1
|
Poudineh M, Mohammadyari F, Parsamanesh N, Jamialahmadi T, Kesharwani P, Sahebkar A. Cell and gene therapeutic approaches in non-alcoholic fatty liver disease. Gene 2025; 956:149466. [PMID: 40189164 DOI: 10.1016/j.gene.2025.149466] [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: 01/06/2025] [Revised: 03/14/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to a range of conditions marked by the buildup of triglycerides in liver cells, accompanied by inflammation, which contributes to liver damage, clinical symptoms, and histopathological alterations. Multiple molecular pathways contribute to NAFLD pathogenesis, including immune dysregulation, endoplasmic reticulum stress, and tissue injury. Both the innate and adaptive immune systems play crucial roles in disease progression, with intricate crosstalk between liver and immune cells driving NAFLD development. Among emerging therapeutic strategies, cell and gene-based therapies have shown promise. This study reviews the pathophysiological mechanisms of NAFLD and explores the therapeutic potential of cell-based interventions, highlighting their immunomodulatory effects, inhibition of hepatic stellate cells, promotion of hepatocyte regeneration, and potential for hepatocyte differentiation. Additionally, we examine gene delivery vectors designed to target NAFLD, focusing on their role in engineering hepatocytes through gene addition or editing to enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | - Negin Parsamanesh
- Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Tananz Jamialahmadi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar, Madhya Pradesh 470003, India.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact and Outcome, Chitkara University, Rajpura 140417, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Jin Y, Li N, Chen SN, Lu YS, Gan Z, Nie P. Transcriptome analysis of head kidney and liver in grass carp (Ctenopharyngodon idella) symptomatically or asymptomatically infected with Flavobacterium columnare. FISH & SHELLFISH IMMUNOLOGY 2025; 161:110293. [PMID: 40122189 DOI: 10.1016/j.fsi.2025.110293] [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: 11/22/2024] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Flavobacterium columnare is an important pathogen causing columnaris disease, which can result in high mortality in freshwater fish worldwide. Understanding the immune response in infection status of fish may be essential for developing effective prevention and treatment strategies. In this study, transcriptomes of liver and head kidney tissues in grass carp (Ctenopharyngodon idella) were compared under symptomatic and asymptomatic statuses following the immersion infection of F. columnare. Significant differences in expression of genes were observed between fish showing disease symptoms and those without symptoms. The number of differentially expressed genes (DEGs) between infected and control groups ranged from 4752 to 8,277, while the DEGs between exposed and control groups ranged from 272 to 1,751, suggesting a strong acute inflammatory response in infected groups. KEGG pathway enrichment analysis of infected groups revealed that among the top 30 enriched pathways, liver and head kidney shared 22 and 16 common pathways, respectively. These common enriched pathways are involved in various functions such as metabolism, diseases, cellular processes, biological systems, and information processing, indicating their roles in the immune response to F. columnare. Notably, we investigated in detail the gene expression profiles associated with complement molecules and three classes of cytokines (interleukin, tumor necrosis factor, and interferon) in different organs/tissues and disease states during the pathogenesis of columnaris disease. The findings highlight the importance of inflammatory responses and complement pathways in the pathogenesis of columnaris disease and suggest potential targets for future research and disease management strategies. The present study thus provides valuable insights into the transcriptomic changes and immune responses in grass carp infected with F. columnare, and sheds light on how highly virulent strains of F. columnare cause morbidity and mortality in the host.
Collapse
Affiliation(s)
- Yong Jin
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, and Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong Province, 518120, China; State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Nan Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Shan Nan Chen
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China
| | - Yi Shan Lu
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, and Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong Province, 518120, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, and Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institute, College of Fishery, Guangdong Ocean University, Zhanjiang, Guangdong Province, 524088, China
| | - Zhen Gan
- Guangdong Provincial Engineering Research Center for Aquatic Animal Health Assessment, and Shenzhen Public Service Platform for Evaluation of Marine Economic Animal Seedings, Shenzhen Institute of Guangdong Ocean University, Shenzhen, Guangdong Province, 518120, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, and Key Laboratory of Control for Disease of Aquatic Animals of Guangdong Higher Education Institute, College of Fishery, Guangdong Ocean University, Zhanjiang, Guangdong Province, 524088, China.
| | - Pin Nie
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, Hubei Province, 430072, China; College of Marine Science and Engineering, Qingdao Agricultural University, Qingdao, Shandong Province, 266109, China.
| |
Collapse
|
3
|
Parolini C. Sepsis and high-density lipoproteins: Pathophysiology and potential new therapeutic targets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167761. [PMID: 40044061 DOI: 10.1016/j.bbadis.2025.167761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/19/2025] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
In 2020, sepsis has been defined a worldwide health major issue (World Health Organization). Lung, urinary tract and abdominal cavity are the preferred sites of sepsis-linked infection. Research has highlighted that the advancement of sepsis is not only related to the presence of inflammation or microbial or host pattern recognition. Clinicians and researchers now recognized that a severe immunosuppression is also a common feature found in patients with sepsis, increasing the susceptibility to secondary infections. Lipopolysaccharides (LPS) are expressed on the cell surface of Gram-negative, whereas Gram-positive bacteria express peptidoglycan (PGN) and lipoteichoic acid (LTA). The main mechanism by which LPS trigger host innate immune responses is binding to TLR4-MD2 (toll-like receptor4-myeloid differentiation factor 2), whereas, PGN and LTA are exogenous ligands of TLR2. Nucleotide-binding oligomerization domain (NOD)-like receptors are the most well-characterized cytosolic pattern recognition receptors, which bind microbial molecules, endogenous by-products and environmental triggers. It has been demonstrated that high-density lipoproteins (HDL), besides their major role in promoting cholesterol efflux, possess diverse pleiotropic properties, ranging from a modulation of the immune system to anti-inflammatory, anti-apoptotic, and anti-oxidant functions. In addition, HDL are able at i) binding LPS, preventing the activating of TLR4, and ii) inducing the expression of ATF3 (Activating transcription factor 3), a negative regulator of the TLR signalling pathways, contributing at justifying their capacity to hamper infection-based illnesses. Therefore, reconstituted HDL (rHDL), constituted by apolipoprotein A-I/apolipoprotein A-IMilano complexed with phospholipids, may be considered as a new therapeutic tool for the management of sepsis.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, "Rodolfo Paoletti", via Balzaretti 9 - Università degli Studi di Milano, 20133 Milano, Italy.
| |
Collapse
|
4
|
Jin Z, Cao J, Liu Z, Gao M, Liu H. Comprehensive profiling of candidate biomarkers and immune infiltration landscape in metabolic dysfunction-associated steatohepatitis. Metabol Open 2025; 26:100366. [PMID: 40292075 PMCID: PMC12032907 DOI: 10.1016/j.metop.2025.100366] [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: 03/26/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025] Open
Abstract
Background The incidence of metabolic dysfunction-associated steatohepatitis (MASH) is increasing, with an incompletely understood pathophysiology involving multiple factors, particularly innate and adaptive immune responses. Given the limited pharmacological treatments available, identification of novel immune metabolic targets is urgently needed. In this study, we aimed to identify hub immune-related genes and potential biomarkers with diagnostic and predictive value for MASH patients. Methods The GSE164760 dataset from the Gene Expression Omnibus was utilized for analysis, and the R package was used to identify differentially expressed genes. Immune-related differentially expressed genes (IR-DEGs) were identified by comparing the overlap of differentially expressed genes with well-known immune-related genes. Furthermore, the biological processes and molecular functions of the IR-DEGs were analyzed. To characterize the hub IR-DEGs, we employed a protein-protein interaction network. The diagnostic and predictive values of these hub IR-DEGs in MASH were confirmed using GSE48452 and GSE63067 datasets. Finally, the significance of the hub IR-DEGs was validated using a mouse model of MASH. Results A total of 91 IR-DEGs were identified, with 61 upregulated and 30 downregulated genes. Based on the protein-protein interaction network, FN1, RHOA, FOS, PDGFRα, CCND1, PIK3R1, CSF1, and FGF3 were identified as the hub IR-DEGs. Moreover, we found that these hub genes are closely correlated with immune cells. Notably, the validation across two independent cohorts as well as a murine MASH model confirmed their high diagnostic potential. Conclusion The hub IR-DEGs, such as FN1, RHOA, FOS, PDGFRα, CCND1, PIK3R1, CSF1, and FGF3, may enhance the diagnosis and prognosis of MASH by modulating immune homeostasis.
Collapse
Affiliation(s)
- Zhangliu Jin
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Jianyun Cao
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, Hunan, 410005, China
| | - Zhaoxun Liu
- Nursing Department, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
- Department of Emergency, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Mei Gao
- Department of Pharmacy, Anhui Chest Hospital, Hefei, Anhui, 230000, China
| | - Hailan Liu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
5
|
Shen H, Liangpunsakul S, Iwakiri Y, Szabo G, Wang H. Immunological mechanisms and emerging therapeutic targets in alcohol-associated liver disease. Cell Mol Immunol 2025:10.1038/s41423-025-01291-w. [PMID: 40399593 DOI: 10.1038/s41423-025-01291-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025] Open
Abstract
Alcohol-associated liver disease (ALD) is a major global health challenge, with inflammation playing a central role in its progression. As inflammation emerges as a critical therapeutic target, ongoing research aims to unravel its underlying mechanisms. This review explores the immunological pathways of ALD, highlighting the roles of immune cells and their inflammatory mediators in disease onset and progression. We also examine the complex interactions between inflammatory cells and non-parenchymal liver cells, as well as their crosstalk with extra-hepatic organs, including the gut, adipose tissue, and nervous system. Furthermore, we summarize current clinical research on anti-inflammatory therapies and discuss promising therapeutic targets. Given the heterogeneity of ALD-associated inflammation, we emphasize the need for precision medicine to optimize treatment strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Haiyuan Shen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - Yasuko Iwakiri
- Section of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| |
Collapse
|
6
|
Chen L, Guillot A, Tacke F. Reviewing the function of macrophages in liver disease. Expert Rev Gastroenterol Hepatol 2025:1-17. [PMID: 40387555 DOI: 10.1080/17474124.2025.2508963] [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: 12/03/2024] [Revised: 04/10/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
INTRODUCTION The liver is a central metabolic organ, but is also hosting a unique immune microenvironment to sustain homeostasis and proper defense measures against injury threats in healthy individuals. Liver macrophages, mostly represented by the tissue-resident Kupffer cells and bone marrow- or monocyte-derived macrophages, are intricately involved in various aspects of liver homeostasis and disease, including tissue injury, inflammation, fibrogenesis and repair mechanisms. AREAS COVERED We review recent findings on defining the liver macrophage landscape and their functions in liver diseases with the aim of highlighting potential targets for therapeutic interventions. A comprehensive literature search in PubMed and Google Scholar was conducted to identify relevant literature up to date. EXPERT OPINION Liver macrophages orchestrate key homeostatic and pathogenic processes in the liver. Thus, targeting liver macrophages represents an attractive strategy for drug development, e.g. to ameliorate liver inflammation, steatohepatitis or fibrosis. However, translation from fundamental research to therapies remains challenging due to the versatile nature of the liver macrophage compartment. Recent and major technical advances such as single-cell and spatially-resolved omics approaches deepened our understanding of macrophage biology at a molecular level. Yet, further studies are needed to identify suitable, etiology- and stage-dependent strategies for the treatment of liver diseases.
Collapse
Affiliation(s)
- Lanlan Chen
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Adrien Guillot
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
7
|
Wang L, Zhang H, Jin M, Zheng A, Lu J, Liu W, Zhang J, Tang B, Huang J, Wang B, Wang Z. Transcriptome-based analysis of the immune response of the four-finger threadfin (Eleutheronema tetradactylum) to Vibrio harveyi infection. FISH & SHELLFISH IMMUNOLOGY 2025; 164:110431. [PMID: 40409697 DOI: 10.1016/j.fsi.2025.110431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 05/18/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
The four-finger threadfin (Eleutheronema tetradactylum), an economically important fish species, has faced significant economic losses due to bacterial diseases, particularly Vibrio harveyi, a Gram-negative bacterium. In this study, we used RNA sequencing (RNA-seq) to analyze transcriptomic responses in the intestine and liver of E. tetradactylum post-V. harveyi infection, comparing control (Cg) and infected (Ig) groups for intestine (Cg-IN vs. Ig-IN) and liver (Cg-LI vs. Ig-LI). The results revealed significant changes in gene expression, with 2783 differentially expressed genes (DEGs) in the intestine (1831 upregulated and 952 downregulated) and 4940 DEGs in the liver (2685 upregulated and 2255 downregulated). KEGG pathway analysis identified immune-related pathways significantly enriched in both tissues. In the intestine, the most enriched pathways were the complement and coagulation cascades and the IL-17 signaling pathway. In the liver, in addition to the complement and coagulation cascades and IL-17 signaling pathway, four additional immune-related pathways were detected, including the NOD-like receptor signaling, Toll and Imd signaling, RIG-I-like receptor signaling and Toll-like receptor signaling pathways. These immune-related pathways highlight the coordinated immune response to V. harveyi infection. Moreover, qPCR validation of nine immune-related DEGs confirmed the accuracy of the RNA-seq data, with consistent expression patterns observed between both methods. This comparative transcriptome analysis of E. tetradactylum infected with V. harveyi provides valuable data and theoretical insights for further studies on the immune system and defense mechanisms in four-finger threadfin.
Collapse
Affiliation(s)
- Linjuan Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Huijuan Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Minxuan Jin
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Anna Zheng
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Jingheng Lu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Weibin Liu
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Jiandong Zhang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Baogui Tang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Jiansheng Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China
| | - Bei Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, Guangdong Province, China
| | - Zhongliang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang, 524088, Guangdong Province, China; Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Zhanjiang, 524088, Guangdong Province, China; Guangdong Marine Fish Science and Technology Innovation Center, Zhanjiang, 524088, Guangdong Province, China; Agro-Tech Extension Center of Guangdong Province, Guangzhou, 510500, China.
| |
Collapse
|
8
|
Tian H, Li P, Lin L, Fan S, Li R, Zhou L, Zhao Q, Zhang J, Tang C. Alleviating Effect and Potential Mechanisms of Selenium Supplementation on Chronic Liver Injury Induced by Oxidized Soybean Oil. Mol Nutr Food Res 2025:e70124. [PMID: 40392050 DOI: 10.1002/mnfr.70124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Lipid oxidation can induce liver oxidative stress and lipotoxic damage, while selenium (Se) possesses detoxification, antioxidation, immunity, and liver protection functions. However, the effects of Se and oxidized lipids on liver oxidative stress and lipid metabolism, along with the underlying mechanisms, remain underexplored. This study aimed to investigate the protective effects of Se against liver injury induced by oxidized soybean oil (OSO) in mice. C57BL/6J mice (n = 60) were randomly divided into Control (0.2 mg/kg Se+7% fresh soybean oil), OSO (0.2 mg/kg Se+7% OSO), and OSO+Se (1.0 mg/kg Se+7% OSO) groups for 10 weeks. The results showed that Se supplementation mitigated the morphological structure and functional impairment, inflammation, and oxidative stress of the liver caused by OSO, and improved changes to the liver fatty acid profile and lipid metabolism disorders. It also reversed the OSO-induced imbalance of liver polyunsaturated fatty acid metabolites and inhibited OSO-induced activation of the PI3K-AKT pathway. Se may activate the Nrf2 pathway and inhibit the PI3K-AKT pathway to improve inflammation, oxidative stress, and fatty acid metabolism disorders, thereby reducing liver injury. These findings highlight the nutritional relevance of Se as a potential therapeutic agent for preventing liver damage from oxidized lipids.
Collapse
Affiliation(s)
- Huihui Tian
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Pingyang Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Luxi Lin
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shijie Fan
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruitong Li
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Longzhu Zhou
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qingyu Zhao
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junmin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chaohua Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
9
|
Wang SQ, Meng YQ, Wu YL, Nan JX, Jin CH, Lian LH. Imidazole-Based ALK5 Inhibitor Attenuates TGF-β/Smad-Mediated Hepatic Stellate Cell Activation and Hepatic Fibrogenesis. Chem Res Toxicol 2025; 38:930-941. [PMID: 40211771 DOI: 10.1021/acs.chemrestox.5c00036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Liver fibrosis resulting from severe liver damage is a major clinical problem for which effective pharmacological drugs and treatment strategies are lacking. TGF-β, a hallmark of liver fibrosis, has been shown to promote ALK5 phosphorylation in an activated state. Hence, the suppression of ALK5 signal transduction has emerged as a promising therapeutic strategy for the treatment of liver fibrosis. In this study, the imidazole derivative J-1149, which exhibited inhibitory activity against ALK5, was synthesized to exert antifibrotic effects, and the inhibition mechanisms were uncovered. Our findings suggested that J-1149 significantly attenuated HSC activation and liver fibrogenesis by acting on the TGF-β/Smad signaling pathway. Concurrently, the potential of J-1149 to impede the P2X7R/NLRP3 axis, curtail the infiltration of macrophages and neutrophils, and reduce liver fibrogenesis was also highlighted. These results demonstrated that J-1149 is a promising candidate for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Si-Qi Wang
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yu-Qing Meng
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Yan-Ling Wu
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Ji-Xing Nan
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Cheng-Hua Jin
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| | - Li-Hua Lian
- Key Laboratory of Traditional Chinese Korean Medicine Research (Yanbian University) of State Ethnic Affairs Commission, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
- Key Laboratory of Natural Medicines of Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, China
| |
Collapse
|
10
|
Zhao R, Zhang X, Liu X, Wang S, Leng S, Peng J, Hu X. Piezo1 deficiency alleviates acute liver failure by inhibiting CpG-ODN induced inflammatory responses. Int Immunopharmacol 2025; 159:114879. [PMID: 40394794 DOI: 10.1016/j.intimp.2025.114879] [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: 02/12/2025] [Revised: 05/11/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025]
Abstract
Macrophages play a critical role in the progression of acute liver failure (ALF), a life-threatening clinical syndrome characterized by the rapid onset of liver injury. Recent research suggests that mechanosensation signaling may be pivotal in modulating acute inflammation. However, its involvement in ALF pathogenesis remains poorly understood. In this study, we found that Piezo1 was highly expressed in liver macrophages during liver injury in both humans and mice. Mice with macrophage-specific Piezo1 depletion were resistant to CpG-ODN/D-GalN-induced ALF, whereas mice pretreated with Yoda1 showed increased susceptibility. Furthermore, Piezo1-deficient macrophages exhibited reduced secretion of inflammatory cytokines upon CpG-ODN stimulation in vitro, while Yoda1 treatment enhanced cytokine secretion. Mechanistically, Piezo1 activation promotes the phosphorylation of CaMKII, which further enhances CpG-ODN-induced NF-κB activation. Additionally, Piezo1 activation promotes the endosomal translocation of TLR9 through the cytoskeleton remodeling. These findings suggest that Piezo1-mediated mechanosensation contributes to the development of CpG-ODN/D-GalN-induced ALF and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Ruxia Zhao
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyu Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyue Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China
| | - Shuwen Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China
| | - Shaoqiu Leng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China.
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China; Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China; State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| | - Xiang Hu
- Shandong Key Laboratory of Hematological Diseases and Immune Microenvironment, Qilu Hospital of Shandong University, Jinan, China; State Key Laboratory for Innovation and Transformation of LuobingTheory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
11
|
Yan X, Arcoverde Cerveira R, Ols S, Lenart K, Hellgren F, Miranda M, Engstrand O, Reinhardt A, Eriksson B, Loré K. Biochemical and hematological reference intervals in rhesus and cynomolgus macaques and implications for vaccine and drug development. Lab Anim (NY) 2025:10.1038/s41684-025-01547-y. [PMID: 40379874 DOI: 10.1038/s41684-025-01547-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 03/26/2025] [Indexed: 05/19/2025]
Abstract
Nonhuman primates have a key role in the evaluation of novel therapeutics including vaccine and drug development. Monitoring biochemical and hematological parameters of macaques is critical to understand toxicity and safety, but general reference intervals following standardized guidelines remain to be determined. Here we compiled multiple internal datasets to define normal ranges of classical biochemical and hematological parameters in Indian and Chinese rhesus macaques as well as cynomolgus macaques. Furthermore, the combination of hematological data with phenotypic information of cells obtained by flow cytometry enabled analyses of specific immune cell subsets. We found that vaccination generally induced transient changes at 24 h in cell frequencies accompanied by fluctuation in selected liver enzymes and metabolites. However, most parameters remained within our identified reference intervals. These deviations did not lead to noticeable side effects. Fluctuation in selected biochemical and hematological parameters was accompanied with differentiation of CD14+CD16+ intermediate monocytes and upregulation of genes associated with interleukin-1 signaling. By contrast, two animals with noticeable side effects showed sustained deviations. This study provides insights into baseline and vaccine-induced biochemical and hematological profiles of healthy macaques, facilitating the interpretation of toxicity and safety assessments in preclinical trials of novel therapies.
Collapse
Affiliation(s)
- Xianglei Yan
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Klara Lenart
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Fredrika Hellgren
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Marcos Miranda
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Olivia Engstrand
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
- Center of Molecular Medicine, Stockholm, Sweden
| | - Bengt Eriksson
- Astrid Fagraeus Laboratory, Comparative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden.
- Center of Molecular Medicine, Stockholm, Sweden.
| |
Collapse
|
12
|
Zhuang X, Shi S, Liu S, Jiao Y, Huang B, Yang Y, Yang L, Yang X, Wang H, Liang C, Song D, Yu H, Zou D, Sun Q, Yang S, Yin C, Li J, Liu Y, Min J, Wang F, Nian Y, Du L, Chu B. Dipyridamole Acts as Clinical Ferroptosis Inhibitor to Prevent from Tissue Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500566. [PMID: 40365742 DOI: 10.1002/advs.202500566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/18/2025] [Indexed: 05/15/2025]
Abstract
Ferroptosis is a newly identified cell death triggered by iron-induced lipid peroxidation. Numerous studies reveal that ferroptosis participates in multiple types of tissue injury including ischaemia-reperfusion (I/R) injury and doxorubicin (Dox)-induced damage. Targeting ferroptosis is a promising approach for disease treatment as the blockade of ferroptosis efficiently alleviates the symptoms. However, no known ferroptosis inhibitors have been used for clinical treatment. Although certain clinical compounds act as ferroptosis inhibitors in vitro, whether these drugs cure tissue injury by suppressing ferroptosis is little known. Here, by screening a large panel of drugs used in the clinic, it is identified that dipyridamole significantly attenuates Dox or I/R-induced cardiac injury. Moreover, dipyridamole can achieve a good therapeutic effect on liver and kidney injury. Mechanistically, dipyridamole-mediated ferroptosis inhibition is strictly dependent on solute carrier family 7 member 11 (SLC7A11). Dipyridamole down-regulates the expression of ring finger protein 126 (RNF126), which is an E3 ligase to ubiquitinate SLC7A11 for proteasome degradation. Deficiency of SLC7A11 largely blocks the protective role of dipyridamole in vitro and in vivo. Together, the findings uncover that dipyridamole acts as a clinical compound to alleviate organ injury via suppressing ferroptosis, providing novel insights into the clinical therapy for ferroptosis-related tissue damage.
Collapse
Affiliation(s)
- Xiao Zhuang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Shuang Shi
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, 250012, China
| | - Shuo Liu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yaqiong Jiao
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Bin Huang
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Yinghong Yang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Li Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, China
| | - Xinquan Yang
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hui Wang
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Chunhui Liang
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Dandan Song
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huaxiang Yu
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Dan Zou
- Department of Emergency Medicine and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Qi Sun
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, 250012, China
| | - Shu Yang
- College of Pharmacy, Nanjing drum tower hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chengqian Yin
- Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China
| | - Jian Li
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, 250012, China
| | - Yiming Liu
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fudi Wang
- The First Affiliated Hospital, The Second Affiliated Hospital, Institute of Translational Medicine, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yong Nian
- College of Pharmacy, Nanjing drum tower hospital, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Lutao Du
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
- Shandong Provincial Key Laboratory of Innovation Technology in Laboratory Medicine, Jinan, 250012, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| |
Collapse
|
13
|
Jiang X, Wang M, Zou R, Fu M, Fan W, Wang Y, Dai C, Swapnil Z, Wang W, Wu H, Xie K, Liu L, Wang Y, Fan Z, Zhao L. Harnessing Kupffer Cell Metabolic Rewiring: Rapamycin-Gliadin Nanoparticle as a Pivotal Strategy for Immune Tolerance in Celiac Disease. ACS NANO 2025; 19:17462-17477. [PMID: 40302617 DOI: 10.1021/acsnano.4c18354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Celiac disease (CeD), triggered by gliadin exposure, necessitates therapeutic strategies that establish an antigen-specific immune tolerance. This study explores the therapeutic efficacy and mechanism of rapamycin-gliadin composite nanoparticles (PLN-GR) for CeD treatment. In vivo analyses demonstrated the efficient uptake of PLN-GR by antigen-presenting cells (APCs), particularly Kupffer cells and splenic dendritic cells (DCs), driving their tolerogenic phenotypic transformation. In a murine CeD model, PLN-GR administration significantly enhanced gluten tolerance and mitigated intestinal inflammation, as indicated by reduced paw edema and improved histopathological parameters. Mechanistically, PLN-GR induced macrophage metabolic reprogramming from glycolysis to oxidative phosphorylation, concomitant with elevated serum itaconate levels. This metabolic shift potentiated interorgan immunoregulatory crosstalk, expanding PD-L1+ tolerogenic splenic DCs while suppressing pathogenic Th1 cell populations. Bone marrow-derived macrophages (BMDMs) from Acod1-/- mice (deficient in itaconate synthesis) failed to induce DC tolerance upon PLN-GR treatment. However, supplementation with the itaconate derivative 4-octyl itaconate (4-OI) restored PD-L1 expression in DC2.4 cells in vitro, revealing that itaconate induces and stabilizes the tolerant DC phenotype. These findings underscore PLN-GR as a novel nanotherapeutic platform for CeD, achieving gliadin-specific tolerance through hepatic-splenic immunometabolic reprogramming and itaconate-dependent PD-L1 regulation, thereby offering a translatable strategy for autoimmune disease management.
Collapse
Affiliation(s)
- Xiaohan Jiang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Min Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Ruihan Zou
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Min Fu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Fourth Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wentao Fan
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Fourth Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yao Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Chenguang Dai
- Department of Gastroenterology, First Afilliated Hospital of Soochow University, Soochow 215000, China
| | - Zaman Swapnil
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wanjun Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Hao Wu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Kunxin Xie
- Pancreas Center, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yan Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili 835800, China
| | - Zhining Fan
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Lili Zhao
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
14
|
Schultheiß C, Binder M. nACh receptors engage a nice ChAT with the liver: B cells serving up regeneration. Immunity 2025; 58:1177-1179. [PMID: 40367919 DOI: 10.1016/j.immuni.2025.04.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/16/2025]
Abstract
Liver regeneration is a remarkable and unique biological process, orchestrated by an intricate cellular crosstalk of (non-)parenchymal, immune, and nerve cells. In this issue of Immunity, Modares et al. uncover the pivotal role of choline acetyltransferase (ChAT+)-expressing B cells as key orchestrators of liver regeneration.
Collapse
Affiliation(s)
- Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland; Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland; Collaborative Research Institute Intelligent Oncology (CRIION), Freiburg, Germany.
| |
Collapse
|
15
|
Xie H, Wang J, Zhao Q. Identification of potential metabolic biomarkers and immune cell infiltration for metabolic associated steatohepatitis by bioinformatics analysis and machine learning. Sci Rep 2025; 15:16596. [PMID: 40360670 PMCID: PMC12075577 DOI: 10.1038/s41598-025-86397-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Metabolic associated steatohepatitis (MASH) represents a severe subtype of metabolic associated fatty liver disease (MASLD), with an increased risk of progression to cirrhosis and hepatocellular carcinoma. The nomenclature shift from nonalcoholic steatohepatitis (NASH)/nonalcoholic fatty liver disease (NAFLD) to MASH/MASLD, underscores the pivotal role of metabolic factors in disease progression. Diagnosis of MASH currently hinges on liver biopsy, a procedure whose invasive nature limits its clinical utility. This study aims to identify and validate metabolism-related genes (MRGs) markers for the non-invasive diagnosis of MASH. METHODS This study extracted multiple datasets from the GEO database to identify metabolism-related differentially expressed genes (MRDEGs). Protein-Protein Interaction (PPI) network and machine learning algorithms, including Least Absolute Shrinkage and Selection Operator (LASSO) regression, Support Vector Machine-Recursive Feature Elimination (SVM-RFE), and Random Forest (RF), were applied to screen for signature MRDEGs. The diagnostic performance of these MRDEGs was evaluated using the Receiver Operating Characteristic (ROC) curve and further validated using independent external datasets. Additionally, enrichment analysis was performed to uncover key driver pathways in MASH. The infiltration levels of various immune cell types were assessed using single sample Gene Set Enrichment Analysis (ssGSEA). Finally, Spearman correlation analysis confirmed the association between signature genes and immune cells. RESULTS We successfully identified seven signature MRDEGs, including CYP7A1, GCK, AKR1B10, HPRT1, GPD1, FADS2, and ENO3, through PPI network analysis and machine learning algorithms. The gene model displayed exceptional diagnostic performance in the training and validation cohorts, as evidenced by the area under ROC curve (AUC) exceeding 0.9. Further enrichment analysis revealed that signature MEDEGs were primarily involved in multiple biological pathways related to glucose and lipid metabolism. Immune infiltration analysis indicated a significant increase in the infiltration levels of activated CD8 T cells, gamma-delta T cells, natural killer cells, and CD56bright NK cells in patients with MASH. CONCLUSION This study successfully identified seven signature MRDEGs as significant diagnostic biomarkers for MASH. The findings not only offer novel strategies for non-invasive diagnosis of MASH but also highlight the substantial role of immune cell infiltration in the progression of MASH.
Collapse
Affiliation(s)
- Haoran Xie
- Hepatobiliary Pancreatic Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Junjun Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyan Zhao
- Department of Gastroenterology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
16
|
Li Y, Cao Z, Lu Y, Lei C, Lyu W. Knowledge landscape of macrophage research in liver fibrosis: a bibliometric review of the literature from WoSCC. Front Pharmacol 2025; 16:1571879. [PMID: 40406489 PMCID: PMC12094998 DOI: 10.3389/fphar.2025.1571879] [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: 03/04/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Recent insights into the immune response in fibrosis have provided valuable perspectives for the treatment of liver fibrosis. Macrophages, as the most abundant immune cells in the liver, are key drivers of liver fibrosis. They are extensively involved in tissue damage, chronic inflammation, and the progression and regression of liver fibrosis. This study aims to conduct a bibliometric analysis and literature review on the mechanisms by which macrophages contribute to liver fibrosis. Specifically, we analyzed a bibliometric dataset comprising 1,312 papers from 59 countries, 1,872 institutions, and 9,784 authors. Keyword co-occurrence analysis identified key research hotspots, including the role of macrophage subtypes in obesity-related metabolic disorders, the crosstalk between macrophages and hepatic stellate cells through mechanoimmunology, emerging strategies for immune modulation targeting macrophages to promote fibrosis regression and liver regeneration, and new discoveries regarding macrophage crosstalk with other immune cells. In conclusion, this study provides a visual analysis of the current research landscape, hotspots, and trends in the field of macrophages and liver fibrosis, and discusses future directions for further exploration in this area.
Collapse
Affiliation(s)
- Yanbo Li
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Zhengmin Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Yanping Lu
- Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Chao Lei
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| | - Wenliang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Chandramoorthy HC, Saleh RO, Altalbawy FMA, Mohammed JS, Ganesan S, Kundlas M, Premkumar J, Ray S, Mustafa YF, Abbas JK. Deciphering cGAS-STING signaling: implications for tumor immunity and hepatocellular carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04240-6. [PMID: 40332552 DOI: 10.1007/s00210-025-04240-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent form of primary liver cancer and poses a significant global health challenge due to its rising incidence and associated mortality. Recent advancements in understanding the cytosolic DNA sensing, the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway have illuminated its critical role in the immune response to HCC. This narrative review deciphers the multifaceted involvement of cGAS-STING in HCC, mainly its function in detecting cytosolic DNA and initiating type I interferon (IFN-I) responses, which are pivotal for antitumor immunity. This immune response is crucial for combating pathogens and can play a role in tumor surveillance. In the context of HCC, the tumor microenvironment (TME) can exhibit immune resistance, which complicates the effectiveness of therapies like immune checkpoint blockade. However, activation of the cGAS-STING pathway has been shown to stimulate antitumor immune responses, enhancing the activity of dendritic cells and cytotoxic T lymphocytes. There is ongoing research into STING agonists as a treatment strategy for HCC, with some studies indicating promising results in prolonging survival and enhancing the immune response against tumors. By summarizing current knowledge and identifying research gaps, this review aims to provide a comprehensive overview of cGAS-STING signaling in HCC and its future directions, emphasizing its potential as a therapeutic target in the fight against HCC. Understanding these mechanisms could pave the way for innovative immunotherapeutic approaches that enhance the efficacy of existing treatments and improve patient prognosis.
Collapse
Affiliation(s)
- Harish C Chandramoorthy
- Department of Microbiology & Clinical Parasitology, College of Medicine & Central Research Laboratories, King Khalid University, Abha, Saudi Arabia
| | - Raed Obaid Saleh
- Department of Medical Laboratories Techniques, College of Health and Medical Techniques, University of Al Maarif, Al Anbar, 31001, Iraq.
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Jaafaru Sani Mohammed
- Medical Analysis Department, Faculty of Applied Science, Tishk International University, Erbil, Iraq
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - J Premkumar
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Jamal K Abbas
- Department of Pharmaceutical, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
18
|
Chen M, He Y, Jia Y, Wu L, Zhao R. Liver transcriptome response to avian pathogenic Escherichia coli infection in broilers with corticosterone treatment. Poult Sci 2025; 104:105020. [PMID: 40088534 PMCID: PMC11937665 DOI: 10.1016/j.psj.2025.105020] [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: 01/02/2025] [Revised: 02/25/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025] Open
Abstract
Avian pathogenic Escherichia coli (APEC) infection has high morbidity and mortality, and multiple stressors encountered during rearing place poultry in a state of stress. However, research on how poultry cope with APEC infection under stress situation is still limited. In this study, we established a broiler stress model by corticosterone (CORT) administration subcutaneously for 7 consecutive days, followed by APEC challenge intramuscularly. CORT treatment significantly reduced body weight (BW) and average daily gain (ADG) while increasing feed conversion ratio (FCR) (P < 0.01). APEC infection significantly decreased ADG (P < 0.01). CORT treatment and APEC infection elevated plasma corticosterone and heterophil to lymphocyte (H/L) ratio (P < 0.05). Additionally, plasma aspartate aminotransferase (AST), AST to alanine aminotransferase (AST/ALT) ratio, and lactate dehydrogenase (LDH) levels increased significantly (P < 0.01). Histopathological analysis revealed structural damage of liver, indicating that CORT treatment and APEC infection induced liver injury. However, CORT pretreatment broilers exhibited a milder histopathological lesions and significantly lower AST, ALT, and LDH levels (P < 0.05) compared to APEC infection alone. CORT treatment and APEC infection increased plasma levels of lysozyme (LZM), total protein (TP), and globulin (GLOB) (P < 0.05), while CORT pretreatment further elevating their concentrations compared to APEC infection alone, suggesting an enhanced innate immune response. Liver transcriptomic analysis identified 768, 335, and 567 differentially expressed genes (DEGs) following CORT, APEC, or both treatments, respectively, enriched in cytokine-cytokine receptor interaction, PPAR signaling pathway, Toll-like receptor signaling pathway, MAPK signaling pathway, steroid hormone biosynthesis pathway, arachidonic acid metabolism, and phagosome pathway, etc., indicating that CORT treatment regulates lipid metabolism and immunity, while APEC infection induces inflammation and disrupts lipid metabolism. Notably, CORT pretreatment may mitigate APEC induced liver injury by enhancing phagosome function. Moreover, glucocorticoid receptor (GR) may regulate DEGs expressions, thus affected broilers response to CORT, APEC, or both treatments. These results suggest that CORT treatment, APEC infection, or both significantly affect the growth performance, immune response and liver function of broilers, while lipid metabolism may play a crucial role.
Collapse
Affiliation(s)
- Mengru Chen
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Yifei He
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Lei Wu
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health & Food Safety, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China; Key Laboratory of Animal Physiology & Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu, 210095, PR China.
| |
Collapse
|
19
|
Liu J, Cheng P, Xu C, Pu K. Molecular probes for in vivo optical imaging of immune cells. Nat Biomed Eng 2025; 9:618-637. [PMID: 39984703 DOI: 10.1038/s41551-024-01275-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/23/2024] [Indexed: 02/23/2025]
Abstract
Advancing the understanding of the various roles and components of the immune system requires sophisticated methods and technology for the detection of immune cells in their natural states. Recent advancements in the development of molecular probes for optical imaging have paved the way for non-invasive visualization and real-time monitoring of immune responses and functions. Here we discuss recent progress in the development of molecular probes for the selective imaging of specific immune cells. We emphasize the design principles of the probes and their comparative performance when using various optical modalities across disease contexts. We highlight molecular probes for imaging tumour-infiltrating immune cells, and their applications in drug screening and in the prediction of therapeutic outcomes of cancer immunotherapies. We also discuss the use of these probes in visualizing immune cells in atherosclerosis, lung inflammation, allograft rejection and other immune-related conditions, and the translational opportunities and challenges of using optical molecular probes for further understanding of the immune system and disease diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Cheng Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
20
|
Rigual MDM, Angulo-Aguado M, Zagorac S, Álvarez-Díaz R, Benítez-Mondéjar M, Yi F, Martínez-Garay C, Santos-de-Frutos K, Kim E, Campos-Olivas R, Djouder N. Macrophages harness hepatocyte glutamate to boost liver regeneration. Nature 2025; 641:1005-1016. [PMID: 40140584 DOI: 10.1038/s41586-025-08778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 02/12/2025] [Indexed: 03/28/2025]
Abstract
Liver regeneration after hepatectomy follows accurate coordination with the body's specific requirements1-3. However, the molecular mechanisms, factors and particular hepatocyte population influencing its efficiency remain unclear. Here we report on a unique regeneration mechanism involving unconventional RPB5 prefoldin interactor 1 (URI1), which exclusively colocalizes with, binds to and activates glutamine synthase (GS) in pericentral hepatocytes. Genetic GS or URI1 depletion in mouse pericentral hepatocytes increases circulating glutamate levels, accelerating liver regeneration after two-third hepatectomy. Conversely, mouse hepatocytic URI1 overexpression hinders liver restoration, which can be reversed by elevating glutamate through supplementation or genetic GS depletion. Glutamate metabolically reprograms bone-marrow-derived macrophages, stabilizing HIF1α, which transcriptionally activates WNT3 to promote YAP1-dependent hepatocyte proliferation, boosting liver regeneration. GS regulation by URI1 is a mechanism that maintains optimal glutamate levels, probably to spatiotemporally fine-tune liver growth in accordance with the body's homeostasis and nutrient supply. Accordingly, in acute and chronic injury models, including in cirrhotic mice with low glutamate levels and in early mortality after liver resection, as well as in mice undergoing 90% hepatectomy, glutamate addition enhances hepatocyte proliferation and survival. Furthermore, URI1 and GS expression co-localize in human hepatocytes and correlate with WNT3 in immune cells across liver disease stages. Glutamate supplementation may therefore support liver regeneration, benefiting patients awaiting transplants or recovering from hepatectomy.
Collapse
Affiliation(s)
- María Del Mar Rigual
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Mariana Angulo-Aguado
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Sladjana Zagorac
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Ruth Álvarez-Díaz
- Bioinformatic Unit, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Marta Benítez-Mondéjar
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Fengming Yi
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Carlos Martínez-Garay
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Karla Santos-de-Frutos
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Eunjeong Kim
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
- KNU G-LAMP Research Center, KNU Institute of Basic Sciences, BK21 FOUR KNU Creative BioResearch Group, Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea
| | - Ramón Campos-Olivas
- Spectroscopy and Nuclear Magnetic Resonance Unit, Structural Biology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Nabil Djouder
- Growth Factors, Nutrients and Cancer Group, Molecular Oncology Programme, Centro Nacional Investigaciones Oncológicas (CNIO), Madrid, Spain.
| |
Collapse
|
21
|
Noda T, Tanaka S, Maruta Y, Haruna M, Mizuguchi S, Fujimoto A, Urashima K, Kohda Y, Kato R. Brigatinib activates inflammasomes: Implication for immune-related adverse events. Toxicol Appl Pharmacol 2025; 498:117310. [PMID: 40122348 DOI: 10.1016/j.taap.2025.117310] [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: 12/26/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKI), including brigatinib, are widely used to treat ALK-positive non-small cell lung cancer. However, severe adverse effects associated with brigatinib, such as interstitial pneumonia and liver dysfunction, may involve immune system activation. The precise mechanisms underlying these immune-related adverse effects remain unclear. In this study, we evaluated the direct activation of inflammasomes by brigatinib and other ALK TKI (crizotinib, alectinib, ceritinib) in differentiated THP-1 cells. Additionally, we analyzed the inflammasome-activating potential of supernatants from functional liver cell (FLC)-4 cells treated with these drugs. Our results demonstrate that brigatinib directly activates inflammasomes in THP-1 cells, inducing the production of interleukin-1β and the activation of caspase-1. In contrast, no inflammasome activation was observed with the other ALK TKIs. Furthermore, supernatants from FLC-4 cells, characterized by high drug-metabolizing activity, were shown to activate inflammasomes in differentiated THP-1 cells following treatment with brigatinib. Brigatinib treatment significantly increased the levels of damage-associated molecular patterns (DAMPs), including heat shock protein 90 and S100A6, in the supernatants of FLC-4 cells. These findings suggest that brigatinib induces the release of DAMPs from hepatocytes, which subsequently activate inflammasomes. This mechanism may be essential for brigatinib-induced immune system activation and the development of immune-related adverse events.
Collapse
Affiliation(s)
- Takumi Noda
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Saori Tanaka
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Yuto Maruta
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Moe Haruna
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Serina Mizuguchi
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Ayumi Fujimoto
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Kazuya Urashima
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Yuka Kohda
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan
| | - Ryuji Kato
- Department of Pharmacotherapeutics and Toxicology, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, Osaka 569-1094, Japan.
| |
Collapse
|
22
|
Wen C, Tao H, Chen H, Pu W, Yan Q, Zou Y, Su SS, Zhou L, Peng Y, Wang G, Xu T, Zheng X, Wu M, Dai Y. Single-cell RNA sequencing and functional analysis reveal the role of altered glycosylation levels of hepatic macrophages in liver cirrhosis. J Gastroenterol 2025; 60:607-620. [PMID: 39888412 DOI: 10.1007/s00535-025-02218-y] [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: 09/02/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Liver cirrhosis represents a critical stage of chronic liver disease, characterized by progressive liver damage, cellular dysfunction, and disrupted cell-to-cell interactions. Glycosylation, an essential post-translational modification, significantly influences cellular behavior and disease progression. Its role in cirrhosis at the single-cell level remains unclear, despite its importance. METHODS This study, based on single-cell glycosylation and transcriptome data, compared the expression of differentially expressed genes in liver tissues from cirrhotic and healthy control samples, identifying changes in glycosylation-related genes and their functional pathway enrichment characteristics. Additionally, it analyzed the composition of immune cells and intercellular interaction features, with a focus on the interaction between macrophages and other immune cells and their potential role in immune regulation. RESULTS The analysis revealed significant changes in immune cell composition and glycosylation patterns in cirrhotic livers. Specifically, the number of macrophages increased substantially, while overall glycosylation levels decreased. Enhanced interactions between macrophages and other cell types were observed, highlighting the central role of macrophages in reshaping the immune microenvironment during cirrhosis progression. Gene expression analysis showed a marked upregulation of FUCA1, a gene encoding a glycosylation-related hydrolase. This change was strongly associated with the observed reduction in glycosylation levels. Functional enrichment analysis further revealed that glycosylation-related genes were primarily involved in immune pathways, including antigen processing and presentation, cytokine signaling, and immune activation. CONCLUSIONS Single-cell glycosylation analysis provides crucial insights into immune cell interactions in cirrhosis. Targeting glycosylation pathways in macrophages may offer new treatment strategies for cirrhosis.
Collapse
Affiliation(s)
- Chunmei Wen
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Huihui Tao
- School of Medicine, Anhui University of Science & Technology, Huainan, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, China.
| | - Huaizhou Chen
- The Organ Transplantation Department of 924th Hospital of Joint Logistic Support Force of PLA, Guilin, China
| | - Wenjun Pu
- Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Qiang Yan
- The Organ Transplantation Department of 924th Hospital of Joint Logistic Support Force of PLA, Guilin, China
| | - Yaoshuang Zou
- The Organ Transplantation Department of 924th Hospital of Joint Logistic Support Force of PLA, Guilin, China
| | - Sheng Sean Su
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Lingling Zhou
- School of Medicine, Anhui University of Science & Technology, Huainan, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, China
| | - Yali Peng
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Guoying Wang
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Tiantian Xu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Xuejia Zheng
- The First Hospital of Anhui University of Science and Technology, Huainan, China
| | - Mengyao Wu
- School of Medicine, Anhui University of Science & Technology, Huainan, China
| | - Yong Dai
- School of Medicine, Anhui University of Science & Technology, Huainan, China.
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, China.
- The Organ Transplantation Department of 924th Hospital of Joint Logistic Support Force of PLA, Guilin, China.
- The First Hospital of Anhui University of Science and Technology, Huainan, China.
| |
Collapse
|
23
|
Tan W, Deng J, Qi L, Tan Z. The role of hepatic sinusoidal microenvironment in NASH: pathogenesis, animal models, and therapeutic prospects. Front Pharmacol 2025; 16:1467950. [PMID: 40356963 PMCID: PMC12066276 DOI: 10.3389/fphar.2025.1467950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 03/28/2025] [Indexed: 05/15/2025] Open
Abstract
The incidence of nonalcoholic steatohepatitis (NASH) is increasing annually, posing a significant threat to human health. NASH is typified by hepatic steatosis, inflammation, and hepatocellular injury, frequently culminating in fibrosis and cirrhosis. Yet, the precise pathogenesis of NASH remains to be fully elucidated. The hepatic sinusoid, which serves as the fundamental structural and functional unit of the liver, is intricately composed of endothelial cells, Kupffer cells, and hepatic stellate cells. Consequently, the homeostasis of the hepatic sinusoidal microenvironment may exert a pivotal influence on the progression and prognosis of NASH. However, the limitations of current NASH animal models have significantly impeded advancements in understanding the disease's pathogenesis and the development of effective therapeutic interventions. In light of these challenges, this review endeavors to delve deeper into the critical role of hepatic sinusoidal microenvironment homeostasis in the pathogenesis of NASH, critically analyze the commonly employed animal models, and comprehensively summarize the most recent and promising developments in drug research and development. It is anticipated that these efforts will collectively expedite the advancement of the field of NASH research and therapeutic innovation.
Collapse
Affiliation(s)
- Wanying Tan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jiangting Deng
- Sichuan Academy of Chinese Medicine Sciences, Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Chengdu, Sichuan, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Lingjun Qi
- Affiliated Sichuan Gem Flower Hospital of North Sichuan Medical College, Chengdu, Sichuan, China
| | - Zhenghuai Tan
- Sichuan Academy of Chinese Medicine Sciences, Sichuan Provincial Key Laboratory of Quality and Innovation Research of Chinese Materia Medica, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Kunduzova O, Oksenych V, Kamyshnyi O. Unlocking the gut-liver axis: microbial contributions to the pathogenesis of metabolic-associated fatty liver disease. Front Microbiol 2025; 16:1577724. [PMID: 40351307 PMCID: PMC12061941 DOI: 10.3389/fmicb.2025.1577724] [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: 02/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex metabolic disorder characterized by hepatic lipid accumulation and subsequent inflammation. This condition is closely linked to metabolic syndrome and obesity, with its prevalence rising due to sedentary lifestyles and high-calorie diets. The pathogenesis of MAFLD involves multiple factors, including insulin resistance, lipotoxicity, oxidative stress, and inflammatory responses. The gut microbiota plays a crucial role in MAFLD development, with dysbiosis contributing to liver inflammation through various mechanisms, such as enhanced intestinal permeability and the translocation of bacterial products like lipopolysaccharide (LPS). Microbial metabolites, including short-chain fatty acids (SCFAs) and bile acids, influence hepatic function and immune responses, with potential implications for disease progression. Specific gut microbiome signatures have been identified in MAFLD patients, offering potential diagnostic and therapeutic targets. Moreover, gut-derived toxins, such as endotoxins, lipopolysaccharides, trimethylamine-N-oxide and bacterial metabolites, significantly influence liver damage and inflammation, highlighting the complex interplay between the gut microbiome and hepatic health. This review comprehensively examines the complex interplay between the gut microbiota and MAFLD, focusing on underlying pathogenic mechanisms, potential biomarkers, and emerging microbiome-targeted therapeutic strategies for disease management.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, Toulouse, France
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
25
|
Puri M, Sonawane S. Liver Sinusoidal Endothelial Cells in the Regulation of Immune Responses and Fibrosis in Metabolic Dysfunction-Associated Fatty Liver Disease. Int J Mol Sci 2025; 26:3988. [PMID: 40362227 PMCID: PMC12071881 DOI: 10.3390/ijms26093988] [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: 03/12/2025] [Revised: 04/08/2025] [Accepted: 04/18/2025] [Indexed: 05/15/2025] Open
Abstract
Liver Sinusoidal Endothelial Cells (LSECs) play a crucial role in maintaining liver homeostasis, regulating immune responses, and fibrosis in liver diseases. This review explores the unique functions of LSECs in liver pathology, particularly their roles in immune tolerance, antigen presentation, and the modulation of hepatic stellate cells (HSCs) during fibrosis. LSECs act as key regulators of immune balance in the liver by preventing excessive immune activation while also filtering antigens and interacting with immune cells, including Kupffer cells and T cells. Metabolic Dysfunction-Associated Fatty Liver Disease(MAFLD) is significant because it can lead to advanced liver dysfunction, such as cirrhosis and liver cancer. The prevalence of Metabolic Associated Steatohepatitis (MASH) is increasing globally, particularly in the United States, and is closely linked to rising rates of obesity and type 2 diabetes. Early diagnosis and intervention are vital to prevent severe outcomes, highlighting the importance of studying LSECs in liver disease. However, during chronic liver diseases, LSECs undergo dysfunction, leading to their capillarization, loss of fenestrations, and promotion of pro-fibrotic signaling pathways such as Transforming growth factor-beta (TGF-β), which subsequently activates HSCs and contributes to the progression of liver fibrosis. The review also discusses the dynamic interaction between LSECs, HSCs, and other hepatic cells during the progression of liver diseases, emphasizing how changes in LSEC phenotype contribute to liver scarring and fibrosis. Furthermore, it highlights the potential of LSECs as therapeutic targets for modulating immune responses and preventing fibrosis in liver diseases. By restoring LSECs' function and targeting pathways associated with their dysfunction, novel therapies could be developed to halt or reverse liver disease progression. The findings of this review reinforce the importance of LSECs in liver pathology and suggest that they hold significant promises as targets for future treatment strategies aimed at addressing chronic liver diseases.
Collapse
Affiliation(s)
- Munish Puri
- Onco-Immunology, Magnit Global, Folsom, CA 95630, USA
| | - Snehal Sonawane
- Department of Pathology, University of Illinois, Chicago, IL 60612, USA;
| |
Collapse
|
26
|
He Z, Tan X, Yuan M, Chen L, Meng Y, Wang Q, Hu J, Qiu Z, Yang Y. Anethole trithione mitigates LPS/D-Gal-induced acute liver injury by suppressing ROS production and NF-κB activity. Int Immunopharmacol 2025; 152:114371. [PMID: 40054324 DOI: 10.1016/j.intimp.2025.114371] [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: 09/04/2024] [Revised: 01/14/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Acute liver injury (ALI) is a prevalent form of hepatic disease associated with significant morbidity and mortality due to medical treatments, exposure to toxins or viral infections. Anethole trithione (ATT) is a heterocyclic sulfur compound recognized for its chemoprotective properties against cancer and drug-induced toxicity. This study aimed to evaluate the effectiveness of ATT in the treatment of ALI. The therapeutic effects of ATT on hepatic injury were evaluated in vivo by inducing ALI in mice through the administration of lipopolysaccharide (LPS) and D-galactosamine (D-Gal). Additionally, HepG2 and Huh7 cells exposed to LPS were utilized to investigate the underlying mechanisms in vitro. The results indicated that ATT significantly reduced the production of reactive oxygen species (ROS), mitigated oxidative stress-related biochemical markers, and inhibited hepatocyte apoptosis in vivo, resulting in marked improvement in ALI in the murine model. Mechanistic studies conducted both in vivo and in vitro demonstrated that ATT alleviates LPS/D-Gal-induced ALI by inhibiting ROS production and the activity of nuclear factor-kappa B (NF-κB). Collectively, these findings underscore the potential therapeutic benefits of ATT in the management of ALI.
Collapse
Affiliation(s)
- Zhen He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yan Meng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qi Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
| |
Collapse
|
27
|
Huang J, Zou W, Lv Z, Han H, Huang J, Su H. Immune cell phenotypes as causal factors in liver disease progression revealed by Mendelian randomization. Sci Rep 2025; 15:12685. [PMID: 40221542 PMCID: PMC11993735 DOI: 10.1038/s41598-025-97429-x] [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: 06/17/2024] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Immune cells are central mediators of the immune response and play critical roles in the pathogenesis and progression of liver diseases. Understanding the specific contributions of immune cells to liver disease progression is essential for developing targeted therapeutic strategies. In this study, we employed a two-sample Mendelian randomization (MR) approach to explore potential causal relationships between peripheral immune cell phenotypes and liver diseases, using genetic instrumental variables from large-scale genome-wide association studies (GWAS). Applying the inverse variance weighted (IVW) methods, we identified that monocyte count(odds ratio (OR) 0.81; 95% confidence interval (CI) 0.74-0.90; P = 5.95 × 10- 5, PFDR = 3.57 × 10- 4), CD3- lymphocyte/lymphocyte (OR 0.59, 95% CI 0.45-0.79; P = 3.29 × 10- 4, PFDR = 5.92 × 10- 3) and SSC-A (Side Scatter Area) on Natural Killer (NK) cells (OR 0.89, 95% CI 0.82-0.95; P = 1.37 × 10- 3, PFDR = 0.0396) acted as protective factors against alcoholic liver disease. Similarly, the trait HLA DR++ monocyte/monocyte was associated with a lower risk of autoimmune hepatitis (OR 0.56, 95% CI 0.41-0.79; P = 7.42 × 10- 4, PFDR = 0.0475). Conversely, an elevated blood monocytic Myeloid-Derived Suppressor Cells (MDSCs) count was associated with a higher risk of chronic hepatitis (OR 1.23, 95% CI 1.11-1.37; P = 1.13 × 10- 4, PFDR = 1.58 × 10- 3). Similarly, higher levels of HLA DR on CD14- CD16+ monocyte (OR 0.84, 95% CI 0.78-0.91; P = 2.07 × 10- 5, PFDR = 1.32 × 10- 3) conferred lower risk for cirrhosis of liver. In hepatic failure, CD39+ resting CD4 regulatory T cell count (OR 0.85, 95% CI 0.79-0.92; P = 1.70 × 10- 5, PFDR = 5.25 × 10- 3) played a protective role and CD28+ CD45RA- CD8dim T cell/CD8dim T cell (OR 1.14, 95% CI 1.06-1.22; P = 2.63 × 10- 4, PFDR = 0.0406) exhibited a risk function. Our findings highlight key immune pathways in liver disease progression and underscore potential immunomodulatory targets for future therapeutic interventions. Further research is warranted to clarify the mechanistic underpinnings of these associations.
Collapse
Affiliation(s)
- Jingtao Huang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wenlu Zou
- Department of Infectious Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, 250012, Shandong, China
| | - Zhihua Lv
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Huan Han
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jiapeng Huang
- Department of Thyroid Surgery, The First Hospital of China Medical University, No. 155 in Nanjing North Street, Heping Distinct, Shenyang, Liaoning, China.
| | - Hanwen Su
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
28
|
Wang J, Gong P, Liu Q, Wang M, Wu D, Li M, Zheng S, Wang H, Long Q. Stimulation of regulatory dendritic cells suppresses cytotoxic T cell function and alleviates DEN-induced liver injury, fibrosis and hepatocellular carcinoma. Front Immunol 2025; 16:1565486. [PMID: 40264769 PMCID: PMC12011597 DOI: 10.3389/fimmu.2025.1565486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Background Dendritic cells (DCs) are versatile professional antigen-presenting cells and play an instrumental role in the generation of antigen-specific T-cell responses. Modulation of DC function holds promise as an effective strategy to improve anti-tumor immunotherapy efficacy and enhance self-antigen tolerance in autoimmune diseases. Methods Wild-type (WT) and TLR2 knockout (KO) mice at 2 weeks of age were injected intraperitoneally (i.p.) with a single dose of diethylnitrosamine (DEN) to induce hepatocellular carcinoma (HCC). Four weeks later, WT and KO mice were randomly divided into control and treatment groups and treated once every two days for 30 weeks with phosphate buffered saline (PBS) and a mix of 4 TLR2-activating lactic acid-producing probiotics (LAP), respectively. Mice were euthanized after 30 weeks of LAP treatment and their liver tissues were collected for gene expression, histological, flow cytometric and single-cell RNA sequencing analyses. Results We demonstrate here that oral administration of a mix of TLR2-activating LAP triggers a marked accumulation of regulatory DCs (rDCs) in the liver of mice. LAP-treated mice are protected from DEN-induced liver injury, fibrosis and HCC in a TLR2-dependent manner. Single-cell transcriptome profiling revealed that LAP treatment determines an immunosuppressive hepatic T-cell program that is characterized by a significantly reduced cytotoxic activity. The observed functional changes of T cells correlated well with the presence of a hepatic DC subset displaying a regulatory or tolerogenic transcriptional signature. Conclusion Overall, these data suggest that stimulation of regulatory dendritic cells (rDCs) in the liver by LAP suppresses cytotoxic T-cell function and alleviates DEN-induced liver damage, fibrosis and tumorigenesis.
Collapse
Affiliation(s)
- Junjie Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Pixu Gong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qingqing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Menglei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Dengfang Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Mengyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Shujie Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
29
|
Wang Y, Hu Y, Wang P, Hu R, Chen Z, Zhang T, Liu J, Noda M, Long J, Peng Y. Distinct Hepatic Metabolic Reprogramming in Acute and Chronic Sleep Deprivation and the Protective Effects of the Chalcone Analogue TAK. Int J Mol Sci 2025; 26:3485. [PMID: 40331934 PMCID: PMC12027424 DOI: 10.3390/ijms26083485] [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: 02/12/2025] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
The prevalence of sleep deprivation is increasing worldwide. Despite the vital roles that the liver plays in metabolism and immune response, hepatic dysfunctions in acute sleep deprivation (ASD) and chronic sleep deprivation (CSD) remain underexplored. Additionally, the effects of the newly developed chalcone analog, 1-(2,3,4-trimethoxyphenyl)-2-(3,4,5-trimethoxyphenyl)-acrylketone (TAK), were evaluated as a potential therapeutic chemical for mitigating SD-induced hepatic damage. A modified multi-platform method was employed to prepare animal models of 72 h ASD and 21-day CSD in rats. TAK (50 mg/kg/day) was administered through irrigation starting one week before the experiment and continuing until the end. ASD triggered hepatic lipid accumulation and inflammation, whereas CSD resulted in pathological portal area expansion and fibrosis, with comparatively fewer disturbances in liver metabolism and inflammation. TAK effectively alleviated ASD-induced disruptions in glycogen synthesis via PI3K/AKT/GSK3/GYS2 pathways, abnormal lipid accumulation via SREBP1/FASN/ACC, liver inflammation by balancing M1 and M2 macrophages, and liver fibrosis induced by ASD/CSD. This study provides valuable insights into the different mechanisms of liver damage induced by severe ASD and mild CSD. Additionally, TAK has been proposed as a potential therapeutic strategy for ultimate SD-related hepatic complications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jiangang Long
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.W.); (Y.H.); (P.W.); (R.H.); (Z.C.); (T.Z.); (J.L.); (M.N.)
| | - Yunhua Peng
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China; (Y.W.); (Y.H.); (P.W.); (R.H.); (Z.C.); (T.Z.); (J.L.); (M.N.)
| |
Collapse
|
30
|
Shi Y, Li X, Liang K, Lu T, Chen Y, Lai Y, Li Y, Wei S, He S, Tang L, Liu D, Li Y. Characteristics and immunoprotective functions of three cysteine proteases from Clonorchis sinensis. Front Immunol 2025; 16:1550775. [PMID: 40248698 PMCID: PMC12003271 DOI: 10.3389/fimmu.2025.1550775] [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: 12/24/2024] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Introduction Cysteine proteases from Clonorchis sinensis, including various proteins, are essential for its pathogenicity and serve as potential vaccine candidates. This study assesses the protective effects of three C. sinensis cysteine proteases (CsCP1-3). Methods Mice immunized with recombinant CsCP1-3 and adjuvants were subsequently infected with C. sinensis metacercariae after three immunization rounds. Liver damage was evaluated through hematoxylin and eosin (H&E), Masson's trichrome, and immunohistochemical analyses. The levels of IgG1, IgG2a antibodies, and cytokines (IFN-g, IL-2, IL-4, and IL-10) were quantified by enzyme-linked immunosorbent assay (ELISA). Results RT-qPCR revealed that CsCP1-2 exhibited the highest expression in newly encysted larvae (NEL), while CsCP3 was predominantly expressed in adult stages. Immunohistochemical localization confirmed that CsCP1-3 are present in the eggshells, syncytial layers of metacercariae, NEL cuticle, and adult intestines. Histological and immunohistochemical analysis demonstrated that the rCsCP1-3-immunized group displayed reduced liver inflammation and biliary fibrosis compared to the control group. The rCsCP1-3 induced a progressive increase in specific IgG1 and IgG2a antibody titers by the second week post-immunization. In the CsCP1-2 group, cytokines IFN-g, IL-2, IL-4, and IL-10 were elevated relative to the control, with particularly high levels of IFN-g and IL-10 in CsCP1, indicating a strong mixed Th1/Th2 immune response. In contrast, the CsCP3 immunization group exhibited a transient increase in cytokines (IFN-g, IL-2, IL-4, and IL-10) three days postinfection, which subsided after one to two weeks. Discussion These findings suggest that CsCP1-3 elicit robust antibody and cellular immune responses, mitigating liver damage caused by C. sinensis infection. CsCP1, in particular, induces a potent mixed Th1/Th2 response, positioning it as a promising vaccine candidate.
Collapse
Affiliation(s)
- Yunliang Shi
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoqin Li
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Department of Medical Laboratory, Shenzhen Longgang District Eighth People’s Hospital, Shenzhen, China
| | - Kai Liang
- Gastroenterology Department, Guangxi Zhuang Autonomous Region People’s Hospital, Nanning, China
| | - Ting Lu
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yu Chen
- Department of Schistosomiasis Prevention and Control, Disease Prevention and Control Center of Hengzhou City, Hengzhou, China
| | - Yashi Lai
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Yaoting Li
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Shuai Wei
- Department of Medical Laboratory, Hechi People’s Hospital, Hechi, China
| | - Shanshan He
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Lili Tang
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
| | - Dengyu Liu
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yanwen Li
- Parasitology Department, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China
- Key Laboratory of Basic Research on Regional Diseases (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
31
|
Devisme C, Stosskopf M, Piquet‐Pellorce C, Palmer G, Gabay C, Seyec JL, Samson M, Raguenes‐Nicol C. Interleukin-18 Binding Protein (IL-18BP) Deficiency Affects Lymphocyte Activation and IL-18 Expression in a Mouse Model of Liver Inflammation. Eur J Immunol 2025; 55:e202451579. [PMID: 40170380 PMCID: PMC11962228 DOI: 10.1002/eji.202451579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/03/2025]
Affiliation(s)
- Christelle Devisme
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| | - Marie Stosskopf
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| | - Claire Piquet‐Pellorce
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| | - Gaby Palmer
- Département de MédecineUniversité de GenèveGeneveSwitzerland
| | - Cem Gabay
- Département de MédecineUniversité de GenèveGeneveSwitzerland
| | - Jacques Le Seyec
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| | - Michel Samson
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| | - Céline Raguenes‐Nicol
- Univ Rennes, Inserm, EHESPIrset (Institut de recherche en santé, environnement et travail) ‐ UMR_S 1085, Rennes, France.RennesFrance
| |
Collapse
|
32
|
Ronca V, Gerussi A, Collins P, Parente A, Oo YH, Invernizzi P. The liver as a central "hub" of the immune system: pathophysiological implications. Physiol Rev 2025; 105:493-539. [PMID: 39297676 DOI: 10.1152/physrev.00004.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 01/16/2025] Open
Abstract
The purpose of this review is to describe the immune function of the liver, guiding the reader from the homeostatic tolerogenic status to the aberrant activation demonstrated in chronic liver disease. An extensive description of the pathways behind the inflammatory modulation of the healthy liver will be provided focusing on the complex immune cell network residing within the liver. The limit of tolerance will be presented in the context of organ transplantation, seizing the limits of homeostatic mechanisms that fail in accepting the graft, progressing eventually toward rejection. The triggers and mechanisms behind chronic activation in metabolic liver conditions and viral hepatitis will be discussed. The last part of the review will be dedicated to one of the greatest paradoxes for a tolerogenic organ, developing autoimmunity. Through the description of the three most common autoimmune liver diseases, the autoimmune reaction against hepatocytes and biliary epithelial cells will be dissected.
Collapse
Affiliation(s)
- Vincenzo Ronca
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paul Collins
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Alessandro Parente
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Ye Htun Oo
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
33
|
Yang J, Wan SY, Song QY, Xie YH, Wan J, Zhou YH, Zhang ZT, Xiao YS, Li X, Chen H, Liu XR, Xu L, You HJ, Hu DS, Petersen RB, Zhang YH, Zheng L, Zhang Y, Huang K. Angiopoietin-like protein 8 directs DNA damage responses towards apoptosis by stabilizing PARP1-DNA condensates. Cell Death Differ 2025; 32:672-688. [PMID: 39592710 PMCID: PMC11982567 DOI: 10.1038/s41418-024-01422-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/14/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024] Open
Abstract
Upon genotoxic stresses, cells employ various DNA damage responses (DDRs), including DNA damage repair or apoptosis, to safeguard genome integrity. However, the determinants among different DDRs choices are largely unknown. Here, we report angiopoietin-like protein 8 (ANGPTL8), a secreted regulator of lipid metabolism, localizes to the nucleus and acts as a dynamic switch that directs DDRs towards apoptosis rather than DNA repair after genotoxin exposure. ANGPTL8 deficiency alleviates DNA damage and apoptosis in cells exposed to genotoxins, as well as in the liver or kidney of mice injured by hepatic ischemia/reperfusion or cisplatin treatment. Mechanistically, ANGPTL8 physically interacts with Poly (ADP-ribose) polymerase 1 (PARP1), in a PARylation-independent manner, and reduces the fluidity of PARP1-DNA condensates, thereby enhancing the pro-apoptotic accumulation of PARP1 and PAR chains on DNA lesions. However, the transcription of ANGPTL8 is gradually decreased following genotoxin treatment, partly due to downregulation of CCAAT enhancer binding protein alpha (CEBPA), presumably to avoid further cytotoxicity. Together, we provide new insights by which genotoxic stress induced DDRs are channeled to suicidal apoptosis to safeguard genome integrity.
Collapse
Affiliation(s)
- Jing Yang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shi-Yuan Wan
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qiu-Yi Song
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yun-Hao Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Jun Wan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yi-Hao Zhou
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zi-Tong Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu-Shuo Xiao
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xi Li
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hong Chen
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin-Ran Liu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Xu
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Juan You
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - De-Sheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
- China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mt. Pleasant, MI, 48858, USA
| | - Yong-Hui Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Yu Zhang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Kun Huang
- School of Pharmacy, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Tongji-Rong Cheng Biomedical Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
34
|
Wang C, Zai W, Zhao K, Li Y, Shi B, Wu M, Zhou X, Kozlowski M, Zhang X, Fang Z, Yuan Z. Potential role of liver-resident CD3 + macrophages in HBV clearance in a mouse hepatitis B model. JHEP Rep 2025; 7:101323. [PMID: 40143948 PMCID: PMC11937660 DOI: 10.1016/j.jhepr.2024.101323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 03/28/2025] Open
Abstract
Background & Aims Chronic HBV infection usually causes cirrhosis and hepatocellular carcinoma. Comparative investigations of acute and chronic HBV cases would help determine the immune responses crucial for viral clearance. Methods A fast-cleared HBV mouse model was established in Alb-Cre mice via hydrodynamic injection of HBV plasmid, while persistent HBV model mice were generated via recombinant covalently closed circular DNA-adeno-associated virus 8 infection. The single-cell transcriptomes of CD45+ intrahepatic non-parenchymal cells from these mice were conducted. Multiplexed immunohistochemistry and flow cytometry were used to confirm the findings from single-cell transcriptomes. Transwell, coculture, and adoptive transfer experiments were performed to study the generation and functions of macrophages. Results Twenty-four clusters of immune cells were identified. Myeloid cells, including granulocytes, monocytes, and dendritic cells, are activated early in HBV fast-cleared mice. Significantly, a cluster of CD3+ macrophages was found in the viral clearance phase, which was confirmed in liver tissue from five acute patients with HBV. These cells highly expressed CXCL1, tumor necrosis factor alpha, and HBsAg-specific T cell receptors. The transwell assay revealed that CD3+ macrophages originate from macrophages (n = 6). T cells and anti-HBsAg antibodies are indispensable for their differentiation, which was further confirmed in T- and/or B-cell-deficient mice. Interestingly, these CD3+ macrophages capable of killing peptide-loaded hepatocytes and engulfing IgG-coated beads were persistently detectable in the mouse liver for 10 weeks after HBV clearance. The expression levels of CD5L and Bcl2, two classical antiapoptotic proteins, increased (p <0.001), suggesting that the CD3+ macrophages are long-term resident populations. Finally, adoptive transfer of CD3+ macrophages accelerated HBV clearance in mice (n = 5, p <0.01). Conclusions We identified long-term polyfunctional CD3+ macrophages residing in HBV fast-cleared livers that could help elucidate the immune responses involved in eliminating HBV. Impact and implications The liver is a special organ with unique immune characteristics and tolerance to foodborne antigens. Chronic infections can develop in newborns after exposure to HBV; however, acute infections usually occur in adults, indicating that immune cells in the liver tissue microenvironment can also effectively fight against the virus. Nevertheless, the mechanisms involved in acute HBV infection have rarely been studied. In this study, we identified a macrophage population with both T cell and macrophage characteristics in the livers of acute HBV model mice and revealed that these macrophages play important roles in HBV clearance. Moreover, we confirmed that this population is derived from macrophages in the presence of virus-specific T cells and antibodies. This finding highlights the complexity of antiviral immune responses in liver microenvironments.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Wenjing Zai
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Kuangjie Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yaming Li
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bisheng Shi
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Min Wu
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaohui Zhou
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Maya Kozlowski
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Xiaonan Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College of Fudan University, Shanghai, China
| | - Zhong Fang
- Liver Cancer Institute of Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, China
| | - Zhenghong Yuan
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Research Unit of Cure of Chronic Hepatitis B Virus Infection (CAMS), Shanghai Frontiers Science Center of Pathogenic Microbes and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Shi JL, Sun T, Li Q, Li CM, Jin JF, Zhang C. Mogroside V protects against acetaminophen-induced liver injury by reducing reactive oxygen species and c-jun-N-terminal kinase activation in mice. World J Hepatol 2025; 17:104520. [PMID: 40177205 PMCID: PMC11959673 DOI: 10.4254/wjh.v17.i3.104520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND High levels of acetaminophen (APAP) consumption can result in significant liver toxicity. Mogroside V (MV) is a bioactive, plant-derived triterpenoid known for its various pharmacological activities. However, the impact of MV on acute liver injury (ALI) is unknown. AIM To investigate the hepatoprotective potential of MV against liver damage caused by APAP and to examine the underlying mechanisms. METHODS Mice were divided into three groups: Saline, APAP and APAP + MV. MV (10 mg/kg) was given intraperitoneally one hour before APAP (300 mg/kg) administration. Twenty-four hours after APAP exposure, serum transaminase levels, liver necrotic area, inflammatory responses, nitrotyrosine accumulation, and c-jun-N-terminal kinase (JNK) activation were assessed. Additionally, we analyzed reactive oxygen species (ROS) levels, JNK activation, and cell death in alpha mouse liver 12 (AML12) cells. RESULTS MV pre-treatment in vivo led to a reduction in the rise of aspartate transaminase and alanine transaminase levels, mitigated liver damage, decreased nitrotyrosine accumulation, and blocked JNK phosphorylation resulting from APAP exposure, without affecting glutathione production. Similarly, MV diminished the APAP-induced increase in ROS, JNK phosphorylation, and cell death in vitro. CONCLUSION Our study suggests that MV treatment alleviates APAP-induced ALI by reducing ROS and JNK activation.
Collapse
Affiliation(s)
- Jia-Lin Shi
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Tian Sun
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Qing Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Chun-Mei Li
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Jun-Fei Jin
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
| | - Chong Zhang
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- China-USA Lipids in Health and Disease Research Center, Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China
- Laboratory of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
36
|
Qi WY, Zheng SH, Li SZ, Wang W, Wang QY, Liu QY, Li XK, Zhang JX, Gan DN, Ye YA, Zao XB. Immune cells in metabolic associated fatty liver disease: Global trends and hotspots (2004-2024). World J Hepatol 2025; 17:103327. [PMID: 40177204 PMCID: PMC11959657 DOI: 10.4254/wjh.v17.i3.103327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/07/2025] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND The interplay between immune cells and metabolic associated fatty liver disease (MAFLD) is a critical research frontier, bridging immunology and hepatology. The bibliometric findings can guide future research and funding priorities in the field by highlighting key areas of focus and potential therapeutic targets. AIM To analyze the literature on immune cells and MAFLD, identifying research trends and future hotspots. METHODS A systematic search in the Web of Science Core Collection from January 1, 2004 to May 20, 2024, yielded 1936 articles on immune cells and MAFLD. Excluding non-research documents, the data were analyzed using R packages Cluster profiler, enrichplot, ggplot2, VOSviewer and CiteSpace. Visualizations were created for countries, institutions, authors, journals, fields, co-cited references, keywords, genes, and diseases, with gene a disease data from Citexs. RESULTS The field gained momentum in 2006, with the United States of America and China as leading contributors. Key research themes included oxidative stress, metabolic syndrome, liver fibrosis, and the role of Kupffer cells. Bioinformatics identified interleukin-6, tumor necrosis factor and signal transducer and activator of transcription 3 as central proteins in immune responses and inflammation, suggesting potential therapeutic targets for MAFLD. Clinically, these hub genes play pivotal roles in the pathogenesis of MAFLD. For instance, targeting the tumor necrosis factor signaling pathway could reduce inflammation, while modulating interleukin-6 and signal transducer and activator of transcription 3 expression may improve metabolic function, offering new strategies for MAFLD therapy. CONCLUSION This bibliometric analysis reports on the research hotspots and emerging trends in the field of immune cells and MAFLD, highlighting key proteins and potential therapeutic strategies through bioinformatics.
Collapse
Affiliation(s)
- Wen-Ying Qi
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Shi-Hao Zheng
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Si-Ze Li
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Wei Wang
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qiu-Yue Wang
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qi-Yao Liu
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiao-Ke Li
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Jia-Xin Zhang
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Da-Nan Gan
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Yong-An Ye
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
| | - Xiao-Bin Zao
- Department of Spleen and Stomach Diseases, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Institute of Hepatology, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital of Beijing University of Chinese Medicine, Beijing 100700, China.
| |
Collapse
|
37
|
Li YH, Zheng CR, Liu Y, Wang K, Zhou FF, Dong X, Yuan T, He QJ, Zhu H, Yang B. The role of calcium signaling in organotropic metastasis of cancer. Acta Pharmacol Sin 2025:10.1038/s41401-025-01537-3. [PMID: 40133629 DOI: 10.1038/s41401-025-01537-3] [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: 12/05/2024] [Accepted: 03/09/2025] [Indexed: 03/27/2025]
Abstract
Tumor metastasis is an important event in cancer progression, representing an enduring and irrevocable hallmark of cancers. The causes of tumor metastasis are complex and diverse. Arising evidence shows that the dysregulation of calcium signaling plays a crucial role in its initiation and progress. Calcium is an essential secondary messenger that regulates signaling pathways associated with tumor metastasis. The transient accumulation of calcium potentially promotes the advancement of tumor metastasis, while calcium-dependent proteins and calcium-related channels also significantly contribute to such malignant process. Thus, compounds specially targeting calcium channels, transporters or pumps may be therapeutic approaches prohibiting tumor metastasis. This review focuses on exploring the roles of calcium ions, calcium-dependent proteins and calcium-related channels in organotropic metastasis of cancer and its clinical applications in the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Yong-Hao Li
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Chu-Run Zheng
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ke Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, 210000, China
| | - Fan-Fan Zhou
- Sydney Pharmacy School, The University of Sydney, Camperdown, NSW, 2050, Australia
| | - Xin Dong
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310015, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310015, China
| | - Qiao-Jun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310015, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310015, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China.
| |
Collapse
|
38
|
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
|
39
|
He F, Du W, Liu Y, Ling Y, Xu M, Liu J, Song P, Fang Z, Yue Z, Duan J, Wang L. Exosome-equipped TNF antisense oligodeoxynucleotide or 2-deoxy-D-glucose ameliorated nonalcoholic steatohepatitis by modulating superoxide dismutase 1 in mice. Redox Biol 2025; 80:103488. [PMID: 39778469 PMCID: PMC11763583 DOI: 10.1016/j.redox.2025.103488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/09/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Inflammatory mediators tumor necrosis factor (TNF) and interleukin 1 beta (IL1β), primarily derived from hepatic macrophages in the liver, play a crucial role in the progression of nonalcoholic steatohepatitis (NASH). Meanwhile, intravenously injected exosomes are mainly distributed in the liver and predominantly taken up by hepatic macrophage. Herein, we aimed to evaluate the feasibility of targeted inhibition of TNF and IL1β expression in hepatic macrophages via exosomes as a potential therapeutic strategy for NASH. In this study, we demonstrated that antisense oligodeoxynucleotide targeting TNF (ASO-TNF) or 2-deoxy-d-glucose (2DG) effectively suppressed the expression of TNF and/or IL1β in macrophages. Exosomes loaded with ASO-TNF or 2DG were able to suppress the expression of TNF and/or IL1β in macrophages in vitro or in vivo. Furthermore, infusion of Exo/ASO-TNF or Exo/2DG significantly attenuated experimental steatohepatitis in choline deficient amino acid-defined (CDAA) or methionine and choline deficient (MCD) diet-fed mice. RNA-seq results showed that treatment with Exo/ASO-TNF or Exo/2DG significantly inhibited pro-inflammatory signaling pathways. Mechanistically, we observed that administration of Exo/ASO-TNF or Exo/2DG could attenuate NASH progression by up-regulating the expression of superoxide dismutase 1 (Sod1). Combined, our findings demonstrated that infusion of exosomes loaded with ASO-TNF or 2DG alleviated experimental steatohepatitis in murine models. Thus, infusion of exosomes loaded with anti-inflammatory agents holds promise as a potential therapeutic strategy for NASH treatment.
Collapse
Affiliation(s)
- Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yingying Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Yuwei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Jingjing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhiqiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Zhensheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Juanli Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, 710032, Xi'an, China.
| |
Collapse
|
40
|
Zhang X, Lau HCH, Yu J. Pharmacological treatment for metabolic dysfunction-associated steatotic liver disease and related disorders: Current and emerging therapeutic options. Pharmacol Rev 2025; 77:100018. [PMID: 40148030 DOI: 10.1016/j.pharmr.2024.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as nonalcoholic fatty liver disease) is a chronic liver disease affecting over a billion individuals worldwide. MASLD can gradually develop into more severe liver pathologies, including metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and liver malignancy. Notably, although being a global health problem, there are very limited therapeutic options against MASLD and its related diseases. While a thyroid hormone receptor agonist (resmetirom) is recently approved for MASH treatment, other efforts to control these diseases remain unsatisfactory. Given the projected rise in MASLD and MASH incidence, it is urgent to develop novel and effective therapeutic strategies against these prevalent liver diseases. In this article, the pathogenic mechanisms of MASLD and MASH, including insulin resistance, dysregulated nuclear receptor signaling, and genetic risk factors (eg, patatin-like phospholipase domain-containing 3 and hydroxysteroid 17-β dehydrogenase-13), are introduced. Various therapeutic interventions against MASH are then explored, including approved medication (resmetirom), drugs that are currently in clinical trials (eg, glucagon-like peptide 1 receptor agonist, fibroblast growth factor 21 analog, and PPAR agonist), and those failed in previous trials (eg, obeticholic acid and stearoyl-CoA desaturase 1 antagonist). Moreover, given that the role of gut microbes in MASLD is increasingly acknowledged, alterations in the gut microbiota and microbial mechanisms in MASLD development are elucidated. Therapeutic approaches that target the gut microbiota (eg, dietary intervention and probiotics) against MASLD and related diseases are further explored. With better understanding of the multifaceted pathogenic mechanisms, the development of innovative therapeutics that target the root causes of MASLD and MASH is greatly facilitated. The possibility of alleviating MASH and achieving better patient outcomes is within reach. SIGNIFICANCE STATEMENT: Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide, and it can progress to more severe pathologies, including steatohepatitis, cirrhosis, and liver cancer. Better understanding of the pathogenic mechanisms of these diseases has facilitated the development of innovative therapeutic strategies. Moreover, increasing evidence has illustrated the crucial role of gut microbiota in the pathogenesis of MASLD and related diseases. It may be clinically feasible to target gut microbes to alleviate MASLD in the future.
Collapse
Affiliation(s)
- Xiang Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
41
|
Duan H, Chang Q, Ding H, Shao W, Wang Y, Lu K, Zhang L, Xu J. GBP1 promotes acute rejection after liver transplantation by inducing Kupffer cells pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167644. [PMID: 39732345 DOI: 10.1016/j.bbadis.2024.167644] [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: 11/02/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Liver transplantation is currently recognized as the most effective treatment for severe liver diseases. Although survival rates after liver transplantation have improved, rejection of the transplanted liver remains a significant cause of morbidity and transplant failure in patients. Our team previously discovered a close association between high GBP1 expression and acute rejection reactions following liver transplantation. Liver biopsies were conducted on patients who experienced acute rejection or successfully achieved immune tolerance post-transplantation. We confirmed that GBP1 was highly expressed in the acute rejection group after transplantation by Immunohistochemistry. This study aims to confirm that GBP1 promotes acute rejection reactions following liver transplantation through inducing pyroptosis in rat transplanted hepatic macrophages (KCs). We knocked down GBP1 in KCs and examined the extent of pyroptosis and the severity of acute rejection in the transplanted liver post-orthotopic liver transplantation in rats and KCs. These data provide new approaches for the study of liver transplant rejection reactions and identify new targets.
Collapse
Affiliation(s)
- Haojiang Duan
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qingyao Chang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Huaxing Ding
- College of Basic Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Wenhao Shao
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Yan Wang
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Kairui Lu
- Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li Zhang
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.
| | - Jun Xu
- Department of Hepatobiliary Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China.
| |
Collapse
|
42
|
Wei Y, Jiang Y, Zhu J, Zhang Z, Li M, Zheng S, Wang X, Sun J, Li C, Shi W, Wang S, Liu X, Lin M, Zhang Z, Zhang D, Sun G. CD36-mediated uptake of oxidized LDL induces double-negative regulatory T cell ferroptosis in metabolic dysfunction-associated steatotic liver disease. Metabolism 2025; 164:156127. [PMID: 39743040 DOI: 10.1016/j.metabol.2024.156127] [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: 09/09/2024] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Metabolic alterations have been shown to instigate liver inflammation in metabolic dysfunction-associated steatotic liver disease (MASLD), but the underlying mechanism is not fully elucidated. During MASLD progression, intrahepatic CD3+TCRαβ+CD4-CD8- double negative T regulatory cells (DNT) decrease cell survival and immunosuppressive function, leading to aggravated liver inflammation. In this study, we aim to reveal the underlying mechanisms that cause changes in DNT during MASLD progression. METHODS The correlation of serum oxidized low-density lipoprotein (oxLDL) levels and DNT from patients with MASLD and MASLD mouse models were evaluated. The mechanisms of oxLDL affecting DNT survival and function were explored through transcriptome sequencing analysis, flow cytometry, and CUT & TAG experiments. RESULTS Serum oxLDL levels are negative correlated with survival and functional molecule expression of circulating DNT in patients with MASLD and intrahepatic DNT in MASLD mouse models. Mechanistically, oxLDL increases DNT CD36 expression through the NF-κB pathway, leading to enhanced uptake of oxLDL and subsequent occurrence of ferroptosis and functional impairment. oxLDL enhances ferroptosis in DNT by upregulating acyl-CoA synthetase long chain family member 4 expression. By transferring CD36-/- DNT into MASLD mice, we observe a significant reduction in ferroptosis and improved immune regulation in CD36-/- DNT compared to wild type DNT. This improvement in DNT results in a notable enhancement of therapeutic efficacy against MASLD. CONCLUSION oxLDL induces a decline in the survival and immune regulatory function of DNT, subsequently weakening their role in maintaining liver immune homeostasis in MASLD. Specific targeting of CD36 to prevent ferroptosis in DNT may provide a novel therapeutic approach for the treatment of MASLD.
Collapse
Affiliation(s)
- Yunxiong Wei
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yuan Jiang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jingjing Zhu
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Zihan Zhang
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Mengyi Li
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shimeng Zheng
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Xiyu Wang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Jie Sun
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Changying Li
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Wen Shi
- Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing 100069, China
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Minjie Lin
- Academic Affairs Department, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhongtao Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| | - Dong Zhang
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; Beijing Laboratory of Oral Health, Capital Medical University School of Basic Medicine, Beijing 100069, China.
| | - Guangyong Sun
- Medical Research Center, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Department of Gastroenterology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China; Immunology Research Center for Oral and Systemic Health, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
43
|
Zhang YZ, Ma Y, Ma E, Chen X, Zhang Y, Yin B, Zhao J. Sophisticated roles of tumor microenvironment in resistance to immune checkpoint blockade therapy in hepatocellular carcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:10. [PMID: 40051497 PMCID: PMC11883234 DOI: 10.20517/cdr.2024.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/13/2025] [Accepted: 02/21/2025] [Indexed: 03/09/2025]
Abstract
Hepatocellular carcinoma (HCC) remains a serious threat to global health, with rising incidence and mortality rates. Therapeutic options for advanced HCC are quite limited, and the overall prognosis remains poor. Recent advancements in immunotherapy, particularly immune-checkpoint blockade (ICB) targeting anti-PD1/PD-L1 and anti-CTLA4, have facilitated a paradigm shift in cancer treatment, demonstrating substantial survival benefits across various cancer types, including HCC. However, only a subset of HCC patients exhibit a favorable response to ICB therapy, and its efficacy is often hindered by the development of resistance. There are many studies to explore the underlying mechanisms of ICB response. In this review, we compiled the latest progression in immunotherapies for HCC and systematically summarized the sophisticated mechanisms by which components of the tumor microenvironment (TME) regulate resistance to ICB therapy. Additionally, we also outlined some scientific rationale strategies to boost antitumor immunity and enhance the efficacy of ICB in HCC. These insights may serve as a roadmap for future research and help improve outcomes for HCC patients.
Collapse
Affiliation(s)
- Yi-Zhe Zhang
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Yunshu Ma
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Authors contributed equally
| | - Ensi Ma
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Institute of Organ Transplantation, Fudan University, Shanghai 200040, China
| | - Xizhi Chen
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yue Zhang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Baobing Yin
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
| | - Jing Zhao
- Hepatobiliary Surgery Center, Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Hepatobiliary surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
- Cancer Metastasis Institute, Fudan University, Shanghai 201206, China
| |
Collapse
|
44
|
Bæk O, Muk T, Wu Z, Ye Y, Khakimov B, Casano AM, Gangadharan B, Bilic I, Brunse A, Sangild PT, Nguyen DN. Altered hepatic metabolism mediates sepsis preventive effects of reduced glucose supply in infected preterm newborns. eLife 2025; 13:RP97830. [PMID: 39992703 PMCID: PMC11850001 DOI: 10.7554/elife.97830] [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] [Indexed: 02/26/2025] Open
Abstract
Preterm infants are susceptible to neonatal sepsis, a syndrome of pro-inflammatory activity, organ damage, and altered metabolism following infection. Given the unique metabolic challenges and poor glucose regulatory capacity of preterm infants, their glucose intake during infection may have a high impact on the degree of metabolism dysregulation and organ damage. Using a preterm pig model of neonatal sepsis, we previously showed that a drastic restriction in glucose supply during infection protects against sepsis via suppression of glycolysis-induced inflammation, but results in severe hypoglycemia. Now we explored clinically relevant options for reducing glucose intake to decrease sepsis risk, without causing hypoglycemia and further explore the involvement of the liver in these protective effects. We found that a reduced glucose regime during infection increased survival via reduced pro-inflammatory response, while maintaining normoglycemia. Mechanistically, this intervention enhanced hepatic oxidative phosphorylation and possibly gluconeogenesis, and dampened both circulating and hepatic inflammation. However, switching from a high to a reduced glucose supply after the debut of clinical symptoms did not prevent sepsis, suggesting metabolic conditions at the start of infection are key in driving the outcome. Finally, an early therapy with purified human inter-alpha inhibitor protein, a liver-derived anti-inflammatory protein, partially reversed the effects of low parenteral glucose provision, likely by inhibiting neutrophil functions that mediate pathogen clearance. Our findings suggest a clinically relevant regime of reduced glucose supply for infected preterm infants could prevent or delay the development of sepsis in vulnerable neonates.
Collapse
Affiliation(s)
- Ole Bæk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Neonatology, RigshospitaletCopenhagenDenmark
| | - Tik Muk
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Ziyuan Wu
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Yongxin Ye
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Food Science, University of CopenhagenCopenhagenDenmark
| | - Bekzod Khakimov
- Department of Food Science, University of CopenhagenCopenhagenDenmark
| | - Alessandra Maria Casano
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Bagirath Gangadharan
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Ivan Bilic
- Plasma-derived therapies, Baxalta Innovations GmbH, part of Takeda Pharmaceuticals LtdViennaAustria
| | - Anders Brunse
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| | - Per Torp Sangild
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
- Department of Neonatology, RigshospitaletCopenhagenDenmark
| | - Duc Ninh Nguyen
- Section for Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of CopenhagenCopenhagenDenmark
| |
Collapse
|
45
|
Guo Y, Guo W, Chen H, Sun J, Yin Y. Mechanisms of sepsis-induced acute liver injury: a comprehensive review. Front Cell Infect Microbiol 2025; 15:1504223. [PMID: 40061452 PMCID: PMC11885285 DOI: 10.3389/fcimb.2025.1504223] [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: 09/30/2024] [Accepted: 01/31/2025] [Indexed: 05/13/2025] Open
Abstract
Sepsis is a severe, often life-threatening form of organ dysfunction that arises from an inappropriately regulated host response to infectious pathogen exposure. As the largest gland in the body, the liver serves as a regulatory hub for metabolic, immune, and detoxification activity. It is also an early sepsis target organ such that hepatic dysfunction is observed in 34-46% of patients with sepsis. The precise mechanisms that give rise to sepsis-induced liver injury, however, remain incompletely understood. Based on the research conducted to date, dysregulated systemic inflammation, microbial translocation, microcirculatory abnormalities, cell death, metabolic dysfunction, and liver inflammation may all contribute to the liver damage that can arise in the context of septicemia. This review was developed to provide an overview summarizing the potential mechanisms underlying sepsis-induced liver injury, informing the selection of potential targets for therapeutic intervention and providing a framework for the alleviation of patient symptoms and the improvement of prognostic outcomes.
Collapse
Affiliation(s)
- Yongjing Guo
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Wanxu Guo
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Huimin Chen
- Department of Neonate, The Second Hospital of Jilin University, Changchun, China
| | - Jian Sun
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| | - Yongjie Yin
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
46
|
Vizioli G, Nicoletti A, Feliciani D, Funaro B, Zileri Dal Verme L, Ponziani FR, Zocco MA, Gasbarrini A, Gabrielli M. Immunotherapy and MASLD-Related HCC: Should We Reconsider the Role of Etiology in the Therapeutic Approach to HCC? APPLIED SCIENCES 2025; 15:2279. [DOI: 10.3390/app15052279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/21/2025]
Abstract
Hepatocellular carcinoma (HCC) accounts for 90% of primary liver cancers and typically arises in the context of chronic liver disease. With the increasing prevalence of metabolic disorders, metabolic dysfunction-associated steatotic liver disease (MASLD) has become the leading cause of chronic liver disease and the most rapidly increasing cause of HCC. The role of dysfunctional innate and adaptive immune responses in the development and progression of HCC is well-established, prompting numerous trials to evaluate the efficacy of immune checkpoint inhibitors (ICIs) in targeting tumor cells. These trials have yielded promising results, and ICIs, in combination with anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, are now approved as first-line therapy for patients with metastatic or unresectable HCC, irrespective of the underlying liver disease. Notably, MASLD itself is characterized by immune system dysfunction, as metabolic inflammation plays a central role in its onset and progression. However, clinical studies and post-hoc analyses suggest that immunotherapy may be less effective in MASLD-associated HCC compared to viral-related HCC. This emerging evidence raises the question of whether the underlying liver disease influences the therapeutic response to ICIs in HCC. It may be time to consider tailoring therapeutic strategies for HCC based on the specific etiological, histological, and genotypical subgroups.
Collapse
Affiliation(s)
- Giuseppina Vizioli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Alberto Nicoletti
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Daniela Feliciani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Barbara Funaro
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lorenzo Zileri Dal Verme
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maria Assunta Zocco
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Maurizio Gabrielli
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
47
|
Millian DE, Arroyave E, Wanninger TG, Krishnan S, Bao D, Zhang JR, Rao A, Spratt H, Ferguson M, Chen V, Stevenson HL, Saldarriaga OA. Alterations in the hepatic microenvironment following direct-acting antiviral therapy for chronic hepatitis C. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.17.25321289. [PMID: 40034770 PMCID: PMC11875275 DOI: 10.1101/2025.02.17.25321289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background and aims. The first direct-acting antivirals (DAAs) to treat the viral hepatitis C (HCV) became available in 2011. Despite numerous clinical studies of patient outcomes after treatment, few have evaluated changes in the liver microenvironment. Despite achieving sustained virologic response (SVR), patients may still experience adverse outcomes like cirrhosis and hepatocellular carcinoma. By comparing gene and protein expression in liver biopsies collected before and after treatment, we sought to determine whether specific signatures correlated with disease progression and adverse clinical outcomes. Methods. Biopsies were collected from 22 patients before and after DAA treatment. We measured ∼770 genes and used multispectral imaging with custom machine learning algorithms to analyze phenotypes of intrahepatic macrophages (CD68, CD14, CD16, MAC387, CD163) and T cells (CD3, CD4, CD8, CD45, FoxP3). Results. Before DAA treatment, patients showed two distinct gene expression patterns: one with high pro-inflammatory and antiviral gene expression and another with weaker expression. Patients with adverse outcomes exhibited significantly (p<0.05) more inflammatory activity and had more advanced fibrosis stages in their baseline biopsies than those with liver disease resolution. Patients who achieved SVR had significantly decreased liver enzymes, reduced inflammatory scores, and restored type 1 interferon pathways similar to controls. However, after DAA treatment, patients with persistently high gene expression (67%, pre-hot) still had significantly worse outcomes (p<0.049) despite achieving SVR. A persistent lymphocytic infiltrate was observed in a subset of these patients (76.5%). After therapy, anti-inflammatory macrophages (CD16+, CD16+CD163+, CD16+CD68+) increased, and T cell heterogeneity was more pronounced, showing a predominance of helper and memory T cells (CD3+CD45RO+, CD4+CD45RO+, CD3+CD4+CD45RO+). Conclusions. Patients who have more inflamed livers and more advanced fibrosis before DAA treatment should be closely followed for the development of adverse outcomes, even after achieving SVR. We can enhance patient risk stratification by integrating gene and protein expression profiles with clinical data. This could identify those who may benefit from more intensive monitoring or alternative therapeutic approaches, inspiring a new era of personalized patient care. Lay Summary Direct-acting antiviral (DAA) therapy has dramatically improved the treatment of chronic HCV, making it curable for most people. This study determined gene and protein expression differences in the liver before and after treatment of HCV. These results will lead to a deeper understanding of the changes in the hepatic immune microenvironment with and without the virus present in the liver in hopes of improving patient surveillance, prognosis, and outcome in the future.
Collapse
|
48
|
Li W, Chen L, Zhou Q, Huang T, Zheng W, Luo F, Luo ZG, Zhang J, Liu J. Liver macrophage-derived exosomal miRNA-342-3p promotes liver fibrosis by inhibiting HPCAL1 in stellate cells. Hum Genomics 2025; 19:9. [PMID: 39910671 PMCID: PMC11800645 DOI: 10.1186/s40246-025-00722-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: 04/13/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND The progression of liver fibrosis involves complex interactions between hepatic stellate cells (HSCs) and multiple immune cells in the liver, including macrophages. However, the mechanism of exosomes in the crosstalk between liver macrophages and HSCs remains unclear. METHOD Exosomes were extracted from primary mouse macrophages and cultured with HSCs, and the differential expression of microRNAs was evaluated using high-throughput sequencing technology. The functions of miR-342-3p in exosomes were verified by qPCR and luciferase reporter gene experiments with HSCs. The function of the target gene Hippocalcin-like protein 1 (HPCAL1) in HSCs was verified by Western blotting, qPCR, cellular immunofluorescence and co-IP in vivo and in vitro. RESULTS We demonstrated that exosomal microRNA-342-3p derived from primary liver macrophages could activate HSCs by inhibiting the expression of HPCAL1 in HSCs. HPCAL1, which is a fibrogenesis suppressor, could inhibit TGF-β signaling in HSCs by regulating the ubiquitination of Smad2 through direct interactions with its EF-hand 4 domain. CONCLUSION This study reveals a previously unidentified profibrotic mechanism of crosstalk between macrophages and HSCs in the liver and suggests an attractive novel therapeutic strategy for treating fibroproliferative liver diseases.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Lirong Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qi Zhou
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Tiansheng Huang
- Department of Digestive Diseases, Shanghai Guanghua Hospital of Integrated Traditional Chinese And Western Medicine, Shanghai, 200040, China
| | - Wanwei Zheng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhong Guang Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
49
|
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
|
50
|
Budkowska M, Ostrycharz-Jasek E, Cecerska-Heryć E, Dołęgowska K, Siennicka A, Nazarewski Ł, Rykowski P, Dołęgowska B. The Impact of Human Liver Transplantation on the Concentration of Fibroblast Growth Factors: FGF19 and FGF21. Int J Mol Sci 2025; 26:1299. [PMID: 39941067 PMCID: PMC11818808 DOI: 10.3390/ijms26031299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
The multitude of processes in which the liver participates makes it vulnerable to many serious diseases, which can lead to chronic organ failure. Modern medicine bases the treatment of end-stage liver failure on liver transplantation. To ensure the proper functioning of the transplanted liver, a balance of cellular and immunological processes and appropriate concentrations of many different factors are necessary, including, among others, fibroblast growth factors (FGFs). Over the last several years, studies have focused on some FGF growth factors, i.e., FGF19 and FGF21. These two growth factors belong to the FGF19 subfamily, and we concentrate on these two factors in our work. These factors diffuse away from the site of secretion into the blood, acting as hormones. FGF19 is a growth factor with a high therapeutic potential, involved in the homeostasis of bile acids necessary to maintain the proper function of the transplanted liver. FGF21, in turn, plays an important role in regulating lipid and glucose homeostasis. This study aimed to evaluate changes in the concentration of growth factors FGF19 and FGF21 in the plasma of 84 patients before, 24 h, and 2 weeks after liver transplantation (ELISA test was used). Additionally, the correlations of the basic laboratory parameters-alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transpeptidase (GGTP), alkaline phosphatase (ALP), total bilirubin, C-reactive protein (CRP), albumin and hemoglobin (Hb)-with FGF19 and FGF21 were determined. Our studies noted statistically significant changes in FGF19 and FGF21 concentrations before, 24 h, and 2 weeks after liver transplantation. The highest values for FGF19 before liver transplantation and the lowest values 24 h after this surgery were observed for FGF21; the highest concentrations were observed the day after liver transplantation, and the lowest were observed immediately before surgery. Observations of increases and decreases in the concentration of the examined factors at individual time points (before and after transplantation) allow us to suspect that FGF19 has an adaptive and protective function toward the transplanted liver. At the same time, FGF21 may affect the regenerative mechanisms of the damaged organ.
Collapse
Affiliation(s)
- Marta Budkowska
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Ewa Ostrycharz-Jasek
- Institute of Biology, University of Szczecin, 71-412 Szczecin, Poland;
- Doctoral School, University of Szczecin, 70-383 Szczecin, Poland
- Molecular Biology and Biotechnology Center, University of Szczecin, 71-412 Szczecin, Poland
| | - Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.C.-H.); (B.D.)
| | - Katarzyna Dołęgowska
- Department of Immunology Diagnostics, Pomeranian Medical University, Al. Powstanców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Aldona Siennicka
- Department of Medical Analytics, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland;
| | - Łukasz Nazarewski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, ul Banacha 1a, 02-097 Warsaw, Poland; (Ł.N.); (P.R.)
| | - Paweł Rykowski
- Department of General, Transplant and Liver Surgery, Medical University of Warsaw, ul Banacha 1a, 02-097 Warsaw, Poland; (Ł.N.); (P.R.)
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Pomeranian Medical University, Al. Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (E.C.-H.); (B.D.)
| |
Collapse
|