1
|
Gruevska A, Leslie J, Perpiñán E, Maude H, Collins AL, Johnson S, Evangelista L, Sabey E, French J, White S, Moir J, Robinson SM, Alrawashdeh W, Thakkar R, Forlano R, Manousou P, Goldin R, Carling D, Hoare M, Thursz M, Mann DA, Cebola I, Posma JM, Safinia N, Oakley F, Hall Z. Spatial lipidomics reveals sphingolipid metabolism as anti-fibrotic target in the liver. Metabolism 2025; 168:156237. [PMID: 40127860 DOI: 10.1016/j.metabol.2025.156237] [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: 01/13/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND AND AIMS Steatotic liver disease (SLD), which encompasses various causes of fat accumulation in the liver, is a major cause of liver fibrosis. Understanding the specific mechanisms of lipotoxicity, dysregulated lipid metabolism, and the role of different hepatic cell types involved in fibrogenesis is crucial for therapy development. METHODS We analysed liver tissue from SLD patients and 3 mouse models. We combined bulk/spatial lipidomics, transcriptomics, imaging mass cytometry (IMC) and analysis of published spatial and single-cell RNA sequencing (scRNA-seq) data to explore the metabolic microenvironment in fibrosis. Pharmacological inhibition of sphingolipid metabolism with myriocin, fumonisin B1, miglustat and D-PDMP was carried out in hepatic stellate cells (HSCs) and human precision cut liver slices (hPCLSs). RESULTS Bulk lipidomics revealed increased glycosphingolipids, ether lipids and saturated phosphatidylcholines in fibrotic samples. Spatial lipidomics detected >40 lipid species enriched within fibrotic regions, notably sphingomyelin (SM) 34:1. Using bulk transcriptomics (mouse) and analysis of published spatial transcriptomics data (human) we found that sphingolipid metabolism was also dysregulated in fibrosis at transcriptome level, with increased gene expression for ceramide and glycosphingolipid synthesis. Analysis of human scRNA-seq data showed that sphingolipid-related genes were widely expressed in non-parenchymal cells. By integrating spatial lipidomics with IMC of hepatic cell markers, we found excellent spatial correlation between sphingolipids, such as SM(34:1), and myofibroblasts. Inhibiting sphingolipid metabolism resulted in anti-fibrotic effects in HSCs and hPCLSs. CONCLUSIONS Our spatial multi-omics approach suggests cell type-specific mechanisms of fibrogenesis involving sphingolipid metabolism. Importantly, sphingolipid metabolic pathways are modifiable targets, which may have potential as an anti-fibrotic therapeutic strategy.
Collapse
Affiliation(s)
- Aleksandra Gruevska
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Elena Perpiñán
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, United Kingdom
| | - Hannah Maude
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Amy L Collins
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Sophia Johnson
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Laila Evangelista
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Eleanor Sabey
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Jeremy French
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Steven White
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - John Moir
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Stuart M Robinson
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Wasfi Alrawashdeh
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Rohan Thakkar
- Department of Hepatobiliary Surgery, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, United Kingdom
| | - Roberta Forlano
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Pinelopi Manousou
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Robert Goldin
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - David Carling
- MRC Laboratory of Medical Sciences, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Matthew Hoare
- Early Cancer Institute, University of Cambridge, Cambridge, United Kingdom
| | - Mark Thursz
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom
| | - Inês Cebola
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Joram M Posma
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Niloufar Safinia
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, United Kingdom
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, University of Newcastle, Newcastle-upon-Tyne, United Kingdom; FibroFind, Unit 26/27, Baker's Yard, Christon Road, Newcastle upon Tyne, United Kingdom
| | - Zoe Hall
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
2
|
Nater M, Brügger M, Cecconi V, Pereira P, Forni G, Köksal H, Dimakou D, Herbst M, Calvanese AL, Lucchiari G, Schneider C, Valenta T, van den Broek M. Hepatic iNKT cells facilitate colorectal cancer metastasis by inducing a fibrotic niche in the liver. iScience 2025; 28:112364. [PMID: 40292307 PMCID: PMC12032931 DOI: 10.1016/j.isci.2025.112364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/05/2025] [Accepted: 04/02/2025] [Indexed: 04/30/2025] Open
Abstract
The liver is an important metastatic organ that contains many innate immune cells, yet little is known about their role in anti-metastatic defense. We investigated how invariant natural killer T (iNKT) cells influence colorectal cancer-derived liver metastasis using different models in immunocompetent mice. We found that hepatic iNKT cells promote metastasis by creating a supportive niche for disseminated cancer cells. Mechanistically, iNKT cells respond to disseminating cancer cells by producing the fibrogenic cytokines interleukin-4 (IL-4) and IL-13 in a T cell receptor-independent manner. Selective abrogation of IL-4 and IL-13 sensing in hepatic stellate cells prevented their transdifferentiation into extracellular matrix-producing myofibroblasts, which hindered metastatic outgrowth of disseminated cancer cells. This study highlights a novel tumor-promoting axis driven by iNKT cells in the initial stages of metastasis.
Collapse
Affiliation(s)
- Marc Nater
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Michael Brügger
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Paulo Pereira
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Geo Forni
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Hakan Köksal
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Despoina Dimakou
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Michael Herbst
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Giulia Lucchiari
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Tomas Valenta
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | |
Collapse
|
3
|
Brown GE, Bodke VV, Ware BR, Khetani SR. Liver portal fibroblasts induce the functions of primary human hepatocytes in vitro. Commun Biol 2025; 8:721. [PMID: 40346200 PMCID: PMC12064700 DOI: 10.1038/s42003-025-08135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
In vitro human liver models are critical to mitigate species-specific differences observed for toxicology, disease modeling, and regenerative medicine. Interactions with mesenchyme (i.e., fibroblasts) can promote phenotypic functions of primary human hepatocytes (PHHs) in culture; however, using liver-derived fibroblasts remains elusive. Portal fibroblasts (PFs) around the portal triad influence bile duct formation during development, but their role in regulating homeostatic hepatic functions remains unknown. Here, we show that human liver PFs induce long-term phenotypic functions in PHHs at higher levels than activated hepatic stellate cells across 2-dimensional and 3-dimensional culture formats. While PF-conditioned media induces some hepatic functions, partly via insulin-like growth factor binding protein-5 signaling, direct contact is necessary to induce optimal functional levels. Inhibiting Notch signaling reduces progenitor-like characteristics of PHHs and further enhances functionality. Overall, this work demonstrates a unique role for PFs in modulating hepatic functions and provides all-human and all-liver coculture strategies for downstream applications.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Vedant V Bodke
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
| | - Brenton R Ware
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Salman R Khetani
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Nirwan A, Saini D, Kaur J, Swain A, Sarkar A, Yadav PP, Mishra DP. Coagulin-L alleviates hepatic stellate cells activation and angiogenesis through modulation of the PI3K/AKT pathway during liver fibrosis. Biochem Pharmacol 2025; 238:116979. [PMID: 40345560 DOI: 10.1016/j.bcp.2025.116979] [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: 03/04/2025] [Revised: 04/08/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Fibrosis, a wound healing response in chronic liver diseases, is a potential therapeutic target during the disease progression. The activation of hepatic stellate cells (HSCs) plays a central role in liver fibrosis and depicts phenotypic change during fibrosis progression. Coagulin-L, a withanolide from Withania coagulans, has shown diverse biological activities, including anti-hyperglycemic and anti-dyslipidemic effects. However, its therapeutic efficacy against HSC activation, pathological angiogenesis and liver fibrosis is unknown. This study investigates the effects of Coagulin-L on HSC activation, pathological angiogenesis and validates these findings in vivo using a methionine and choline-deficient (MCD) diet-induced liver fibrosis model in C57BL/6 mice. Therapeutic efficacy of Coagulin-L was studied using transforming growth factor beta (TGF-β) activated HSC cell line LX-2 in vitro. Network pharmacology was used for target prediction, followed by Human umbilical vein endothelial cells (HUVEC) cell based angiogenic assays. The validation studies were carried out in a mice model of MCD diet induced liver fibrosis using serum biochemistry, histopathological assessment and immunohistochemistry methods. We found that Coagulin-L mitigated TGF-β induced activation of stellate cells and exhibited anti-angiogenic effects by downregulation of vascular endothelial growth factor (VEGF) expression and secretion from stellate cells with inhibition of the PI3K/AKT signaling pathway. In the MCD diet-induced liver fibrosis model, Coagulin-L alleviated liver injury, improved liver function, and reduced collagen deposition. Collectively, our results underscored the anti-fibrotic and anti-angiogenic effects of Coagulin-L in vitro and in vivo liver fibrosis models, thereby indicating its therapeutic potential in chronic liver diseases including metabolic dysfunctional-associated steatohepatitis (MASH).
Collapse
Affiliation(s)
- Abhishek Nirwan
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepika Saini
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Jaspreet Kaur
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India
| | - Abinash Swain
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India
| | - Abhisek Sarkar
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Prem Prakash Yadav
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Durga Prasad Mishra
- Cell Death Research Laboratory, Endocrinology Division, CSIR-Central Drug Research Institute, B.S. 10/1, Sector-10, Jankipuram Extension Lucknow, Uttar Pradesh 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Chen C, Wang J, Zhu X, Zhang S, Yuan X, Hu J, Liu C, Liu L, Zhang Z, Li J. Lactylation as a metabolic epigenetic modification: Mechanistic insights and regulatory pathways from cells to organs and diseases. Metabolism 2025:156289. [PMID: 40324589 DOI: 10.1016/j.metabol.2025.156289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/20/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
In recent years, lactylation, a novel post-translational modification, has demonstrated a unique role in bridging cellular metabolism and epigenetic regulation. This modification exerts a dual-edged effect in both cancer and non-cancer diseases by dynamically integrating the supply of metabolic substrates and the activity of modifying enzymes: on one hand, it promotes tissue homeostasis and repair through the activation of repair genes; on the other, it exacerbates pathological progression by driving malignant phenotypes. In the field of oncology, lactylation regulates key processes such as metabolic reprogramming, immune evasion, and therapeutic resistance, thereby shaping the heterogeneity of the tumor microenvironment. In non-cancerous diseases, including neurodegeneration and cardiovascular disorders, its aberrant activation can lead to mitochondrial dysfunction, fibrosis, and chronic inflammation. Existing studies have revealed a dynamic regulatory network formed by the cooperation of modifying and demodifying enzymes, and have identified mechanisms such as subcellular localization and RNA metabolism intervention that influence disease progression. Nevertheless, several challenges remain in the field. This article comprehensively summarizes the disease-specific regulatory mechanisms of lactylation, with the aim of providing a theoretical foundation for its targeted therapeutic application.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Jie Wang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shan Zhang
- Department of Endocrinology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiandun Yuan
- Department of Rheumatology and Immunology, Peking University Third Hospital, Beijing 100096, China
| | - Jun Hu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Chao Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Lanchun Liu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China
| | - Zhenpeng Zhang
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| | - Jun Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Beijing 100053, China.
| |
Collapse
|
6
|
Oñate MK, Oon C, Bhattacharyya S, Low V, Chen C, Zhao X, Arnold F, Yan Z, Pramod S, Hang Y, Ho YJ, Lowe SW, Kim SK, Xia Z, Sherman MH. Stromal KITL/SCF Maintains Pancreas Tissue Homeostasis and Restrains Tumor Progression. Cancer Discov 2025; 15:913-929. [PMID: 39918337 PMCID: PMC12046321 DOI: 10.1158/2159-8290.cd-24-1079] [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: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 02/12/2025]
Abstract
SIGNIFICANCE By analyzing transcriptional programs in healthy and tumor-associated pancreatic mesenchyme, we find that a subpopulation of mesenchymal cells in healthy pancreas tissue expresses the paracrine signaling factor KITL. The loss of mesenchymal KITL is an accompanying and permissive feature of pancreas tumor evolution, with potential implications for cancer interception. See related article by Dolskii and Cukierman, p. 872.
Collapse
Affiliation(s)
- Maria Kathrina Oñate
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Chet Oon
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Sohinee Bhattacharyya
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Vivien Low
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Canping Chen
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Xiaofan Zhao
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Frank Arnold
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Ziqiao Yan
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
| | - Sneha Pramod
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, New York, New York
| | - Yan Hang
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
| | - Yu-Jui Ho
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott W. Lowe
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Seung K. Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, California
- Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, California
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Department of Pediatrics, Stanford University School of Medicine, Stanford, California
| | - Zheng Xia
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon
| | - Mara H. Sherman
- Cancer Biology & Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
7
|
Nie YM, Zhou WQ, Niu T, Mao MF, Zhan YX, Li Y, Wang KP, Li MX, Ding K. Peptidoglycan isolated from the fruit of Lycium barbarum alleviates liver fibrosis in mice by regulating the TGF-β/Smad7 signaling and gut microbiota. Acta Pharmacol Sin 2025; 46:1329-1344. [PMID: 39833303 PMCID: PMC12032012 DOI: 10.1038/s41401-024-01454-x] [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: 07/15/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
The hepatoprotective effect of the fruit of Lycium barbarum has been documented in China over millennia. Lycium barbarum polysaccharides (LBPs) were the first macromolecules reported to mitigate liver fibrosis in carbon tetrachloride (CCl4)-treated mice. Herein, a neutral peptidoglycan, named as LBPW, was extracted from the fruit of Lycium barbarum. In this study, we investigated the hepatoprotective mechanisms of LBPW. CCl4-induced liver fibrosis mice were administered LBPW (50, 100, 200 mg ·kg-1 ·d-1, i.p.) or (100, 200, 300 mg· kg-1 ·d-1, i.g.) for 6 weeks. We showed that either i.p. or i.g. administration of LBPW dose-dependently attenuated liver damage and fibrosis in CCl4-treated mice. Pharmacokinetic analysis showed that cyanine 5.5 amine (Cy5.5)-labeled LBPW (Cy5.5-LBPW) could be detected in the liver through i.p. and i.g. administration with i.g.-administered Cy5.5-LBPW mainly accumulating in the intestine. In TGF-β1-stimulated LX-2 cells as well as in the liver of CCl4-treated mice, we demonstrated that LBPW significantly upregulated Smad7, a negative regulator of TGF-β/Smad signaling, to retard the activation of hepatic stellate cells (HSCs) and prevent liver fibrosis. On the other hand, LBPW significantly boosted the abundance of Akkermansia muciniphila (A. muciniphila) and fortified gut barrier function. We demonstrated that A. muciniphila might be responsible for the efficacy of LBPW since decreasing the abundance of this bacterium by antibiotics (Abs) blocked the effectiveness of LBPW. Overall, our results show that LBPW may exert the hepatoprotective effect via rebalancing TGF-β/Smad7 signaling and propagating gut commensal A. muciniphila, suggesting that LBPW could be leading components to be developed as new drug candidates or nutraceuticals against liver fibrosis.
Collapse
Affiliation(s)
- Ying-Min Nie
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wan-Qi Zhou
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China
- Lingang Laboratory, Shanghai, 201203, China
| | - Ting Niu
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Meng-Fei Mao
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yu-Xue Zhan
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun Li
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai-Ping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Mei-Xia Li
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Kan Ding
- Carbohydrate-Based Drug Research Center, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
- Lingang Laboratory, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, SSIP Healthcare and Medicine Demonstration Zone, Zhongshan Tsuihang New District, Zhongshan, 528400, China.
| |
Collapse
|
8
|
Du Y, Zhao Y, Li J, Wang J, You S, Zhang Y, Zhang L, Yang J, Alinejad-Rokny H, Cheng S, Shao C, Zou D, Ye Y. PLXDC1 + Tumor-Associated Pancreatic Stellate Cells Promote Desmoplastic and Immunosuppressive Niche in Pancreatic Ductal Adenocarcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2415756. [PMID: 40091495 DOI: 10.1002/advs.202415756] [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: 11/26/2024] [Revised: 01/27/2025] [Indexed: 03/19/2025]
Abstract
Pancreatic stellate cells (PSCs) contribute to pancreatic ductal adenocarcinoma (PDAC) progression and therapeutic resistance, yet their detailed functions remain unclear. This study combined RNA sequencing and assay for transposase-accessible chromatin using sequencing (ATAC-seq) on sorted PSCs from adjacent normal and PDAC tissues to investigate their transcriptional and epigenetic activation. PSCs heterogeneity and functions are characterized through bulk, single-cell, and spatial transcriptomes, as well as in situ sequencing. The clinical relevance of PSCs in immunotherapy is assessed using an in-house immune-checkpoint blockade (ICB) treatment cohort. Findings showed that stress and hypoxia signaling activated PSCs in PDAC. Three common PSCs (CPSCs) and four tumor-associated PSCs (TPSCs) are identified, each with distinct functions. CPSCs differentiated into CCL19+ TPSCs in immune-enriched regions, MYH11+ TPSCs in the stromal region, and PLXDC1+ TPSCs, which exhibited cancer-associated myofibroblasts (myCAFs) phenotype linked to poor prognosis. Notably, PLXDC1+ TPSCs, located near aggressive LRRC15+ myCAFs and SPP1+ macrophages, formed a desmoplastic and immunosuppressive niche around the tumor boundary, promoting CD8 T cell exhaustion. Single-cell transcriptomics of PDAC patients treated with ICB revealed that PLXDC1+ TPSCs correlated with poor immunotherapy efficacy. Overall, this study provides key insights into PSCs in PDAC and potential therapeutic targets.
Collapse
Affiliation(s)
- Yanhua Du
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Yizhou Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Judong Li
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jiaxin Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Shenglan You
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Yao Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Li Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Jihong Yang
- Department of Hepatobiliary Surgery, Hebei Key Laboratory of General Surgery for Digital Medicine, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Hamid Alinejad-Rokny
- UNSW BioMedical Machine Learning Lab (BML), School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, 2052, Australia
| | - Shujie Cheng
- Department of Hepatobiliary Surgery, Hebei Key Laboratory of General Surgery for Digital Medicine, Affiliated Hospital of Hebei University, Baoding, 071000, China
| | - Chenghao Shao
- Department of Pancreatic-biliary Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Duowu Zou
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
| | - Youqiong Ye
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200001, China
- Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, State Key Laboratory of Systems Medicine for Cancer, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| |
Collapse
|
9
|
Zhang X, Liu H, Cai P, Huang Z, Ma J, Luo L. Mdka produced by the activated HSCs drives bipotential progenitor cell redifferentiation during zebrafish biliary-mediated liver regeneration. Hepatology 2025; 81:1400-1415. [PMID: 39188045 DOI: 10.1097/hep.0000000000001031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/09/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND AND AIMS After extensive hepatocyte loss or impaired hepatocyte proliferation, liver regeneration occurs through trans-differentiation of biliary epithelial cells (BECs), which involves dedifferentiation of biliary epithelial cells into bipotential progenitor cells (BP-PCs) and subsequent redifferentiation of BP-PCs into nascent hepatocytes and biliary epithelial cells. Despite several studies on the redifferentiation process of BP-PCs into nascent hepatocytes, the contributions of nonparenchymal cells in this process remain poorly understood. APPROACH AND RESULTS Using the zebrafish severe liver injury model, we observed specific expression of midkine a (Mdka) in the activated HSCs through single-cell analyses and fluorescence in situ hybridization. Genetic mutation, pharmacological inhibition, whole-mount in situ hybridizations, and antibody staining demonstrated an essential role of mdka in the redifferentiation of BP-PCs during liver regeneration. Notably, we identified Nucleolin (Ncl), the potential receptor for Mdka, specifically expressed in BP-PCs, and its mutant recapitulated the mdka mutant phenotypes with impaired BP-PC redifferentiation. Mechanistically, the Mdka-Ncl axis drove Erk1 activation in BP-PCs during liver regeneration. Furthermore, overexpression of activated Erk1 partially rescued the defective liver regeneration in the mdka mutant. CONCLUSIONS The activated HSCs produce Mdka to drive the redifferentiation process of BP-PCs through activating Erk1 during the biliary-mediated liver regeneration, implying previously unappreciated contributions of nonparenchymal cells to this regeneration process.
Collapse
Affiliation(s)
- Xintao Zhang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Huijuan Liu
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Pengcheng Cai
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Zhuofu Huang
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Jianlong Ma
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Liver Cancer Institute of Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
10
|
Kuchay MS, Choudhary NS, Ramos-Molina B. Pathophysiological underpinnings of metabolic dysfunction-associated steatotic liver disease. Am J Physiol Cell Physiol 2025; 328:C1637-C1666. [PMID: 40244183 DOI: 10.1152/ajpcell.00951.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 01/31/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is emerging as the leading cause of chronic liver disease worldwide, reflecting the global epidemics of obesity, metabolic syndrome, and type 2 diabetes. Beyond its strong association with excess adiposity, MASLD encompasses a heterogeneous population that includes individuals with normal body weight ("lean MASLD") highlighting the complexity of its pathogenesis. This disease results from a complex interplay between genetic susceptibility, epigenetic modifications, and environmental factors, which converge to disrupt metabolic homeostasis. Adipose tissue dysfunction and insulin resistance trigger an overflow of lipids to the liver, leading to mitochondrial dysfunction, oxidative stress, and hepatocellular injury. These processes promote hepatic inflammation and fibrogenesis, driven by cross talk among hepatocytes, immune cells, and hepatic stellate cells, with key contributions from gut-liver axis perturbations. Recent advances have unraveled pivotal molecular pathways, such as transforming growth factor-β signaling, Notch-induced osteopontin, and sphingosine kinase 1-mediated responses, that orchestrate fibrogenic activation. Understanding these interconnected mechanisms is crucial for developing targeted therapies. This review integrates current knowledge on the pathophysiology of MASLD, emphasizing emerging concepts such as lean metabolic dysfunction-associated steatohepatitis (MASH), epigenetic alterations, hepatic extracellular vesicles, and the relevance of extrahepatic signals. It also discusses novel therapeutic strategies under investigation, aiming to provide a comprehensive and structured overview of the evolving MASLD landscape for both basic scientists and clinicians.
Collapse
Affiliation(s)
| | - Narendra Singh Choudhary
- Institute of Digestive and Hepatobiliary Sciences, Medanta-The Medicity Hospital, Gurugram, India
| | - Bruno Ramos-Molina
- Group of Obesity, Diabetes & Metabolism, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
11
|
Yang C, Gao Z, Tang R, Zhou L, Zhou P, Shi W, Ren D, Chen H, Zhang Z, Xie X, Lin J, Ye Y, Feng Z, Feng X, He Y, Zhi Y, Liu D, Zhang X, Jiang L. POU6F2 promotes liver metastasis of gastric adenocarcinoma by dual mechanism of transcriptional upregulation of SNAI1 and IGF2/PI3K/AKT signaling-induced conversion of hepatic stellate cells into cancer-associated fibroblasts. Br J Cancer 2025:10.1038/s41416-025-03017-1. [PMID: 40301686 DOI: 10.1038/s41416-025-03017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 03/10/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Activation of cancer-associated fibroblasts (CAFs) plays an important role in tumor metastasis. The purpose of this study is to investigate the role of POU6F2 in conversion of hepatic stellate cells (HSCs) into CAFs in liver metastasis of gastric adenocarcinoma (GAC). METHODS POU6F2 expression was examined by real-time PCR, Western blot and immunohistochemical staining. The functional roles of POU6F2 in GAC liver metastasis were investigated both cellular experiments in vitro and in vivo using a mouse model of subcutaneous splenic injection. ChIP and ELISA assays were used to explore the underlying molecular mechanism of POU6F2 in liver metastasis of GAC. RESULTS Here we reported that POU6F2 was upregulated in GAC tissue with liver metastasis, which predicted poor early liver metastasis. Upregulating POU6F2 promoted EMT, invasion and migration of GAC cells in vitro, and the liver metastasis of GAC cells in vivo. Mechanic investigation further revealed that upregulating POU6F2 promoted the invasion and metastasis of GAC by transcriptional upregulation of EMT-inducer SNAI1, and promoting the conversion of HSCs into CAFs dependent on transcriptional upregulation of IGF2-induced activation of PI3K/AKT signaling. CONCLUSION Our findings uncover a novel dual mechanism by which POU6F2 promotes liver metastasis of GAC.
Collapse
Affiliation(s)
- Chunxiao Yang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhiqing Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
- Department of Molecular Medicine, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Ruiming Tang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lihuan Zhou
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Ping Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Wangpan Shi
- Department of Pathology, University of California San Diego Health System, San Diego, CA, USA
| | - Dong Ren
- Departments of Pathology, University of California Irvine Medical Center, Orange, CA, USA
| | - Han Chen
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhuojun Zhang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyi Xie
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Jiaqian Lin
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Yingming Ye
- Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhengfu Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Xiaoli Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Yaofeng Zhi
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China
| | - Dongmei Liu
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Precision and Clinical Translation Medicine, Jiangmen Clinical Medical College of Guangdong Medical University, Jiangmen Central Hospital, Jiangmen, China.
| | - Lili Jiang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Terrell JA, Chen C. Extracellular Matrix Microstructures Modulate Hepatic Methionine Cycle and Methylations. Biomacromolecules 2025. [PMID: 40298277 DOI: 10.1021/acs.biomac.4c01748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The field of mechanobiology has grown in the past decade, but limited studies investigate how the extracellular matrix affects the cell metabolome. The methionine cycle involves the catabolism and regeneration of methionine through the donation and recovery of a single methyl group; this methyl group can methylate DNA, RNA, and proteins to alter gene expression and protein-protein interactions. Through studying cells cultured on fibrous (mimicking healthy extracellular matrice (ECM)) and flat (mimicking severely fibrotic ECM) substrates, we observed an increase in methionine cycle enzyme expression in cells on the flat substrate. We also present how the methionine cycle is modulated by the ECM through transmembrane protein integrin β1. By inhibiting integrin activation through the ligand-mimicking peptide RGD, we observed that the methionine cycle was protected from alteration. The results presented provide insight into possible therapeutic targets for fibrotic diseases and knowledge of mechanisms by which the ECM alters cell processes.
Collapse
Affiliation(s)
- John A Terrell
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| | - Chengpeng Chen
- Department of Chemistry and Biochemistry, University of Maryland Baltimore County, Baltimore, Maryland 21250, United States
| |
Collapse
|
13
|
Vijayan V, M Unagolla J, Panchal D, John JE, Menon SS, Menon JU. Biomimetic nanoparticles for targeted therapy of liver disease. RSC PHARMACEUTICS 2025:d5pm00044k. [PMID: 40321406 PMCID: PMC12045541 DOI: 10.1039/d5pm00044k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025]
Abstract
Liver fibrosis is a progressive and fatal condition characterized by stiffness and scarring of the liver due to excessive buildup of extracellular matrix (ECM) proteins. If left untreated, it can progress to liver cirrhosis and hepatocellular carcinoma (HCC)-one of the fastest-rising causes of cancer mortality in the United States. Despite the increased prevalence of liver fibrosis due to infections, exposure to toxins, and unhealthy lifestyles, there are no effective treatments available. Recent advances in nanomedicine can lead to more targeted and effective strategies for treating liver diseases than existing treatments. In particular, the use of biomimetic nanoparticles (NPs) such as liposomes and cell-membrane-coated NPs is of interest. NPs functionalized with cell membranes mimic the properties of the source cell used and provide inherent immune evasion ability, homologous adhesion, and prolonged circulation. This review explores the types of biomimetic coatings, different cargoes delivered through biomimetic NPs for various treatment modalities, and the type of core NPs used for targeting liver fibrosis and HCC.
Collapse
Affiliation(s)
- Veena Vijayan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Janitha M Unagolla
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Dhruvisha Panchal
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | - Judith Eloyi John
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
| | | | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island Kingston RI 02881 USA
- Department of Chemical Engineering, University of Rhode Island Kingston RI 02881 USA
| |
Collapse
|
14
|
Runtian Z, Wenqiang H, Zimeng S, Tianyu W, Jingquan Z. AEBP1 or ACLP, which is the key factor in inflammation and fibrosis? Int J Biol Macromol 2025; 310:143554. [PMID: 40294683 DOI: 10.1016/j.ijbiomac.2025.143554] [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: 02/01/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
Adipocyte enhancer-binding protein 1 (AEBP1) and Aortic carboxypeptidase-like protein (ACLP) are two protein isoforms produced by the AEBP1 gene. AEBP1, originally discovered in preadipocytes, functions as a transcriptional repressor and is involved in promoting inflammation, proliferation, and migration through various signaling pathways. ACLP is an extracellular matrix protein linked to Ehlers-Danlos syndrome, a genetic disorder characterized by defective connective tissue development. Structurally, AEBP1 and ACLP share many similarities, and both participate in critical physiological or pathological processes, such as cancer and fibrosis, by influencing pathways like NK-κB, WNT, and TGF-β. In recent years, research on AEBP1 and ACLP has expanded to include major organs such as the brain, kidneys, and lungs, with a particular focus on the cardiovascular system, where they show potential as novel drug targets. However, most studies do not clearly distinguish between AEBP1 and ACLP. For instance, AEBP1 is implicated in myocardial fibrosis in hypertrophic cardiomyopathy models, whereas ACLP is associated with fibrosis in other organs. Additionally, literature on the relationship between AEBP1 and fibrosis is often contradictory. Clarifying the distinct roles of AEBP1 and ACLP and their different functions in various cell types would greatly benefit further research. Current research suggests that the AEBP1 gene encodes two proteins, AEBP1 and ACLP, which have been reported to exhibit distinct functions in different studies. However, many studies do not differentiate between these two proteins, potentially leading to misconceptions. Therefore, we have conducted a comprehensive review of the existing literature to elucidate the functions of the AEBP1 gene and its encoded proteins in detail.
Collapse
Affiliation(s)
- Zhang Runtian
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Han Wenqiang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Shen Zimeng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Wang Tianyu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Zhong Jingquan
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China.
| |
Collapse
|
15
|
Becker J, Domenger C, Choksi P, Krämer C, Baumgartl C, Maiakovska O, Kim JJ, Weinmann J, Huber G, Schmidt F, Thirion C, Müller OJ, Willenbring H, Grimm D. Identification of a robust promoter in mouse and human hepatocytes by in vivo biopanning of a barcoded AAV library. Mol Ther 2025:S1525-0016(25)00301-6. [PMID: 40263935 DOI: 10.1016/j.ymthe.2025.04.027] [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: 01/21/2025] [Revised: 03/21/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are leading vectors for in vivo human gene therapy. An integral vector element is promoters, which control transgene expression in either a ubiquitous or cell-type-selective manner. Identifying optimal capsid-promoter combinations is challenging, especially when considering on- versus off-target expression. Here, we report a pipeline for in vivo promoter biopanning in AAV building on our AAV capsid barcoding technology and illustrate its potential by screening 53 promoters in 16 murine tissues using an AAV9 vector. Surprisingly, the 2.2-kb human glial fibrillary acidic protein (GFAP) promoter was the top hit in the liver, where it outperformed robust benchmarks such as the human α-1-antitrypsin promoter or the clinically used liver-specific promoter 1 (LP1). Analysis of hepatic cell populations revealed preferred GFAP promoter activity in hepatocytes. Notably, the GFAP promoter also surpassed the LP1 and cytomegalovirus promoters in human hepatocytes engrafted in an immune-deficient mouse. These findings establish the GFAP promoter as an exciting alternative for research and clinical applications requiring efficient and specific transgene expression in hepatocytes. Our pipeline expands the arsenal of technologies for high-throughput in vivo screening of viral vector components and is compatible with capsid barcoding, facilitating the combinatorial interrogation of complex AAV libraries.
Collapse
Affiliation(s)
- Jonas Becker
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Claire Domenger
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Pervinder Choksi
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Chiara Krämer
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Conradin Baumgartl
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Olena Maiakovska
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Jae-Jun Kim
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Jonas Weinmann
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | - Georg Huber
- Revvity Gene Delivery GmbH, 82166 Gräfelfing, Germany
| | - Florian Schmidt
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany
| | | | - Oliver J Müller
- Department of Internal Medicine V, University Hospital Schleswig-Holstein and University of Kiel, 24105 Kiel, Germany; German Centre for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, 24105 Kiel, Germany
| | - Holger Willenbring
- Department of Surgery, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty, University of Heidelberg, 69120 Heidelberg, Germany; BioQuant, Center for Integrative Infectious Diseases (CIID), University of Heidelberg, 69120 Heidelberg, Germany; German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Heidelberg, 69120 Heidelberg, Germany; Faculty of Engineering Sciences, University of Heidelberg, 69120 Heidelberg, Germany.
| |
Collapse
|
16
|
Ma X, Li B, Liu Y, Guo X. An inverted U-shaped association between high-sensitivity C-reactive protein and the albumin ratio and hepatic steatosis and liver fibrosis: a population-based study. Front Nutr 2025; 12:1534200. [PMID: 40303878 PMCID: PMC12037389 DOI: 10.3389/fnut.2025.1534200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Background The high-sensitivity C-reactive protein to albumin (CAR) ratio is a comprehensive measure of inflammation in vivo. Hepatic steatosis and fibrosis are significantly correlated with inflammation. The present study aimed to explore the possible associations between CAR and hepatic steatosis and fibrosis in the American population. Methods The study population involved the National Health and Nutrition Examination Survey (NHANES) participants from 2017 to 2020. The natural logarithm of CAR, calculated as Ln(CAR) with base "e," was used for further analyses. The relationships between Ln(CAR) and the controlled attenuation parameter (CAP) and between Ln(CAR) and liver stiffness measurement (LSM) were investigated through multivariate linear regression analysis. Interaction and subgroup analysis identified factors affecting these variables. Nonlinear relationships were elucidated by smoothing curves and threshold effect analysis. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive performance of the CAR for non-alcoholic fatty liver disease (NAFLD). The results were adjusted for U.S. population estimates. Results The study included a total of 7,404 individuals. Ln(CAR) was positively correlated with CAP in the fully adjusted model, with an effect value of β = 1.827 (95% CI, 0.611, 3.042). A more pronounced positive association was observed among participants with a BMI ≥ 25 kg/m2 in the subgroup analysis. An inverted U-shaped association was shown between Ln(CAR) and CAP through smooth curve fitting and a two-segment linear regression model, with an inflection point of (-9.594). ROC curve analysis showed that CAR had a moderate predictive value for NAFLD (AUC = 0.6895), with a sensitivity of 0.7276 and a specificity of 0.6092. No significant association was detected between Ln(CAR) and the LSM. Conclusion We demonstrate an inverted U-shaped relationship between Ln(CAR) and CAP risk within the U.S. demographic. Our results suggest that CAR may serve as a valuable diagnostic tool for NAFLD. Further prospective research is necessary to validate this conclusion.
Collapse
Affiliation(s)
| | | | | | - Xiaoyan Guo
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
17
|
Habibi M, Ferguson D, Eichler SJ, Chan MM, Fu C, Pietka TA, Bredemeyer AL, LaPoint A, Shew TM, He M, Liss KHH, Lutkewitte AJ, Cho K, Schilling JD, Patti GJ, Finck BN. A Critical Role for the Mitochondrial Pyruvate Carrier in Hepatic Stellate Cell Activation. Cell Mol Gastroenterol Hepatol 2025:101517. [PMID: 40239806 DOI: 10.1016/j.jcmgh.2025.101517] [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: 05/20/2024] [Revised: 04/04/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND & AIMS Hepatic stellate cells (HSCs) are non-parenchymal cells of the liver that produce the extracellular matrix that forms fibrotic lesions in chronic liver disease, including metabolic dysfunction-associated steatohepatitis (MASH). The mitochondrial pyruvate carrier (MPC) catalyzes the transport of pyruvate from the cytosol into the mitochondrial matrix, which is a critical step in pyruvate metabolism. An MPC inhibitor has shown promise as a novel therapeutic for MASH and HSC activation, but a mechanistic understanding of the direct effects of MPC inhibition on HSC activation is lacking. METHODS Stable lines of LX2 cells expressing short hairpin RNA against MPC2 were established and examined in a series of studies to assess HSC metabolism and activation. Mice with conditional, HSC-specific MPC2 deletion were generated and their phenotypes assessed in the context of diets that cause hepatic steatosis, injury, and early-stage fibrosis. RESULTS Genetic suppression of MPC activity markedly decreased expression of markers of HSC activation in vitro. MPC knockdown reduced the abundance of several intermediates of the tricarboxylic acid cycle and attenuated HSC activation by suppressing hypoxia inducible factor-1α signaling. Supplementing alpha-ketoglutarate to replenish the tricarboxylic acid cycle intermediates was sufficient to overcome the effects of MPC inhibition on hypoxia inducible factor-1α and HSC activation. On high-fat diets, mice with HSC-specific MPC deletion exhibited reduced circulating transaminases, numbers of HSCs, and hepatic expression of markers of HSC activation and inflammation compared with wild-type mice. CONCLUSIONS These data suggest that MPC inhibition modulates HSC metabolism to attenuate activation and illuminate mechanisms by which MPC inhibitors could prove therapeutically beneficial for treating MASH.
Collapse
Affiliation(s)
- Mohammad Habibi
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel Ferguson
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Sophie J Eichler
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mandy M Chan
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Christina Fu
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Terri A Pietka
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Andrea L Bredemeyer
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew LaPoint
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Trevor M Shew
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Mai He
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Kim H H Liss
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri; (4)Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Andrew J Lutkewitte
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kevin Cho
- Department of Chemistry, Washington University, St. Louis, Missouri
| | - Joel D Schilling
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Gary J Patti
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Chemistry, Washington University, St. Louis, Missouri
| | - Brian N Finck
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
18
|
Wang SS, Yuan J, Tang XT, Yin X, Fang K, Chen LV, Ren Z, Zhou BO. Periductal fibroblasts participate in liver homeostasis, fibrosis, and tumorigenesis. J Exp Med 2025; 222:e20232123. [PMID: 39888328 PMCID: PMC11784584 DOI: 10.1084/jem.20232123] [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: 11/17/2023] [Revised: 09/20/2024] [Accepted: 01/08/2025] [Indexed: 02/01/2025] Open
Abstract
Hepatic fibroblasts comprise groups of stromal cells in the liver that are phenotypically distinct from hepatic stellate cells. However, their physiology is poorly understood. By single-cell RNA sequencing, we identified Cd34 and Dpt as hepatic fibroblast-specific genes. Cd34-CreER labeled periportal-venous and periductal fibroblasts, but few pericentral-venous fibroblasts. Cd34+ fibroblasts generated ∼25% of myofibroblasts in periportal fibrosis and ∼40% of cancer-associated fibroblasts (CAFs) in intrahepatic cholangiocarcinoma (ICC). Myofibroblast formation by Cd34+ fibroblasts required Tgfbr2. Depletion of Cd34+ fibroblasts increased the frequency of the ductal epithelial cells under homeostasis and accelerated the progression of ICC. Dpt-CreER labeled periportal- and pericentral-venous fibroblasts, but much less periductal fibroblasts. Dpt+ cells generated ∼15% of myofibroblasts in periportal fibrosis, but few myofibroblasts in pericentral fibrosis or CAFs in ICC. Thus, an orthogonal combination of Cd34-CreER and Dpt-CreER dissected the fates of periductal, periportal-venous, and pericentral-venous fibroblasts. Both periductal and periportal-venous fibroblasts contribute to liver fibrosis. Periductal fibroblasts also contribute to ductal homeostasis and ICC progression.
Collapse
Affiliation(s)
- Shan-Shan Wang
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Yuan
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xinyu Thomas Tang
- Key Laboratory of Multi-Cell System, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiujuan Yin
- Key Laboratory of Multi-Cell System, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Ke Fang
- Key Laboratory of Multi-Cell System, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Lin Veronica Chen
- Key Laboratory of Multi-Cell System, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhenggang Ren
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Bo O. Zhou
- Key Laboratory of Multi-Cell System, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
19
|
Jiao X, Lai L, Sun B, Qian Y, Yang W. The transcription factor mesenchyme homeobox 1 exacerbates hepatic fibrosis by transcriptional activation of connective tissue growth factor. Exp Cell Res 2025; 447:114513. [PMID: 40073959 DOI: 10.1016/j.yexcr.2025.114513] [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: 12/03/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
As a chronic condition, liver fibrosis is characterized by diverse etiological factors, and the pivotal event to its pathogenesis is the activation of quiescent hepatic stellate cells (HSCs) into myofibroblasts. Mesenchyme homeobox 1 (MEOX1) is a transcription factors central to cellular development and differentiation. However, the role of MEOX1 signaling in hepatic fibrosis still remains largely unknown. In this study, we investigated the potential role and mechanism of MEOX1 in liver fibrosis using different models in vivo and in vitro. The hepatic expression of MEOX1 exhibited a positive correlation with the degree of fibrosis in patients diagnosed with non-alcoholic steatohepatitis (NASH), as determined through bioinformatics analysis. Furthermore, MEOX1 demonstrated high expression levels in activated HSCs and fibrotic liver tissues induced by methionine and choline-deficient diet (MCD), thioacetamide (TAA), or carbon tetrachloride (CCl4) treatment in C57/BL6 mice. Mechanistically, MEOX1 facilitated HSC activation, proliferation, and migration. The comprehensive analysis of transcriptome sequencing and chromatin immunoprecipitation sequencing data revealed that connective tissue growth factor (CTGF) served as a target gene for MEOX1 in HSCs. Specifically, MEOX1 bound to the promoter region of CTGF and enhanced its transcriptional activity, thereby mediating the exacerbating effect of MEOX1 on hepatic fibrosis. In conclusion, our current findings elucidate the role of MEOX1 in exacerbating hepatic fibrosis progression through transcriptional activation of CTGF. Our findings provide valuable insights into the therapeutic potential of targeting MEOX1 for the treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Jiao
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Linying Lai
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Bo Sun
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Yiting Qian
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China
| | - Wenzhuo Yang
- Department of Gastroenterology and Hepatology, Institute of Digestive Disease, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, PR China.
| |
Collapse
|
20
|
Sugimoto A, Saito Y, Wang G, Sun Q, Yin C, Lee KH, Geng Y, Rajbhandari P, Hernandez C, Steffani M, Qie J, Savage T, Goyal DM, Ray KC, Neelakantan TV, Yin D, Melms J, Lehrich BM, Yasaka TM, Liu S, Oertel M, Lan T, Guillot A, Peiseler M, Filliol A, Kanzaki H, Fujiwara N, Ravi S, Izar B, Brosch M, Hampe J, Remotti H, Argemi J, Sun Z, Kendall TJ, Hoshida Y, Tacke F, Fallowfield JA, Blockley-Powell SK, Haeusler RA, Steinman JB, Pajvani UB, Monga SP, Bataller R, Masoodi M, Arpaia N, Lee YA, Stockwell BR, Augustin HG, Schwabe RF. Hepatic stellate cells control liver zonation, size and functions via R-spondin 3. Nature 2025; 640:752-761. [PMID: 40074890 PMCID: PMC12003176 DOI: 10.1038/s41586-025-08677-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 01/21/2025] [Indexed: 03/14/2025]
Abstract
Hepatic stellate cells (HSCs) have a central pathogenetic role in the development of liver fibrosis. However, their fibrosis-independent and homeostatic functions remain poorly understood1-5. Here we demonstrate that genetic depletion of HSCs changes WNT activity and zonation of hepatocytes, leading to marked alterations in liver regeneration, cytochrome P450 metabolism and injury. We identify R-spondin 3 (RSPO3), an HSC-enriched modulator of WNT signalling, as responsible for these hepatocyte-regulatory effects of HSCs. HSC-selective deletion of Rspo3 phenocopies the effects of HSC depletion on hepatocyte gene expression, zonation, liver size, regeneration and cytochrome P450-mediated detoxification, and exacerbates alcohol-associated and metabolic dysfunction-associated steatotic liver disease. RSPO3 expression decreases with HSC activation and is inversely associated with outcomes in patients with alcohol-associated and metabolic dysfunction-associated steatotic liver disease. These protective and hepatocyte-regulating functions of HSCs via RSPO3 resemble the R-spondin-expressing stromal niche in other organs and should be integrated into current therapeutic concepts.
Collapse
Affiliation(s)
- Atsushi Sugimoto
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Yoshinobu Saito
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Guanxiong Wang
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Qiuyan Sun
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Chuan Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Ki Hong Lee
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yana Geng
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Presha Rajbhandari
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Celine Hernandez
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Marcella Steffani
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Jingran Qie
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Thomas Savage
- Department of Microbiology & Immunology, Columbia University, New York, NY, USA
| | - Dhruv M Goyal
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Kevin C Ray
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Taruna V Neelakantan
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
| | - Deqi Yin
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Johannes Melms
- Department of Medicine, Columbia University, New York, NY, USA
| | - Brandon M Lehrich
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tyler M Yasaka
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Silvia Liu
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Oertel
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tian Lan
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Aveline Filliol
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Hiroaki Kanzaki
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Naoto Fujiwara
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samhita Ravi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Benjamin Izar
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Mario Brosch
- Department of Internal Medicine I, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Jochen Hampe
- Department of Internal Medicine I, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Helen Remotti
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Josepmaria Argemi
- Liver Unit and RNA Biology and Therapies Program, Cima Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, Madrid, Spain
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Timothy J Kendall
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Yujin Hoshida
- Liver Tumour Translational Research Program, Harold C. Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Storm K Blockley-Powell
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | - Rebecca A Haeusler
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
| | | | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, NY, USA
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Institute of Human Nutrition, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, Pittsburgh Liver Research Center, and Organ Pathobiology and Therapeutics Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ramon Bataller
- Liver Unit,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Nicholas Arpaia
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Columbia Center for Translational Immunology, Department of Medicine, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Youngmin A Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Brent R Stockwell
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA
- Department of Biological Sciences and Department of Chemistry, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis Research, German Cancer Research Center, Heidelberg, Germany.
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY, USA.
- Columbia University Digestive and Liver Disease Research Center, New York, NY, USA.
- Department of Hepatology & Gastroenterology, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- Institute of Human Nutrition, New York, NY, USA.
- Burch-Lodge Center for Human Longevity, Columbia University, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, New York, NY, USA.
| |
Collapse
|
21
|
Carra D, Maas SCE, Seoane JA, Alonso-Curbelo D. Exposomal determinants of non-genetic plasticity in tumor initiation. Trends Cancer 2025; 11:295-308. [PMID: 40023688 DOI: 10.1016/j.trecan.2025.01.010] [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: 11/26/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 03/04/2025]
Abstract
The classical view of cancer as a genetically driven disease has been challenged by recent findings of oncogenic mutations in phenotypically healthy tissues, refocusing attention on non-genetic mechanisms of tumor initiation. In this context, gene-environment interactions take the stage, with recent studies showing how they unleash and redirect cellular and tissue plasticity towards protumorigenic states in response to the exposome, the ensemble of environmental factors impinging on tissue homeostasis. We conceptualize tumor-initiating plasticity as a phenotype-transforming force acting at three levels: cell-intrinsic, focusing on mutant epithelial cells' responses to environmental variation; reprogramming of non-neoplastic cells of the host, leading to protumor micro- and macroenvironments; and microbiome ecosystem dynamics. This perspective highlights cell, tissue, and organismal plasticity mechanisms underlying tumor initiation that are shaped by the exposome, and how their functional investigation may provide new opportunities to prevent, detect, and intercept cancer-promoting plasticity.
Collapse
Affiliation(s)
- Davide Carra
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Silvana C E Maas
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Direna Alonso-Curbelo
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| |
Collapse
|
22
|
Gupta V, Sehrawat TS, Pinzani M, Strazzabosco M. Portal Fibrosis and the Ductular Reaction: Pathophysiological Role in the Progression of Liver Disease and Translational Opportunities. Gastroenterology 2025; 168:675-690. [PMID: 39251168 PMCID: PMC11885590 DOI: 10.1053/j.gastro.2024.07.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/27/2024] [Accepted: 07/20/2024] [Indexed: 09/11/2024]
Abstract
A consistent feature of chronic liver diseases and the hallmark of pathologic repair is the so-called "ductular reaction." This is a histologic abnormality characterized by an expansion of dysmorphic cholangiocytes inside and around portal spaces infiltrated by inflammatory, mesenchymal, and vascular cells. The ductular reaction is a highly regulated response based on the reactivation of morphogenetic signaling mechanisms and a complex crosstalk among a multitude of cell types. The nature and mechanism of these exchanges determine the difference between healthy regenerative liver repair and pathologic repair. An orchestrated signaling among cell types directs mesenchymal cells to deposit a specific extracellular matrix with distinct physical and biochemical properties defined as portal fibrosis. Progression of fibrosis leads to vast architectural and vascular changes known as "liver cirrhosis." The signals regulating the ecology of this microenvironment are just beginning to be addressed. Contrary to the tumor microenvironment, immune modulation inside this "benign" microenvironment is scarcely known. One of the reasons for this is that both the ductular reaction and portal fibrosis have been primarily considered a manifestation of cholestatic liver disease, whereas this phenomenon is also present, albeit with distinctive features, in all chronic human liver diseases. Novel human-derived cellular models and progress in "omics" technologies are increasing our knowledge at a fast pace. Most importantly, this knowledge is on the edge of generating new diagnostic and therapeutic advances. Here, we will critically review the latest advances, in terms of mechanisms, pathophysiology, and treatment prospects. In addition, we will delineate future avenues of research, including innovative translational opportunities.
Collapse
Affiliation(s)
- Vikas Gupta
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Tejasav S Sehrawat
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Massimo Pinzani
- UCL Institute for Liver & Digestive Health, Royal Free Hospital, London, United Kingdom; University of Pittsburgh Medical Center-Mediterranean Institute for Transplantation and Highly Specialized Therapies, Palermo, Italy
| | - Mario Strazzabosco
- Liver Center and Section of Digestive Diseases, Department of Internal Medicine, Yale University, New Haven, Connecticut.
| |
Collapse
|
23
|
Ni X, Wei Z, Peng Y, Zheng L, Shang J, Liu F, Li Y, Liu J. Triclosan exposure induces liver fibrosis in mice: The heterogeneous nuclear ribonucleoprotein A1/pyruvate kinase M2 axis drives hepatic stellate cell activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 294:118113. [PMID: 40157328 DOI: 10.1016/j.ecoenv.2025.118113] [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: 11/07/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Triclosan (TCS) is an effective broad-spectrum antibacterial agent. TCS possesses a stable structure, can easily accumulate in the environment, and may have numerous negative impacts on human health. One organ particularly susceptible to TCS damage is the liver; however, the molecular mechanisms underlying TCS-induced liver damage remain unclear. A long-term TCS exposure model was established in C57BL/6 mice through maternal administration from gestation to postnatal 8-week-old. The offspring were randomly assigned to three groups (0, 50, and 100 mg/kg TCS) with six animals per group, ensuring an equal gender distribution (3 males and 3 females). The results showed that TCS-exposed mice exhibited serum aspartate aminotransferase, alanine aminotransferase, and alkaline phosphatase enzyme activities increased by 1.5-2 times when compared with vehicle-treated mice, along with features of liver fibrosis. In the LX-2 cell line, used as an in vitro model, TCS promoted proliferation and migration and induced the activation of hepatic stellate cells (HSCs). The level of pyruvate kinase M2 (PKM2) dimer increased by 200 % in LX-2 cells treated with TCS. PKM2 dimer overexpression stimulated HSC activation, whereas treatment with TEPP-46 (a PKM2 dimer inhibitor) significantly decreased the activation process. The expression of heterogeneous ribonucleoprotein particle A1 (hnRNPA1) was upregulated in the TCS treatment group and promoted the PKM2 expression. Moreover, disruption of the hnRNPA1/PKM2 axis reduced HSC proliferation and migration activated by TCS. Overall, our findings highlighted that TCS could cause liver fibrosis by stimulating the proliferation and migration of HSCs activated via the hnRNPA1/PKM2 axis, providing promising treatment options for TCS-related liver damage.
Collapse
Affiliation(s)
- Xiao Ni
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Ziyun Wei
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yuxuan Peng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Linlin Zheng
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Jianing Shang
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Fu Liu
- Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China
| | - Yunwei Li
- Department of Anorectal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, PR China.
| | - Jieyu Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, PR China; Department of Health Laboratory Technology, School of Public Health, China Medical University, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
24
|
Hansen D, Jensen JER, Andersen CAT, Jakobsgaard PR, Havelund J, Lauritsen L, Mandacaru S, Siersbaek M, Shackleton OL, Inoue H, Brewer JR, Schwabe RF, Blagoev B, Færgeman NJ, Salmi M, Ravnskjaer K. Hepatic stellate cells regulate liver fatty acid utilization via plasmalemma vesicle-associated protein. Cell Metab 2025; 37:971-986.e8. [PMID: 40037362 DOI: 10.1016/j.cmet.2025.01.022] [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: 12/12/2023] [Revised: 11/26/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025]
Abstract
The liver is essential for normal fatty acid utilization during fasting. Circulating fatty acids are taken up by hepatocytes and esterified as triacylglycerols for either oxidative metabolization and ketogenesis or export. Whereas the regulation of fatty acid oxidation in hepatocytes is well understood, the uptake and retention of non-esterified fatty acids by hepatocytes is not. Here, we show that murine hepatic stellate cells (HSCs) and their abundantly expressed plasmalemma vesicle-associated protein (PLVAP) control hepatic substrate preference for fasting energy metabolism. HSC-specific ablation of PLVAP in mice elevated hepatic insulin signaling and improved glucose tolerance. Fasted HSC PLVAP knockout mice showed suppressed hepatic fatty acid esterification into di- and triacylglycerols, shifting fasting metabolism from fatty acid oxidation to reliance on carbohydrates. By super-resolution microscopy, we localized HSC PLVAP to caveolae residing along the sinusoidal lumen, supporting a role for HSCs and PLVAP-diaphragmed caveolae in normal fasting metabolism of the liver.
Collapse
Affiliation(s)
- Daniel Hansen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Jasmin E R Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Christian A T Andersen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Peter R Jakobsgaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Jesper Havelund
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Line Lauritsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Samuel Mandacaru
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Majken Siersbaek
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Oliver L Shackleton
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa 920-8641, Ishikawa, Japan
| | - Jonathan R Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Robert F Schwabe
- Department of Medicine, Columbia University, New York, NY 10032, USA; Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Blagoy Blagoev
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Marko Salmi
- MediCity Research Laboratory, University of Turku, 20014 Turku, Finland; Institute of Biomedicine, University of Turku, 20014 Turku, Finland; InFLAMES Research Flagship Centre, University of Turku, 20014 Turku, Finland
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark; Center for Functional Genomics and Tissue Plasticity (ATLAS), University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|
25
|
Xiong H, Guo J. Targeting Hepatic Stellate Cells for the Prevention and Treatment of Liver Cirrhosis and Hepatocellular Carcinoma: Strategies and Clinical Translation. Pharmaceuticals (Basel) 2025; 18:507. [PMID: 40283943 PMCID: PMC12030350 DOI: 10.3390/ph18040507] [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: 02/19/2025] [Revised: 03/24/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Hepatic stellate cells (HSC) are the major source of myofibroblasts (MFB) in fibrosis and cancer- associated fibroblasts (CAF) in both primary and metastatic liver cancer. Over the past few decades, there has been significant progress in understanding the cellular and molecular mechanisms by which liver fibrosis and HCC occur, as well as the key roles of HSC in their pathogenesis. HSC-targeted approaches using specific surface markers and receptors may enable the selective delivery of drugs, oligonucleotides, and therapeutic peptides that exert optimized anti-fibrotic and anti-HCC effects. Recent advances in omics, particularly single-cell sequencing and spatial transcriptomics, hold promise for identifying new HSC targets for diagnosing and treating liver fibrosis/cirrhosis and liver cancer.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Diseases, Fudan University, Shanghai 200032, China;
- Department of Internal Medicine, Shanghai Medical College, Fu Dan University, Shanghai 200032, China
| | - Jinsheng Guo
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Shanghai Institute of Liver Diseases, Fudan University, Shanghai 200032, China;
- Department of Internal Medicine, Shanghai Medical College, Fu Dan University, Shanghai 200032, China
| |
Collapse
|
26
|
Cheng Y, Song T, Yao J, Wang Q, Meng C, Feng F. Study on the mechanism of hsa_circ_0074763 regulating the miR-3667-3P/ACSL4 axis in liver fibrosis. Sci Rep 2025; 15:10548. [PMID: 40148434 PMCID: PMC11950437 DOI: 10.1038/s41598-025-91393-2] [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: 08/09/2024] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
This study aimed to investigate the involvement of hsa_circ_0074763 in the activation of HSCs (hepatic stellate cells ) and liver fibrosis. Additionally, it aimed to conduct a preliminary analysis of the molecular mechanism targeting miR-3667-3p/ACSL4 (Long-chain acyl-CoA synthetase 4), thereby providing novel molecular targets for liver fibrosis. The GEO database was utilized to identify differentially expressed hsa_circ_0074763 and determined its subcellular localization in LX-2 cells using fluorescence in situ hybridization. Bioinformatics analysis was employed for result prediction, and the interaction between hsa_circ_0074763 and miR-3667-3P was confirmed using dual-luciferase reporter gene assay. ACSL4 mediated ferroptosis was detected with kit. Hsa_circ_0074763 exhibits high expression levels in the fibrosis model. Validation through dual-luciferase reporter gene assays confirms the interaction between hsa_circ_0074763 and miR-3667-3P. Functional cell experiments demonstrate that overexpression of hsa_circ_0074763 promotes proliferation of LX-2 cells, elevates inflammation levels, and inhibits apoptosis. Additionally, ACSL4 has been identified as a direct target of miR-3667-3P, with overexpression of hsa_circ_0074763 counteracting the inhibitory effect on ACSL4 by suppressing miR-3667-3P. Overexpression of ACSL4 increased the expression levels of ROS (Lipid Oxidation), Iron (Ferro Orange) and MDA (Malondialdehyde), and decreased the expression levels of GPX4 (Glutathione peroxidase 4) and GSH (Glutathione). Our finding suggests that overexpression of hsa_circ_0074763 likely enhances the HSC activation through modulation of the miR-3667-3P/ACSL4 axis. Therefore, hsa_circ_0074763 holds potential as a therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Yanling Cheng
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Tiantian Song
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Jiachen Yao
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Qirong Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Chunyan Meng
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China.
| | - Fumin Feng
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China.
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, Tangshan, China.
- Hebei Coordinated Innovation Center of Occupational Health and Safety, Tangshan, China.
| |
Collapse
|
27
|
Ning M, Lu D, Liang D, Ren PG. Single-cell RNA sequencing advances in revealing the development and progression of MASH: the identifications and interactions of non-parenchymal cells. Front Mol Biosci 2025; 12:1513993. [PMID: 40201243 PMCID: PMC11976672 DOI: 10.3389/fmolb.2025.1513993] [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: 10/19/2024] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Developing drugs for the treatment of Metabolic Associated Steatohepatitis (MASH) has always been a significant challenge. Researchers have been dedicated to exploring drugs and therapeutic strategies to alleviate disease progression, but treatments remain limited. This is partly due to the complexity of the pathophysiological processes, and inadequate knowledge of the cellular and molecular mechanisms in MASH. Especially, the liver non-parenchymal cells (NPCs) like Kupffer cells, hepatic stellate cells and sinusoidal endothelial cells which play critical roles in live function, immune responses, fibrosis and disease progression. Deciphering how these cells function in MASH, would help understand the pathophysiological processes and find potential drug targets. In recent years, new technologies have been developed for single-cell transcriptomic sequencing, making cell-specific transcriptome profiling a reality in healthy and diseased livers. In this review, we discussed how the use of single-cell transcriptomic sequencing provided us with an in-depth understanding of the heterogeneous, cellular interactions among non-parenchymal cells and tried to highlight recent discoveries in MASH by this technology. It is hoped that the summarized features and markers of various subclusters in this review could provide a technical reference for further experiments and a theoretical basis for clinical applications.
Collapse
Affiliation(s)
- Meng Ning
- Department of Endocrinology, Peking University Shenzhen Hospital, Shenzhen, China
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Endocrinology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Donghui Lu
- Department of Endocrinology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dong Liang
- Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Pei-Gen Ren
- Center for Cancer Immunology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
28
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [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: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
29
|
Pita-Juarez Y, Karagkouni D, Kalavros N, Melms JC, Niezen S, Delorey TM, Essene AL, Brook OR, Pant D, Skelton-Badlani D, Naderi P, Huang P, Pan L, Hether T, Andrews TS, Ziegler CGK, Reeves J, Myloserdnyy A, Chen R, Nam A, Phelan S, Liang Y, Gregory M, He S, Patrick M, Rane T, Wardhani A, Amin AD, Biermann J, Hibshoosh H, Veregge M, Kramer Z, Jacobs C, Yalcin Y, Phillips D, Slyper M, Subramanian A, Ashenberg O, Bloom-Ackermann Z, Tran VM, Gomez J, Sturm A, Zhang S, Fleming SJ, Warren S, Beechem J, Hung D, Babadi M, Padera RF, MacParland SA, Bader GD, Imad N, Solomon IH, Miller E, Riedel S, Porter CBM, Villani AC, Tsai LTY, Hide W, Szabo G, Hecht J, Rozenblatt-Rosen O, Shalek AK, Izar B, Regev A, Popov YV, Jiang ZG, Vlachos IS. A single-nucleus and spatial transcriptomic atlas of the COVID-19 liver reveals topological, functional, and regenerative organ disruption in patients. Genome Biol 2025; 26:56. [PMID: 40087773 PMCID: PMC11907808 DOI: 10.1186/s13059-025-03499-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/07/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND The molecular underpinnings of organ dysfunction in severe COVID-19 and its potential long-term sequelae are under intense investigation. To shed light on these in the context of liver function, we perform single-nucleus RNA-seq and spatial transcriptomic profiling of livers from 17 COVID-19 decedents. RESULTS We identify hepatocytes positive for SARS-CoV-2 RNA with an expression phenotype resembling infected lung epithelial cells, and a central role in a pro-fibrotic TGFβ signaling cell-cell communications network. Integrated analysis and comparisons with healthy controls reveal extensive changes in the cellular composition and expression states in COVID-19 liver, providing the underpinning of hepatocellular injury, ductular reaction, pathologic vascular expansion, and fibrogenesis characteristic of COVID-19 cholangiopathy. We also observe Kupffer cell proliferation and erythrocyte progenitors for the first time in a human liver single-cell atlas. Despite the absence of a clinical acute liver injury phenotype, endothelial cell composition is dramatically impacted in COVID-19, concomitantly with extensive alterations and profibrogenic activation of reactive cholangiocytes and mesenchymal cells. CONCLUSIONS Our atlas provides novel insights into liver physiology and pathology in COVID-19 and forms a foundational resource for its investigation and understanding.
Collapse
Affiliation(s)
- Yered Pita-Juarez
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dimitra Karagkouni
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nikolaos Kalavros
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Spatial Technologies Unit, HMS Initiative for RNA Medicine / Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Johannes C Melms
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Sebastian Niezen
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Toni M Delorey
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adam L Essene
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Olga R Brook
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Deepti Pant
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Disha Skelton-Badlani
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Pourya Naderi
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Pinzhu Huang
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Liuliu Pan
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Tallulah S Andrews
- Ajmera Transplant Centre, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Carly G K Ziegler
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Program in Computational & Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Andriy Myloserdnyy
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rachel Chen
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Andy Nam
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Yan Liang
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Shanshan He
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Tushar Rane
- NanoString Technologies, Inc., Seattle, WA, USA
| | | | - Amit Dipak Amin
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Jana Biermann
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Center for Translational Immunology, New York, NY, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Molly Veregge
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Zachary Kramer
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Christopher Jacobs
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Yusuf Yalcin
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Devan Phillips
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Michal Slyper
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | | | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zohar Bloom-Ackermann
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Victoria M Tran
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - James Gomez
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexander Sturm
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shuting Zhang
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephen J Fleming
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Deborah Hung
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Mehrtash Babadi
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Precision Cardiology Laboratory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Sonya A MacParland
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA
- Department of Immunology, University of Toronto, Medical Sciences Building, 1 King's College Circle, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, Toronto, ON, Canada
| | - Nasser Imad
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Isaac H Solomon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Eric Miller
- NanoString Technologies, Inc., Seattle, WA, USA
| | - Stefan Riedel
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Caroline B M Porter
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandra-Chloé Villani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Linus T-Y Tsai
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Boston Nutrition and Obesity Research Center Functional Genomics and Bioinformatics Core, Boston, MA, USA
| | - Winston Hide
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Gyongyi Szabo
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jonathan Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA
| | - Alex K Shalek
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Program in Health Sciences & Technology, Harvard Medical School & Massachusetts Institute of Technology, Boston, MA, USA.
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
- Program in Computational & Systems Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA.
- Columbia Center for Translational Immunology, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA.
- Program for Mathematical Genomics, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, USA.
| | - Yury V Popov
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Z Gordon Jiang
- Harvard Medical School, Boston, MA, USA.
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | - Ioannis S Vlachos
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Spatial Technologies Unit, HMS Initiative for RNA Medicine / Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Cancer Research Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Harvard Medical School Initiative for RNA Medicine, Boston, MA, USA.
| |
Collapse
|
30
|
Manns MP, Bergquist A, Karlsen TH, Levy C, Muir AJ, Ponsioen C, Trauner M, Wong G, Younossi ZM. Primary sclerosing cholangitis. Nat Rev Dis Primers 2025; 11:17. [PMID: 40082445 DOI: 10.1038/s41572-025-00600-x] [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: 02/07/2025] [Indexed: 03/16/2025]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic biliary inflammation associated with periductular fibrosis of the intrahepatic and extrahepatic bile ducts leading to strictures, bacterial cholangitis, decompensated liver disease and need for liver transplantation. This rare focal liver disease affects all races and ages, with a predominance of young males. There is an up to 88% association with inflammatory bowel disease. Although the aetiology is unknown and the pathophysiology is poorly understood, PSC is regarded as an autoimmune liver disease based on a strong immunogenetic background. Further, the associated risk for various malignancies, particularly cholangiocellular carcinoma, is also poorly understood. No medical therapy has been approved so far nor has been shown to improve transplant-free survival. However, ursodeoxycholic acid is widely used since it improves the biochemical parameters of cholestasis and is safe at low doses. MRI of the biliary tract is the primary imaging technology for diagnosis. Endoscopic interventions of the bile ducts should be limited to clinically relevant strictures for balloon dilatation, biopsy and brush cytology. End-stage liver disease with decompensation is an indication for liver transplantation with recurrent PSC in up to 38% of patients. Several novel therapeutic strategies are in various stages of development, including apical sodium-dependent bile acid transporter and ileal bile acid transporter inhibitors, integrin inhibitors, peroxisome proliferator-activated receptor agonists, CCL24 blockers, recombinant FGF19, CCR2/CCR5 inhibitors, farnesoid X receptor bile acid receptor agonists, and nor-ursodeoxycholic acid. Manipulation of the gut microbiome includes faecal microbiota transplantation. This article summarizes present knowledge and defines unmet medical needs to improve quality of life and survival.
Collapse
Affiliation(s)
- Michael P Manns
- Hannover Medical School (MHH) and Centre for Individualised Infection Medicine (CiiM), Hannover, Germany.
| | - Annika Bergquist
- Division of Hepatology, Department of Upper Gastrointestinal Disease, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Clinic of Surgery and Specialized medicine, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cynthia Levy
- Division of Digestive Health and Liver Diseases, University of Miami School of Medicine, Miami, FL, USA
| | - Andrew J Muir
- Division of Gastroenterology, Duke University School of Medicine, Durham, NC, USA
| | - Cyriel Ponsioen
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Grace Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Union Hospital, Hong Kong SAR, China
| | | |
Collapse
|
31
|
Li W, Sparks RP, Sun C, Yang Y, Pantano L, Kirchner R, Arghiani N, Weilheimer A, Toles BJ, Chen JY, Moran SP, Barrera V, Li Z, Zhou P, Brassil ML, Wrobel D, Ho Sui SJ, Aspnes G, Schuler M, Smith J, Medoff BD, Zhou C, Boustany-Kari CM, Rippmann JF, Santos DM, Doerner JF, Mullen AC. Screening the human druggable genome identifies ABHD17B as an anti-fibrotic target in hepatic stellate cells. Nat Commun 2025; 16:2109. [PMID: 40025044 PMCID: PMC11873113 DOI: 10.1038/s41467-025-56900-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/03/2025] [Indexed: 03/04/2025] Open
Abstract
Hepatic stellate cells (HSCs) are activated with chronic liver injury and transdifferentiate into myofibroblasts, which produce excessive extracellular matrices that form the fibrotic scar. While the progression of fibrosis is understood to be the cause of end-stage liver disease, there are no approved therapies directed at interfering with the activity of HSC myofibroblasts. Here, we perform a high-throughput small interfering RNA (siRNA) screen in primary human HSC myofibroblasts to identify gene products necessary for the fibrotic phenotype of HSCs. We find that depletion of ABHD17B promotes the inactivation of HSCs, characterized by reduced COL1A1 and ACTA2 expression and accumulation of lipid droplets. Mice deficient in Abhd17b are also protected from fibrosis in the setting of in vivo liver injury. While ABHD17B is a depalmitoylase, our data suggest that ABHD17B promotes fibrosis through pathways independent of depalmitoylation that include interaction with MYO1B to modulate gene expression and HSC migration. Together, our results provide an analysis of the phenotypic consequences for siRNAs targeting RNAs from >9500 genes in primary human HSCs and identify ABHD17B as a potential therapeutic target to inhibit liver fibrosis.
Collapse
Affiliation(s)
- Wenyang Li
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Robert P Sparks
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Cheng Sun
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yang Yang
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Lorena Pantano
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Rory Kirchner
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Nahid Arghiani
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Arden Weilheimer
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Benjamin J Toles
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jennifer Y Chen
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Liver Center, Department of Medicine, University of California, San Francisco, CA, USA
| | - Sean P Moran
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Victor Barrera
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zixiu Li
- Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Peng Zhou
- Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Meghan L Brassil
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA, USA
- UMass Cancer Center, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - David Wrobel
- ICCB-Longwood Screening Facility, Harvard Medical School, Boston, MA, USA
| | - Shannan J Ho Sui
- Harvard Chan Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gary Aspnes
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riss, Biberach, Germany
| | - Michael Schuler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riss, Biberach, Germany
| | - Jennifer Smith
- ICCB-Longwood Screening Facility, Harvard Medical School, Boston, MA, USA
| | - Benjamin D Medoff
- Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Chan Zhou
- Population and Quantitative Health Sciences, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | - Jörg F Rippmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riss, Biberach, Germany
| | | | - Julia F Doerner
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach a.d. Riss, Biberach, Germany
| | - Alan C Mullen
- Division of Gastroenterology, Massachusetts General Hospital, Boston, MA, USA.
- Division of Gastroenterology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
- Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
32
|
Tuffs C, Dupovac M, Richter K, Holten S, Schaschinger T, Marg O, Poljo A, Tasdemir AN, Harnoss JM, Billeter A, Schneider M, Strowitzki MJ. Genetic Loss of HIF-Prolyl-Hydroxylase 1, but Not Pharmacological Inhibition, Mitigates Hepatic Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:480-493. [PMID: 39566823 DOI: 10.1016/j.ajpath.2024.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 10/09/2024] [Accepted: 10/16/2024] [Indexed: 11/22/2024]
Abstract
Liver fibrosis is characterized by excessive deposition of extracellular matrix due to chronic inflammation of the liver. Hepatic stellate cells (HSCs) become activated and produce increased amounts of extracellular matrix. Loss of HIF-prolyl-hydroxylase 1 (PHD1) attenuates HSC activation and fibrotic tissue remodeling in a murine model of biliary liver fibrosis. Herein, the protective effect of PHD1 deficiency (PHD1-/-) in an additional (toxic) model of liver fibrosis was validated and the effect of dimethyloxalylglycine (DMOG), a pan-HIF-prolyl-hydroxylase inhibitor, on the development of liver fibrosis, was evaluated. Liver fibrosis was induced utilizing carbon tetrachloride in wild-type (WT) and PHD1-/- mice treated with either vehicle or DMOG. To assess fibrosis development, expression of profibrotic genes in the livers was analyzed by Sirius red staining. When compared with WT mice, PHD1-/- mice developed less-severe liver fibrosis. DMOG treatment did not prevent this liver fibrosis. PHD1-/- mice had fewer α-SMA+ cells and less macrophage infiltration compared with WT mice. Expression of profibrogenic and proinflammatory genes was reduced in livers from carbon tetrachloride-exposed PHD1-/- mice. In vitro analyses of PHD1-deficient human HSCs revealed attenuated mRNA levels of profibrotic genes, as well as impaired migration and invasion. Although PHD1 deficiency attenuated activation of HSCs, pharmacologic PHD inhibition did not ameliorate fibrosis development. These data indicate that selective PHD1 inhibitors could prove effective in preventing and treating liver fibrosis.
Collapse
Affiliation(s)
- Christopher Tuffs
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Mareen Dupovac
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Katrin Richter
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany; Department of Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | - Sophia Holten
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Thomas Schaschinger
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Oliver Marg
- Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adisa Poljo
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Ayse Nur Tasdemir
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany
| | - Jonathan M Harnoss
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Adrian Billeter
- Clarunis University Digestive Healthcare Center Basel, Basel, Switzerland
| | - Martin Schneider
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany
| | - Moritz J Strowitzki
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University, Heidelberg, Germany; Department of General, Visceral, Thoracic, and Transplantation Surgery, University of Giessen, Giessen, Germany.
| |
Collapse
|
33
|
Affὸ S, Sererols-Viñas L, Garcia-Vicién G, Cadamuro M, Chakraborty S, Sirica AE. Cancer-Associated Fibroblasts in Intrahepatic Cholangiocarcinoma: Insights into Origins, Heterogeneity, Lymphangiogenesis, and Peritoneal Metastasis. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:378-396. [PMID: 39117110 DOI: 10.1016/j.ajpath.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/10/2024]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) denotes a rare, highly malignant, and heterogeneous class of primary liver adenocarcinomas exhibiting phenotypic characteristics of cholangiocyte differentiation. Among the distinctive pathological features of iCCA, one that differentiates the most common macroscopic subtype (eg, mass-forming type) of this hepatic tumor from conventional hepatocellular carcinoma is a prominent desmoplastic reaction manifested as a dense fibro-collagenous-enriched tumor stroma. Cancer-associated fibroblasts (CAFs) represent the most abundant mesenchymal cell type in the desmoplastic reaction. Although the protumor effects of CAFs in iCCA have been increasingly recognized, more recent cell lineage tracing studies, advanced single-cell RNA sequencing, and expanded biomarker analyses have provided new awareness into their ontogeny, as well as underscored their biological complexity as reflected by the presence of multiple subtypes. In addition, evidence supports CAFs' potential to display cancer-restrictive roles, including immunosuppression. However, CAFs also play important roles in facilitating metastasis, as exemplified by lymph node metastasis and peritoneal carcinomatosis, which are common in iCCA. Herein, the authors provide a timely appraisal of the origins and phenotypic and functional complexity of CAFs in iCCA, together with providing mechanistic insights into lymphangiogenesis and peritoneal metastasis relevant to this lethal human cancer.
Collapse
Affiliation(s)
- Silvia Affὸ
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | - Sanjukta Chakraborty
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Alphonse E Sirica
- Department of Pathology (Emeritus), Virginia Commonwealth University School of Medicine, Richmond, Virginia.
| |
Collapse
|
34
|
Sun C, Zhou C, Daneshvar K, Ben Saad A, Kratkiewicz AJ, Toles BJ, Arghiani N, Hess A, Chen JY, Pondick JV, York SR, Li W, Moran SP, Gentile SD, Rahman RU, Li Z, Zhou P, Sparks RP, Habboub T, Kim BM, Choi MY, Affo S, Schwabe RF, Popov YV, Mullen AC. Conserved long noncoding RNA TILAM promotes liver fibrosis through interaction with PML in HSCs. Hepatology 2025; 81:853-869. [PMID: 38563629 PMCID: PMC11825499 DOI: 10.1097/hep.0000000000000822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 02/01/2024] [Indexed: 04/04/2024]
Abstract
BACKGROUND AND AIMS Fibrosis is the common end point for all forms of chronic liver injury, and the progression of fibrosis leads to the development of end-stage liver disease. Activation of HSCs and their transdifferentiation into myofibroblasts results in the accumulation of extracellular matrix proteins that form the fibrotic scar. Long noncoding RNAs regulate the activity of HSCs and provide targets for fibrotic therapies. APPROACH AND RESULTS We identified long noncoding RNA TILAM located near COL1A1 , expressed in HSCs, and induced with liver fibrosis in humans and mice. Loss-of-function studies in human HSCs and human liver organoids revealed that TILAM regulates the expression of COL1A1 and other extracellular matrix genes. To determine the role of TILAM in vivo, we annotated the mouse ortholog ( Tilam ), generated Tilam- deficient green fluorescent protein-reporter mice, and challenged these mice in 2 different models of liver fibrosis. Single-cell data and analysis of single-data and analysis of Tilam-deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Tilam -deficient reporter mice revealed that Tilam is induced in murine HSCs with the development of fibrosis in vivo. Furthermore, loss of Tilam expression attenuated the development of fibrosis in the setting of in vivo liver injury. Finally, we found that TILAM interacts with promyelocytic leukemia nuclear body scaffold protein to regulate a feedback loop by which TGF-β2 reinforces TILAM expression and nuclear localization of promyelocytic leukemia nuclear body scaffold protein to promote the fibrotic activity of HSCs. CONCLUSIONS TILAM is activated in HSCs with liver injury and interacts with promyelocytic leukemia nuclear body scaffold protein to drive the development of fibrosis. Depletion of TILAM may serve as a therapeutic approach to combat the development of end-stage liver disease.
Collapse
Affiliation(s)
- Cheng Sun
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Chan Zhou
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Kaveh Daneshvar
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Amel Ben Saad
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Arcadia J. Kratkiewicz
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin J. Toles
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Nahid Arghiani
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Anja Hess
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer Y. Chen
- Department of Medicine, Liver Center, University of California, San Francisco, California, USA
| | - Joshua V. Pondick
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel R. York
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wenyang Li
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sean P. Moran
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan D. Gentile
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Raza Ur Rahman
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| | - Zixiu Li
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Peng Zhou
- Department of Population and Quantitative Health Sciences, Chan Medical School, University of Massachusetts, Worcester, Massachusetts USA
| | - Robert P. Sparks
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
| | - Tim Habboub
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Byeong-Moo Kim
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Michael Y. Choi
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Silvia Affo
- Department of Liver, Digestive System, and Metabolism, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Robert F. Schwabe
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Yury V. Popov
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan C. Mullen
- Department of Medicine, Division of Gastroenterology, Chan Medical School, University of Massachusetts, Worcester, Massachusetts, USA
- Broad Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
35
|
Xia Y, Wang Y, Xiong Q, He J, Wang H, Islam M, Zhou X, Kim A, Zhang H, Huang H, Tsung A. Neutrophil extracellular traps promote MASH fibrosis by metabolic reprogramming of HSC. Hepatology 2025; 81:947-961. [PMID: 38266270 PMCID: PMC11881075 DOI: 10.1097/hep.0000000000000762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND AND AIMS Metabolic dysfunction-associated steatohepatitis (MASH) fibrosis is a reversible stage of liver disease accompanied by inflammatory cell infiltration. Neutrophils extrude a meshwork of chromatin fibers to establish neutrophil extracellular traps (NETs), which play important roles in inflammatory response regulation. Our previous work demonstrated that NETs promote HCC in MASH. However, it is still unknown if NETs play a role in the molecular mechanisms of liver fibrosis. APPROACH AND RESULTS Following 12 weeks of Western diet/carbon tetrachloride, MASH fibrosis was identified in C57BL/6 mice with increased NET formation. However, NET depletion using DNase I treatment or mice knocked out for peptidyl arginine deaminase type IV significantly attenuated the development of MASH fibrosis. NETs were demonstrated to induce HSCs activation, proliferation, and migration through augmented mitochondrial and aerobic glycolysis to provide additional bioenergetic and biosynthetic supplies. Metabolomic analysis revealed markedly an altered metabolic profile upon NET stimulation of HSCs that were dependent on arachidonic acid metabolism. Mechanistically, NET stimulation of toll-like receptor 3 induced cyclooxygenase-2 activation and prostaglandin E2 production with subsequent HSC activation and liver fibrosis. Inhibiting cyclooxygenase-2 with celecoxib reduced fibrosis in our MASH model. CONCLUSIONS Our findings implicate NETs playing a critical role in the development of MASH hepatic fibrosis by inducing metabolic reprogramming of HSCs through the toll-like receptor 3/cyclooxygenase-2/cyclooxygenase-2 pathway. Therefore, NET inhibition may represent an attractive treatment target for MASH liver fibrosis.
Collapse
Affiliation(s)
- Yujia Xia
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Yu Wang
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Xiong
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiayi He
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Mozaffarul Islam
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Xinyu Zhou
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Alex Kim
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Hongji Zhang
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Allan Tsung
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
36
|
Meng X, Wang D, Zhang H, Kang T, Meng X, Liang S. Portulaca oleracea L. extract relieve mice liver fibrosis by inhibiting TLR-4/NF-κB, Bcl-2/Bax and TGF-β1/Smad2 signalling transduction. Nat Prod Res 2025; 39:1435-1443. [PMID: 38164691 DOI: 10.1080/14786419.2023.2300034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Portulaca oleracea L. are annual herb, which has various pharmacological effects including hepatoprotective property. However, the effect of Portulaca oleracea L. (POL-1) in mice with carbon tetrachloride (CCl4)-induced liver fibrosis and its mechanism of action have not been clarified. POL-1 ameliorated the CCl4-induced liver fibrosis in mice, as shown by decreased collagen deposition and the decreased expression of liver fibrosis marker collagen I and α-smooth muscle actin (α-SMA) mRNA. In addition, treatment with POL-1 suppressed the proliferation of activated human hepatic stellate cell line (LX-2). POL-1 inhibited the oxidative stress and inflammation in fibrotic livers of mice. Mechanistically, POL-1 inhibited the CCl4-induced expression of toll-like receptor-4 (TLR4), myeloid differentiation factor 88 (MyD88), nuclear factor kappa-B (NF-κBp65) p65, Bcl2-associated X (Bax), transforming growth factor-β1 (TGF-β1) and drosophila mothers against decapentaplegic 2 (Smad2) proteins, upregulated B-cell lymphoma -2 (Bcl-2) proteins in livers of mice. These findings suggested that POL-1 attenuated liver fibrosis.
Collapse
Affiliation(s)
- Xianqun Meng
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Dan Wang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Hui Zhang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Tingguo Kang
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xiansheng Meng
- Department of Traditional Chinese Medicine Identification, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Shanshan Liang
- Plant Polysaccharide Research Center, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
37
|
Sererols-Viñas L, Garcia-Vicién G, Ruiz-Blázquez P, Lee TF, Lee YA, Gonzalez-Sanchez E, Vaquero J, Moles A, Filliol A, Affò S. Hepatic Stellate Cells Functional Heterogeneity in Liver Cancer. Semin Liver Dis 2025; 45:33-51. [PMID: 40043738 DOI: 10.1055/a-2551-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Hepatic stellate cells (HSCs) are the liver's pericytes, and play key roles in liver homeostasis, regeneration, fibrosis, and cancer. Upon injury, HSCs activate and are the main origin of myofibroblasts and cancer-associated fibroblasts (CAFs) in liver fibrosis and cancer. Primary liver cancer has a grim prognosis, ranking as the third leading cause of cancer-related deaths worldwide, with hepatocellular carcinoma (HCC) being the predominant type, followed by intrahepatic cholangiocarcinoma (iCCA). Moreover, the liver hosts 35% of all metastatic lesions. The distinct spatial distribution and functional roles of HSCs across these malignancies represent a significant challenge for universal therapeutic strategies, requiring a nuanced and tailored understanding of their contributions. This review examines the heterogeneous roles of HSCs in liver cancer, focusing on their spatial localization, dynamic interactions within the tumor microenvironment (TME), and emerging therapeutic opportunities, including strategies to modulate their activity, and harness their potential as targets for antifibrotic and antitumor interventions.
Collapse
Affiliation(s)
- Laura Sererols-Viñas
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Gemma Garcia-Vicién
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paloma Ruiz-Blázquez
- University of Barcelona, Barcelona, Spain
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Ting-Fang Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Youngmin A Lee
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ester Gonzalez-Sanchez
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Javier Vaquero
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
- HepatoBiliary Tumours Lab, Centro de Investigación del Cáncer and Instituto de Biología Molecular y Celular del Cáncer, CSIC-Universidad de Salamanca, Salamanca, Spain
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Anna Moles
- Tissue Remodeling Fibrosis and Cancer Group, Institute of Biomedical Research of Barcelona, Spanish National Research Council, Barcelona, Spain
- Institute of Biomedical Research of Barcelona (IDIBAPS), Barcelona, Spain
- CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Aveline Filliol
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Silvia Affò
- Tumor Microenvironment Plasticity and Heterogeneity Research Group, Institut d'Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
38
|
Wong T, Kang R, Yun K. The multi-faceted immune modulatory role of S100A4 in cancer and chronic inflammatory disease. Front Immunol 2025; 16:1525567. [PMID: 40078995 PMCID: PMC11897520 DOI: 10.3389/fimmu.2025.1525567] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
S100A4 is a Ca2+-binding protein involved in multiple chronic inflammatory and neoplastic conditions. This review focuses on recent advances in the understanding of S100A4 function in immune cells, comparing and contrasting S100A4 regulation of immune responses in cancer and chronic inflammatory diseases. We provide evidence that S100A4 regulation of immune cell function has a profound role in promoting the pathogenesis of cancer and pro-inflammatory conditions. Finally, we discuss relevant future directions to target S100A4 therapeutically in different disease states.
Collapse
Affiliation(s)
- Thomas Wong
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
- College of Medicine, Texas A&M University, Bryan, TX, United States
| | - Reece Kang
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
| | - Kyuson Yun
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, United States
- Department of Neurology, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
39
|
Wang T, Liu B, Huang J, Zhao Q, Shen H, Bi T, Liu Z, Dai Y, Sun Q. IFN-γ-mediated inhibition of JAK/STAT signaling via nano-scutellarin treatment is an efficient strategy for ameliorating liver fibrosis. J Transl Med 2025; 23:195. [PMID: 39962553 PMCID: PMC11834254 DOI: 10.1186/s12967-025-06155-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/18/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatohepatitis (MASH) is a large group of metabolic diseases that are hazardous to human health. Endothelial-to-mesenchymal transition (EndMT) mediated myofibroblast activation is an important factor that aggravates the development of liver fibrosis during MASH. However, the limited understanding of the underlying molecular mechanisms that drive EndMT in MASH has hindered the development of molecularly targeted therapies specifically targeting this pathological process. METHODS We employed wild-type and ifn-γ-deficient mice, MASH models were induced repeated CCl4 injections and a high-fat diet to verify the significance of IFN-γ role in vivo and its impact in EndMT. Male mice models of MASH were used to further analyze the effect of Scutellarin@BSA on the improvement of liver fibrosis during MASH in vivo and HUVECs were used to assess IFN-γ effect on EndMT and its interaction with JAK signaling pathway in vitro. RESULTS The results showed that IFN-γ is revealed as a key regulator of EndMT during MASH, as evidenced by the significantly lower levels of EndMT and reduced pathological damage in the livers of ifn-γ knockout mice. Furthermore, our research has led to the development of Scutellarin@BSA therapy, which targets and mitigates IFN-γ-driven EndMT, which showed excellent therapeutic effects on EndMT and liver fibrosis in vivo and in vitro during MASH. Mechanistically, IFN-γ can directly bind to the JAK protein and activate downstream STAT1 transcription factors, exerting transcriptional activity and further driving the expression of EndMT-associated proteins. Notably, Scutellarin@BSA treatment effectively diminishes the hallmarks of liver fibrosis by modulating the canonical JAK/STAT1 signaling pathway. CONCLUSIONS IFN-γ was identified as a key regulator of EndMT, and Scutellarin@BSA, as an emerging treatment, has been found to effectively inhibit EndMT by directly targeting the regulatory influence of the IFN-γ signaling. This result demonstrates significant therapeutic efficacy in alleviating hepatic fibrosis during MASH, highlighting its great potential as an innovative liver fibrosis treatment.
Collapse
Affiliation(s)
- Ting Wang
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Bangguo Liu
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Juan Huang
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Qixin Zhao
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Hongping Shen
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Tao Bi
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
- State Key Laboratories for Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 853, China
| | - Zengjin Liu
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yong Dai
- Sichuan Police College, Luzhou, 646000, Sichuan, China.
| | - Qin Sun
- Drug Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Institute of Integrated Chinese and Western Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
40
|
Yang YM, Kim J, Wang Z, Kim J, Kim SY, Cho GJ, Lee JH, Kim SM, Tsuchiya T, Matsuda M, Pandyarajan V, Pandol SJ, Lewis MS, Gangi A, Noble PW, Jiang D, Merchant A, Posadas EM, Bhowmick NA, Lu SC, You S, Xu AM, Seki E. Metastatic tumor growth in steatotic liver is promoted by HAS2-mediated fibrotic tumor microenvironment. J Clin Invest 2025; 135:e180802. [PMID: 39946200 PMCID: PMC11957696 DOI: 10.1172/jci180802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Steatotic liver enhances liver metastasis of colorectal cancer (CRC), but this process is not fully understood. Steatotic liver induced by a high-fat diet increases cancer-associated fibroblast (CAF) infiltration and collagen and hyaluronic acid (HA) production. We investigated the role of HA synthase 2 (HAS2) in the fibrotic tumor microenvironment in steatotic liver using Has2ΔHSC mice, in which Has2 is deleted from hepatic stellate cells. Has2ΔHSC mice had reduced steatotic liver-associated metastatic tumor growth of MC38 CRC cells, collagen and HA deposition, and CAF and M2 macrophage infiltration. We found that low-molecular weight HA activates Yes-associated protein (YAP) in cancer cells, which then releases connective tissue growth factor to further activate CAFs for HAS2 expression. Single-cell analyses revealed a link between CAF-derived HAS2 and M2 macrophages and CRC cells through CD44; these cells were associated with exhausted CD8+ T cells via programmed death-ligand 1 and programmed cell death protein 1 (PD-1). HA synthesis inhibitors reduced steatotic liver-associated metastasis of CRC, YAP expression, and CAF and M2 macrophage infiltration, and improved response to anti-PD-1 antibody. In conclusion, steatotic liver modulates a fibrotic tumor microenvironment to enhance metastatic cancer activity through a bidirectional regulation between CAFs and metastatic tumors, enhancing the metastatic potential of CRC in the liver.
Collapse
Affiliation(s)
- Yoon Mee Yang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pharmacy
- Multidimensional Genomics Research Center, and
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | - Jieun Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zhijun Wang
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jina Kim
- Samuel Oschin Comprehensive Cancer Institute
- Department of Urology
- Department of Computational Biomedicine, and
| | - So Yeon Kim
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Gyu Jeong Cho
- Department of Pharmacy
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | | | - Sun Myoung Kim
- Department of Pharmacy
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, South Korea
| | - Takashi Tsuchiya
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michitaka Matsuda
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vijay Pandyarajan
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Stephen J. Pandol
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Michael S. Lewis
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Pathology, Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California, USA
| | | | - Paul W. Noble
- Department of Medicine and Women’s Guild Lung Institute
| | - Dianhua Jiang
- Department of Medicine and Women’s Guild Lung Institute
| | - Akil Merchant
- Samuel Oschin Comprehensive Cancer Institute
- Division of Hematology and Cellular Therapy, Department of Medicine
| | - Edwin M. Posadas
- Samuel Oschin Comprehensive Cancer Institute
- Division of Medical Oncology, Department of Medicine, and
| | - Neil A. Bhowmick
- Samuel Oschin Comprehensive Cancer Institute
- Division of Medical Oncology, Department of Medicine, and
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute
- Department of Urology
- Department of Computational Biomedicine, and
| | - Alexander M. Xu
- Samuel Oschin Comprehensive Cancer Institute
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, Maryland, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Sharip A, Kunz J. Mechanosignaling via Integrins: Pivotal Players in Liver Fibrosis Progression and Therapy. Cells 2025; 14:266. [PMID: 39996739 PMCID: PMC11854242 DOI: 10.3390/cells14040266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/26/2025] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury, represents a major global health burden and is the leading cause of liver failure, morbidity, and mortality. The pathological hallmark of this condition is excessive extracellular matrix deposition, driven primarily by integrin-mediated mechanotransduction. Integrins, transmembrane heterodimeric proteins that serve as primary ECM receptors, orchestrate complex mechanosignaling networks that regulate the activation, differentiation, and proliferation of hepatic stellate cells and other ECM-secreting myofibroblasts. These mechanical signals create self-reinforcing feedback loops that perpetuate the fibrotic response. Recent advances have provided insight into the roles of specific integrin subtypes in liver fibrosis and revealed their regulation of key downstream effectors-including transforming growth factor beta, focal adhesion kinase, RhoA/Rho-associated, coiled-coil containing protein kinase, and the mechanosensitive Hippo pathway. Understanding these mechanotransduction networks has opened new therapeutic possibilities through pharmacological manipulation of integrin-dependent signaling.
Collapse
Affiliation(s)
- Aigul Sharip
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
- Laboratory of Bioinformatics and Systems Biology, National Laboratory Astana, Astana 020000, Kazakhstan
| | - Jeannette Kunz
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana 020000, Kazakhstan;
| |
Collapse
|
42
|
Martínez García de la Torre RA, Vallverdú J, Xu Z, Ariño S, Ferrer-Lorente R, Zanatto L, Mercado-Gómez M, Aguilar-Bravo B, Ruiz-Blázquez P, Fernandez-Fernandez M, Navarro-Gascon A, Blasco-Roset A, Sànchez-Fernàndez-de-Landa P, Pera J, Romero-Moya D, Ayuso Garcia P, Martínez Sánchez C, Sererols Viñas L, Cantallops Vilà P, Cárcamo Giráldez CI, McQuillin A, Morgan MY, Moya-Rull D, Montserrat N, Eberlé D, Staels B, Antoine B, Azkargorta M, Lozano JJ, Martínez-Chantar ML, Giorgetti A, Elortza F, Planavila A, Varela-Rey M, Woodhoo A, Zorzano A, Graupera I, Moles A, Coll M, Affo S, Sancho-Bru P. Trajectory analysis of hepatic stellate cell differentiation reveals metabolic regulation of cell commitment and fibrosis. Nat Commun 2025; 16:1489. [PMID: 39929812 PMCID: PMC11811062 DOI: 10.1038/s41467-025-56024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Defining the trajectory of cells during differentiation and disease is key for uncovering the mechanisms driving cell fate and identity. However, trajectories of human cells remain largely unexplored due to the challenges of studying them with human samples. In this study, we investigate the proteome trajectory of iPSCs differentiation to hepatic stellate cells (diHSCs) and identify RORA as a key transcription factor governing the metabolic reprogramming of HSCs necessary for diHSCs' commitment, identity, and activation. Using RORA deficient iPSCs and pharmacologic interventions, we show that RORA is required for early differentiation and prevents diHSCs activation by reducing the high energetic state of the cells. While RORA knockout mice have enhanced fibrosis, RORA agonists rescue multi-organ fibrosis in in vivo models. Notably, RORA expression correlates negatively with liver fibrosis and HSCs activation markers in patients with liver disease. This study reveals that RORA regulates cell metabolic plasticity, important for mesoderm differentiation, pericyte quiescence, and fibrosis, influencing cell commitment and disease.
Collapse
Affiliation(s)
| | - Julia Vallverdú
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Zhenqing Xu
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Silvia Ariño
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Raquel Ferrer-Lorente
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Laura Zanatto
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Maria Mercado-Gómez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Beatriz Aguilar-Bravo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Paloma Ruiz-Blázquez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona, Spanish National Research Council (IIBB-CSIC), Barcelona, Spain
| | - Maria Fernandez-Fernandez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona, Spanish National Research Council (IIBB-CSIC), Barcelona, Spain
| | - Artur Navarro-Gascon
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain
- CIBER Fisitopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Barcelona, Spain
| | - Albert Blasco-Roset
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain
- CIBER Fisitopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Barcelona, Spain
| | - Paula Sànchez-Fernàndez-de-Landa
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Joan Pera
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Damia Romero-Moya
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Paula Ayuso Garcia
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Celia Martínez Sánchez
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Laura Sererols Viñas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | - Paula Cantallops Vilà
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
| | | | - Andrew McQuillin
- Molecular Psychiatry Laboratory, Division of Psychiatry, University College London, London, WC1E 6DE, UK
| | - Marsha Y Morgan
- UCL Institute for Liver & Digestive Health, Division of Medicine, Royal Free Campus, University College London, London, NW3 2PF, UK
| | - Daniel Moya-Rull
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, Spain
| | - Núria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, Barcelona, Spain
| | - Delphine Eberlé
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, 59000, Lille, France
| | - Bénédicte Antoine
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Mikel Azkargorta
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Proteomics Platform, CIC BioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Science and Technology Park, Derio, Spain
| | - Juan-José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Maria L Martínez-Chantar
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, Derio, Spain
| | - Alessandra Giorgetti
- Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Barcelona University, Barcelona, Spain
| | - Félix Elortza
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Proteomics Platform, CIC BioGUNE, Basque Research and Technology Alliance (BRTA), Bizkaia Science and Technology Park, Derio, Spain
| | - Anna Planavila
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain
- CIBER Fisitopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Barcelona, Spain
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Barcelona University, Barcelona, Spain
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
- Oportunius Research Professor at CIMUS/USC, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| | - Antonio Zorzano
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Institut de Biomedicina de la Universitat de Barcelona, Universitat de Barcelona, Barcelona, Spain
- CIBER Fisitopatologia de la Obesidad y Nutrición, Instituto de Salud Carlos III, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Isabel Graupera
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Liver Unit, Hospital Clínic, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Anna Moles
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona, Spanish National Research Council (IIBB-CSIC), Barcelona, Spain
| | - Mar Coll
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- University of Barcelona, Barcelona, Spain
- Medicine Department, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Silvia Affo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Pau Sancho-Bru
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain.
- Medicine Department, Faculty of Medicine, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
43
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [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: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
44
|
Kim J, Seki E. Inflammation and Immunity in Liver Neoplasms: Implications for Future Therapeutic Strategies. Mol Cancer Ther 2025; 24:188-199. [PMID: 39365846 PMCID: PMC11794036 DOI: 10.1158/1535-7163.mct-23-0726] [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: 06/04/2024] [Revised: 07/28/2024] [Accepted: 08/09/2024] [Indexed: 10/06/2024]
Abstract
Over the past two decades, the "hallmarks of cancer" have revolutionized cancer research and highlighted the crucial roles of inflammation and immunity. Protumorigenic inflammation promotes cancer development along with inhibition of antitumor immunity, shaping the tumor microenvironment (TME) toward a tumor-permissive state and further enhancing the malignant potential of cancer cells. This immunosuppressive TME allows tumors to evade immunosurveillance. Thus, understanding the complex interplay between tumors and the immune system within the TME has become pivotal, especially with the advent of immunotherapy. Although immunotherapy has achieved notable success in many malignancies, primary liver cancer, particularly hepatocellular carcinoma, presents unique challenges. The hepatic immunosuppressive environment poses obstacles to the effectiveness of immunotherapy, along with high mortality rates and limited treatment options for patients with liver cancer. In this review, we discuss current understanding of the complex immune-mediated mechanisms underlying liver neoplasms, focusing on hepatocellular carcinoma and liver metastases. We describe the molecular and cellular heterogeneity within the TME, highlighting how this presents unique challenges and opportunities for immunotherapy in liver cancers. By unraveling the immune landscape of liver neoplasms, this review aims to contribute to the development of more effective therapeutic interventions, ultimately improving clinical outcomes for patients with liver cancer.
Collapse
Affiliation(s)
- Jieun Kim
- Karsh Division of Gastroenterology Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekihiro Seki
- Karsh Division of Gastroenterology Hepatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
45
|
Neumeyer V, Chavan P, Steiger K, Ebert O, Altomonte J. Cross-Talk Between Tumor Cells and Stellate Cells Promotes Oncolytic VSV Activity in Intrahepatic Cholangiocarcinoma. Cancers (Basel) 2025; 17:514. [PMID: 39941881 PMCID: PMC11816849 DOI: 10.3390/cancers17030514] [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: 12/19/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
As the mechanisms underlying tumorigenesis become better understood, the dynamic roles of cellular components of the tumor microenvironment, and their cross-talk with tumor cells, have come to light as key drivers of disease progression and have emerged as important targets of new cancer therapies. In the field of oncolytic virus (OV) therapy, stromal cells have been considered as potential barriers to viral spread, thus limiting virus replication and therapeutic outcome. However, new evidence indicates that intratumoral fibroblasts could support virus replication. We have demonstrated in a rat model of stromal-rich intrahepatic cholangiocarcinoma (CCA) that vesicular stomatitis virus (VSV) can be localized within intratumoral hepatic stellate cells (HSCs), in addition to tumor cells, when the virus was applied via hepatic arterial infusion. Furthermore, VSV was shown to efficiently kill CCA cells and activated HSCs, and co-culture of CCA and HSCs increased viral titers. Interestingly, this effect is also observed when each cell type is cultured alone in a conditioned medium of the other cell type, indicating that secreted cell factors are at least partially responsible for this phenomenon. Partial reduction in sensitivity to type I interferons was observed in co-culture systems, providing a possible mechanism for the increased viral titers. Together, the results indicate that targeting activated HSCs with VSV could provide an additional mechanism of OV therapy, which, until now has not been considered. Furthermore, these findings suggest that VSV is a potentially powerful therapeutic agent for stromal-rich tumors, such as CCA and pancreatic cancer, both of which are very difficult to treat with conventional therapy and have a very poor prognosis.
Collapse
Affiliation(s)
- Victoria Neumeyer
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Purva Chavan
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- Department of Pathology, Technical University of Munich, 81675 Munich, Germany
| | - Oliver Ebert
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| | - Jennifer Altomonte
- Department of Internal Medicine 2, University Hospital of the Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
46
|
Markovic J, Li R, Khanal R, Peng Q, Möbus S, Yuan Q, Engel B, Taubert R, Vondran FWR, Bantel H, Singh MK, Cantz T, Büning H, Wedemeyer H, Ott M, Balakrishnan A, Sharma AD. Identification and functional validation of miR-190b-5p and miR-296-3p as novel therapeutic attenuators of liver fibrosis. J Hepatol 2025; 82:301-314. [PMID: 39218230 DOI: 10.1016/j.jhep.2024.08.014] [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: 01/14/2024] [Revised: 07/30/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND & AIMS Liver fibrosis and its end-stage form cirrhosis contribute to millions of deaths annually. The lack of robust antifibrotic molecules is in part attributed to the absence of any functional screens to identify molecular regulators using patient-derived primary human hepatic myofibroblasts, which are key drivers of fibrosis. METHODS Here, to identify robust regulators of fibrosis, we performed functional microRNA screenings in primary human hepatic myofibroblasts followed by in vivo validation in three independent mouse models of fibrosis (toxin, cholestasis and MASH). RESULTS We identified miR-190b-5p and miR-296-3p as robust antifibrotic miRNAs that suppress liver fibrosis. Notably, the expression of miR-190b-5p and miR-296-3p was found to be significantly reduced in human livers with fibrosis. Mechanistically, we discovered hyaluronan synthase 2 (HAS2) and integrin alpha-6 (ITGA6) as novel targets of miR-190b-5p and miR-296-3p, respectively. Furthermore, we demonstrated that the antifibrotic properties of miR-190b-5p and miR-296-3p are, at least in part, dependent on HAS2 and ITGA6. Finally, we showed the antifibrotic function of both miRNAs in a human liver bud model, which mimics multiple features of the human liver. CONCLUSIONS Collectively, in our study we discovered miR-190b-5p and miR-296-3p as two novel antifibrotic miRNAs, and that HAS2 and ITGA6 contribute to miR-190b-5p- and miR-296-3p-mediated inhibition of liver fibrosis. These results provide a foundation for future research to explore the clinical utility of miR-190b-5p and miR-296-3p in fibrosis. IMPACT AND IMPLICATIONS Liver fibrosis and cirrhosis contribute to millions of deaths worldwide and remain unmet medical needs. In this study, we discovered two microRNAs, miR-190b-5p and miR-296-3p, which suppress liver fibrosis in preclinical mouse models and a human liver bud model. Our promising results encourage further studies that aim to develop both miRNAs for the treatment of liver fibrosis in patients.
Collapse
Affiliation(s)
- Jovana Markovic
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Ruomeng Li
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Rajendra Khanal
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Qi Peng
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Selina Möbus
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Qinggong Yuan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Bastian Engel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Richard Taubert
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Florian W R Vondran
- Department of General, Visceral, Pediatric and Transplant Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Heike Bantel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Manvendra K Singh
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, 8 College Road, Office 08-15, Singapore 169857, Singapore
| | - Tobias Cantz
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Hildegard Büning
- Laboratory for Infection Biology and Gene Transfer, Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Michael Ott
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Asha Balakrishnan
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany.
| | - Amar Deep Sharma
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany; Research Group RNA Therapeutics & Liver Regeneration, REBIRTH-Research Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
47
|
Lam WLM, Gabernet G, Poth T, Sator-Schmitt M, Oquendo MB, Kast B, Lohr S, de Ponti A, Weiß L, Schneider M, Helm D, Müller-Decker K, Schirmacher P, Heikenwälder M, Klingmüller U, Schneller D, Geisler F, Nahnsen S, Angel P. RAGE is a key regulator of ductular reaction-mediated fibrosis during cholestasis. EMBO Rep 2025; 26:880-907. [PMID: 39747668 PMCID: PMC11811172 DOI: 10.1038/s44319-024-00356-7] [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: 02/28/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025] Open
Abstract
Ductular reaction (DR) is the hallmark of cholestatic diseases manifested in the proliferation of bile ductules lined by biliary epithelial cells (BECs). It is commonly associated with an increased risk of fibrosis and liver failure. The receptor for advanced glycation end products (RAGE) was identified as a critical mediator of DR during chronic injury. Yet, the direct link between RAGE-mediated DR and fibrosis as well as the mode of interaction between BECs and hepatic stellate cells (HSCs) to drive fibrosis remain elusive. Here, we delineate the specific function of RAGE on BECs during DR and its potential association with fibrosis in the context of cholestasis. Employing a biliary lineage tracing cholestatic liver injury mouse model, combined with whole transcriptome sequencing and in vitro analyses, we reveal a role for BEC-specific Rage activity in fostering a pro-fibrotic milieu. RAGE is predominantly expressed in BECs and contributes to DR. Notch ligand Jagged1 is secreted from activated BECs in a Rage-dependent manner and signals HSCs in trans, eventually enhancing fibrosis during cholestasis.
Collapse
Affiliation(s)
- Wai-Ling Macrina Lam
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Ruprecht Karl University of Heidelberg, Heidelberg, Germany
| | - Gisela Gabernet
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Tanja Poth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Melanie Sator-Schmitt
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Morgana Barroso Oquendo
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Bettina Kast
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sabrina Lohr
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Aurora de Ponti
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Lena Weiß
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Martin Schneider
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Protein Analysis Unit, Genomics and Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karin Müller-Decker
- Tumor Models Unit, Center for Preclinical Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ursula Klingmüller
- Division of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Doris Schneller
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Fabian Geisler
- TUM School of Medicine and Health, Department of Clinical Medicine - Clinical Department for Internal Medicine II, University Medical Center, Technical University of Munich, München, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Peter Angel
- Division of Signal Transduction and Growth Control, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
48
|
Wang Z, Gao H, Ma X, Zhu D, Zhao L, Xiao W. Adrenic acid: A promising biomarker and therapeutic target (Review). Int J Mol Med 2025; 55:20. [PMID: 39575474 PMCID: PMC11611323 DOI: 10.3892/ijmm.2024.5461] [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: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 01/05/2025] Open
Abstract
Adrenic acid is a 22‑carbon unsaturated fatty acid that is widely present in the adrenal gland, liver, brain, kidney and vascular system that plays a regulatory role in various pathophysiological processes, such as inflammatory reactions, lipid metabolism, oxidative stress, vascular function, and cell death. Adrenic acid is a potential biomarker for various ailments, including metabolic, neurodegenerative and cardiovascular diseases and cancer. In addition, adrenic acid is influenced by the pharmacological properties of several natural products, such as astragaloside IV, evodiamine, quercetin, kaempferol, Berberine‑baicalin and prebiotics, so it is a promising new target for clinical treatment and drug development. However, the molecular mechanisms by which adrenic acid exerts are unclear. The present study systematically reviewed the biosynthesis and metabolism of adrenic acid, focusing on intrinsic mechanisms that influence the progression of metabolic, cardiovascular and neurological disease. These mechanisms regulate several key processes, including immuno‑inflammatory response, oxidative stress, vascular function and cell death. In addition, the present study explored the potential clinical translational value of adrenic acid as a biomarker and therapeutic target. To the best of our knowledge, the present study is first systematic summary of the mechanisms of action of adrenic acid across a range of diseases. The present study provides understanding of the wide range of metabolic activities of adrenic acid and a basis for further exploring the pathogenesis and therapeutic targets of various diseases.
Collapse
Affiliation(s)
- Ze Wang
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Haoyang Gao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Xiaotong Ma
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Danlin Zhu
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| | - Linlin Zhao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
- School of Physical Education, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Weihua Xiao
- Shanghai Key Laboratory of Human Performance, Shanghai University of Sport, Shanghai 200438, P.R. China
| |
Collapse
|
49
|
Chen SY, Chen X, Zhu S, Xu JJ, Li XF, Yin NN, Xiao YY, Huang C, Li J. miR-324-3p Suppresses Hepatic Stellate Cell Activation and Hepatic Fibrosis Via Regulating SMAD4 Signaling Pathway. Mol Biotechnol 2025; 67:673-688. [PMID: 38407690 PMCID: PMC11711260 DOI: 10.1007/s12033-024-01078-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
In hepatic fibrosis (HF), hepatic stellate cells (HSCs) form the extracellular matrix (ECM), and the pathological accumulation of ECM in the liver leads to inflammation. Our previous research found that miR-324-3p was down-regulated in culture-activated human HSCs. However, the precise effect of miR-324-3p on HF has not been elucidated. In this study, the HF mouse models were induced through directly injecting carbon tetrachloride (CCl4) into mice; the HF cell models were constructed using TGF-β1-treated LX-2 cells. Next, real-time-quantitative polymerase chain reaction (RT-qPCR), western blot (WB) and immunohistochemistry (IHC) were applied to assess the expression levels of miR-324-3p, α-smooth muscle actin (α-SMA), Vimentin or SMAD4; hematoxylin and eosin (H&E), Masson' s trichrome and Sirius red staining to evaluate the liver injury; luciferase reporter assay to verify the targeting relationship between miR-324-3p and SMAD4; enzyme-linked immunosorbent assay (ELISA) to determine the levels of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST); and cell counting kit-8 (CCK-8) and flow cytometry to evaluate the effects of miR-324-3p on cell proliferation and cycle/apoptosis, respectively. The experimental results showed a reduction in miR-324-3p level in CCl4-induced HF mice as well as transforming growth factor (TGF)-β1-activated HSCs. Interestingly, the miR-324-3p level was rescued following the HF recovery process. In HF mice induced by CCl4, miR-324-3p overexpression inhibited liver tissue damage, decreased serum ALT and AST levels, and inhibited fibrosis-related biomarkers (α-SMA, Vimentin) expression, thereby inhibiting HF. Similarly, miR-324-3p overexpression up-regulated α-SMA and Vimentin levels in HF cells, while knockdown of miR-324-3p had the opposite effect. Besides, miR-324-3p played an antifibrotic role through inhibiting the proliferation of hepatocytes. Further experiments confirmed that miR-324-3p targeted and down-regulated SMAD4 expression. SMAD4 was highly expressed in HF cells, and silencing SMAD4 significantly decreased the α-SMA and Vimentin levels in HF cells. Collectively, the miR-324-3p may suppress the activation of HSCs and HF by targeting SMAD4. Therefore, miR-324-3p is identified as a potential and novel therapeutic target for HF.
Collapse
Affiliation(s)
- Si-Yu Chen
- Department of Pharmacy, Hefei BOE Hospital, Intersection of Dongfang Avenue and Wenzhong Road, Hefei, China
| | - Xin Chen
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Sai Zhu
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Jin-Jin Xu
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Xiao-Feng Li
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Na-Na Yin
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Yan-Yan Xiao
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, 81 Mei Shan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
50
|
Feng Q, Zhao Q, Qu S, Zhao Y, Li K, Yuan B, Chang Q, Xu J, Wang H, Zhu Y, Fu K, Liu J. Qianggan Ruanjian Pill ameliorates liver fibrosis through regulation of the TGF-β1/Smad and PI3K/AKT signalling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118893. [PMID: 39362322 DOI: 10.1016/j.jep.2024.118893] [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: 06/15/2024] [Revised: 08/17/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver fibrosis is a critical pathological process in the progression of chronic liver injury, ultimately resulting in cirrhosis, for which currently available therapeutic interventions remain inadequate. Among these, the Qianggan Ruanjian Pill (QGRJP) has emerged as a clinically experienced formula with notable therapeutic efficacy against liver fibrosis. However, the precise underlying mechanisms require further investigation. AIM OF THE STUDY In this study, we investigated the key pathways and target genes of QGRJP that attenuate liver fibrosis and elucidated the underlying mechanisms. MATERIALS AND METHODS High-performance liquid chromatography-mass spectrometry (HPLC-MS) was used to identify the major components of the QGRJP. Mouse models of liver fibrosis were established by injecting olive oil containing 25% carbon tetrachloride (CCl4), which was administered at different doses of QGRJP by gavage. Liver damage and function were assessed using serum biochemical detection, ultrasound imaging, and histopathological examination. The anti-fibrosis effect was assessed using immunohistochemistry, western blotting, and quantitative real-time PCR (qRT-PCR). The in vivo safety of the QGRJP was evaluated using weight monitoring and biopsy. Potential anti-liver fibrosis signalling pathways and key targets of QGRJP were identified using RNA-seq analysis and network pharmacology. The predicted targets and pathways were validated using in vitro and in vivo experiments. RESULTS QGRJP significantly ameliorated CCl4-induced liver fibrosis, and its mechanism was correlated with the inhibition of hepatic stellate cell (HSC) activation and the inflammatory response via inhibition of the TGF-β1/Smad and PI3K/AKT pathways, leading to a significant reduction in the expression of collagen and other fibrosis-related proteins. Additionally, no obvious toxic side effects were observed in the major organs of the mice or in activated HSCs (aHSCs). CONCLUSION This study demonstrated that QGRJP mitigated liver injury, inflammation, and fibrosis by inhibiting the TGF-β1/Smad and PI3K/AKT signalling pathways.
Collapse
Affiliation(s)
- Qunying Feng
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Qinghua Zhao
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Shuaiyong Qu
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Yiju Zhao
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Kunlun Li
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Bo Yuan
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Qinzheng Chang
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Jingjiang Xu
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Hongxia Wang
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Yongqin Zhu
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Kai Fu
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China
| | - Jingsheng Liu
- Kaifeng Hospital of Traditional Chinese Medicine, Kaifeng, Henan, 475000, China.
| |
Collapse
|