1146/annurev-bioeng-103023-122327] [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: 05/03/2025]
Abstract
Over the last decade, a plethora of organoid models have been generated to recapitulate aspects of human development, disease, tissue homeostasis, and repair. Organoids representing multiple tissues have emerged and are typically categorized based on their origin. Tissue-derived organoids are established directly from tissue-resident stem/progenitor cells of either adult or fetal origin. Starting from pluripotent stem cells (PSCs), PSC-derived organoids instead recapitulate the developmental trajectory of a given organ. Gene editing technologies, particularly the CRISPR-Cas toolbox, have greatly facilitated gene manipulation experiments with considerable ease and scalability, revolutionizing organoid-based human biology research. Here, we review the recent adaptation of CRISPR-based screenings in organoids. We examine the strategies adopted to perform CRISPR screenings in organoids, discuss different screening scopes and readouts, and highlight organoid-specific challenges. We then discuss individual organoid-based genome screening studies that have uncovered novel genes involved in a variety of biological processes. We close by providing an outlook on how widespread adaptation of CRISPR screenings across the organoid field may be achieved, to ultimately leverage our understanding of human biology.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands; ,
| | | |
Collapse
|
16
|
John S, Bhowmick K, Park A, Huang H, Yang X, Mishra L. Recent advances in targeting obesity, with a focus on TGF-β signaling and vagus nerve innervation. Bioelectron Med 2025; 11:10. [PMID: 40301996 PMCID: PMC12042417 DOI: 10.1186/s42234-025-00172-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/31/2025] [Indexed: 05/01/2025] Open
Abstract
Over a third of the global population is affected by obesity, fatty liver disease (Metabolic Dysfunction-Associated Steatotic Liver Disease, MASLD), and its severe form, MASH (Metabolic Dysfunction-Associated Steatohepatitis), which can ultimately progress to hepatocellular carcinoma (HCC). Recent advancements include therapeutics such as glucagon-like peptide 1 (GLP-1) agonists and neural/vagal modulation strategies for these disorders. Among the many pathways regulating these conditions, emerging insights into transforming growth factor-β (TGF-β) signaling highlight potential future targets through its role in pathophysiological processes such as adipogenesis, inflammation, and fibrosis. Vagus nerve innervation in the gastrointestinal tract is involved in satiety regulation and energy homeostasis, and vagus nerve stimulation has been applied in weight loss and diabetes. This review explores clinical trials in obesity, novel therapeutic targets, and the role of TGF-β signaling and vagus nerve modulation in obesity-related liver diseases and HCC.
Collapse
Affiliation(s)
- Sahara John
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Krishanu Bhowmick
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Andrew Park
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Hai Huang
- Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA
| | - Xiaochun Yang
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| | - Lopa Mishra
- Institute for Bioelectronic Medicine, Divisions of Gastroenterology and Hepatology, Department of Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, 11030, USA.
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
- Department of Surgery, George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
17
|
Menzel JP, Birrer FE, Stroka D, Masoodi M. Skylite: Skyline-Based Lipid Isomer Retention Time Evaluation for Lipidomics in Metabolic Dysfunction-Associated Steatohepatitis. Anal Chem 2025; 97:8791-8800. [PMID: 40226872 DOI: 10.1021/acs.analchem.4c06503] [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: 04/15/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent liver disorder worldwide and can progress to steatohepatitis. Elevated de novo lipogenesis (DNL) is a key contributor to hepatic steatosis. Fatty acid (FA) desaturation produces several unsaturated lipid isomers that are structurally very similar but have diverse biological functions. However, due to their structural similarity, many conventional mass spectrometry approaches cannot detect such metabolic alterations. Thus, we introduce the Skylite (Skyline-based lipid isomer retention time evaluation) workflow using conventional liquid chromatography-mass spectrometry (LC-MS) to identify important isomer features. Retention times of isomeric phosphatidylcholines are compared with the well-characterized human plasma reference standard NIST 1950. Retention time trends correlate well with fixed-charge derivatized FA in liquid chromatography and ozone-induced dissociation mass spectrometry data. The interpretation is supported by double bond diagnostic fragments in LC-MS/MS experiments of epoxidized hydrolyzed fatty acids. We investigate hepatic lipid profiles, focusing on esterified fatty acids in two mouse models of metabolic dysfunction-associated steatohepatitis (MASH). Out of 37 phosphatidylcholine sum compositions, the workflow identifies 123 lipid features. Importantly, CCl4-induced and melanocortin-4 receptor knockout mice on a western diet (WD) have significantly higher levels of mead acid, branched-chain fatty acid, and n-7 PUFA incorporated into phosphatidylcholines. While the MASH mouse liver tissues contain notable amounts of n-7 PUFA, no n-10 PUFA were detected, potentially indicating a unique desaturation pattern. The screening for altered lipid isomer profiles bridges the gap between high-throughput analyses and specialized structure-resolved techniques.
Collapse
Affiliation(s)
- Jan Philipp Menzel
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Fabienne E Birrer
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Mojgan Masoodi
- Department of Clinical Chemistry, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| |
Collapse
|
18
|
Iakovleva V, de Jong YP. Gene-based therapies for steatotic liver disease. Mol Ther 2025:S1525-0016(25)00298-9. [PMID: 40254880 DOI: 10.1016/j.ymthe.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Advances in nucleic acid delivery have positioned the liver as a key target for gene therapy, with adeno-associated virus vectors showing long-term effectiveness in treating hemophilia. Steatotic liver disease (SLD), the most common liver condition globally, primarily results from metabolic dysfunction-associated and alcohol-associated liver diseases. In some individuals, SLD progresses from simple steatosis to steatohepatitis, cirrhosis, and eventually hepatocellular carcinoma, driven by a complex interplay of genetic, metabolic, and environmental factors. Genetic variations in various lipid metabolism-related genes, such as patatin-like phospholipase domain-containing protein 3 (PNPLA3), 17β-hydroxysteroid dehydrogenase type 13 (HSD17B13), and mitochondrial amidoxime-reducing component 1 (MTARC1), impact the progression of SLD and offer promising therapeutic targets. This review largely focuses on genes identified through clinical association studies, as they are more likely to be effective and safe for therapeutic intervention. While preclinical research continues to deepen our understanding of genetic factors, early-stage clinical trials involving gene-based SLD therapies, including transient antisense and small-molecule approaches, are helping prioritize therapeutic targets. Meanwhile, hepatocyte gene editing technologies are advancing rapidly, offering alternatives to transient methods. As such, gene-based therapies show significant potential for preventing the progression of SLD and enhancing long-term liver health.
Collapse
Affiliation(s)
- Viktoriia Iakovleva
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
19
|
Liu K, Chen X, Fan Z, Ren F, Liu J, Hu B. From organoids to organoids-on-a-chip: Current applications and challenges in biomedical research. Chin Med J (Engl) 2025; 138:792-807. [PMID: 39994843 PMCID: PMC11970821 DOI: 10.1097/cm9.0000000000003535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Indexed: 02/26/2025] Open
Abstract
ABSTRACT The high failure rates in clinical drug development based on animal models highlight the urgent need for more representative human models in biomedical research. In response to this demand, organoids and organ chips were integrated for greater physiological relevance and dynamic, controlled experimental conditions. This innovative platform-the organoids-on-a-chip technology-shows great promise in disease modeling, drug discovery, and personalized medicine, attracting interest from researchers, clinicians, regulatory authorities, and industry stakeholders. This review traces the evolution from organoids to organoids-on-a-chip, driven by the necessity for advanced biological models. We summarize the applications of organoids-on-a-chip in simulating physiological and pathological phenotypes and therapeutic evaluation of this technology. This section highlights how integrating technologies from organ chips, such as microfluidic systems, mechanical stimulation, and sensor integration, optimizes organoid cell types, spatial structure, and physiological functions, thereby expanding their biomedical applications. We conclude by addressing the current challenges in the development of organoids-on-a-chip and offering insights into the prospects. The advancement of organoids-on-a-chip is poised to enhance fidelity, standardization, and scalability. Furthermore, the integration of cutting-edge technologies and interdisciplinary collaborations will be crucial for the progression of organoids-on-a-chip technology.
Collapse
Affiliation(s)
- Kailun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xiaowei Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhen Fan
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fei Ren
- State Key Lab of Processors, Institute of Computing Technology, Chinese Academy of Sciences, Beijing 100190, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101 China
| |
Collapse
|
20
|
Shinozaki K, Honda T, Yamaji K, Nishijima E, Ichi I, Yamane D. Impaired ApoB secretion triggers enhanced secretion of ApoE to maintain triglyceride homeostasis in hepatoma cells. J Lipid Res 2025; 66:100795. [PMID: 40180213 DOI: 10.1016/j.jlr.2025.100795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 03/22/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025] Open
Abstract
Apolipoprotein B (ApoB) is essential for the assembly and secretion of triglyceride (TG)-rich VLDL particles, and its dysfunction is linked to metabolic disorders, including dyslipidemia and liver steatosis. However, less attention has been paid to whether and how other apolipoproteins play redundant or compensatory roles when the ApoB function is compromised. Here, we investigated the effects of microsomal triglyceride transfer protein (MTP), which mediates lipidation of nascent ApoB, on ApoE function. We observed a paradoxical increase in ApoE secretion resulting from increased expression in MTP inhibitor (MTPi)-treated human hepatoma cells. This phenotype was recapitulated in APOB-knockout cells and was associated with impaired ApoB secretion. While MTP-dependent transfer of neutral lipids is dispensable for ApoE secretion, TG biosynthesis, redundantly catalyzed by DGAT1 and DGAT2, is required for efficient ApoE secretion in hepatoma cells. ApoE colocalizes with lipid droplets near the Golgi apparatus and mediates TG export in an ApoB-independent fashion. We found that simultaneous inhibition of both ApoE and ApoB, but not inhibition of either alone, led to TG accumulation in hepatoma cells, indicating that both proteins function redundantly to control TG content. Validation studies in primary human hepatocytes (PHHs) demonstrated DGAT2-dependent secretion of ApoE. While MTPi treatment did not elevate ApoE secretion, it induced increased sialylation of ApoE in the supernatants of PHHs. These results show that enhanced ApoE secretion compensates for the impaired ApoB function to maintain the lipid homeostasis, providing an alternative route to modulate lipid turnover in hepatoma cells.
Collapse
Affiliation(s)
- Kotomi Shinozaki
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Department of Nutrition and Food Science, Ochanomizu University, Tokyo, Japan
| | - Tomoko Honda
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kenzaburo Yamaji
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Emi Nishijima
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Ikuyo Ichi
- Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan
| | - Daisuke Yamane
- Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan; Graduate School of Humanities and Sciences, Ochanomizu University, Tokyo, Japan.
| |
Collapse
|
21
|
Piscopo F, Gasparrini B, van Halderen R, Brouwers JF, van den Broek J, van Tol HTA, Vos PLAM, Aardema H. Periparturient oleic acid-rich fat supplementation affects the lipid profile in blood and results in an increased oocyte yield in postpartum dairy cows. Theriogenology 2025; 236:33-44. [PMID: 39904068 DOI: 10.1016/j.theriogenology.2025.01.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/06/2025]
Abstract
In high-producing dairy cows periparturient negative energy balance (NEB) triggers body fat mobilization, resulting in elevated blood levels of non-esterified fatty acids (NEFAs). Blood is dominated by the saturated fatty acids (SFA) palmitic (C16:0) and stearic acid (C18:0), which are associated with a negative effect on oocyte developmental competence. In contrast, the monounsaturated fatty acid (MUFA) oleic acid is harmless and is able to counteract the negative effect of saturated NEFAs on in vitro maturing oocytes. Since preantral follicles lack oleic acid-rich follicular fluid, we hypothesized that preantral follicles and oocytes may benefit from oleic acid-rich fat supplementation during NEB. Eight-month pregnant Holstein Friesian heifers were randomly divided in two groups to receive a standard, palmitic acid-rich (CTR, n = 5), or rumen-protected oleic acid-rich (UNSAT, n = 6), periparturient fat supplementation until 4 weeks post-calving. NEFA, β-Hydroxybutyric acid and haptoglobin profiles in blood were monitored, and cumulus-oocyte-complexes (COCs) were via transvaginal ovum pick-up (OPU) collected at 8, 12, and 16 weeks postpartum for in vitro maturation, fertilization (day 0), and culture until day 8. Oleic acid supplementation increased C18:1 and reduced C16:0 levels in blood in comparison to CTR, during the peripartum period. Interestingly, the UNSAT group exhibited a 1.6-times higher oocyte yield in comparison to the CTR, but no difference in oocyte developmental competence between the groups. These findings suggest that peripartum oleic acid supplementation supports follicles and oocytes during NEB. Potential long-term benefits of oleic acid on fertility in dairy cows, in a higher number of animals, warrant further investigation.
Collapse
Affiliation(s)
- F Piscopo
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Naples, Italy
| | - B Gasparrini
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Naples, Italy
| | - R van Halderen
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - J F Brouwers
- Research Group Analysis Techniques in the Life Sciences, Academy for Life Sciences and Technology, Avans University of Applied Sciences, Breda, the Netherlands
| | - J van den Broek
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - H T A van Tol
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - P L A M Vos
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - H Aardema
- Farm Animal Health, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
22
|
Zhang F, Yue K, Sun S, Lu S, Jia G, Zha Y, Zhang S, Chou CJ, Liao C, Li X, Duan Y. Targeting Histone Deacetylase 11 with a Highly Selective Inhibitor for the Treatment of MASLD. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412903. [PMID: 39976110 PMCID: PMC12005767 DOI: 10.1002/advs.202412903] [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: 10/14/2024] [Revised: 01/26/2025] [Indexed: 02/21/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) represents the most prevalent chronic liver disorder globally. Due to its intricate pathogenesis and the current lack of efficacious pharmacological interventions, there is a pressing need to discover novel therapeutic targets and agents for MASLD treatment. Herein, it is found that histone deacetylase 11 (HDAC11), a subtype of HDAC family, is markedly overexpressed in both in vitro and in vivo models of MASLD. Furthermore, the knockdown of HDAC11 is observed to mitigate lipid accumulation in hepatic cells. A highly selective HDAC11 inhibitor, B6, which exhibits favorable pharmacokinetic property and liver distribution, is further designed and synthesized. Integrating RNA-seq data with in vivo and in vitro experiments, B6 is found to inhibit de novo lipogenesis (DNL) and promote fatty acid oxidation, thus mitigating hepatic lipid accumulation and pathological symptoms in MASLD mice. Further omics analysis and experiments reveal that B6 enhances the phosphorylation of AMPKα1 at Thr172 through the inhibition of HDAC11, consequently modulating DNL and fatty acid oxidation in the liver. In summary, this study identifies HDAC11 as a potential therapeutic target in MASLD and reports the discovery of a highly selective HDAC11 inhibitor with favorable drug-like properties for the treatment of MASLD.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Kairui Yue
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Simin Sun
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Shengyuan Lu
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Geng Jia
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
| | - Yang Zha
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Shuang Zhang
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - C. James Chou
- Department of Drug Discovery and Biomedical SciencesCollege of PharmacyMedical University of South CarolinaCharlestonSC29425USA
| | - Chenzhong Liao
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education InstitutesHefei University of TechnologyHefei230031China
| | - Xiaoyang Li
- Key Laboratory of Marine DrugsChinese Ministry of EducationSchool of Medicine and PharmacyOcean University of China5 Yushan RoadQingdao266003China
- Marine Biomedical Research Institute of QingdaoQingdaoShandong266071China
| | - Yajun Duan
- Department of Cardiologythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
23
|
Zhao J, Zhi Y, Ren H, Wang J, Zhao Y. Emerging biotechnologies for engineering liver organoids. Bioact Mater 2025; 45:1-18. [PMID: 39588483 PMCID: PMC11585797 DOI: 10.1016/j.bioactmat.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/27/2024] Open
Abstract
The engineering construction of the liver has attracted enormous attention. Organoids, as emerging miniature three-dimensional cultivation units, hold significant potential in the biomimetic simulation of liver structure and function. Despite notable successes, organoids still face limitations such as high variability and low maturity. To overcome these challenges, engineering strategies have been established to maintain organoid stability and enhance their efficacy, laying the groundwork for the development of advanced liver organoids. The present review comprehensively summarizes the construction of engineered liver organoids and their prospective applications in biomedicine. Initially, we briefly present the latest research progress on matrix materials that maintain the three-dimensional morphology of organoids. Next, we discuss the manipulative role of engineering technologies in organoid assembly. Additionally, we outline the impact of gene-level regulation on organoid growth and development. Further, we introduce the applications of liver organoids in disease modeling, drug screening and regenerative medicine. Lastly, we overview the current obstacles and forward-looking perspectives on the future of engineered liver organoids. We anticipate that ongoing innovations in engineered liver organoids will lead to significant advancements in medical applications.
Collapse
Affiliation(s)
- Junqi Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yue Zhi
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
24
|
Hu Y, Peng Z, Qiu M, Xue L, Ren H, Wu X, Zhu X, Ding Y. Developing biotechnologies in organoids for liver cancer. BIOMEDICAL TECHNOLOGY 2025; 9:100067. [DOI: 10.1016/j.bmt.2024.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
25
|
Barcena-Varela M, Monga SP, Lujambio A. Precision models in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2025; 22:191-205. [PMID: 39663463 DOI: 10.1038/s41575-024-01024-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) represents a global health challenge, and ranks among one of the most prevalent and deadliest cancers worldwide. Therapeutic advances have expanded the treatment armamentarium for patients with advanced HCC, but obstacles remain. Precision oncology, which aims to match specific therapies to patients who have tumours with particular features, holds great promise. However, its implementation has been hindered by the existence of numerous 'HCC influencers' that contribute to the high inter-patient heterogeneity. HCC influencers include tumour-related characteristics, such as genetic alterations, immune infiltration, stromal composition and aetiology, and patient-specific factors, such as sex, age, germline variants and the microbiome. This Review delves into the intricate world of HCC, describing the most innovative model systems that can be harnessed to identify precision and/or personalized therapies. We provide examples of how different models have been used to nominate candidate biomarkers, their limitations and strategies to optimize such models. We also highlight the importance of reproducing distinct HCC influencers in a flexible and modular way, with the aim of dissecting their relative contribution to therapy response. Next-generation HCC models will pave the way for faster discovery of precision therapies for patients with advanced HCC.
Collapse
Affiliation(s)
- Marina Barcena-Varela
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
26
|
Dwyer BJ, Tirnitz-Parker JEE. Patient-derived organoid models to decode liver pathophysiology. Trends Endocrinol Metab 2025; 36:235-248. [PMID: 39191607 DOI: 10.1016/j.tem.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/10/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
Liver diseases represent a growing global health challenge, and the increasing prevalence of obesity and metabolic disorders is set to exacerbate this crisis. To meet evolving regulatory demands, patient-specific in vitro liver models are essential for understanding disease mechanisms and developing new therapeutic approaches. Organoid models, which faithfully recapitulate liver biology, can be established from both non-malignant and malignant liver tissues, offering insight into various liver conditions, from acute injuries to chronic diseases and cancer. Improved understanding of liver microenvironments, innovative biomaterials, and advanced imaging techniques now facilitate comprehensive and unbiased data analysis, paving the way for personalised medicine. In this review, we discuss state-of-the-art patient-derived liver organoid models, recent technological advancements, and strategies to enhance their clinical impact.
Collapse
Affiliation(s)
- Benjamin J Dwyer
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| | - Janina E E Tirnitz-Parker
- Curtin Medical School and Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia; Liver Cancer Collaborative, Perth, WA, Australia; www.livercancercollaborative.au.
| |
Collapse
|
27
|
Silva B, Bragança J. Induced pluripotent stem cell-derived mesenchymal stem cells for modeling and treating metabolic associated fatty liver disease and metabolic associated steatohepatitis: Challenges and opportunities. World J Stem Cells 2025; 17:99331. [DOI: 10.4252/wjsc.v17.i2.99331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/21/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
The potential of induced pluripotent stem cells (iPSCs) for modeling and treating metabolic associated fatty liver disease (MAFLD) and metabolic associated steatohepatitis (MASH) is emerging. MAFLD is a growing global health concern, currently with limited treatment options. While primary mesenchymal stem cells hold promise, iPSCs offer a versatile alternative due to their ability to differentiate into various cell types, including iPSC-derived mesenchymal stem cells. However, challenges remain, including optimizing differentiation protocols, ensuring cell safety, and addressing potential tumorigenicity risks. In addition, iPSCs offer the possibility to generate complex cellular models, including three-dimensional organoid models, which are closer representations of the human disease than animal models. Those models would also be valuable for drug discovery and personalized medicine approaches. Overall, iPSCs and their derivatives offer new perspectives for advancing MAFLD/MASH research and developing novel therapeutic strategies. Further research is needed to overcome current limitations and translate this potential into effective clinical applications.
Collapse
Affiliation(s)
- Bárbara Silva
- Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- PhD Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
| | - José Bragança
- Algarve Biomedical Center, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, Algarve Biomedical Center-Research Institute, University of Algarve, Faro 8005-139, Portugal
- Champalimaud Research Program, Champalimaud Centre for the Unknown, Lisbon 1000-001, Portugal
| |
Collapse
|
28
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
29
|
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
|
30
|
Wu H, Wang J, Liu S, Wang Y, Tang X, Xie J, Wang N, Shan H, Chen S, Zhang X, Zeng W, Chen C, Fu Y, Lai L, Duan Y. Large-Scale Production of Expandable Hepatoblast Organoids and Polarised Hepatocyte Organoids From hESCs Under 3D Static and Dynamic Suspension Conditions. Cell Prolif 2025:e70001. [PMID: 39921573 DOI: 10.1111/cpr.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
To date, generating viable and functional hepatocytes in large scale remains challenge. By employing 3D suspension condition with the support of low concentration Matrigel, a novel culture system was developed to generate expandable hepatoblast organoids (HB-orgs) and mature polarised hepatocyte organoids (P-hep-orgs) from human embryonic stem cells (hESCs) in both dishes and bioreactors. scRNA-seq and functional assays were used to characterise HB-orgs and P-hep-orgs. hESC-derived HB-orgs could proliferate at least for 15 passages, leading to 1012 in total cells in 4 weeks. P-hep-orgs differentiated from HB-orgs displayed characteristics of mature hepatocytes with polarisation. Moreover, single-cell RNA sequencing exhibited that over 40% of cells in P-hep-orgs were highly fidelity with human primary hepatocytes. Eventually, large-scale production of P-hep-orgs could be generated from massively expanded HB-orgs within 1 week with similar number in bioreactors, which were achieved by the enhancements in energy metabolism contribute to the expansion of HB-orgs and maturation of P-hep-orgs in bioreactors. By providing a cost-efficient and robust platform, our study represents a significant step toward manufacturing large-scale functioning hESC-derived hepatocytes for cell-based therapeutics, disease modelling, pharmacology and toxicology studies.
Collapse
Affiliation(s)
- Haibin Wu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jue Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Shoupei Liu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yiyu Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xianglian Tang
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jinghe Xie
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Ning Wang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huanhuan Shan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Sen Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xueyan Zhang
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Weiping Zeng
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuxin Chen
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yinjie Fu
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
| | - Liangxue Lai
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell, Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institute for Medical Research, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Laboratory of Stem Cells and Translational Medicine, Institute for Life Science, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- The Innovation Centre of Ministry of Education for Development and Diseases, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
31
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
32
|
Verstegen MMA, Coppes RP, Beghin A, De Coppi P, Gerli MFM, de Graeff N, Pan Q, Saito Y, Shi S, Zadpoor AA, van der Laan LJW. Clinical applications of human organoids. Nat Med 2025; 31:409-421. [PMID: 39901045 DOI: 10.1038/s41591-024-03489-3] [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: 05/31/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Organoids are innovative three-dimensional and self-organizing cell cultures of various lineages that can be used to study diverse tissues and organs. Human organoids have dramatically increased our understanding of developmental and disease biology. They provide a patient-specific model to study known diseases, with advantages over animal models, and can also provide insights into emerging and future health threats related to climate change, zoonotic infections, environmental pollutants or even microgravity during space exploration. Furthermore, organoids show potential for regenerative cell therapies and organ transplantation. Still, several challenges for broad clinical application remain, including inefficiencies in initiation and expansion, increasing model complexity and difficulties with upscaling clinical-grade cultures and developing more organ-specific human tissue microenvironments. To achieve the full potential of organoid technology, interdisciplinary efforts are needed, integrating advances from biology, bioengineering, computational science, ethics and clinical research. In this Review, we showcase pivotal achievements in epithelial organoid research and technologies and provide an outlook for the future of organoids in advancing human health and medicine.
Collapse
Affiliation(s)
- Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Rob P Coppes
- Departments of Biomedical Sciences and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne Beghin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Research and Engineering in Space Technology, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mattia F M Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, UK
| | - Nienke de Graeff
- Department of Medical Ethics and Health Law, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Shaojun Shi
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| |
Collapse
|
33
|
Kim Y, Kang M, Mamo MG, Adisasmita M, Huch M, Choi D. Liver organoids: Current advances and future applications for hepatology. Clin Mol Hepatol 2025; 31:S327-S348. [PMID: 39722609 PMCID: PMC11925438 DOI: 10.3350/cmh.2024.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
The creation of self-organizing liver organoids represents a significant, although modest, step toward addressing the ongoing organ shortage crisis in allogeneic liver transplantation. However, researchers have recognized that achieving a fully functional whole liver remains a distant goal, and the original ambition of organoid-based liver generation has been temporarily put on hold. Instead, liver organoids have revolutionized the field of hepatology, extending their influence into various domains of precision and molecular medicine. These 3D cultures, capable of replicating key features of human liver function and pathology, have opened new avenues for human-relevant disease modeling, CRISPR gene editing, and high-throughput drug screening that animal models cannot accomplish. Moreover, advancements in creating more complex systems have led to the development of multicellular assembloids, dynamic organoid-on-chip systems, and 3D bioprinting technologies. These innovations enable detailed modeling of liver microenvironments and complex tissue interactions. Progress in regenerative medicine and transplantation applications continues to evolve and strives to overcome the obstacles of biocompatibility and tumorigenecity. In this review, we examine the current state of liver organoid research by offering insights into where the field currently stands, and the pivotal developments that are shaping its future.
Collapse
Affiliation(s)
- Yohan Kim
- Department of MetaBioHealth, Sungkyunkwan University, Suwon, Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Korea
| | - Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Michael Girma Mamo
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Michael Adisasmita
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
34
|
Kimura H, Nishikawa M, Kutsuzawa N, Tokito F, Kobayashi T, Kurniawan DA, Shioda H, Cao W, Shinha K, Nakamura H, Doi K, Sakai Y. Advancements in Microphysiological systems: Exploring organoids and organ-on-a-chip technologies in drug development -focus on pharmacokinetics related organs. Drug Metab Pharmacokinet 2025; 60:101046. [PMID: 39847980 DOI: 10.1016/j.dmpk.2024.101046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 01/25/2025]
Abstract
This study explored the evolving landscape of Microphysiological Systems (MPS), with a focus on organoids and organ-on-a-chip (OoC) technologies, which are promising alternatives to animal testing in drug discovery. MPS technology offers in vitro models with high physiological relevance, simulating organ function for pharmacokinetic studies. Organoids composed of 3D cell aggregates and OoCs mimicking in vivo environments based on microfluidic platforms represent the forefront of MPS. This paper provides a comprehensive overview of their application in studying the gut, liver, and kidney and their challenges in becoming reliable alternatives to in vivo models. Although MPS technology is not yet fully comparable to in vivo systems, its continued development, aided by in silico, automation, and AI approaches, is anticipated to bring about further advancements. Collaboration across multiple disciplines and ongoing regulatory discussions will be crucial in driving MPS toward practical and ethical applications in biomedical research and drug development.
Collapse
Affiliation(s)
- Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan.
| | - Masaki Nishikawa
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Naokata Kutsuzawa
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan; Division of Pulmonary Medicine, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara, Kanagawa, 259-1143, Japan
| | - Fumiya Tokito
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Takuma Kobayashi
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Dhimas Agung Kurniawan
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Hiroki Shioda
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Wenxin Cao
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| | - Kenta Shinha
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Kotaro Doi
- Division of Nephrology and Endocrinology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8655, Japan
| | - Yasuyuki Sakai
- Department of Chemical System Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo, Tokyo, 113-8654, Japan
| |
Collapse
|
35
|
Shao W, Xu H, Zeng K, Ye M, Pei R, Wang K. Advances in liver organoids: replicating hepatic complexity for toxicity assessment and disease modeling. Stem Cell Res Ther 2025; 16:27. [PMID: 39865320 PMCID: PMC11771052 DOI: 10.1186/s13287-025-04139-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 01/13/2025] [Indexed: 01/28/2025] Open
Abstract
The lack of in vivo accurate human liver models hinders the investigation of liver-related diseases, injuries, and drug-related toxicity, posing challenges for both basic research and clinical applications. Traditional cellular and animal models, while widely used, have significant limitations in replicating the liver's complex responses to various stressors. Liver organoids derived from human pluripotent stem cells, adult stem cells primary cells, or tissues can mimic diverse liver cell types, major physiological functions, and architectural features. Recent advancements in the field have shown that some liver organoids have sufficient accuracy to replicate specific aspects of the human liver's complexity. This review highlights recent progress in liver organoid research, with a particular emphasis on their potential for toxicity assessment and disease modeling. The intrinsic advantages of liver organoids include higher sensitivity and suitability for long-term studies, which enhance the predictive value in drug and nanomaterial toxicity testing. The integration of liver organoids with microfluidic devices enables the simulation of the liver microenvironment and facilitates high-throughput drug screening. The liver organoids also serve as ideal platforms for studying liver diseases such as hepatitis, liver fibrosis, viral liver diseases, and monogenic diseases. Additionally, this review discusses the advantages and limitations of liver organoids along with potential avenues for future research.
Collapse
Affiliation(s)
- Weidong Shao
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
- China Pharmaceutical University, 639 Longmian Rd, Nanjing, Jiangsu, 210009, China
| | - Hui Xu
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Kanghua Zeng
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Mingzhou Ye
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China
| | - Renjun Pei
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| | - Kai Wang
- Organoid Innovation Center, Suzhou Institute of Nanotech and Nano-bionics, Chinese Academy of Sciences, 398 Ruoshui Rd, Suzhou, Jiangsu, 215123, China.
| |
Collapse
|
36
|
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
|
37
|
Li D, Chen Y, Zhu X, Yang Y, Li H, Zhao RC. A novel human specific lncRNA MEK6-AS1 regulates adipogenesis and fatty acid biosynthesis by stabilizing MEK6 mRNA. J Biomed Sci 2025; 32:6. [PMID: 39773638 PMCID: PMC11708274 DOI: 10.1186/s12929-024-01098-3] [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: 03/26/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Obesity is becoming one of the major non-communicable diseases with increasing incidence and risks that cannot be ignored. However effective and safe clinical treatment strategies still need to be deeply explored. Increased number and volume of adipocytes lead to overweight and obesity. The aim of our work is to identify lncRNAs that have important regulatory in differentiation of human mesenchymal stem cells (MSCs) into adipocytes, and to provide effective targets for clinical prevention and treatment of obesity and related metabolic disorders. METHODS We extracted primary MSCs from human adipose tissue, and conducted expression profile analysis of lncRNAs during adipogenic differentiation of MSCs to screen changed lncRNAs. Characteristics of lncRNA were revealed mainly by RACE and RNA FISH. Loss- and gain-of function experiments in vivo and in vitro were used to analyze effects of lncRNA. Targeted metabolomics was utilized to detect levels of free fatty acids. RNA pull-down, mRNA stability tests, etc. were employed to explore mechanisms of lncRNA. RESULTS Human-specific lncRNA, we named it MEK6-AS1, was the most up-regulated transcript during adipogenic differentiation of MSCs. MEK6-AS1 was highly expressed in adipose tissue samples from individuals with BMI ≥ 25 and positively correlated with adipogenic marker genes in these samples. Knocking down lncRNA inhibited expression of adipogenic differentiation markers and ectopic adipogenesis, reducing contents of various free fatty acids, as well as promoting osteogenic differentiation. Overexpression of lncRNA had the opposite effects to the above processes. We also found that MEK6-AS1 was elevated during hepatic steatosis organoid generation. Mechanistically, MEK6-AS1 worked partially through stabilization of MEK6 mRNA by NAT10. CONCLUSIONS We have identified a human-specific lncRNA (MEK6-AS1) with position information in the genomic database but has not been extensively reported. We demonstrated that MEK6-AS1 as a novel lncRNA involved in adipogenic differentiation and adipogenesis, fatty acid metabolism, and osteogenic differentiation. We found that MEK6-AS1 may exert its effect by enhancing MEK6 mRNA stability through NAT10. Our study may provide insights into implication of lncRNAs in stem cell biology and offer a new potential therapeutic target for the prevention and treatment of obesity and other related disease.
Collapse
Affiliation(s)
- Di Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Yunhua Chen
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Xingyu Zhu
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China
| | - Yanlei Yang
- Clinical Biobank, Department Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongling Li
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China.
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China.
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China.
- Center for Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China.
- State Key Laboratory of Common Mechanism Research for Major Diseases, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China.
| |
Collapse
|
38
|
Zhang Y, Lian X, Xu H, Zhu S, Zhang H, Ni Z, Fu T, Liu S, Tao L, Zhou Y, Zhu F. OrgXenomics: an integrated proteomic knowledge base for patient-derived organoid and xenograft. Nucleic Acids Res 2025; 53:D504-D515. [PMID: 39373514 PMCID: PMC11701540 DOI: 10.1093/nar/gkae861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024] Open
Abstract
Patient-derived models (PDMs, particularly organoids and xenografts) are irreplaceable tools for precision medicine, from target development to lead identification, then to preclinical evaluation, and finally to clinical decision-making. So far, PDM-based proteomics has emerged to be one of the cutting-edge directions and massive data have been accumulated. However, such PDM-based proteomic data have not been provided by any of the available databases, and proteomics profiles of all proteins in proteomic study are also completely absent from existing databases. Herein, an integrated database named 'OrgXenomics' was thus developed to provide the proteomic data for PDMs, which was unique in (a) explicitly describing the establishment detail for a wide array of models, (b) systematically providing the proteomic profiles (expression/function/interaction) for all proteins in studied proteomic analysis and (c) comprehensively giving the raw data for diverse organoid/xenograft-based proteomic studies of various diseases. Our OrgXenomics was expected to server as one good complement to existing proteomic databases, and had great implication for the practice of precision medicine, which could be accessed at: https://idrblab.org/orgxenomics/.
Collapse
Affiliation(s)
- Yintao Zhang
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| | - Xichen Lian
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Hangwei Xu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Sisi Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Hao Zhang
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ziheng Ni
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Tingting Fu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Shuiping Liu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Lin Tao
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China
| | - Ying Zhou
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 330110, China
| |
Collapse
|
39
|
Youhanna S, Kemas AM, Wright SC, Zhong Y, Klumpp B, Klein K, Motso A, Michel M, Ziegler N, Shang M, Sabatier P, Kannt A, Sheng H, Oliva‐Vilarnau N, Büttner FA, Seashore‐Ludlow B, Schreiner J, Windbergs M, Cornillet M, Björkström NK, Hülsmeier AJ, Hornemann T, Olsen JV, Wang Y, Gramignoli R, Sundström M, Lauschke VM. Chemogenomic Screening in a Patient-Derived 3D Fatty Liver Disease Model Reveals the CHRM1-TRPM8 Axis as a Novel Module for Targeted Intervention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407572. [PMID: 39605182 PMCID: PMC11744578 DOI: 10.1002/advs.202407572] [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: 07/05/2024] [Revised: 09/19/2024] [Indexed: 11/29/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a leading cause of chronic liver disease with few therapeutic options. To narrow the translational gap in the development of pharmacological MASH treatments, a 3D liver model from primary human hepatocytes and non-parenchymal cells derived from patients with histologically confirmed MASH was established. The model closely mirrors disease-relevant endpoints, such as steatosis, inflammation and fibrosis, and multi-omics analyses show excellent alignment with biopsy data from 306 MASH patients and 77 controls. By combining high-content imaging with scalable biochemical assays and chemogenomic screening, multiple novel targets with anti-steatotic, anti-inflammatory, and anti-fibrotic effects are identified. Among these, activation of the muscarinic M1 receptor (CHRM1) and inhibition of the TRPM8 cation channel result in strong anti-fibrotic effects, which are confirmed using orthogonal genetic assays. Strikingly, using biosensors based on bioluminescence resonance energy transfer, a functional interaction along a novel MASH signaling axis in which CHRM1 inhibits TRPM8 via Gq/11 and phospholipase C-mediated depletion of phosphatidylinositol 4,5-bisphosphate can be demonstrated. Combined, this study presents the first patient-derived 3D MASH model, identifies a novel signaling module with anti-fibrotic effects, and highlights the potential of organotypic culture systems for phenotype-based chemogenomic drug target identification at scale.
Collapse
|
40
|
Ando M, Lee-Okada HC, Yokomizo T. [Molecular Mechanisms by Which Polyunsaturated Fatty Acids Suppress the Pathogenesis and Progression of NAFLD]. YAKUGAKU ZASSHI 2025; 145:177-182. [PMID: 40024729 DOI: 10.1248/yakushi.24-00177-2] [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: 03/04/2025]
Abstract
In recent years, the number of people suffering from lifestyle diseases such as hyperlipidemia and fatty liver disease has increased rapidly due to westernization of dietary patterns. Among fatty liver diseases, those that are not caused by alcohol are referred to as nonalcoholic fatty liver disease (NAFLD). Some NAFLD can progress to nonalcoholic steatohepatitis (NASH), and further progression of NAFLD can lead to cirrhosis and liver cancer. Although numerous studies have demonstrated the efficacy of dietary polyunsaturated fatty acids (PUFAs), particularly omega-3 PUFAs eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), against NAFLD, the detailed mechanisms by which these PUFAs exert their protective effects on the pathogenesis and progression of NAFLD are not well understood. Recent studies using knockout mouse models and genome-wide association studies have suggested a potential role for the enzymes responsible for the biosynthesis of PUFAs (FADS1, FADS2, ELOVL2, and ELOVL5) and their incorporation into phospholipids (LPCAT3/MBOAT5/LPLAT12 and LPIAT1/MBOAT7/LPLAT11) in the development of NAFLD. In this review, we summarize recent findings on the association of NAFLD and PUFAs with a focus on PUFA biosynthetic and metabolic enzymes to discuss the potential role of PUFAs in the prevention of NAFLD.
Collapse
Affiliation(s)
- Misa Ando
- Department of Biochemistry, Juntendo University Graduate School of Medicine
- Department of Breast Oncology, Juntendo University School of Medicine
| | | | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University Graduate School of Medicine
| |
Collapse
|
41
|
Brenner DA. Alternatives to animal testing to assess MASH drugs and hepatotoxicity. Hepatology 2025; 81:304-311. [PMID: 37934631 PMCID: PMC11070445 DOI: 10.1097/hep.0000000000000669] [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: 10/03/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023]
Abstract
The Food and Drug Administration (FDA) Modernization Act 2.0 "allows for alternatives to animal testing for purposes of drug and biological product applications." This provides an opportunity to develop and improve alternatives to animal studies to assess drugs in the liver. Two-dimensional cultures of liver cells fail to maintain their differentiated state and fail to reproduce liver disease phenotypes. Therefore, several platforms using human liver cells are being developed either to (1) assess hepatotoxicity of drugs or (2) create "diseases in a dish" to assess the effectiveness of drugs in treating liver diseases, primarily focused on treating MASH. The technological approaches include precision cut liver slices, human liver spheroids, human liver organoids, bioprinted human liver tissues, and microphysiological systems. This review evaluates each of these technologies and their role in providing alternatives to testing in animals.
Collapse
Affiliation(s)
- David A Brenner
- Sanford Burnham Prebys and UC San Diego, La Jolla, California, USA
| |
Collapse
|
42
|
Hadefi A, Leprovots M, Dinsart G, Marefati M, Vermeersch M, Monteyne D, Pérez-Morga D, Lefort A, Libert F, Verset L, Liefferinckx C, Moreno C, Devière J, Trépo E, Garcia MI. Duodenal Organoids From Metabolic Dysfunction-Associated Steatohepatitis Patients Exhibit Absorptive and Barrier Alterations. GASTRO HEP ADVANCES 2024; 4:100599. [PMID: 39996241 PMCID: PMC11849614 DOI: 10.1016/j.gastha.2024.100599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/07/2024] [Indexed: 02/26/2025]
Abstract
Background and Aims Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disease that can lead to fibrosis, cirrhosis, and hepatocellular carcinoma. Though MASH is closely tied to metabolic risk factors, the underlying pathogenic mechanisms remain scarcely understood. Recent research has emphasized the importance of the gut-liver axis in its pathogenesis, an aspect less explored in human studies. Here, we investigated whether the duodenal epithelium of MASH patients could exhibit intrinsic dysfunctions. Methods Duodenal epithelial organoids were generated from 16 MASH patients and 14 healthy controls. Biopsies and patient-derived organoid transcriptomes were then analyzed to evaluate if specific intestinal pathways were differentially modulated in MASH subjects. Functional assays were performed to assess the duodenal epithelial absorptive potential and barrier functionality. Results Organoid formation efficiency was similar between control-derived duodenal epithelial organoids and MASH-derived duodenal epithelial organoids (MDEOs) (71% and 69%, respectively). Despite global heterogeneity in growth patterns, MDEOs frequently exhibited cystic spheroid morphology. MDEOs displayed altered digestive potential associated with reduced mature absorptive cell fate, but they retained their lipid metabolic capacity, possibly mediated by lipid oxidation in stem/progenitor cells. Additionally, MDEOs misexpressed components of tight and adherens junctions and desmosomes compared to controls. However, MDEOs maintained pore and leak pathway integrity, indicating that the duodenal epithelial barrier remained functionally preserved under tested conditions. Conclusion This study provides evidence that the duodenal epithelium of MASH patients exhibits significant alterations in its nutrition-related and barrier functions. This study sheds light on the intricate dynamics of duodenal epithelial alterations in MASH, highlighting potential therapeutic avenues for restoring intestinal functions.
Collapse
Affiliation(s)
- Alia Hadefi
- IRIBHM, Jacques E. Dumont, Faculty of Medicine, Université Libre de Bruxelles ULB, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Morgane Leprovots
- IRIBHM, Jacques E. Dumont, Faculty of Medicine, Université Libre de Bruxelles ULB, Brussels, Belgium
| | - Gilles Dinsart
- IRIBHM, Jacques E. Dumont, Faculty of Medicine, Université Libre de Bruxelles ULB, Brussels, Belgium
| | - Maryam Marefati
- IRIBHM, Jacques E. Dumont, Faculty of Medicine, Université Libre de Bruxelles ULB, Brussels, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Charleroi, Belgium
| | - Daniel Monteyne
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Charleroi, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles (ULB), Charleroi, Belgium
| | - Anne Lefort
- BRIGHTcore ULB-VUB and Institute of Interdisciplinary Research in Human and Molecular Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Frédérick Libert
- BRIGHTcore ULB-VUB and Institute of Interdisciplinary Research in Human and Molecular Biology (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Laurine Verset
- Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Centre d’Anatomie pathologique, rue Meylermeersch, Brussels, Belgium
| | - Claire Liefferinckx
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Christophe Moreno
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Jacques Devière
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, CUB Hôpital Erasme, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Marie-Isabelle Garcia
- IRIBHM, Jacques E. Dumont, Faculty of Medicine, Université Libre de Bruxelles ULB, Brussels, Belgium
| |
Collapse
|
43
|
Shin DS, Son MJ, Bae M, Kim H. Local Stiffness Measurement of Hepatic Steatosis Model Liver Organoid by Fluorescence Imaging-Assisted Probe Indentation. ACS Biomater Sci Eng 2024; 10:7386-7393. [PMID: 39562163 DOI: 10.1021/acsbiomaterials.4c01242] [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: 11/21/2024]
Abstract
Mechanical stiffness of liver organoid is a key indicator for the progress of hepatic steatosis. Probe indentation is a noninvasive methodology to measure Young's modulus (YM); however, the inhomogeneous nature of the liver organoid induces measurement uncertainty requiring a large number of indentations covering a wide scanning area. Here, we demonstrate that lipid-stained fluorescence imaging-assisted probe indentation significantly reduces the number of measurements by specifying the highly lipid-induced area. Lipid-stained hepatic steatosis model liver organoid shows broad fluorescence distributions that are spatially correlated with a decreased YM on a lipid-filled region with bright fluorescence compared with that measured on a blank region with dark fluorescence. The organoid viability remained robust even after exposure to an ambient condition up to 6 h, showing that probe indentations can be noninvasive methods for liver organoid stiffness measurements.
Collapse
Affiliation(s)
- Dae-Seop Shin
- Drug Discovery Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Myung Jin Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Functional Genomics, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Myungae Bae
- Drug Discovery Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Hyunwoo Kim
- Drug Discovery Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| |
Collapse
|
44
|
Cao Y, Li X, Pan Y, Wang H, Yang S, Hong L, Ye L. CRISPR-based genetic screens advance cancer immunology. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2554-2562. [PMID: 39048715 DOI: 10.1007/s11427-023-2571-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 03/18/2024] [Indexed: 07/27/2024]
Abstract
CRISPR technologies have revolutionized research areas ranging from fundamental science to translational medicine. CRISPR-based genetic screens offer a powerful platform for unbiased screening in various fields, such as cancer immunology. Immune checkpoint blockade (ICB) therapy has been shown to strongly affect cancer treatment. However, the currently available ICBs are limited and do not work in all cancer patients. Pooled CRISPR screens enable the identification of previously unknown immune regulators that can regulate T-cell activation, cytotoxicity, persistence, infiltration into tumors, cytokine secretion, memory formation, T-cell metabolism, and CD4+ T-cell differentiation. These novel targets can be developed as new immunotherapies or used with the current ICBs as new combination therapies that may yield synergistic efficacy. Here, we review the progress made in the development of CRISPR technologies, particularly technological advances in CRISPR screens and their application in novel target identification for immunotherapy.
Collapse
Affiliation(s)
- Yuanfang Cao
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Xueting Li
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Yumu Pan
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Huahe Wang
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Siyu Yang
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Lingjuan Hong
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China
| | - Lupeng Ye
- Institute of Modern Biology, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
45
|
Tong L, Cui W, Zhang B, Fonseca P, Zhao Q, Zhang P, Xu B, Zhang Q, Li Z, Seashore-Ludlow B, Yang Y, Si L, Lundqvist A. Patient-derived organoids in precision cancer medicine. MED 2024; 5:1351-1377. [PMID: 39341206 DOI: 10.1016/j.medj.2024.08.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/11/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024]
Abstract
Organoids are three-dimensional (3D) cultures, normally derived from stem cells, that replicate the complex structure and function of human tissues. They offer a physiologically relevant model to address important questions in cancer research. The generation of patient-derived organoids (PDOs) from various human cancers allows for deeper insights into tumor heterogeneity and spatial organization. Additionally, interrogating non-tumor stromal cells increases the relevance in studying the tumor microenvironment, thereby enhancing the relevance of PDOs in personalized medicine. PDOs mark a significant advancement in cancer research and patient care, signifying a shift toward more innovative and patient-centric approaches. This review covers aspects of PDO cultures to address the modeling of the tumor microenvironment, including extracellular matrices, air-liquid interface and microfluidic cultures, and organ-on-chip. Specifically, the role of PDOs as preclinical models in gene editing, molecular profiling, drug testing, and biomarker discovery and their potential for guiding personalized treatment in clinical practice are discussed.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Weiyingqi Cui
- Chemical Biology Consortium Sweden, Science for Life Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Boya Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Pedro Fonseca
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Qian Zhao
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Ping Zhang
- Organcare (Shenzhen) Biotechnology Company, Shenzhen, China
| | - Beibei Xu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qisi Zhang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhen Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | | | - Ying Yang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Zhejiang, China
| | - Longlong Si
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
46
|
Velliou RI, Giannousi E, Ralliou C, Kassi E, Chatzigeorgiou A. Ex Vivo Tools and Models in MASLD Research. Cells 2024; 13:1827. [PMID: 39594577 PMCID: PMC11592755 DOI: 10.3390/cells13221827] [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: 09/24/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/28/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MASLD) presents a growing global health challenge with limited therapeutic choices. This review delves into the array of ex vivo tools and models utilized in MASLD research, encompassing liver-on-a-chip (LoC) systems, organoid-derived tissue-like structures, and human precision-cut liver slice (PCLS) systems. Given the urgent need to comprehend MASLD pathophysiology and identify novel therapeutic targets, this paper aims to shed light on the pivotal role of advanced ex vivo models in enhancing disease understanding and facilitating the development of potential therapies. Despite challenges posed by the elusive disease mechanism, these innovative methodologies offer promise in reducing the utilization of in vivo models for MASLD research while accelerating drug discovery and biomarker identification, thereby addressing critical unmet clinical needs.
Collapse
Affiliation(s)
- Rallia-Iliana Velliou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece; (R.-I.V.); (E.G.); (C.R.)
| | - Eirini Giannousi
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece; (R.-I.V.); (E.G.); (C.R.)
| | - Christiana Ralliou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece; (R.-I.V.); (E.G.); (C.R.)
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str., 11527 Athens, Greece; (R.-I.V.); (E.G.); (C.R.)
| |
Collapse
|
47
|
Jeong EY, Kim HJ, Lee S, Park Y, Kim YM. Label-free long-term measurements of adipocyte differentiation from patient-driven fibroblasts and quantitative analyses of in situ lipid droplet generation. JOURNAL OF THE OPTICAL SOCIETY OF AMERICA. A, OPTICS, IMAGE SCIENCE, AND VISION 2024; 41:C125-C136. [PMID: 39889084 DOI: 10.1364/josaa.528703] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/20/2024] [Indexed: 02/02/2025]
Abstract
The visualization and tracking of adipocytes and their lipid droplets (LDs) during differentiation are pivotal in developmental biology and regenerative medicine studies. Traditional staining or labeling methods, however, pose significant challenges due to their labor-intensive sample preparation, potential disruption of intrinsic cellular physiology, and limited observation timeframe. This study introduces a novel method for long-term visualization and quantification of biophysical parameters of LDs in unlabeled adipocytes, utilizing the refractive index (RI) distributions of LDs and cells. We employ low-coherence holotomography (HT) to systematically investigate and quantitatively analyze the 42-day redifferentiation process of fat cells into adipocytes. This technique yields three-dimensional, high-resolution refractive tomograms of adipocytes, enabling precise segmentation of LDs based on their elevated RI values. Subsequent automated analysis quantifies the mean concentration, volume, projected area, and dry mass of individual LDs, revealing a gradual increase corresponding with adipocyte maturation. Our findings demonstrate that HT is a potent tool for non-invasively monitoring live adipocyte differentiation and analyzing LD accumulation. This study, therefore, offers valuable insights into adipogenesis and lipid research, establishing HT and image-based analysis as a promising approach in these fields.
Collapse
|
48
|
Cederroth CR, Dyhrfjeld-Johnsen J, Canlon B. Pharmacological Approaches to Hearing Loss. Pharmacol Rev 2024; 76:1063-1088. [PMID: 39164117 PMCID: PMC11549935 DOI: 10.1124/pharmrev.124.001195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 08/22/2024] Open
Abstract
Hearing disorders pose significant challenges to individuals experiencing them and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Current treatment options often focus on amplification devices, cochlear implants, or other rehabilitative therapies, leaving a substantial gap regarding effective pharmacological interventions. Advancements in our understanding of the molecular and cellular mechanisms involved in hearing disorders induced by noise, aging, and ototoxicity have opened new avenues for drug development, some of which have led to numerous clinical trials, with promising results. The development of optimal drug delivery solutions in animals and humans can also enhance the targeted delivery of medications to the ear. Moreover, large genome studies contributing to a genetic understanding of hearing loss in humans combined with advanced molecular technologies in animal studies have shown a great potential to increase our understanding of the etiologies of hearing loss. The auditory system exhibits circadian rhythms and temporal variations in its physiology, its vulnerability to auditory insults, and its responsiveness to drug treatments. The cochlear clock rhythms are under the control of the glucocorticoid system, and preclinical evidence suggests that the risk/benefit profile of hearing disorder treatments using chronopharmacological approaches would be beneficial. If translatable to the bedside, such approaches may improve the outcome of clinical trials. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug formulation and delivery as well as optimized timing of drug administration, holds great promise of more effective treatments. SIGNIFICANCE STATEMENT: Hearing disorders pose significant challenges to individuals and their overall quality of life, emphasizing the critical need for advanced pharmacological approaches to address these conditions. Ongoing research into the molecular and genetic basis of auditory disorders, coupled with advancements in drug delivery procedures and optimized timing of drug administration, holds the promise of more effective treatments.
Collapse
Affiliation(s)
- Christopher R Cederroth
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Jonas Dyhrfjeld-Johnsen
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| | - Barbara Canlon
- Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden (C.R.C., B.C.); Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany (C.R.C.); and Acousia Therapeutics GmbH, Tübingen, Germany (J.D.-J.)
| |
Collapse
|
49
|
Zhou B, Luo Y, Bi H, Zhang N, Ma M, Dong Z, Ji N, Zhang S, Wang X, Liu Y, Guo X, Wei W, Xie C, Wu L, Wan X, Zheng MH, Zhao B, Li Y, Hu C, Lu Y. Amelioration of nonalcoholic fatty liver disease by inhibiting the deubiquitylating enzyme RPN11. Cell Metab 2024; 36:2228-2244.e7. [PMID: 39146936 DOI: 10.1016/j.cmet.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/16/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), including its more severe manifestation nonalcoholic steatohepatitis (NASH), is a global public health challenge. Here, we explore the role of deubiquitinating enzyme RPN11 in NAFLD and NASH. Hepatocyte-specific RPN11 knockout mice are protected from diet-induced liver steatosis, insulin resistance, and steatohepatitis. Mechanistically, RPN11 deubiquitinates and stabilizes METTL3 to enhance the m6A modification and expression of acyl-coenzyme A (CoA) synthetase short-chain family member 3 (ACSS3), which generates propionyl-CoA to upregulate lipid metabolism genes via histone propionylation. The RPN11-METTL3-ACSS3-histone propionylation pathway is activated in the livers of patients with NAFLD. Pharmacological inhibition of RPN11 by Capzimin ameliorated NAFLD, NASH, and related metabolic disorders in mice and reduced lipid contents in human hepatocytes cultured in 2D and 3D. These results demonstrate that RPN11 is a novel regulator of NAFLD/NASH and that suppressing RPN11 has therapeutic potential for the treatment.
Collapse
Affiliation(s)
- Bing Zhou
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunchen Luo
- Department of Endocrinology and Metabolism, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Hanqi Bi
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ni Zhang
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mingyue Ma
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixia Dong
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nana Ji
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Zhang
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoye Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuejun Liu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wei Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinjian Wan
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yao Li
- Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, China.
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
50
|
Yao Q, Cheng S, Pan Q, Yu J, Cao G, Li L, Cao H. Organoids: development and applications in disease models, drug discovery, precision medicine, and regenerative medicine. MedComm (Beijing) 2024; 5:e735. [PMID: 39309690 PMCID: PMC11416091 DOI: 10.1002/mco2.735] [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: 04/02/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
Organoids are miniature, highly accurate representations of organs that capture the structure and unique functions of specific organs. Although the field of organoids has experienced exponential growth, driven by advances in artificial intelligence, gene editing, and bioinstrumentation, a comprehensive and accurate overview of organoid applications remains necessary. This review offers a detailed exploration of the historical origins and characteristics of various organoid types, their applications-including disease modeling, drug toxicity and efficacy assessments, precision medicine, and regenerative medicine-as well as the current challenges and future directions of organoid research. Organoids have proven instrumental in elucidating genetic cell fate in hereditary diseases, infectious diseases, metabolic disorders, and malignancies, as well as in the study of processes such as embryonic development, molecular mechanisms, and host-microbe interactions. Furthermore, the integration of organoid technology with artificial intelligence and microfluidics has significantly advanced large-scale, rapid, and cost-effective drug toxicity and efficacy assessments, thereby propelling progress in precision medicine. Finally, with the advent of high-performance materials, three-dimensional printing technology, and gene editing, organoids are also gaining prominence in the field of regenerative medicine. Our insights and predictions aim to provide valuable guidance to current researchers and to support the continued advancement of this rapidly developing field.
Collapse
Affiliation(s)
- Qigu Yao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Sheng Cheng
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Qiaoling Pan
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jiong Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Guoqiang Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Lanjuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Hongcui Cao
- State Key Laboratory for the Diagnosis and Treatment of Infectious DiseasesNational Clinical Research Center for Infectious DiseasesCollaborative Innovation Center for Diagnosis and Treatment of Infectious DiseasesNational Medical Center for Infectious DiseasesThe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- Zhejiang Key Laboratory for Diagnosis and Treatment of Physic‐Chemical and Aging‐Related InjuriesHangzhouChina
| |
Collapse
|