1
|
Hao S, Lv Y, Wang Y, Liu K, Yu X, Tan M. Potential benefits of orally deliverable silymarin-loaded spirulina platensis in mitigating alcoholic liver disease. Food Res Int 2025; 212:116508. [PMID: 40382056 DOI: 10.1016/j.foodres.2025.116508] [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: 12/09/2024] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025]
Abstract
Alcoholic liver disease (ALD), caused by excessive alcohol abuse, encompasses a battery of liver conditions including fatty liver to more severe conditions such as alcoholic hepatitis and cirrhosis. Silymarin derived from the dried fruits of Silybum marianum can shield the liver cells from alcohol damage and prevent toxins from penetrating and damaging the liver. Overcoming the challenges of silymarin's poor solubility, low bioavailability, and limited absorption is crucial to fully realize its health benefits. Herein, spirulina platensis (SP) was used as a natural carrier for silymarin delivery to alleviate ALD in mice. The functional ingredient silymarin was loaded into SP via a single step to construct the hybrid of silymarin@SP. The inherent chlorophyll gives silymarin@SP incredible fluorescence imaging ability for noninvasive monitoring. The orally deliverable silymarin@SP markedly increased silymarin's blood concentration from 2.04 to 4.12 μg/mL and enhanced its hepatic retention. The helical structure of SP carriers allowed silymarin@SP to improve the biological effectiveness of silymarin, significantly enhancing its effectiveness in alleviating ALD. Treatment with silymarin@SP significantly decreased proinflammatory cytokine levels. The potential benefits of orally deliverable silymarin@SP suggest that the SP carrier is effective in enhancing the biological efficacy of encapsulated silymarin in treating alcohol-induced liver damage. The integration of SP with silymarin may also provide combinatorial effects in protecting against and repairing the ALD.
Collapse
Affiliation(s)
- Sijia Hao
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China; College Food Science & Engineering, Bohai University, Jinzhou 121013, Liaoning, China
| | - Yueqi Lv
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Yuxiao Wang
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Kangjing Liu
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaoting Yu
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Mingqian Tan
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
2
|
Ai R, Tian M, Sun J, He S, Cui Z, Yang Y, Hou X, Zhao Y, Dou T, Chen X, Wang J. Mogroside V prevents ethanol-induced hangover and liver damage by reducing oxidative stress, steatosis and inflammation. Biochem Biophys Res Commun 2025; 766:151912. [PMID: 40306161 DOI: 10.1016/j.bbrc.2025.151912] [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: 01/30/2025] [Revised: 04/04/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Excessive alcohol consumption is a leading cause of alcohol-associated liver disease (ALD). Previous studies presented Mogroside V (MV) have protective effects on against nonalcoholic fatty liver disease. however, the effects of MV on ethanol-induced hangover and liver damage remains to be elucidated. Herein, we investigated the potential effects of MV in relieving hangover and mitigating liver injury induced by ethanol. MV significantly reduced blood ethanol, liver histological alterations and serum ALT, AST、TG levels in ethanol-treated mice. Moreover, MV accelerates alcohol metabolism by inhibiting the upregulation of CYP2E1 induced by ethanol, while enhancing the activity of ADH and ALDH, as well as upregulating the expression of ADH1 and ALDH2. MV mitigates oxidative stress by decreased hepatic malondialdehyde (MDA) levels, restored glutathione (GSH)、superoxide dismutase (SOD) and catalase (CAT) content in ethanol-induced mice. Mechanistically, MV activated the p-AMPK/SREBP-1/FASN pathway to decreased hepatic lipid accumulation and alleviated steatosis. Additionally, MV promoted nuclear translocation of Nrf-2 to attenuates oxidative stress and suppressed TLR4/MyD88/NF-κB signaling pathway to reduce Inflammatory responses triggered by ethanol in mice. In summary, this study highlights mogroside V's hangover relieving effect and its protective effects against ethanol-related liver damage through its lipid metabolism regulation, antioxidative action and anti-inflammatory properties. These results suggest that mogroside V could be developed as a potential therapeutic agent against ethanol-induced liver damage.
Collapse
Affiliation(s)
- Rui Ai
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Muzhao Tian
- Faculty of Basic Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Jiawang Sun
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Shuying He
- Faculty of Clinical Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Zhi Cui
- Faculty of Basic Medicine, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Yizhuang Yang
- School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Xinyue Hou
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Yue Zhao
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China
| | - Tong Dou
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; School of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Macau, Taipa, 999078, China
| | - Xu Chen
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China.
| | - Juan Wang
- Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; School of Pharmacy, Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541199, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Zhuang Autonomous Region, 541001, China.
| |
Collapse
|
3
|
Capone F, Vacca A, Bidault G, Sarver D, Kaminska D, Strocchi S, Vidal-Puig A, Greco CM, Lusis AJ, Schiattarella GG. Decoding the Liver-Heart Axis in Cardiometabolic Diseases. Circ Res 2025; 136:1335-1362. [PMID: 40403112 DOI: 10.1161/circresaha.125.325492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
The liver and heart are closely interconnected organs, and their bidirectional interaction plays a central role in cardiometabolic disease. In this review, we summarize current evidence linking liver dysfunction-particularly metabolic dysfunction-associated steatotic liver disease, alcohol-associated liver disease, and cirrhosis-with an increased risk of heart failure and other cardiovascular diseases. We discuss how these liver conditions contribute to cardiac remodeling, systemic inflammation, and hemodynamic stress and how cardiac dysfunction in turn impairs liver perfusion and promotes hepatic injury. Particular attention is given to the molecular mediators of liver-heart communication, including hepatokines and cardiokines, as well as the emerging role of advanced research methodologies, including omics integration, proximity labeling, and organ-on-chip platforms, that are redefining our understanding of interorgan cross talk. By integrating mechanistic insights with translational tools, this review aims to support the development of multiorgan therapeutic strategies for cardiometabolic disease.
Collapse
Affiliation(s)
- Federico Capone
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Department of Medicine, Unit of Internal Medicine III, Padua University Hospital, University of Padua, Padova, Italy (F.C.)
- Department of Biomedical Sciences, University of Padova, Italy (F.C.)
| | - Antonio Vacca
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Clinica Medica, Department of Medicine, University of Udine, Italy (A.V.)
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
| | - Dylan Sarver
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Dorota Kaminska
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
| | - Stefano Strocchi
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, United Kingdom (G.B., A.V.-P.)
- Centro de Investigacion Principe Felipe, Valencia, Spain (A.V.-P.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Milan, Italy (C.M.G.)
- IRCCS Humanitas Research Hospital, Milan, Italy (C.M.G.)
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine (D.S., D.K., A.J.L.), University of California, Los Angeles
- Department of Microbiology, Immunology and Molecular Genetics (D.S., A.J.L.), University of California, Los Angeles
- Department of Human Genetics (D.S., A.J.L.), University of California, Los Angeles
| | - Gabriele G Schiattarella
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (F.C., A.V., S.S., G.G.S.)
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, Germany (S.S., G.G.S.)
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany (G.G.S.)
- Friede Springer Cardiovascular Prevention Center at Charité-Universitätsmedizin Berlin, Germany (G.G.S.)
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin Berlin and Max Delbruck Center for Molecular Medicine, Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy (G.G.S.)
| |
Collapse
|
4
|
Ye RQ, Chen YF, Ma C, Cheng X, Guo W, Li S. Advances in identifying risk factors of metabolic dysfunction-associated alcohol-related liver disease. Biomed Pharmacother 2025; 188:118191. [PMID: 40408808 DOI: 10.1016/j.biopha.2025.118191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/13/2025] [Accepted: 05/21/2025] [Indexed: 05/25/2025] Open
Abstract
Metabolic dysfunction-associated alcohol-related liver disease (MetALD) is an emerging clinical entity that reflects the coexistence of metabolic dysfunction and alcohol-related liver injury. Unlike classical alcoholic liver disease (ALD), MetALD patients often present with lower to moderate alcohol consumption alongside metabolic risk factors such as obesity, insulin resistance, and dyslipidemia. These factors can synergistically worsen liver injury even at lower alcohol intake levels. Alcohol abuse remains a major global health concern, with the liver being the primary target of alcohol's toxic effects. Long-term alcohol exposure, especially when compounded by metabolic dysfunction, can accelerate the progression from steatosis to inflammation, fibrosis, cirrhosis, and hepatocellular carcinoma. Besides alcohol itself, various factors, including genetic predispositions, gender, type of alcoholic beverage, drinking patterns, and co-morbidities such as viral infections (HBV, HCV) modulate disease susceptibility and severity. This review summarizes current knowledge of risk factors contributing to MetALD, highlights the synergistic interactions between metabolic dysfunction and alcohol consumption, and discusses potential strategies for disease prevention and management.
Collapse
Affiliation(s)
- Rui-Qi Ye
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430022, China; Xinhua Clinical Medical College, Shanghai Jiao Tong University, Shanghai 200135, China
| | - Yi-Fan Chen
- College of Basic Medical Sciences, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Chang Ma
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xi Cheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wei Guo
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430022, China.
| | - Sha Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
5
|
Zhang Y, Pan K, Xu A, Sun S, Huang Q, Wang Y, Wang H, Han Q, Li D, Ding Q, Li J. n-3 polyunsaturated fatty acids-enriched fish oil attenuates chronic alcohol-induced liver injury via a mechanism involving the upregulation of Retsat. J Nutr Biochem 2025:109971. [PMID: 40409513 DOI: 10.1016/j.jnutbio.2025.109971] [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: 11/19/2024] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
This study aimed to delineate the protective role of fish oil against alcoholic liver disease (ALD), identify the principal active component between eicosapentaenoic acid (EPA, C20:5 n-3) and docosahexaenoic acid (DHA, C22:6 n-3), and elucidate the molecular mechanisms. C57BL/6J mice were randomly assigned to receive either an alcohol-fed (AF) or pair-fed control (PF) diet, enriched with fish oil (FO) or corn oil (CO) for four weeks. Additionally, a series of in vitro experiments were performed using AML-12 cells to further investigate potential mechanisms. The results showed that plasma levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were significantly lower in the AF-FO group compared to the AF-CO group, indicating that fish oil alleviated alcohol-induced liver damage. Hepatic antioxidant markers, including glutathione (GSH), superoxide dismutase (SOD), and catalase (CAT) were also higher in the AF-FO group than in the AF-CO group. Transcriptomic analysis revealed FO supplementation significantly affected genes involved in oxidoreductase activity and lipid metabolism pathways, with Retsat being the most up-regulated gene. The in vitro experiments indicated that DHA, but not EPA, markedly increased Retsat expression, cell viability, and the expression of genes related to oxidoreductase activity and lipid metabolism, compared to linoleic acid (LA, C18:2 n-6). Notably, knocking down Retsat abolished the protective effects of DHA. In conclusion, dietary fish oil mitigated chronic alcohol-induced liver injury primarily through DHA by upregulating Retsat and downstream genes associated with oxidoreductase function and lipid metabolism.
Collapse
Affiliation(s)
- Yuxuan Zhang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Kaixin Pan
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Angcheng Xu
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Shuzhen Sun
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Qingling Huang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Yicheng Wang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Hao Wang
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Qiang Han
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao, 266071, China
| | - Qinchao Ding
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China
| | - Jiaomei Li
- School of Public Health, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou, 310051, China.
| |
Collapse
|
6
|
Twum E, Ofosu-Boateng M, Nnamani DO, Gebreyesus LH, Yadak N, Kharbanda KK, Gonzalez FJ, Gyamfi MA. Blockade of the estrogen receptor alpha-pregnane X receptor axis protects ovariectomized mice against ethanol-induced hepatotoxicity. J Biol Chem 2025:110238. [PMID: 40381698 DOI: 10.1016/j.jbc.2025.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/20/2025] Open
Abstract
Women develop alcohol-associated liver disease (ALD) faster than men at any level of alcohol consumption, implicating estrogen as a contributing factor. However, the precise mechanism remains unknown. Therefore, 12-weeks-old female C57BL/6N mice were subjected to either bilateral ovariectomy (OVX) or sham surgery. After a three-week recovery period, the mice were fed either a 5% ethanol (EtOH)-containing liquid diet or paired-fed control diet for 10 days followed by a single gavage dose of EtOH (5 g/kg, 30% EtOH solution). The mice were examined for serum biochemical parameters, hepatotoxicity, histology, expression of xenobiotic nuclear receptors PXR and CAR, and their target gene mRNAs and proteins in hepatic and perigonadal white adipose tissues (pgWAT). While OVX mice on a control diet significantly gained weight, EtOH significantly increased hepatotoxicity, residual EtOH levels, lipid peroxidation, and oxidative stress in sham-operated mice but not in their OVX counterparts. Additionally, in the livers and pgWAT of the sham mice, EtOH significantly increased the mRNA and/or protein levels of the major estrogen receptor ERα, PXR, CAR, and their target genes, proinflammatory cytokines and chemokines, lipogenic genes, and FGF21 levels, a predictive biomarker for ALD severity in humans, but inhibited NRF2 and its targets genes encoding NQO1 and BHMT. Unexpectedly, all these changes were attenuated in the EtOH-fed OVX mice by the upregulation of NRF2 and aryl hydrocarbon receptor (AhR) and their downstream antioxidant target genes. Together these results suggest the existence of an estrogen-regulated ERα-PXR-NRF2-signaling axis in liver and pgWAT which contributes to sexual dimorphism in ALD.
Collapse
Affiliation(s)
- Elizabeth Twum
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Malvin Ofosu-Boateng
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Daniel O Nnamani
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Lidya H Gebreyesus
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Nour Yadak
- Department of Pathology and Laboratory Medicine, The University of Tennessee Health Science Center, Memphis, TN, 38163
| | - Kusum K Kharbanda
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105 USA; Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Frank J Gonzalez
- Center for Cancer Research, National Cancer Institute, Building 37, Room 3106, Bethesda, MD, 20892, USA
| | - Maxwell A Gyamfi
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
7
|
Cheng M, Lu H, Wu Y, Jia L, Xiang T, Deng L, Zhao G, Feng J. Sex differences in alcohol inhibits bone formation and promotes bone resorption in young male and female rats by altering intestinal flora, metabolites, and bone microenvironment. PLoS One 2025; 20:e0323222. [PMID: 40338892 PMCID: PMC12061194 DOI: 10.1371/journal.pone.0323222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/04/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Long-term alcohol intake has toxic effects on osteoblasts and osteoclasts, resulting in decreased bone density, which directly disrupts the composition of the gut microbiota and affects bone metabolism and immune activity. The effects of alcohol on the bones may be closely related to sex. This study investigated the effects of long-term alcohol consumption on bone status in different sexes by examining the gut microbiota, bone metabolism, and immune activity. METHODS Young male and female rats were administered a Bio-Serv liquid diet containing 5% alcohol. The effects of alcohol metabolism capacity, bone morphology, bone formation, bone resorption, bone marrow immune activity, gut microbiota, and metabolite differences were analyzed in male and female rats using hematoxylin and eosin staining, micro-computed tomography, enzyme-linked immunosorbent assay, western blotting, 16S rRNA sequencing, and untargeted metabolomics. RESULTS Chronic alcohol consumption resulted in excessive osteoclast activation and decreased bone mineral density. Furthermore, alcohol reduced bone metabolism and formation while increasing bone resorption. Bone loss was significantly more severe in female rats than in male rats, indicating that the effects of alcohol on rat bones are related to sex. Chronic alcohol consumption also led to polarization of bone marrow immunoreactivity toward the M1 phenotype. In addition, chronic alcohol consumption affected the composition of gut microbiota, reduced the richness and diversity of intestinal microbiota, and decreased the ratio of Firmicutes/Bacteroidetes. Long-term alcohol consumption also affected fecal metabolites, and 754 differentially expressed metabolites were identified. CONCLUSIONS Chronic alcohol consumption increased bone resorption, inhibited bone formation, and affected bone marrow immunoreactivity in young male and female rats. Alcohol can also affect gut microbiota composition and fecal metabolism. Female rats were more susceptible to alcohol, possibly because young female rats have a lower alcohol metabolism, immunomodulatory capacity, and gut microbiota diversity than young male rats.
Collapse
Affiliation(s)
- Ming Cheng
- School of Sports Medicine and Health, Chengdu Sport University, Sichuan, China
- Department of Rehabilitation, Jinniu District People’s Hospital of Chengdu, Sichuan, China
| | - Hua Lu
- Operating room, Sichuan Academy of Medical Sciences& Sichuan Provincial People’s Hospital, Sichuan, China
| | - Yangling Wu
- Department of Rehabilitation, Jinniu District People’s Hospital of Chengdu, Sichuan, China
| | - Long Jia
- Department of Rehabilitation, Jinniu District People’s Hospital of Chengdu, Sichuan, China
| | - Tao Xiang
- Department of Rehabilitation, Jinniu District People’s Hospital of Chengdu, Sichuan, China
| | - L.i Deng
- Department of Orthopaedics, Sichuan Academy of Medical Sciences& Sichuan Provincial People’s Hospital, Sichuan, China
| | - Guanlan Zhao
- Department of Orthopaedics, Sichuan Academy of Medical Sciences& Sichuan Provincial People’s Hospital, Sichuan, China
| | - Junwei Feng
- Department of Orthopaedics, Sichuan Academy of Medical Sciences& Sichuan Provincial People’s Hospital, Sichuan, China
| |
Collapse
|
8
|
Xue W, Guo N, Shan L, Zhang Z, Sun Y, Wang Y, Fang X, Liu X, Liu J, Hu C. Caveolin-1 protects against liver injury and lipid accumulation in alcoholic fatty liver via ferroptosis resistance. Mol Immunol 2025; 181:53-65. [PMID: 40073697 DOI: 10.1016/j.molimm.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/14/2025]
Abstract
Alcoholic fatty liver (AFL) is one of the most common chronic liver diseases globally with complex and controversial pathogenesis. Recent evidence suggests that iron overload and lipid peroxidation are risk factors for AFL. Caveolin-1 (CAV1) is an important signal platform that can maintain lipid homeostasis during the development of non-alcoholic fatty liver. Here, we studied the effect of CAV1 on ferroptosis in AFL. The AFL mouse model was established by chronic-plus-binge alcohol feeding. In vitro, AML-12 cells were incubated with ethanol and oleic acid for 48 h. We found alcohol-induced AFL triggered ferroptosis and decreased CAV1 expression. Overexpression of CAV1 by CAV1 scaffolding domain peptides (CSD) attenuated liver injury and hepatic steatosis, as well as inhibited ferroptosis in AFL mice. Additionally, the effects of CAV1 on ferroptosis-related protein levels (such as SLC7A11, GPX4, and ACSL4) and lipid accumulation were reversed by its small interfering RNA administration. Ferroptosis agonist (Erastin) treatment abrogated CAV1 plasmid-mediated ferroptosis resistance and steatosis alleviation. Collectively, the results revealed a crucial role of CAV1 in preventing hepatic steatosis and ferroptosis in alcohol-induced liver injury, which may identify potential targets for the treatment of AFL.
Collapse
Affiliation(s)
- Weiju Xue
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Ning Guo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Liang Shan
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Zhengsheng Zhang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Yuquan Sun
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yong Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Xing Fang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Xiuzhen Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Jianjun Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China.
| | - Chengmu Hu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, Anhui, China; Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
9
|
Wolf T, Moss L, Hudson C, Winters AM, Abdelmaboud SS, Avlas M, Wohlfahrt J, Guergues J, Bickford PC, Stevens SM. Chronic alcohol exposure during young adulthood attenuates microglial reactivity and downstream immune response pathways in a mouse model of tauopathy later in life. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:985-1000. [PMID: 40114609 PMCID: PMC12097940 DOI: 10.1111/acer.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder marked by the buildup of amyloid-β and tau protein tangles. Alcohol use has been identified as a risk factor for AD; however, the molecular mechanisms underlying this potential causal link remain elusive. An emerging area of research focuses on the role of microglia, the brain's innate immune cells, in AD pathogenesis, with evidence suggesting that alcohol exposure may prime microglia to exhibit an exaggerated immune response when they are subsequently exposed to proinflammatory stimuli. METHODS We used a single 10-day chronic-plus-binge alcohol exposure model in male and female C57BL/6J mice aged 8-10 weeks One month later, tauopathy was induced via adenoviral vector (AAV)-mediated overexpression of h-p301L Tau. After 2.5 months, the mice underwent behavioral and cognitive testing. Two weeks later, microglia were collected using fluorescence-activated cell sorting (FACS) and processed for unbiased, mass spectrometry-based proteomic analysis to determine the molecular pathways related to microglial reactivity. RESULTS Microglia from mice exposed to alcohol in young adulthood exhibited a blunted immune response when challenged with AAV-mediated delivery and accumulation of human tau later in life. This was characterized by decreased expression of MHC II- and interferon-associated proteins and bioinformatic prediction of inhibited inflammation-related pathways in the absence of gross histological, behavioral, or cognitive deficits. These results demonstrate unique, temporally specific microglial reactivity to tau that is modulated by early adulthood alcohol exposure, implicating a microglial response that could negatively affect the mechanisms necessary for tau clearance and potentially exacerbate tau pathogenesis. CONCLUSIONS This study provides novel insights into the long-term effects of alcohol exposure in early adulthood on microglial function and the complexity of context-dependent microglial involvement in tauopathy. Consideration of early-adulthood environmental factors is critical for understanding and potentially mitigating the risk of neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Tiara Wolf
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Lauren Moss
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Charles Hudson
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Alexis M. Winters
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Salma S. Abdelmaboud
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Marta Avlas
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Jessica Wohlfahrt
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Jennifer Guergues
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| | - Paula C. Bickford
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Research Service, Tampa, FL, USA
| | - Stanley M. Stevens
- Department of Molecular Biosciences, University of South Florida, Tampa, FL, USA
| |
Collapse
|
10
|
Chen Y, Zhang D, Wu Y, Jiang W, Guo L, Pan D, He Q, Yin Z, Sun L, Wang S. Chronic intermittent hypoxia alleviates alcohol-related liver injury via downregulation of hepatic hypoxia-inducible factor-2α. Am J Physiol Gastrointest Liver Physiol 2025; 328:G610-G623. [PMID: 40243734 DOI: 10.1152/ajpgi.00283.2024] [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: 09/06/2024] [Revised: 11/17/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
Alcohol-related liver disease (ALD) is one of the leading causes of alcohol-related morbidity and mortality worldwide. Unfortunately, limited therapeutic options are currently available, due to the complex risk factors involved as well as the lack of information on the molecular mechanisms driving its progression. Interestingly, chronic, excessive alcohol intake has been reported to exacerbate the severity of obstructive sleep apnea (OSA), a respiratory disorder typically characterized by chronic intermittent hypoxia (CIH). However, this relationship between alcohol-enhanced OSA and ALD development/progression remains to be elucidated. As an approach to investigate this relationship, in vivo Gao-binge ALD and CIH mouse models were established. Alcohol-related liver injury, hepatic steatosis, inflammation, and oxidative stress were then assessed in these models. In addition, lipopolysaccharide (LPS) and ethanol-cotreated mouse normal hepatocyte cell line AML12 served as an in vitro model to investigate the mechanisms through which CIH affects ethanol-induced liver injury. CIH intervention ameliorated alcohol-related liver injury, reduced hepatic lipid accumulation and oxidative stress, and alleviated liver inflammation. Mechanistically, in the liver of these Gao-binge mice, CIH intervention inhibited alcohol-induced upregulation and activation of hypoxia-inducible factor 2α (HIF-2α), a protein which plays a key role in hepatic lipid metabolism and liver injury. Similar to these effects observed in response to CIH intervention, treatment of Gao-binge mice with the selective inhibitor of HIF-2α, PT2385, alleviated alcohol-related liver injury and steatosis while inhibiting oxidative stress and inflammation. Additional findings from our in vitro model revealed that CIH downregulated HIF-2α by promoting calpains protein expression, thereby reducing the accumulation of lipid droplets and decreasing reactiveoxygenspecies (ROS) production in AML12 cells co-challenged with LPS and ethanol. The above results provide important, new evidence that reconceptualizes the role of alcohol-enhanced OSA in ALD progression. Moreover, these findings can serve as the foundation for the development of HIF-2α inhibitors for use in the prevention and treatment of ALD.NEW & NOTEWORTHY Chronic intermittent hypoxia (CIH) intervention mitigated hepatic lipid accumulation and reduced hepatic injury, inflammation, and oxidative stress in alcohol-related liver disease (ALD) mice. CIH alleviates ALD and is likely linked to the downregulation of hypoxia-inducible factor 2α (HIF-2α) expression mediated by calpains. This study presents a new possibility for ALD treatment and lays a theoretical foundation for the clinical treatment of ALD.
Collapse
Affiliation(s)
- Yunling Chen
- Science and Technology Innovation Center, Shandong First Medical University, Jinan, People's Republic of China
- Medical School, Nankai University, Tianjin, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Shandong University, Jinan, People's Republic of China
| | - Dongyuan Zhang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, People's Republic of China
| | - Yunxiao Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Shandong University, Jinan, People's Republic of China
| | - Wenshan Jiang
- Science and Technology Innovation Center, Shandong First Medical University, Jinan, People's Republic of China
| | - Luoting Guo
- Science and Technology Innovation Center, Shandong First Medical University, Jinan, People's Republic of China
| | - Di Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Shandong University, Jinan, People's Republic of China
| | - Qiao He
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Shandong University, Jinan, People's Republic of China
| | - Zhaoqing Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Shandong University, Jinan, People's Republic of China
| | - Lichao Sun
- Emergency Department, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Shuanglian Wang
- Science and Technology Innovation Center, Shandong First Medical University, Jinan, People's Republic of China
| |
Collapse
|
11
|
Zhou S, Liang L, Zhong W, Chen J, Xiao L. Kaempferol ameliorated central nervous injury induced by alcohol uptake through improving intestinal barrier function. Neuroreport 2025:00001756-990000000-00355. [PMID: 40298627 DOI: 10.1097/wnr.0000000000002170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Excessive neuroinflammation resulting from chronic alcohol intake is an important risk factor for central nervous system injury. The aim of this study was to investigate the effect of kaempferol (KAE) on alcohol-induced neural injury and its underlying mechanism. C57BL/6 N mice were employed to develop a binge-on-chronic alcohol exposure model, with different doses of KAE as an interventional drug for 6 weeks. Neuronal damage and microglial activation in the brain, as well as colonic tissue damage and serum lipopolysaccharide (LPS) concentrations, were systematically assessed. Additionally, Caco-2 cells were exposed to alcohol to induce intestinal epithelial injury in vitro. Chronic alcohol exposure let to significant neuronal damage in the cortex and hippocampus of mice. KAE treatment effectively attenuated microglial activation and reduced neuronal damage in the brains of alcohol-exposed mice. Analysis of colonic tissues revealed that KAE administration inhibited miRNA-122a expression, alleviated pathological damage, and enhanced occludin expression, thereby significantly lowing serum LPS concentrations in alcohol-fed mice. In vitro, KAE markedly decreased miRNA-122a expression and enhanced occludin levels in Caco-2 cells treated with alcohol. Furthermore, overexpression of miRNA-122a was found to diminish occludin protein production in Caco-2 cells, which was significantly counteracted by KAE treatment. KAE treatment enhanced intestinal barrier function to alleviate neuronal damage caused by microglial activation mediated by gut-derived LPS under alcohol expose. This effect of KAE was involved in the enhance of intestinal occludin expression by inhibiting the expression of miRNA-122a. This suggested that KAE had the potential to prevent alcohol-induced neurological damage.
Collapse
Affiliation(s)
- Shinan Zhou
- College of Basic Medical Sciences, China Three Gorges University, YiChang, China and
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, YiChang, China
| | - Lu Liang
- College of Basic Medical Sciences, China Three Gorges University, YiChang, China and
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, YiChang, China
| | - Wenyan Zhong
- College of Basic Medical Sciences, China Three Gorges University, YiChang, China and
| | - Jingjing Chen
- College of Basic Medical Sciences, China Three Gorges University, YiChang, China and
| | - Li Xiao
- College of Basic Medical Sciences, China Three Gorges University, YiChang, China and
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, YiChang, China
| |
Collapse
|
12
|
Che Z, Cai M, Dong X, Yuan Y, Wang Y, Xiao L, Song Y, Zhong J, Luo P, Wang H, Lu G, Sun Y, Xiao J. Angiotensinogen inhibition concurrently mitigates alcohol-associated hepatic and muscle injury. Metabolism 2025; 169:156275. [PMID: 40311841 DOI: 10.1016/j.metabol.2025.156275] [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: 11/14/2024] [Revised: 04/16/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
AIMS The organ communication mechanisms driven by alcohol-associated liver disease (ALD) remain inadequately understood. This study explores the endocrine roles of the hepatokine angiotensinogen (AGT) and the renin-angiotensin system (RAS) in ALD. METHODS AND RESULTS Hepatokine screening tests revealed that chronic-binge ethanol consumption upregulates hepatic AGT production, triggering downstream RAS activation. Hepatocyte-specific knockout of Agt (AGTΔHep) significantly alleviated ALD-induced liver injury. In organ screening between AGTflox/flox (AGTf/f) and AGTΔHep mice, skeletal muscle exhibited the most pronounced improvement in alcoholic myopathy (AM)-related phenotypes, including reduced muscle mass, enhanced oxidative stress, and mitochondrial dysfunction post-ethanol administration. Mechanistically, the renin-angiotensin axis transmits damaging signals from AGT to their membrane receptor AGTR1 in both hepatocytes and myocytes. Pharmacological inhibition of AGT, renin, and angiotensin-converting enzyme, as well as specific knockdown of Agtr1 in hepatocytes or myocytes, effectively attenuated both conditions. Activation of the counteractive axis of the RAS-AGTR1 pathway, involving Ang (1-7) and its membrane receptor MAS1, ameliorated the alcoholic injury of both the liver and muscle. Conversely, specific knockdown of Mas1 in hepatocytes and myocytes exacerbated these injuries. CONCLUSIONS Our work demonstrates that hepatokine AGT promotes ALD and AM through the activation of the RAS-AGTR1 axis and the inhibition of the Ang(1-7)-MAS1 axis, offering a foundation for concurrent therapeutic strategies for both diseases.
Collapse
Affiliation(s)
- Zhaodi Che
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Mingxiang Cai
- Clinical Research Platform for Interdiscipline of Stomatology, The First Affiliated Hospital of Jinan University, Department of Stomatology, College of Stomatology, Jinan University, Guangzhou 510630, China
| | - Xiaowu Dong
- Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China
| | - Yuan Yuan
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Aier School of Ophthalmology, Central South University, Changsha 410083, China
| | - Yaodong Wang
- Kunshan Hospital of Chinese Medicine, Kunshan Affiliated Hospital of Yangzhou University, Suzhou 215000, China
| | - Lu Xiao
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yali Song
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiajun Zhong
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Pingping Luo
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Wang
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Guotao Lu
- Institute of Digestive Diseases, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou 225009, China.
| | - Yao Sun
- Department of Oral Implantology, School of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai 200092, China.
| | - Jia Xiao
- Department of Anesthesiology and Clinical Research Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266000, China.
| |
Collapse
|
13
|
Li Q, Huang J, Zhao Q, Li F. FXR as a pivotal role linking JNK and G0s2 mitigates triptolide-induced hepatotoxicity through the regulation of metabolic disorder of liver. Pharmacol Res 2025; 216:107738. [PMID: 40288593 DOI: 10.1016/j.phrs.2025.107738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Triptolide (TP), as a principal bioactive compound derived from Tripterygium wilfordii Hook. f., exhibits significant anti-tumor, anti-inflammatory, and immunomodulatory properties. However, the serious adverse reactions and hepatotoxicity of TP limit its clinical application. Therefore, in this study, an intraperitoneal injection was employed to establish a TP-induced hepatotoxicity model, characterized by elevated levels of transaminases (AST and ALT) and metabolic disorders. The administration of the JNK inhibitor SP600125 effectively mitigated the elevated transaminases and inflammation induced by TP. The resistance of SP600125 to metabolic disturbances induced by TP was contingent upon Fxr, as demonstrated through the use of Fxr knockout mice. Supplementation of GW4064 restored the concentrations of bile acids, long-chain fatty acids, and carnitine disrupted by TP. Transcriptomic data suggested that G0s2 was one of the genes most severely disrupted by TP, and the ameliorative effects of SP600125 and GW4064 were accompanied by the upregulation of G0s2. The expression of G0s2 was disrupted by siRNA in vitro, thereby intensifying the cytotoxicity of TP. A comparative analysis of the impact of TP on the G0s2 gene in two mouse models revealed that a smaller reduction in wild-type mice compared to Fxr-/- mice, indicating that Fxr mitigates the inhibitory effect of TP on G0s2. The aberrant JNK/Fxr/G0s2 signaling plays a key role in TP-induced hepatotoxicity. Targeting Fxr might be a potential strategy for alleviating the liver toxicity of TP.
Collapse
Affiliation(s)
- Qinmei Li
- Department of Pharmacy and Laboratory of Hepato-Intestinal Diseases and Metabolism, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jianfeng Huang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China; Shanwei Institute for Food and Drug Control, Shanwei, Guangdong Province 516622, China
| | - Qi Zhao
- Department of Pharmacy and Laboratory of Hepato-Intestinal Diseases and Metabolism, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fei Li
- Department of Pharmacy and Laboratory of Hepato-Intestinal Diseases and Metabolism, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Qu J, Xu X, Yang J, Zhang Q, Zhang Y, Xu L, Xu H, Li Q. Establishment of an efficacy-oriented quality grading framework for herbal medicines: Phyllanthus emblica as an example. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119632. [PMID: 40089198 DOI: 10.1016/j.jep.2025.119632] [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: 01/27/2025] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Quality control is a powerful method for ensuring the effectiveness and safety of herbal medicines. Phyllanthus emblica L. fruit (PE) has been extensively used in both Ayurvedic and traditional Chinese medicine. However, the current Indian and Chinese pharmacopeias set a minimum concentration threshold of gallic acid to identify qualified PE samples, without providing a clear framework to distinguish superior-quality PE samples. AIM OF THE STUDY To establish an efficacy-oriented quality grading framework for herbal medicines, using PE, a medicinal plant known for its hepatoprotective activity, as an example. METHODS First, a mouse model of alcohol-induced liver injury was developed to evaluate the hepatoprotective effects of PE. Second, a combined strategy of serum pharmacochemistry, network pharmacology, metabolomics and experimental validation was employed to identify key quality markers (Q-markers) linked to the hepatoprotective effects of PE. Finally, PE samples from different sources were collected to assess their hepatoprotective activities and Q-marker concentrations. A discriminant analysis model was then developed to classify PE samples into different quality grades by using Q-marker concentration as the predictive factor and hepatoprotective activity as the evaluation criterion. RESULTS PE significantly alleviated liver damage, as evidenced by a reduction in pathological abnormalities and serum aminotransferase levels. Six hepatoprotective Q-markers in PE were identified and verified, including gallic acid, methyl gallate, corilagin, chebulagic acid, ellagic acid and quercitrin. Significant variability in Q-marker concentrations and hepatoprotective effects was observed among different sources of PF samples, and a discriminant analysis model accurately classified PE samples into distinct quality grades. CONCLUSIONS This study successfully established an efficacy-oriented quality grading framework for PE, providing a methodological approach for the quality classification of herbal medicines.
Collapse
Affiliation(s)
- Jiameng Qu
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xuege Xu
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Junjie Yang
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qian Zhang
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yiwen Zhang
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Li Xu
- College of Basic Medicine, Dali University, Dali, 671003, China
| | - Huarong Xu
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Qing Li
- National and Local Joint Engineering Laboratory for Key Technology of Chinese Material Medica Quality Control, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
15
|
Chatti IB, Kosksi T, Selmi S, Selmi A, Rejeb M, Debbabi N, Jaziri SK, Ghedira LC. Antioxidant, Antigenotoxic, and Anti-Inflammatory Properties of Rhamnus alaternus Leaf Extract Against Ethanol-Induced Liver Injury in Rat Model. Chem Biodivers 2025:e202500115. [PMID: 40267411 DOI: 10.1002/cbdv.202500115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/24/2025] [Accepted: 04/23/2025] [Indexed: 04/25/2025]
Abstract
The purpose of our study was to investigate the curative effect of an enriched total oligomer flavonoid (TOF) extract from Rhamnus alaternus on ethanol-induced rat hepatic injury. We investigated the effects of R. alaternus in adult Wistar rats administered the drug (20 mg/kg) along with 4 g/kg of 40% ethanol. The findings noted that treatment with ethanol caused an increase in serum liver function parameters (alanine transferases, aspartate transaminase, and total bilirubin), in tumor necrosis factor alpha (TNF-α), in interleukin 1 beta (IL-1β), in granulocyte-macrophage colony-stimulating factor (GM-CSF), and in malondialdehyde (MDA) levels and a decrease in hepatic antioxidant activities, including superoxide dismutase, glutathione peroxidase, and glutathione. In addition, it intensified histopathological changes in the liver, as revealed by mononuclear cells' infiltration and steatosis compared to control. When compared to the ethanol-alone-treated group, the rat receiving ethanol plus TOF extract exhibited significant decrease in serum liver function parameters, in TNF-α, in IL-1β in GM-CSF and MDA levels, and increased hepatic antioxidant activities. Histopathological observations confirmed the curative effect of TOF against ethanol-induced liver injury in rats. These results suggest that TOF extract from R. alaternus has hepatoprotective effect by elevating antioxidative potentials and levels of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Ines Bouhlel Chatti
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
- Department of Biology and Geology, Preparatory Institute for Engineering Studies in Gabès, University of Gabès, Gabès, Tunisia
| | - Tahsine Kosksi
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Salima Selmi
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Arem Selmi
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Marwa Rejeb
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Nawres Debbabi
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Soumaya Kilani Jaziri
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| | - Leila Chekir Ghedira
- Faculty of Dental Medicine, Unity of Bioactive Natural Substances and Biotechnology, University of Monastir, Monastir, Tunisia
| |
Collapse
|
16
|
Iakovleva V, de Jong YP. Gene-based therapies for steatotic liver disease. Mol Ther 2025:S1525-0016(25)00298-9. [PMID: 40254880 DOI: 10.1016/j.ymthe.2025.04.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 03/26/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025] Open
Abstract
Advances in nucleic acid delivery have positioned the liver as a key target for gene therapy, with adeno-associated virus vectors showing long-term effectiveness in treating hemophilia. Steatotic liver disease (SLD), the most common liver condition globally, primarily results from metabolic dysfunction-associated and alcohol-associated liver diseases. In some individuals, SLD progresses from simple steatosis to steatohepatitis, cirrhosis, and eventually hepatocellular carcinoma, driven by a complex interplay of genetic, metabolic, and environmental factors. Genetic variations in various lipid metabolism-related genes, such as patatin-like phospholipase domain-containing protein 3 (PNPLA3), 17β-hydroxysteroid dehydrogenase type 13 (HSD17B13), and mitochondrial amidoxime-reducing component 1 (MTARC1), impact the progression of SLD and offer promising therapeutic targets. This review largely focuses on genes identified through clinical association studies, as they are more likely to be effective and safe for therapeutic intervention. While preclinical research continues to deepen our understanding of genetic factors, early-stage clinical trials involving gene-based SLD therapies, including transient antisense and small-molecule approaches, are helping prioritize therapeutic targets. Meanwhile, hepatocyte gene editing technologies are advancing rapidly, offering alternatives to transient methods. As such, gene-based therapies show significant potential for preventing the progression of SLD and enhancing long-term liver health.
Collapse
Affiliation(s)
- Viktoriia Iakovleva
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
17
|
Kim DY, Shin SC, Kim GJ, Eom JI, Han CH, Pan CH, Lee JK. The Beneficial Effects of Ethanol Extract of the Microalga Phaeodactylum tricornutum in Alcoholic Liver Disease. Int J Mol Sci 2025; 26:3851. [PMID: 40332498 PMCID: PMC12027534 DOI: 10.3390/ijms26083851] [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: 02/24/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Alcoholic liver disease (ALD) is a condition resulting from liver damage linked to excessive drinking over a brief duration. It poses a significant public health challenge globally, with its prevalence and morbidity rising annually due to escalating rates of alcohol abuse, which adversely affect human health. Phaeodactylum tricornutum (PT), a diatom species of microalgae, is reported to possess active components that provide anti-inflammatory and antioxidant benefits. This study aimed to investigate the preventive and therapeutic effects of PT extract on ALD. To address our purpose, we used ethanol diet induced live disease model. Mice fed an ethanol diet showed less weight gain and higher levels of AST and ALT compared to those fed a regular diet. PT extract suppressed the inhibition of weight gain and the increase in AST/ALT levels caused by an ethanol diet. In addition, PT extract also prevented liver tissue damage caused by an ethanol diet. Thus, the effect of PT on ALD was found to be related to the inhibition of mitogen-activated protein kinase (MAP kinases) phosphorylation and TNF-α production.
Collapse
Affiliation(s)
- Dae Yoon Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Republic of Korea;
| | - Seung Cheol Shin
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju 28160, Republic of Korea; (S.C.S.); (G.J.K.)
| | - Gab Jung Kim
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju 28160, Republic of Korea; (S.C.S.); (G.J.K.)
| | - Jae-In Eom
- Microalgae Ask Us Co., Ltd., Gangneung 25441, Republic of Korea; (J.-I.E.); (C.-H.H.)
| | - Cheol-Ho Han
- Microalgae Ask Us Co., Ltd., Gangneung 25441, Republic of Korea; (J.-I.E.); (C.-H.H.)
| | - Cheol-Ho Pan
- Microalgae Ask Us Co., Ltd., Gangneung 25441, Republic of Korea; (J.-I.E.); (C.-H.H.)
| | - Jae Kwon Lee
- Department of Biology Education, College of Education, Chungbuk National University, Cheongju 28160, Republic of Korea; (S.C.S.); (G.J.K.)
| |
Collapse
|
18
|
Thimme Gowda C, Siraganahalli Eshwaraiah M, Wang J, Lim Y, Tomasi ML, Mavila N, Ramani K. The AKAP12-PKA axis regulates lipid homeostasis during alcohol-associated liver disease. Signal Transduct Target Ther 2025; 10:109. [PMID: 40199859 PMCID: PMC11979000 DOI: 10.1038/s41392-025-02202-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Disrupted lipogenic signaling and steatosis are key features of alcohol-associated liver disease (ALD). A-kinase anchoring protein 12 (AKAP12) is a scaffolding partner of the cAMP-dependent protein kinase, PKA that controls its spatiotemporal localization. Activation of PKA by cAMP inhibits lipogenesis and facilitates fatty acid oxidation (FAO). The goal of this work is to examine how AKAP12's PKA-anchoring ability regulates outcomes of alcohol-associated steatosis. Crosslinking proteomics identified PKA and its lipogenic substrates as interacting partners of AKAP12. Alcohol exposure diminished AKAP12's interaction with PKA regulatory subunits and PKA substrates, acetyl CoA carboxylase (ACC1), pyruvate dehydrogenase (PDHA) and adipose triglyceride lipase (ATGL). Alcohol inhibited PKA activity and increased triglyceride content in human hepatocytes. Forced expression of AKAP12 restored alcohol suppressed PKA activation and inhibited lipid accumulation, whereas silencing had the reverse effect. Since AKAP12 sustained PKA activity, we evaluated whether the AKAP12-PKA scaffold was important in lipid homeostasis. Inhibition of AKAP12-PKA interaction by CRISPR deletion of AKAP12's PKA binding domain in cultured hepatocytes or in mouse models of ALD dramatically suppressed PKA activity, enhanced ACC1 activity demonstrated by reduced inhibitory phosphorylation, increased lipid accumulation and reduced FAO in hepatocytes. Overexpression of AKAP12 in mouse livers sustained PKA activation, diminished basal and alcohol potentiated triglyceride content, and regulated inflammatory signaling altered by alcohol. Mechanistically, we discovered that alcohol enhanced the inhibitory activity of a kinase, serine/threonine-protein kinase 25 (STK25) on PKA that regulated its interaction with AKAP12. In conclusion, the AKAP12-PKA scaffold controls lipogenic signaling, disruption of which favors steatosis during ALD.
Collapse
Affiliation(s)
- Chandana Thimme Gowda
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Jiaohong Wang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Youngyi Lim
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maria Lauda Tomasi
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Nirmala Mavila
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Komal Ramani
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Applied Cell Biology Division, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
19
|
Maccioni L, Guan Y, Kim M, Parra MA, Peiffer B, Fu Y, Wang Y, Lin YH, Mackowiak B, Feng D, Cameron A, Sun Z, Kunos G, Stärkel P, Gao B. Opposite regulation of intestinal and intrahepatic CD8 + T cells controls alcohol-associated liver disease progression. Gut 2025:gutjnl-2024-334412. [PMID: 40199574 DOI: 10.1136/gutjnl-2024-334412] [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: 11/25/2024] [Accepted: 02/15/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Gut-liver crosstalk plays an important role in alcohol-associated liver disease (ALD) pathogenesis; but underlying mechanisms remain obscure. OBJECTIVE We examined the regulation of intestinal and intrahepatic CD8+ T lymphocytes and their contribution to ALD. DESIGN ALD patients were recruited for evaluation of intestinal and liver T cells. Single-cell RNA sequencing (scRNA seq) was performed to analyse intrahepatic and peripheral T cells in ALD. Wildtype, CD8-specific Bcl2 transgenic (Cd8 Bcl-2), and Cd8 -/- mice were subjected to chronic-plus-binge ethanol feeding. RESULTS In ALD patients, duodenal CD8+ T cells were selectively reduced and negatively correlated with liver injury and bacterial translocation markers, while intrahepatic CD8+ T cells were markedly increased. ScRNA seq analysis of ALD patient livers revealed several populations of CD8+ T cells expressing activation and survival genes (eg, Bcl2). Transcriptomics and functional studies revealed a key role of prosurvival BCL2 in this opposite regulation of CD8+ T cells. Mechanistically, chronic-plus-binge ethanol feeding reduced CD8+ T cells specifically in the duodenum where ethanol levels are high. Inducing BCL2 in CD8+ T cells reversed ethanol-induced loss of duodenal CD8+ T cells, improved gut barrier function and ameliorated ALD, while CD8 deficiency was linked to enhanced neutrophil and macrophage infiltration in the liver, exacerbating ALD in mice. CONCLUSIONS ALD is associated with loss of duodenal CD8+ T cells but elevation of intrahepatic CD8+ T cells, which aggravates and ameliorates ALD, respectively. Restoration of survival and functions of intestinal and intrahepatic CD8+ T cells may represent a novel therapeutic strategy for ALD patients.
Collapse
Affiliation(s)
- Luca Maccioni
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yukun Guan
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Mariia Kim
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia, USA
| | - Maria A Parra
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Brandon Peiffer
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yaojie Fu
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yang Wang
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Yu-Hong Lin
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Bryan Mackowiak
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Dechun Feng
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Andrew Cameron
- Surgery - Division of Transplant Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| | - Peter Stärkel
- Department of Hepato-Gastroenterology, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
- Laboratory of Hepato-gastroenterology (GAEN), Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Bin Gao
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA
| |
Collapse
|
20
|
Xu L, Zhao Y, Yang Y, Qi E, Liu B, Zhuang P, Song S, Chang T, Chen Z, Kang X, Xiong X. Constitutive Hepatic mTORC1 Activation Aggravates Alcohol-Induced Liver Injury via Endoplasmic Reticulum Stress-Mediated Ferroptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00106-3. [PMID: 40204188 DOI: 10.1016/j.ajpath.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
Alcohol-related liver disease (ALD), a consequence of excessive alcohol use, manifests across a broad spectrum of liver damage, ranging from steatosis to cirrhosis. DEPDC5 (DEP domain-containing protein 5) is a component of the GATOR1 (gap activity towards rags 1) complex, which functions as a repressor of the amino acid-sensing branch of the mammalian target of rapamycin complex 1 (mTORC1) pathway. In this study, hepatocyte-specific Depdc5 knockout mice (Depdc5△Hep) were generated, and it was found that aberrant activation of mTORC1 caused by Depdc5 deletion led to exacerbated endoplasmic reticulum (ER) stress and hepatocyte ferroptosis in the livers of ethanol-fed mice. Torin-1, an ATP-competitive mTOR inhibitor, suppressed the mTORC1 activity and reversed the effects of Depdc5 deletion on ER stress and ferroptosis in ethanol-fed mouse livers. Furthermore, pharmacologic relief of ER stress using tauroursodeoxycholic acid or inhibition of ferroptosis with liproxstatin-1 both alleviated the liver abnormalities induced by Depdc5 ablation in ethanol-fed mice. In addition, the research uncovered that ER stress functions as an upstream signal of ferroptosis in the progression of ALD. These findings provide novel in vivo evidence that sustained mTORC1 activation leads to alcoholic liver injury by inducing ER stress and ferroptosis, suggesting that targeting these pathways may represent a potential therapeutic strategy for ALD.
Collapse
Affiliation(s)
- Lin Xu
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; School of Medical Technology, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yuanyuan Zhao
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Yang Yang
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Enbo Qi
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Boao Liu
- Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Peili Zhuang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Shiyi Song
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Tingmin Chang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhiguo Chen
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaohong Kang
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Department of Radiation Oncology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China.
| | - Xiwen Xiong
- Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China; Xinxiang Key Laboratory of Metabolism and Integrative Physiology, School of Forensic Medicine, Xinxiang Medical University, Xinxiang, Henan, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China.
| |
Collapse
|
21
|
Chen L, Liu Q, Li X, Zhang L, Dong W, Li Q, Su H, Luo G, Huang Y, Yang X. The diabetes medication Canagliflozin attenuates alcoholic liver disease by reducing hepatic lipid accumulation via SIRT1-AMPK-mTORC1 signaling pathway. Eur J Pharmacol 2025; 992:177320. [PMID: 39929419 DOI: 10.1016/j.ejphar.2025.177320] [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: 09/30/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND AND AIMS Chronic consumption of large amounts of alcohol can lead to hepatic lipid accumulation and mitochondrial oxidative stress, resulting in alcoholic liver disease (ALD). Canagliflozin (Cana), an oral antidiabetic drug, regulates blood glucose by inhibiting sodium-glucose cotransporter-2 in renal tubulars, which also improves lipid metabolism and alleviates oxidative stress in hepatocyte. This study aims to determine the therapeutic effects of Cana on alcoholic liver injury and to explore the mechanistic pathways involved. METHODS C57BL/6J male mice at 8 weeks were used to construct a model of alcoholic fatty liver disease using the chronic-plus-binge alcohol feeding model. Primary hepatocytes and AML12 cell lines were used as in vitro models. The effects and mechanisms of Cana on alcoholic liver injury were investigated by using immunofluorescence, ELISA, H&E and Oil Red O staining, RT-PCR, and western blotting analysis. RESULTS Cana treatment reduced hepatic lipid accumulation, decreased glutathione and TNF-α levels, alleviated oxidative stress and inflammation. Mechanistic studies revealed that Cana reduced FAS expression in the liver, decreasing hepatic fatty acid synthesis, and increased PPARα expression, promoting fatty acid oxidation. Additionally, Cana increased mitochondrial content and promoted mitophagy. These effects were mediated by the SIRT1-AMPK-mTORC1 signaling pathway. CONCLUSIONS Cana activates the SIRT1-AMPK-mTORC1 signaling pathway, inhibiting alcohol-induced fatty acid synthesis, promoting fatty acid degradation, thereby alleviating alcoholic liver injury.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qinhui Liu
- Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiangyu Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Liaoyun Zhang
- Department of Pharmacy, Sichuan Provincial Maternity and Child Health Care Hospital & Women's and Children's Hospital, Chengdu, Sichuan, 610000, China
| | - Wenjie Dong
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Qiuyu Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Hao Su
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Gang Luo
- Department of Gastroenterology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China; School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
22
|
Wang Y, Zhou X, Chen H, Li Z. Molecular mechanisms of alcohol-associated liver disease-ferroptosis and autophagy crosstalk. Mol Biol Rep 2025; 52:361. [PMID: 40183835 DOI: 10.1007/s11033-025-10443-0] [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: 12/28/2024] [Accepted: 03/17/2025] [Indexed: 04/05/2025]
Abstract
Alcohol-associated liver disease (ALD) is a chronic liver injury caused by prolonged heavy drinking and its pathogenesis is extremely complex. According to current researches, ethanol metabolism and the generation of some of its related metabolites, including acetaldehyde and reactive oxygen species, are significant contributors to hepatocyte toxicity. These substances-induced lipid metabolism disorders, inflammatory response, mitochondrial damage, and cellular oxidative stress are important factors that lead to liver injury. Ethanol has been shown in numerous studies to exacerbate ALD by disrupting autophagy via a variety of mechanisms. ALD can be somewhat alleviated by activating autophagy, which plays a significant role in the development of ALD by removing accumulated protein polymers, damaged mitochondria, and excess lipid droplets from hepatocytes. Furthermore, persistent alcohol use raises serum iron levels, which in turn causes hepatocytes to absorb more iron. This, in turn, encourages iron loading in the liver's and other organs' parenchymal and nonparenchymal cells, finally resulting in ferroptosis. Both ferroptosis and autophagy are significant types of controlled cell death, and new research has revealed that cellular autophagy and a variety of signaling pathways play a key role in the initiation and progression of ferroptosis. Alcohol and iron both have the ability to cause oxidative stress on their own, thus their combined effects hasten liver damage. Iron loading, on the other hand, accelerates the development of ALD by triggering mitochondrial oxidative stress and activating signaling pathways and proteins linked to Ferritinophagy. Thus, we think that a new approach to treating ALD in the future will involve examining the interaction between ferroptosis and mitochondrial autophagy based on iron overload.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xin Zhou
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China
| | - Hui Chen
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Zhi Li
- Department of Spleen and Stomach Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan Province, China.
- School of Integrated Traditional Chinese and Western Clinical Medicine, North Sichuan Medical College, Nanchong, Sichuan, China.
| |
Collapse
|
23
|
Feng X, Wang Y, Zhu C, Huai Q, Cui J. Porphyromonas gingivalis aggravates alcohol-related liver injury via gut microbiome-HO-1-ACSL4-dependent ferroptosis. Front Microbiol 2025; 16:1554703. [PMID: 40241734 PMCID: PMC12000935 DOI: 10.3389/fmicb.2025.1554703] [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: 01/09/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
Background Alcoholic liver disease (ALD) is a common liver condition caused by long-term alcohol consumption, and its specific molecular mechanism remains unclear. It may be influenced to some extent by ferroptosis and Porphyromonas gingivalis (P.g), which is an important pathogen of periodontitis. Materials and methods C57BL/6 J mice and AML12 cells were selected as the study subjects. The periodontitis model was induced using P.g, and the alcoholic liver model was created. Pathological analysis was performed on the liver, intestine, and periodontal tissues. 16S rRNA sequencing was used to analyze changes in the intestinal flora and intestinal gap junction protein (zonula occludens-1 (ZO-1) and occludin) levels in each group. Ferroptosis indices were detected in the liver tissues and AML12 cells. Results Oral exposure to P.g induced mice periodontitis and exacerbated alcohol-related liver injury. Both alcohol and P.g caused intestinal flora disturbance, damage to the intestinal epithelial barrier, increased permeability, and activation of mouse hepatocyte ferroptosis. Furthermore, P.g aggravated such alcohol-induced liver damage. Conclusion Both alcohol and P.g can lead to intestinal flora disturbance, damage to the intestinal epithelial barrier, increased permeability, and the activation of mouse hepatocyte ferroptosis, and P.g can aggravate such alcohol-induced liver damage. Acyl-CoA synthetase long-chain family member 4 (ACSL4) and heme oxygenase-1 (HO-1) play important roles in the exacerbation of alcoholic liver injury by P.g.
Collapse
Affiliation(s)
- Xuezhe Feng
- Department of Stomatology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Wang
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cheng Zhu
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qian Huai
- Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Juanjuan Cui
- Department of Stomatology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
24
|
Docherty J. Therapeutic potential of faecal microbiota transplantation for alcohol use disorder, a narrative synthesis. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111354. [PMID: 40185194 DOI: 10.1016/j.pnpbp.2025.111354] [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: 08/12/2024] [Revised: 02/04/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Faecal microbiota transplantation is proposed as an alternative therapy to treat alcohol use disorder and has completed a Phase 1 clinical trial, with a Phase 2 clinical trial underway. Alcohol, a modifiable risk factor for noncommunicable diseases, resulted in approximately 3 million global deaths (5 %) in 2016 according to the World Health Organization. AIMS A narrative synthesis examines the effects of alcohol and faecal microbiota transplantation on gut microbiota and how gut microbiota impacts the gut-brain axis, leading to certain behavioural symptoms of alcohol use disorder. These behavioural symptoms are alcohol craving and relapse in humans; and preference for alcohol, anxiety and depression in rodents. SEARCH METHODS AND RESULTS Electronic databases PubMed, Embase, and Scopus were searched in January 2024 using the terms: faecal microbiota trans* AND alcohol AND microbio*. Ten studies out of 964 met the inclusion criteria of published primary studies with faecal microbiota transplantation as an intervention to study the gut-brain axis in alcohol use disorder. RESULTS The gut microbiota is altered in alcohol use disorder, which can be modified with faecal microbiota transplantation. Behavioural symptoms such as alcohol craving and relapse are associated with inflammation due to a loss of intestinal barrier function. Beneficial microbiota produce short-chain fatty acids that maintain intestinal barrier function and reduce inflammation. Studies also reported anxiety and depression-like behaviours, in addition to a preference for alcohol in alcohol-naïve rodents after faecal microbiota transplantation from patients with alcohol use disorder. CONCLUSIONS Faecal microbiota transplantation may moderate the behavioural symptoms of alcohol use disorder by altering gut microbiota, affecting intestinal permeability and inflammation, however, specific gut microbiota composition and long-term treatment outcomes require further clinical studies.
Collapse
Affiliation(s)
- Jennifer Docherty
- Formerly, Institute of Psychiatry, Psychology & Neuroscience, King's College London, United Kingdom..
| |
Collapse
|
25
|
Meijnikman AS, Fondevila MF, Arrese M, Kisseleva T, Bataller R, Schnabl B. Towards more consistent models and consensual terminology in preclinical research for steatotic liver disease. J Hepatol 2025; 82:760-766. [PMID: 39581500 DOI: 10.1016/j.jhep.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024]
Abstract
Steatotic liver disease (SLD) is one of the most prevalent liver conditions globally and a leading cause of liver transplantation, yet therapeutic advances have not kept pace with its major impact on global morbidity and mortality. This underscores the critical importance of developing and refining relevant preclinical animal models. However, preclinical research has faced significant challenges, with concerns about the translational validity of animal models, as findings often fail to accurately reflect human disease. With the recent adoption of new nomenclature for SLD in humans, questions have arisen about how to integrate these changes into preclinical models. Here, we offer suggestions on how to improve preclinical models, including the incorporation of factors such as diet, alcohol, and other metabolic stressors, to better replicate the complexity of human disease. While implementing these improvements presents practical challenges, doing so is essential for enhancing the translational relevance and reproducibility of animal studies, and advancing therapeutic discoveries. Furthermore, we address the persisting inconsistency in terminology used in animal studies and propose clinically meaningful terms that can be applied consistently to preclinical research.
Collapse
Affiliation(s)
- Abraham S Meijnikman
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 BK Amsterdam, the Netherlands
| | - Marcos F Fondevila
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marco Arrese
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Ramon Bataller
- Liver Unit, Hospital Clinic. Institut d'Investigacions Biomediques August Pi i Sunyer (IDI-BAPS). Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA, USA.
| |
Collapse
|
26
|
Ma Q, Huang S, Li MY, Luo QH, Chen FM, Hong CL, Yan HH, Qiu J, Zhao KL, Du Y, Zhao JK, Zhou LQ, Lou DY, Efferth T, Li CY, Qiu P. Dihydromyricetin regulates the miR-155-5p/SIRT1/VDAC1 pathway to promote liver regeneration and improve alcohol-induced liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156522. [PMID: 39986231 DOI: 10.1016/j.phymed.2025.156522] [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: 05/31/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Alcohol-related liver disease (ALD) has become an increasingly serious global health issue. In recent years, growing evidence has highlighted the restoration of liver regenerative capacity as an effective therapeutic strategy for improving ALD. Previous studies have demonstrated the protective effect of dihydromyricetin (DMY) in alcohol-induced liver injury, but its pharmacological role in ALD-related liver regeneration impairment remains poorly understood. OBJECTIVE This study aims to explore the therapeutic potential and molecular mechanisms of DMY in the context of liver regeneration impairment in ALD. METHODS The classic Lieber-DeCarli alcohol liquid diet was used to establish an ALD model in vivo. DMY (75 and 150 mg/kg/day) and silybin (200 mg/kg) were administered for 7 weeks to assess the hepatoprotective effects of DMY. First, biochemical markers and liver histopathology were used to evaluate liver inflammation and steatosis in ALD mice. Second, we explored the potential molecular mechanisms by which DMY improves ALD through serum untargeted metabolomics, hepatic transcriptomics, and single-cell sequencing data. Furthermore, in vivo and in vitro experiments, combined with Western blotting, dual-luciferase reporter assays, and immunofluorescence, were conducted to elucidate the protective mechanisms underlying DMY's effects on ALD. RESULTS In vivo studies showed that DMY significantly ameliorated ALT/AST abnormalities, liver inflammation, and steatosis in ALD mice. Multi-omics and bioinformatics analyses revealed that DMY may exert its anti-ALD effects by regulating the miR-155-5p/SIRT1/VDAC1 pathway, thereby mitigating cellular senescence. Notably, knockdown of miR-155 provided partial protection against ethanol-induced liver damage. Additionally, clinical ALD samples and in vivo and in vitro experiments further confirmed that excessive alcohol exposure induces the production of miR-155-5p in liver Kupffer cells. miR-155-5p targets and inhibits SIRT1, promoting the expression of mitochondrial VDAC1, leading to mitochondrial DNA leakage, thereby accelerating hepatocyte senescence and inflammation. However, DMY improved the disruption of the miR-155-5p/SIRT1/VDAC1 pathway and hepatocyte senescence, thereby restoring liver regenerative function and exerting anti-ALD effects. CONCLUSION In this study, we provide the first evidence that DMY improves liver inflammation and cellular senescence by regulating the miR-155-5p/SIRT1/VDAC1 positive feedback loop, promoting liver regeneration to improve ALD. In summary, our work provides important research evidence and theoretical support for DMY as a promising candidate drug for the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Qing Ma
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China; School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shuo Huang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Mei-Ya Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qi-Han Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Fang-Ming Chen
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chun-Lan Hong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Hong-Hao Yan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiang Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Kang-Lu Zhao
- Zhejiang Rehabilitation Medical Center, Rehabilitation Hospital Area of the Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou Zhejiang, China; The Fourth Affiliated Hospital Zhejiang University, School of Medicine, Yiwu Zhejiang, China
| | - Yu Du
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Jin-Kai Zhao
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Li-Qin Zhou
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Da-Yong Lou
- Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| | - Chang-Yu Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ping Qiu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310000, China.
| |
Collapse
|
27
|
Li W, Liu L, Qian S, Chen Y, Ya R, Ma N, Hao Y, Ge S, Zhang X, Yang L, He Y. Hepatic microRNA-320 restrains ferroptosis to mitigate acute-on-chronic alcohol-induced liver injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167748. [PMID: 40015214 DOI: 10.1016/j.bbadis.2025.167748] [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: 11/04/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Alcohol-associated Liver Disease (ALD) is one of the major chronic liver diseases worldwide and has high mortality and high incidence rate. microRNA-320 (miR-320), a highly conserved and widely expressed miRNA, has been reported to be involved in lipid metabolism; however, whether miR-320 affects the progression of ALD remains unclear. In this study, we demonstrated that hepatic miR-320 was significantly downregulated in chronic-plus-binge alcohol-fed mice. Interestingly, such downregulation might accelerate ALD progression as evidenced that hepatocyte-specific miR-320 deficient mice displayed higher susceptibility to acute-on-chronic alcohol feeding-induced steatosis and inflammation. Moreover, restoration of hepatic miR-320 ameliorated acute-on-chronic alcohol-induced hepatocyte damage and steatosis. Mechanistically, miR-320 inhibited alcohol-induced ferroptosis by targeting Transferrin Receptor 1 (TFRC) to suppress iron accumulation. Moreover, silencing of Tfrc in hepatocytes attenuated ethanol-induced iron accumulation, thus inhibiting ferroptosis and ultimately mitigating ALD. Taken together, these findings suggest that miR-320 plays an important role in limiting ALD progression via inhibiting ferroptosis, providing a therapeutic target for the treatment of ALD.
Collapse
Affiliation(s)
- Wenjun Li
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Li Liu
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Shengying Qian
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yingfen Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ru Ya
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ningning Ma
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yawen Hao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shujun Ge
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Xiaoxiao Zhang
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China
| | - Liu Yang
- Department of Pharmacology, School of Basic Medical Science, Nanjing Medical University, Nanjing, China.
| | - Yong He
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
28
|
Staller DW, Panigrahi SS, Jayasinghe YP, Dong Y, Mahto S, Kumar V, Ronning DR, Mahato RI. A novel phosphodiesterase inhibitor for the treatment of chronic liver injury and metabolic diseases. Hepatology 2025; 81:1288-1303. [PMID: 38950389 PMCID: PMC11922481 DOI: 10.1097/hep.0000000000000999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND AND AIMS Chronic liver disease leads to ~2 million deaths annually. Cyclic AMP (cAMP) signaling has long been studied in liver injury, particularly in the regulation of fatty acid (FA) β-oxidation and pro-inflammatory polarization of tissue-resident lymphocytes. Phosphodiesterase 4B inhibition has been explored as a therapeutic modality, but these drugs have had limited success and are known to cause significant adverse effects. The PDE4 inhibitor 2-(4-([2-(5-Chlorothiophen-2-yl)-5-ethyl-6-methylpyrimidin-4-yl]amino)phenyl)acetic acid) (known as A-33) has yet to be explored for the treatment of metabolic diseases. APPROACH AND RESULTS Herein, we evaluated the efficacy of A-33 in the treatment of animal models of alcohol-associated liver disease and steatotic liver disease. We demonstrated that A-33 effectively ameliorated the signs and symptoms of chronic liver disease, resulting in significant decreases in serum alanine aminotransferase and aspartate aminotransferase levels, decreased overall fat and collagen deposition in the liver, decreased intrahepatic triglyceride concentrations, and normalized expression of genes related to β-oxidation of fatty acids, inflammation, and extracellular matrix deposition. We also designed and synthesized a novel analog of A-33, termed MDL3, which inhibited both phosphodiesterase 4B and PDE5A and was more effective in ameliorating pathophysiological signs and symptoms of liver injury and inflammation. In addition, MDL3 re-sensitized obese mice to glucose and significantly inhibited the pathological remodeling of adipose tissue, which was not observed with A-33 administration. CONCLUSIONS In conclusion, we synthesized and demonstrated that MDL3, a novel phosphodiesterase 4B and PDE5A inhibitor, presents a promising avenue of exploration for treating chronic liver disease.
Collapse
Affiliation(s)
- Dalton W. Staller
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanjali S. Panigrahi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yahani P. Jayasinghe
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yuxiang Dong
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sohan Mahto
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Virender Kumar
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Donald R. Ronning
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- UNMC Center for Drug Design and Innovation, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ram I. Mahato
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
29
|
Zhu YQ, Wang LL, Li ZH, Qian SS, Xu Z, Zhang J, Song YH, Pan XS, Du N, Abou-Elnour A, Tay LJ, Zhang JR, Li MX, Shen YX, Huang Y. Acid-sensing ion channel 1a promotes alcohol-associated liver disease in mice via regulating endoplasmic reticulum autophagy. Acta Pharmacol Sin 2025; 46:989-1001. [PMID: 39592735 PMCID: PMC11950321 DOI: 10.1038/s41401-024-01423-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Alcohol-associated liver disease (ALD) is a hepatocyte dysfunction disease caused by chronic or excessive alcohol consumption, which can lead to extensive hepatocyte necrosis and even liver failure. Currently, the pathogenesis of ALD and the anti-ALD mechanisms have not been fully elucidated yet. In this study, we investigated the effects of endoplasmic reticulum autophagy (ER-phagy) in ALD and the role of acid-sensing ion channel 1a (ASIC1a) in ER stress-mediated ER-phagy. A mouse model of ALD was established using the Gao-Binge method and the AML12 cell line treated with alcohol was used as an in vitro model. We showed that ASIC1a expression was significantly increased and ER-phagy was activated in both the in vivo and in vitro models. In alcohol-treated AML12 cells, we showed that blockade of ASIC1a with PcTx-1 or knockdown of ASIC1a reduced alcohol-induced intracellular Ca2+ accumulation and ER stress. In addition, inhibition of ER stress with 4-PBA reduced the level of ER-phagy. Furthermore, knockdown of the ER-phagy receptor family with sequence similarity 134 member B (FAM134B) alleviated alcohol-triggered hepatocyte injury and apoptosis. In conclusion, this study demonstrates that alcohol activates ER stress-induced ER-phagy and liver injury by increasing ASIC1a expression and ASIC1a-mediated Ca2+ influx, providing a novel strategy for the treatment of ALD.
Collapse
Affiliation(s)
- Yue-Qin Zhu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
- Office of Clinical Trial Institution, Anhui Provincial Cancer Hospital, Hefei, 230031, China
| | - Li-Li Wang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zi-Hao Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Shi-Shun Qian
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Zhou Xu
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Jin Zhang
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Yong-Hu Song
- The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Xue-Sheng Pan
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Na Du
- Department of Pharmacy, Shanghai Songjiang District Central Hospital, Shanghai, 201600, China
| | - Amira Abou-Elnour
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Lynn Jia Tay
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Jing-Rong Zhang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Meng-Xue Li
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yu-Xian Shen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Yan Huang
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
30
|
Tang XH, Pesola G, Chen Q, Miller D, Nagy LE, McMullen MR, Schwartz RE, Tsoy S, Lim C, Chikara S, Gross SS, Trasino SE, Gudas LJ, Melis M. Ethanol causes rapid decreases in the hepatic retinoid levels shaping the early steps of alcohol-associated liver disease. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:754-770. [PMID: 40016864 PMCID: PMC12014373 DOI: 10.1111/acer.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Chronic alcohol drinking causes hepatic vitamin A (retinoids and derivatives) decreases, which correlate with the progression and severity of alcohol-associated liver disease (ALD). However, the effects of short-term ethanol (EtOH) intake on liver retinoids and ALD are still undefined. METHODS Using high-performance liquid chromatography and high-performance liquid chromatography coupled with tandem mass spectrometry (HPLC, HPLC-MS/MS), and molecular biology techniques in mice and cultured human hepatocytes, we investigated the temporal EtOH effects on retinoids and ALD. RESULTS In female and male mice, acute EtOH intake caused hepatic retinol (ROL) and retinyl palmitate (RP) decreases within hours, whereas it did not significantly change the retinoic acid (RA) levels, and those of the RA catabolism metabolite, 4-oxo-RA. After EtOH withdrawal, the liver recovered the ROL and RP levels within 48 h, whereas RA and 4-oxo-RA levels remained almost undetectable by this time point. Compared with control diet-fed mice, hepatic ROL and RP levels remained decreased in the 10-day and 3-week-long EtOH treatments, while retinyl oleate and linoleate increased. Interestingly, some of the RA signaling receptors, Rarβ, along with Cyp26a1, revealed dramatic transcript increases during the 10-day-long experiments that attenuated over time (up to 8 weeks), reflecting impaired RA signaling. Our work also showed that primary human hepatocytes serve as a model to better define the role of EtOH in retinoid biology. CONCLUSIONS This work reveals that acute and short-term exposures to EtOH disrupt retinoid homeostasis, identifying key events in the early pathogenesis of ALD.
Collapse
Affiliation(s)
- Xiao-Han Tang
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Glen Pesola
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Qiuying Chen
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Dawson Miller
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Laura E. Nagy
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Megan R. McMullen
- Department of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, USA
| | - Robert E. Schwartz
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Sergey Tsoy
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Christine Lim
- Division of Gastroenterology & Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Shireen Chikara
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Steven S. Gross
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Steven E. Trasino
- Department of Nutrition and Public Health, Hunter College, City University of New York, New York, NY, USA
| | - Lorraine J. Gudas
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Marta Melis
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
31
|
Xu H, Wu Z, Qin J, Li X, Xu F, Wang W, Zhang H, Yin H, Zhu S, Zhang W, Yang Y, Wei Y, Gao L, Liu J, Gao Y, Zheng MH, Zhou H, Qi T, Chen J, Gao Y, Zuo L, Chen J, Liangpunsakul S, Li J, Wang H. Stressed hepatocyte sustains alcohol-associated hepatitis progression by producing leukocyte cell-derived chemotaxin 2. Gut 2025:gutjnl-2024-334318. [PMID: 40139745 DOI: 10.1136/gutjnl-2024-334318] [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: 11/13/2024] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Neutrophil infiltration and hepatocyte damage are indispensable hallmarks in alcohol-associated hepatitis (AH), yet the underlying crosstalk between neutrophils and hepatocytes and its role in AH pathogenesis remain unclear. OBJECTIVE We investigate the regulatory role of leucocyte cell-derived chemotaxin 2 (LECT2) in hepatocyte-neutrophil interaction and its impact on AH progression. DESIGN We used bulk and single-cell RNA sequencing to identify hepatocyte-secreted factors targeting neutrophils. We analysed serum and liver samples from AH patients and employed genetically modified mice alongside in vitro studies. RESULTS RNA-sequencing analysis identified several neutrophil chemokines that are elevated in hepatocytes from AH patients, including LECT2 whose role in AH remains largely unknown. AH patients exhibited increased levels of LECT2 in hepatocytes, positively correlating with the severity of AH. Ethanol-fed mice also exhibited elevated liver LECT2, which was abolished by inhibiting endoplasmic reticulum stress. Functional studies revealed that ethanol-induced liver injury was ameliorated in Lect2-deficient mice but was exacerbated in mice with hepatic overexpression of Lect2. Furthermore, LECT2 exacerbated ethanol-induced liver injury by promoting reactive oxygen species (ROS) through its interaction with prohibitin 2 (PHB2), a neutrophil membrane protein. By directly binding to PHB2, LECT2 disrupts the stable structure of PHB1/PHB2 heterodimerisation, consequently leading to PHB2 degradation, ROS accumulation, neutrophil activation and neutrophil extracellular trap formation. Moreover, therapeutic intervention of LECT2 via Lect2 shRNA ameliorated ethanol-induced liver injury. CONCLUSION Our studies identified a novel vicious cycle between neutrophils and hepatocytes through the LECT2-PHB2 interaction, presenting a promising therapeutic intervention by targeting LECT2 to mitigate AH in patients.
Collapse
Affiliation(s)
- Honghai Xu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zihao Wu
- Department of Geriatrics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
- Anhui Key Laboratory of Geriatric Immunology and Nutrition Therapy, Hefei, Anhui, China
| | - Jiangfeng Qin
- Department of Infectious Diseases, the People's Hospital of Xuancheng City, Xuancheng, Anhui, China
| | - Xutong Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feng Xu
- Department of Intensive Care Unit & Central Laboratory, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Wei Wang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hui Zhang
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - HeHe Yin
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
| | - Shiwei Zhu
- Department of Pathology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenzhe Zhang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China
| | - Yuanru Yang
- Department of Blood Transfusion, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, Beijing, China
| | - Yuanyuan Wei
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Long Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jiatao Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yufeng Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, Zhejiang, China
| | - Haoxiong Zhou
- Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tingting Qi
- Hepatology Unit, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, Guangdong, China
| | - Jinjun Chen
- State Key Laboratory of Organ Failure Research; Key Laboratory of Infectious Diseases Research in South China, Ministry of Education; Guangdong Provincial Key Laboratory for Prevention and Control of Major Liver Diseases, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Guangdong Institute of Hepatology, Guangdong Provincial Research Center for Liver Fibrosis Engineering and Technology, Guangzhou, Guangdong, China
- Hepatology Unit, Department of Infectious Diseases, Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yanhang Gao
- Department of Hepatology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-Products, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Anhui Center for Surveillance of Bacterial Resistance, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
32
|
Fu Z, Zhou J, Pan H, Yang S, Pan Z, Shen Y, Yao J, Hu J. QingGan LiDan capsules improved alcoholic liver injury by regulating liver lipid transport and oxidative stress in mice. Front Pharmacol 2025; 16:1575280. [PMID: 40206071 PMCID: PMC11979125 DOI: 10.3389/fphar.2025.1575280] [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: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Background The QingGan LiDan capsule (QGLD) consists of five traditional Chinese herbs, which have been used for hepatobiliary diseases such as jaundice. However, the effects and mechanisms by which QGLD prevent alcoholic liver diseases (ALD) remain unknown. Aim of the study Investigate the therapeutic potential of QingGan Lidan capsule (QGLD) in alleviating alcohol-induced liver injury. Materials and Methods Acute alcoholic liver injury model and chronic and Binge ethanol Feeding Model (NIAAA) model were established. Mice were administered QGLD (360, 720, 1,440 mg/kg) or vehicle. Liver function indicators (ALT, AST), serum lipid (TC, TG), antioxidant markers (SOD, GSH, MDA), lipid metabolism/transport genes relative expression levels, liver and ileal villus morphology were analyzed. Network pharmacology analysis was also performed to identify potential targets and pathways of QGLD. Results QGLD reduced serum ALT, AST, hepatic TC, TG, and lipid droplet accumulation in both models. It upregulated antioxidant enzymes (SOD, GSH) and downregulated MDA. QGLD regulated the mRNA levels of genes related to the NRF2/KEAP1 pathway and lipid transport. Network pharmacology identified 221 potential targets. Conclusion QGLD mitigates alcohol-induced liver injury by reducing lipid accumulation, regulating lipid transport and enhancing antioxidant capacity. This supports its potential application in ALD management.
Collapse
Affiliation(s)
- Zhiwen Fu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Jiangxi Conba Traditional Chinese Medicine Co.,Ltd, Shangrao, China
| | - Jiafeng Zhou
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
| | - Hongye Pan
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
- Zhejiang Key Laboratory of Major TCM Cultivation and TCM Innovation, Hangzhou, China
| | - Song Yang
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
| | - Zhenzhen Pan
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
| | - Yujia Shen
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
| | - Jianbiao Yao
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
- Zhejiang Key Laboratory of Major TCM Cultivation and TCM Innovation, Hangzhou, China
| | - Jiangning Hu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Conba Pharmaceutical Co.,Ltd, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Pharmaceutical Technology, Hangzhou, China
- Zhejiang Key Laboratory of Major TCM Cultivation and TCM Innovation, Hangzhou, China
| |
Collapse
|
33
|
Chandrashekar DV, Jagadeesan N, Abdullah T, Chang R, Steinberg RA, Sanchez F, Khal E, Yang J, Cribbs DH, Han D, Sumbria RK. Effect of chronic alcohol feeding using the Lieber-DeCarli diet on Alzheimer's disease pathology in Tg2576 mice. Front Aging Neurosci 2025; 17:1526571. [PMID: 40196176 PMCID: PMC11973299 DOI: 10.3389/fnagi.2025.1526571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/25/2025] [Indexed: 04/09/2025] Open
Abstract
Background Chronic alcohol drinking is a modifiable risk factor for Alzheimer's disease (AD), but underlying mechanisms remain poorly understood. Most studies of alcohol feeding to AD mice have utilized young mice and delivered alcohol in drinking water without controlling nutritional intake. Objective To study the impact of Lieber-DeCarli (LDC) liquid alcohol diet, which balances nutritional intake, on AD pathology of aged Tg2576 and wild-type (WT) mice, which is unexplored. Methods 13-month-old male and female Tg2576 or WT mice were fed LDC diet (5% ethanol or control) for six weeks (n = 11-13/group). Exploration (open-field test) and spatial reference memory (Y-maze test) were assessed after six weeks, and brains and livers were studied for Aβ levels, and Aβ synthesis and transport proteins (APP and LRP-1). Neuroinflammation, blood-brain barrier function, and synaptic health were studied using immunoassays. Results LDC alcohol feeding significantly reduced survival (p < 0.05) and spatial memory (p < 0.05) in Tg2576 mice, but not in WT mice. Alcohol feeding increased (p < 0.001) insoluble endogenous mouse Aβ1-42 and reduced microgliosis (p < 0.05) in Tg2576 mice brains, but not in WT mice. LDC alcohol feeding to Tg2576 mice caused mild liver injury, and important amyloidosis-relevant hepatic proteins (LRP-1 and APP) were largely unaltered. However, brain Aβ and microgliosis were positively correlated (p < 0.05) with serum alanine aminotransferase, a marker of liver injury, in Tg2576 mice. Conclusion Chronic alcohol intake, resulting in mild liver injury, caused modest but significant AD-relevant changes in aged Tg2576 mice, which correlated with liver injury; the latter suggests significant liver-brain crosstalk in an AD model of moderate alcohol intake.
Collapse
Affiliation(s)
- Devaraj V. Chandrashekar
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Nataraj Jagadeesan
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Tamara Abdullah
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Rudy Chang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - Ross A. Steinberg
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Frankey Sanchez
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Elias Khal
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Joshua Yang
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
| | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Derick Han
- School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Rachita K. Sumbria
- Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy, Chapman University, Irvine, CA, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
34
|
Rodrigo-Torres D, Kilpatrick AM, Ferreira-Gonzalez S, Aird RE, Atkinson SR, Gadd VL, Man TY, Tyson LD, Dhondalay GKR, Vergis N, Arteel GE, Thursz MR, Martinez-Gili L, Forbes SJ. Longitudinal paired liver biopsies and transcriptome profiling in alcohol-associated hepatitis reveal dynamic changes in cellular senescence. Gut 2025:gutjnl-2024-334094. [PMID: 40122595 DOI: 10.1136/gutjnl-2024-334094] [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: 10/14/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND AND AIMS Alcohol-associated hepatitis (AH) is an acute form of alcohol-related liver disease (ALD) with high mortality rate. AH is histologically characterised by cellular processes, including steatosis, inflammation and cell death. Apoptosis is the most studied form of cell death in AH; however, the role of cellular senescence, another response to cellular injury, in AH is unknown. Here, we explore the mechanisms of ALD pathophysiology and describe the role of senescence in AH. METHODS We performed RNA sequencing and bioinformatics analysis of 0- and 28-day transjugular liver biopsies (n=65) from patients with AH participating in the IL-1 Signal Inhibition In Alcoholic Hepatitis (ISAIAH) clinical trial. Additional bioinformatics reanalysis of existing AH transcriptomic datasets was conducted to confirm our findings. We also performed multiomic analysis of an in vitro model of AH with ethanol-treated hepatocytes overexpressing ethanol-metabolising enzymes. RESULTS Our longitudinal analysis revealed that senescence and inflammation were reduced at transcriptomic level following AH resolution; the expression of hepatocyte markers was increased. We identified two senescence-associated protein complexes, cytochrome c oxidase and the proteasome, which may act as senescence-induction mechanisms. We confirmed that senescence markers and pathways were increasingly expressed in hepatocytes as ALD progressed towards AH; this was partially reversed following AH resolution. Our in vitro model revealed that ethanol directly induces senescence and was dependent on ethanol metabolism. CONCLUSIONS Our results suggest a possible pathogenic role for senescence in AH and indicate cellular senescence as a potential therapeutic target in early ALD to limit AH severity.
Collapse
Affiliation(s)
- Daniel Rodrigo-Torres
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
| | - Sofia Ferreira-Gonzalez
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Rhona E Aird
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Stephen Rahul Atkinson
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
| | - Victoria L Gadd
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Tak Yung Man
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Luke D Tyson
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
| | - Gopal Krishna R Dhondalay
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Nikhil Vergis
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
| | - Gavin E Arteel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mark R Thursz
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
| | - Laura Martinez-Gili
- Division of Digestive Diseases, Department of Metabolism Digestion and Reproduction, Imperial College London, London, UK
- Section of Bioinformatics, Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
35
|
Xie H, Zhang P, Yang S, Du J, Ren Y, Gao X, Li N, Yang T, Ma Y, Hou X. Myeloid-derived MANF ameliorates ethanol-induced liver injury by enhancing microRNA-223 expression. J Gastroenterol 2025:10.1007/s00535-025-02240-0. [PMID: 40111540 DOI: 10.1007/s00535-025-02240-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Myeloid cells play a pivotal role in the pathogenesis of alcoholic liver disease (ALD), yet the mechanisms regulating their function and specific contributions to ALD remain inadequately understood. This study aims to investigate the role of mesencephalic astrocyte-derived neurotrophic factor (MANF) in the development of ALD. METHODS Myeloid-specific Manf knockout mice and wild-type controls were fed an ethanol-based diet for 10 days, followed by a single ethanol binge. Hepatic MANF levels, along with the correlation between MANF and inflammatory factors in patients with alcoholic hepatitis, were analyzed using the GSE28619 dataset. RESULTS Our study demonstrated that myeloid MANF expression in the liver was upregulated following chronic-plus-binge ethanol exposure. Deletion of the Manf gene in myeloid cells, including neutrophils, exacerbated ethanol-induced liver injury, steatosis, neutrophil infiltration, and reactive oxygen species production. Mechanistic analysis revealed that MANF promotes neutrophil miR-223 expression, a key anti-inflammatory factor in these cells. MANF enhances miR-223 transcription by increasing the expression of the transcription factor PU.1 via p38 mitogen-activated protein kinase signaling. In addition, hepatic MANF levels were elevated in patients with alcoholic hepatitis and correlated with IL-6, IL-1β, and phagocytic oxidase (phox) p47phoxlevels. CONCLUSION Myeloid-derived MANF mitigates alcohol-induced liver injury by upregulating the neutrophilic p38-PU.1-miR-223 axis.
Collapse
Affiliation(s)
- Huiyuan Xie
- Department of Laboratory Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, China
| | - Pingping Zhang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shanru Yang
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Jia Du
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yan Ren
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Xianxian Gao
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Tao Yang
- Department of General Surgery, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yang Ma
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Xin Hou
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
36
|
Pietrantonio AF, Urian RA, Hardy DB, Allman BL, Willmore KE. Hyperactivity in male and female mice manifests differently following early, acute prenatal alcohol exposure and mild juvenile stress. Front Behav Neurosci 2025; 19:1501937. [PMID: 40170739 PMCID: PMC11958967 DOI: 10.3389/fnbeh.2025.1501937] [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: 09/25/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Chronic prenatal alcohol exposure (PAE) and severe juvenile stress independently contribute to hyperactive and depressive behavioral phenotypes, with their combination exacerbating these effects. However, while chronic PAE and traumatic juvenile stress are well-studied, little is known about the impact of early, acute PAE and mild juvenile stress on hyperactivity and depression. This knowledge gap is clinically relevant, as these milder early-life insults are common in Western societies. Here, we provide the first investigation into the effects of early, acute PAE and juvenile sub-chronic, unpredictable, mild stress (SUMS)-both independently and in combination-on hyperactivity and depressive-like behaviors in mice throughout the lifespan. Methods We assessed hyperactivity through movement-related measures (i.e., distance traveled, thigmotaxis, and rearing), whereas depressive-like behaviors were evaluated using the u-shaped two-choice field and forced swim tests. Behavioural testing was performed on equivalent numbers of male and female offspring and repeated at juvenile, adolescent, and adult timepoints to enable assessment of sex and age effects. Results Neither early, acute PAE, juvenile SUMS, nor their combination induced depressive-like behaviors at any age; findings in contrast to the more severe chronic PAE and stress insults used in previous studies. However, these milder early-life insults did result in various hyperactivity phenotypes in both the male and female offspring. For example, juvenile SUMS had the strongest impact on hyperactive behaviors across both sexes, but only the adolescent females exhibited increased emotionality-associated activity. Moreover, early, acute PAE-both alone and in combination with juvenile SUMS significantly increased movement during adolescence and adulthood exclusively in male offspring. Discussion Thus, our collective findings not only indicate that early, acute PAE and juvenile SUMS influence hyperactivity in a sex- and age-dependent manner, but also highlight that their influence on hyperactive and depressive phenotypes do not simply mirror those of the more severe early-life insults. Given the potential prevalence of early, acute alcohol exposure and juvenile stress in Western society, further research is warranted to fully understand their long-term behavioral consequences.
Collapse
Affiliation(s)
- Amy F. Pietrantonio
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Raluca A. Urian
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
| | - Daniel B. Hardy
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Department of Obstetrics and Gynecology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Children’s Health Research Institute, London, ON, Canada
| | - Brian L. Allman
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Children’s Health Research Institute, London, ON, Canada
| | - Katherine E. Willmore
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, ON, Canada
- Children’s Health Research Institute, London, ON, Canada
| |
Collapse
|
37
|
Zhang JP, Li ML, Ren DL, Yang QL, Mao J, Liu SP. High-throughput screening of probiotics in fermented foods and their potential application in alleviating alcohol-induced damage. Food Funct 2025; 16:2564-2576. [PMID: 40035662 DOI: 10.1039/d4fo05735j] [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: 03/06/2025]
Abstract
This study is dedicated to screening strains with high ethanol degradation capabilities from various fermented products, aiming to mitigate the adverse health effects of alcohol consumption. The research began with the isolation of target strains from pickled vegetables, sauerkraut, enzymes, and spoiled yellow wine, and then established a high-throughput screening method based on the color reaction of WST-8 and NADH. By utilizing automated liquid handling and imaging technologies, rapid transfer and cultivation of single colonies, as well as efficient assessment of ethanol degradation activity, were achieved. The selected strains were rescreened, and their ethanol tolerance, and the ability to degrade ethanol and acetaldehyde were comprehensively evaluated. Ultimately, the potential protective effects of the target strains against alcoholic liver injury were assessed through animal experiments. The study successfully constructed a high-throughput screening platform, providing an effective technical strategy for the rapid identification and evaluation of strains capable of degrading ethanol.
Collapse
Affiliation(s)
- Jun-Pu Zhang
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ming-Liang Li
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Dong-Liang Ren
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qi-Lin Yang
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jian Mao
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
| | - Shuang-Ping Liu
- School of Food Science and Technology, Jiangnan university, Wuxi, Jiangsu 214122, China.
- National Engineering Research Centre of Cereal Fermentation and Food Biomanufacturing, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Shaoxing Key Laboratory of Traditional Fermentation Food and Human Health, Jiangnan University (Shaoxing) Industrial Technology Research Institute, Shaoxing, Zhejiang 312000, China
| |
Collapse
|
38
|
Ma X, Han R, Wang J, Zhang B, Ruan M, Zhao W, Zhang J. Novel NIR fluorescent probe based on BODIPY for diagnosis and treatment evaluation of alcoholic liver disease via visualizing HClO fluctuation. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 328:125497. [PMID: 39615092 DOI: 10.1016/j.saa.2024.125497] [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: 07/22/2024] [Revised: 11/20/2024] [Accepted: 11/24/2024] [Indexed: 12/10/2024]
Abstract
Alcoholic liver disease (ALD) is gradually becoming common due to the increasing number of drinkers worldwide, which is a serious threat to human physical and mental health. In the process of ALD, it is often accompanied by the occurrence of inflammation, which induce high expression of reactive oxygen species including HClO. In this work, we successfully fabricated a NIR fluorescent probe BDP-ENE-Fur-HClO for real-time imaging alcoholic liver disease via tracing HClO. The probe displayed good sensitivity and specificity, rapid recognition speed and NIR emitting (700 nm) for detection of HClO in vitro. Based on the remarkable performances, probe was capable of tracing endogenous/exogenous HClO in living cells without interference from other ROS as well as in ALD cell model. Additionally, probe could monitor the exogenous HClO in normal mice and high expression of HClO in the peritonitis mice, that accomplishing the diagnosis of inflammation. What's more, one simulated hazardous drinking ALD mice model and simulated excessive drinking (a type of alcohol use disorder) ALD mice model were developed, probe could image the alcoholic liver injury of mice by monitoring the HClO fluctuation in ALD mice, which affording a valid instrument for the diagnosis of ALD. Ultimately, after hepatoprotective drug administrating to the models, probe could triumphantly evaluate the treatment effect of drug on ALD.
Collapse
Affiliation(s)
- Xiaoteng Ma
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China
| | - Ruiqi Han
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China
| | - Jiamin Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, State Key Laboratory of Antiviral Drugs, Henan University, Kaifeng 475004, PR China.
| | - Bo Zhang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China
| | - Minghao Ruan
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China
| | - Weili Zhao
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China; School of Pharmacy, Institutes of Integrative Medicine, Fudan University, Shanghai 201203, PR China.
| | - Jian Zhang
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Nanoscience and Materials Engineering, Henan University, Kaifeng 475004, PR China.
| |
Collapse
|
39
|
Chen F, Zhou Z, Fu J, Gao C. Selenomethionine Alleviates Alcohol-Induced Liver Injury by Inhibiting Ferroptosis. Dig Dis Sci 2025:10.1007/s10620-025-08960-w. [PMID: 40029572 DOI: 10.1007/s10620-025-08960-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND AND AIM Selenomethionine (Se-Met) has been reported to reduce oxidative stress (OS) and hepatic injury; however, its role in alcoholic liver disease (ALD), particularly with ferroptosis, remains poorly understood. METHODS Oxidative stress was induced using ethanol, and ferroptosis was inhibited with ferrostatin-1 (fer-1) in L-02 and LX2 cell lines, respectively. The effects of Se-Met on alcohol-induced hepatocyte damage were evaluated in vitro by examining cell viability, lipid peroxidation, and the level of key ferroptosis-associated markers. In vivo, the interaction between Se-Met and ferroptosis was examined via an ALD mouse model through analyses of liver histology, lipid peroxidation, liver function, and ferroptosis-related indices. RESULTS In vitro and in vivo experiments indicated that both Se-Met and fer-1 have a significant protective role against alcohol-induced hepatocyte death and liver injury. Treatment with Se-Met or fer-1 can promote hepatocyte proliferation, ameliorate the typical symptoms of lipid peroxidation (e.g., glutathione depletion, superoxide dismutase enzyme activity, intracellular reactive oxygen species (ROS) level, malonaldehyde (MDA) content), and altered the expression of ferroptosis-related factors. Moreover, the findings indicated that the administration of Se-Met or fer-1 significantly ameliorated the pathological alterations and improved liver function indices associated with alcohol-induced liver damage in mice. These effects may collectively suppress the deleterious impact of ethanol on hepatic tissue. CONCLUSION This study concluded that the ferroptosis pathway regulated alcohol-induced hepatocyte injury. The administration of selenomethionine protects ALD by partially inhibiting the ferroptosis pathway, providing a novel therapeutic approach for ALD.
Collapse
Affiliation(s)
- Feng Chen
- Division of Gastroenterology, The Seventh Affiliated Hospital of Sun Yat-Sen University, No.628, Zhenyuan Road, Shenzhen, 518107, China.
| | - Zhenhua Zhou
- Department of Neurology, The Air Force Hospital of Southern Theater Command, PLA, Guangzhou, 510062, China
| | - Jinxian Fu
- Division of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Chang Gao
- Division of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| |
Collapse
|
40
|
Wang Y, Yuan M, Li S, Tang J, Wan Y, Liang X, Guo Y, Guo L. Multifunctional Liposome Delivery System Based on Ursodeoxycholic Acid Sodium for the Encapsulation of Silibinin and Combined Treatment of Alcoholic Liver Injury. Mol Pharm 2025; 22:1480-1497. [PMID: 39931930 DOI: 10.1021/acs.molpharmaceut.4c01197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Alcohol liver disease (ALD) is a chronic liver disorder resulting from long-term heavy alcohol consumption. The pathogenesis of ALD is multifactorial, and existing therapeutic agents primarily target specific aspects of the disease while presenting significant side effects, including drug-induced liver injury and hepatobiliary disease. Silibinin (SLB) has attracted widespread attention for its hepatoprotective effects and favorable safety profile. However, inherent limitations associated with SLB, such as poor solubility and bioavailability, have significantly limited its clinical application. Drug delivery systems, including liposomes, offer promising potential for the delivery of hydrophobic drugs. However, the selection of an appropriate delivery vehicle requires optimization. Ursodeoxycholic acid sodium (UAS) serves as a promising alternative to cholesterol in liposomal formulations, offering a potential strategy to mitigate the health risks associated with cholesterol. In this study, UAS was employed as the liposomal membrane material to prepare a UAS liposome loaded with SLB (SUL), and its efficacy and mechanism of action in alcoholic-induced liver injury were subsequently evaluated. The experimental results demonstrated that SUL exhibited a uniform particle size distribution, good stability, and an effective release profile in vitro. Following oral administration, SUL effectively inhibited alcohol-induced liver damage, oxidative stress, and fat accumulation. In addition, SUL regulated the expression of the kelch-1ike ECH- associated protein l (Keap1), nuclear factor erythroid 2-related factor 2 (Nrf2), and heme oxygenase 1 (HO-1) proteins, thereby exerting antioxidative stress effects. Furthermore, it also modulated apoptosis-related factors, including B-cell lymphoma-2 (Bcl-2), BCL-2-associated X (Bax), cysteinyl aspartate specific proteinase-3 (Caspase-3), and cleaved caspase-3, to mitigate hepatocyte apoptosis. In summary, SUL demonstrates enhanced therapeutic efficacy against ALD, offering a novel approach for the clinical application of SLB in the prevention and treatment of ALD.
Collapse
Affiliation(s)
- Yulu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Minghao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Sihui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Jiamei Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Yan Wan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Xue Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Yiping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611100, China
| |
Collapse
|
41
|
Kang J, Park SH, Khanam M, Park SB, Shin S, Seo W. Impact of binge drinking on alcoholic liver disease. Arch Pharm Res 2025; 48:212-223. [PMID: 40035998 DOI: 10.1007/s12272-025-01537-1] [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: 05/30/2024] [Accepted: 02/13/2025] [Indexed: 03/06/2025]
Abstract
Numerous studies have examined the pathophysiological changes induced by chronic alcohol (ethanol) consumption and the underlying mechanisms, while much less attention has been devoted to understanding the health impacts of binge drinking. Binge drinking is defined as the excessive consumption of alcohol within a single drinking episode, and is the typical consumption pattern among young people in Western countries. While most young binge drinkers are not clinically alcohol dependent, binge drinking has emerged as a significant social and public health concern. The circulating alcohol consumed during binge episodes permeates cellular membranes throughout the body, exerting profound effects on multiple organs, and signaling pathways. Regular binge drinking eventually induces hepatic steatosis (fatty liver), initiates acute inflammation, and accelerates neutrophil infiltration, de novo lipogenesis, adipocyte death/lipolysis, and the production of nonoxidative alcohol metabolites, processes that synergize to damage liver tissue and impair liver function. Metabolic abnormalities such as diabetes and obesity can also exacerbate the progression of alcohol-related liver disease among binge drinkers. Several animal models have been developed to evaluate the pathophysiological changes resulting from binge drinking; however, the pathogenesis of binge drinking is not fully understood due to differences in alcohol metabolism between animal models and humans. Thus, given the high prevalence and severe health implications of binge drinking, there is an urgent need for comprehensive experimental and clinical investigations to unravel the associated pathophysiological changes. This review summarizes recent research findings on the impact of binge drinking, specifically focusing on its contributions to alcoholic liver injury.
Collapse
Affiliation(s)
- Jisoo Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seol Hee Park
- Department of Companion Animal Health, Hanyang Women's University, Seoul, 04763, Republic of Korea
| | - Mushira Khanam
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seo Bhin Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sumin Shin
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Wonhyo Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
42
|
Wu Q, Yang D, Liu C, Xu T. Alcohol Plus Additional Risk Factors: Rodent Model of Liver Injury. Semin Liver Dis 2025; 45:81-98. [PMID: 39719149 DOI: 10.1055/a-2490-4278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2024]
Abstract
Alcohol-associated liver disease (ALD), primarily caused by chronic excessive alcohol consumption, is a leading cause of chronic liver disease worldwide. ALD includes alcohol-associated steatotic liver, alcohol-associated hepatitis (AH), fibrosis, cirrhosis, and can even progress to hepatocellular carcinoma (HCC). Existing research indicates that the risk factors of ALD are quite numerous. In addition to drinking patterns, factors such as aldehyde dehydrogenase 2 (ALDH2) deficiency, smoking, medication administration, high-fat diet (HFD), hepatitis virus infection, and disruption of circadian rhythms can also increase susceptibility to ALD. However, there is limited understanding regarding the exacerbation of liver injury by alcohol plus additional risk factors. This review presents rodent models of EtOH + "X," which simulate the synergistic effects of alcohol and additional risk factors in causing liver injury. These models offer a further exploration of the interactions between alcohol and additional risk factors, advancing the simulation of human ALD and providing a more reliable platform for studying disease mechanisms and exploring therapeutic interventions. We summarize the modeling methods, relevant indicators of liver injury, and focus on the targets of the synergistic effects as well as the associated mechanisms.
Collapse
Affiliation(s)
- Qixiang Wu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Dashuai Yang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| | - Chixiang Liu
- Department of Blood Transfusion, Southern Medical University, Nanfang Hospital, Guangzhou, P.R. China
- School of Laboratory and Biotechnology, Institute of Antibody Engineering, Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, Anhui, P.R. China
- Institute for Liver Diseases of Anhui Medical University, Hefei, Anhui, P.R. China
| |
Collapse
|
43
|
Tongtong P, Yujuan S, Ting L, Fangfang Y, Shijia W, Yilun H, Huadong Z, Qiongying Z, Yongping C, Dazhi C. Decoding ferroptosis in alcoholic hepatitis: A bioinformatics approach to hub gene identification. Genomics 2025; 117:111009. [PMID: 39864635 DOI: 10.1016/j.ygeno.2025.111009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/25/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Ferroptosis is associated with alcoholic hepatitis (AH); however, the underlying mechanisms remain unclear. METHODS Changes in iron content and oxidative stress in AH patients and in vivo and in vitro models were analyzed. Iron homeostasis pathways in the livers of patients with AH were investigated using RNA sequencing. AH-associated ferroptosis-related genes (FRGs) were identified using weighted gene co-expression network analysis. Hub genes were identified using machine learning methods, and their diagnostic potential for AH was assessed. The correlation between FRGs and the immune microenvironment was analyzed, and the underlying regulatory mechanism was explored. FRG expression was validated in clinical samples and in vitro and in vivo models. The role of FRGs in AH-related ferroptosis was explored through gene-silencing experiments. RESULTS Significant iron deposits and oxidative stress were detected in clinical samples and in vivo and in vitro AH models. Bioinformatics identified GCLC, NQO1, and ULK1 as key FRGs linked to the immune microenvironment and AH-related pathogenic genes. A nomogram based on these FRGs accurately assessed AH risk, as validated using the calibration curve. A regulatory network involving 154 miRNAs and 136 transcription factors was mapped for FRGs. In AH patients, NQO1 was upregulated in the liver, whereas GCLC and ULK1 were downregulated. Silencing GCLC and ULK1 reduced cell viability and increased oxidative stress and ferroptosis, whereas silencing NQO1 had the opposite effect. CONCLUSIONS Therefore, GCLC, NQO1, and ULK1 are key AH-related FRGs, potentially serving as targets for diagnosing and treating AH.
Collapse
Affiliation(s)
- Pan Tongtong
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Shen Yujuan
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Wuxing District, Huzhou 313000, China
| | - Li Ting
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yi Fangfang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Shijia
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Huang Yilun
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhang Huadong
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhang Qiongying
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou 325035, China
| | - Chen Yongping
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Chen Dazhi
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Hangzhou Medical College, Linan District, Hangzhou 311300, China.
| |
Collapse
|
44
|
Brigagão Pacheco da Silva C, Nascimento-Silva EA, Zaramela LS, da Costa BRB, Rodrigues VF, De Martinis BS, Carlos D, Tostes RC. Drinking pattern and sex modulate the impact of ethanol consumption on the mouse gut microbiome. Physiol Genomics 2025; 57:179-194. [PMID: 39918827 DOI: 10.1152/physiolgenomics.00031.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 01/28/2025] [Indexed: 03/04/2025] Open
Abstract
Gut microbiota impacts host homeostasis and diseases. Chronic plus binge ethanol consumption has been linked to increased injuries than chronic or binge ethanol intake alone. We hypothesized that distinct shapes in gut microbiota composition are induced by chronic, binge, and the association of these treatments, thereby affecting host functions and contributing to sex-based differences in alcohol use disorders. Male and female C57BL/6J mice were submitted to chronic, binge, or chronic plus binge ethanol feeding. DNA was extracted from fecal microbiota, followed by analysis of the V3-V4 region of the 16S rRNA gene and sequencing on an Illumina platform. Gut microbiome analysis was performed using QIIME v2022.2.0. Functional profiling of the gut microbiome was performed using PICRUSt2. Ethanol differentially affected the gut microbiota of female and male mice. Decreased α diversity was observed in male and female mice from the chronic plus binge and chronic groups, respectively. The genera Faecalibaculum, Lachnospiraceae, and Alistipes were identified as major potential biomarkers for gut dysbiosis induced by ethanol consumption. In addition, ethanol-induced gut dysbiosis altered several metabolic pathways. Ethanol consumption modifies the mouse gut microbiome in a drinking pattern- and sex-dependent manner, potentially leading to different susceptibility to ethanol-related diseases. Chronic plus binge ethanol intake induces a more pronounced gut dysbiosis in male mice. Conversely, chronic ethanol is linked to a greater degree of gut dysbiosis in female mice. The changed gut microbiome may be potentially targeted to prevent, mitigate, or treat alcohol use disorders.NEW & NOTEWORTHY Ethanol alters the mouse gut microbiome in a drinking pattern- and sex-dependent manner. Chronic plus binge ethanol intake induces a more severe gut dysbiosis in male mice, whereas chronic ethanol consumption appears to be a more potent inductor of gut dysbiosis in female mice. Ethanol-induced gut dysbiosis alters several pathways linked to metabolism, genetic and environmental information processing, cellular processes, organism systems, and neurological human diseases.
Collapse
Affiliation(s)
| | | | - Lívia Soares Zaramela
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Ruiz Brandão da Costa
- Department of Clinical, Toxicological and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Fernandes Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Bruno Spinosa De Martinis
- Department of Chemistry, Faculty of Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Rita C Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
45
|
Yoon HJ, Doyle MA, Altemus ME, Bethi R, Lago SH, Winder DG, Calipari ES. Operant ethanol self-administration behaviors do not predict sex differences in continuous access home cage drinking. Alcohol 2025; 123:87-99. [PMID: 39218047 PMCID: PMC12034132 DOI: 10.1016/j.alcohol.2024.08.004] [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: 04/12/2024] [Revised: 07/03/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Understanding sex differences in disease prevalence is critical to public health, particularly in the context of alcohol use disorder (AUD). The goal of this study was to understand sex differences in ethanol drinking behavior and define the precise conditions under which sex differences emerge. Consistent with prior work, C57BL/6J females drank more than males under continuous access two-bottle choice conditions. However, using ethanol self-administration - where an operant response results in access to an ethanol sipper for a fixed time period - we found no sex differences in operant response rates or ethanol consumption (volume per body weight consumed, as well as lick behavior). This remained true across a wide range of parameters including acquisition, when the ethanol sipper access period was manipulated, and when the concentration of the ethanol available was scaled. The only sex differences observed were in total ethanol consumption, which was explained by differences in body weight between males and females, rather than by sex differences in motivation to drink. Using dimensionality reduction approaches, we found that drinking behavior in the operant context did not cluster by sex, but rather clustered by high and low drinking phenotypes. Interestingly, these high and low drinking phenotypes in the operant context showed no correlation with those same categorizations in the home cage context within the same animals. These data underscore the complexity of sex differences in ethanol consumption, highlighting the important role that drinking conditions/context plays in the expression of these differences.
Collapse
Affiliation(s)
- Hye Jean Yoon
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Marie A Doyle
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Neurobiology, UMass Chan Medical School, Worcester, MA, USA
| | - Megan E Altemus
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| | - Rishik Bethi
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Sofia H Lago
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
| | - Danny G Winder
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Neurobiology, UMass Chan Medical School, Worcester, MA, USA
| | - Erin S Calipari
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA; Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
46
|
Ritzenthaler JD, Ekuban A, Horsman B, Roman J, Watson WH. Alcohol-induced liver injury is mediated via α4-containing nicotinic acetylcholine receptors expressed in hepatocytes. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025; 49:515-525. [PMID: 39853711 PMCID: PMC11928250 DOI: 10.1111/acer.15533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
BACKGROUND Our previous study demonstrated that alcohol induced the expression of the α4 subunit of nicotinic acetylcholine receptors (nAChRs) in the livers of wild type mice (WT), and that whole-body α4 nAChR knockout mice (α4KO) showed protection against alcohol-induced steatosis, inflammation, and injury. Based on these findings, we hypothesized that hepatocyte-specific α4 nAChRs may directly contribute to the detrimental effects of alcohol on the liver. METHODS Hepatocyte-specific α4 knockout mice (α4HepKO) were generated, and the absence of α4 nAChR was confirmed through PCR of genomic DNA. Female WT and α4HepKO mice were exposed to alcohol in the NIAAA chronic + binge model. After 10 days on the Lieber-DeCarli liquid diet containing 5% (vol/vol) alcohol or isocaloric maltose-dextrin, the mice were gavaged with a single dose of alcohol or isocaloric maltose-dextrin. The mice were euthanized 9 h later and their organs harvested. Additionally, hepatocytes were isolated from WT, α4HepKO, α4floxed, and α4KO mice and exposed to 80 mM alcohol in vitro for 24 h. Steatosis, inflammation, and cell injury were assessed in both liver and isolated hepatocytes. RESULTS In WT mice, alcohol exposure resulted in hepatic steatosis, inflammation, and injury as evidenced by increased liver triglycerides, neutrophil infiltration, and serum concentrations of liver enzymes. All of these responses were markedly lower in α4HepKO mice. mRNA expression of genes involved in lipogenesis (Srebf1, Fasn, and Dgat2) and inflammation (TNFα, Cxcl5, Cxcl1, and Serpine1) were increased in the livers of WT mice exposed to alcohol in vivo and in WT hepatocytes exposed to alcohol in vitro. These changes were not observed in liver or hepatocytes from mice lacking α4 nAChRs. CONCLUSIONS α4 nAChRs expressed in hepatocytes mediate alcohol-associated hepatoxicity. Therefore, the development of therapeutic strategies targeting hepatocyte α4-containing nAChRs could help reduce the burden of ALD.
Collapse
Affiliation(s)
- Jeffrey D. Ritzenthaler
- Division of Pulmonary, Allergy and Critical Care Medicine and the Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA USA
| | - Abigail Ekuban
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Benjamin Horsman
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jesse Roman
- Division of Pulmonary, Allergy and Critical Care Medicine and the Jane & Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, PA USA
| | - Walter H. Watson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
47
|
Wang Z, Liang G, Peng J, Gu Y, Zhang X, Ding C, Yu T, Li Z. Sirtuin 7 Promotes Alcohol-Associated Liver Injury via Modulating Myeloid Cell Chemokine (C-C Motif) Ligand 2 Secretion through the NF-κB Signaling Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:575-588. [PMID: 39746506 DOI: 10.1016/j.ajpath.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 12/07/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
The pathogenesis of alcohol-associated liver disease (ALD) involves ethanol-induced enhancement of gut permeability, bacterial products released from intestine and intrahepatic inflammation, and liver damage. Hepatic macrophages play a crucial role in mediating inflammatory response by alcohol. Sirtuin 7 (SIRT7), a NAD+-dependent type III histone deacetylase, is being recognized as a therapeutic target in various human diseases. Emerging evidence shows that SIRT7 participates in immune regulation, but whether it is involved in ALD remains elusive. In the present study, myeloid cell-specific Sirt7 knockout mice (Lyz2-Sirt7-/-) were used to show that knockout Sirt7 in myeloid cells significantly ameliorated alcohol-induced liver injury, inflammation, and cell infiltration, while only mildly affecting lipid metabolism pathways. Chemokine (C-C motif) ligand 2 (CCL2) was identified as the main target impaired by Sirt7 knockout after alcohol. In vitro studies confirmed that Sirt7 knockout impaired macrophages' ability of CCL2 secretion and monocyte recruiting, and exogenous CCL2 reversed this impairment. At the molecular level, knockout of Sirt7 significantly impaired lipopolysaccharide-induced p65 phosphorylation and nuclear localization. More importantly, the SIRT7 inhibitor 40569 sufficiently decreased alcohol-induced liver injury and hepatic inflammation via preventing CCL2 in vivo. The current data thus uncovered a previously undescribed role of myeloid SIRT7 in mediating ALD via promoting CCL2 secretion through the NF-κB signaling pathway. Targeting SIRT7 might offer novel mechanism-based therapeutic options for ALD.
Collapse
Affiliation(s)
- Zhiqiang Wang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China; Human Anatomy Teaching and Experimental Center, School of Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Gaoshuang Liang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Jinying Peng
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Yiying Gu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Xiangwen Zhang
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Cong Ding
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Tingzi Yu
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China
| | - Zhuan Li
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, The Key Laboratory of Model Animals and Stem Cell Biology of Hunan Province, Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University School of Pharmaceutical Science, Changsha, China.
| |
Collapse
|
48
|
Le XN, Long DP, Yin SS, Qing RY, Chi ZZ, Gao MQ, Zhu MQ. The efficient separation of bioactive components from Eucommia ulmoides Oliver using membrane filtration technology and its mechanisms in preventing alcoholic liver disease. Carbohydr Polym 2025; 351:123100. [PMID: 39779014 DOI: 10.1016/j.carbpol.2024.123100] [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: 07/03/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025]
Abstract
The efficient extraction and purification of active components from Eucommia ulmoides Oliver (EUO) are crucial for their utilization. The structure and properties of the prepared EUO leaf polysaccharides (ELPs) and extractum (ELE) were comprehensively characterized in this study, and the intervention mechanism of the EUO polysaccharides and extractum in alcoholic liver disease (ALD) were investigated. The yield of EUO extractum was 24.82 %, from which nine active components were identified. The yield of EUO leaf polysaccharides was 8.06 %, and the polysaccharides were fractionated into three components ELP1, ELP2, and ELP3 through ultrafiltration technology, with yields of 4.19 %, 1.26 %, and 2.59 %, respectively. Ultrafiltration significantly reduced protein content, enhanced polysaccharide homogeneity, and altered monosaccharide composition. ELP3 exhibited higher scavenging efficacy on •OH and ABTS•+ than ELP1 and ELP2, reaching 82.53 % and 88.41 % respectively. ELP3 and ELE intervention preserved liver integrity, mitigated lipid accumulation and inflammation, and regulated hepatic oxidative stress. Moreover, they maintained intestinal barrier function, suppressed harmful bacteria (Escherichia-Shigella, and UBA1819), and promoted beneficial bacteria (Dubosiella, Monoglobus, and Lachnospiraceae). Thirteen hallmark differential metabolites were identified, and KEGG pathway enrichment analysis suggested that ELP3 and ELE may ameliorate ALD through pathways like longevity regulation, choline metabolism in cancer, oxidative phosphorylation, and AMPK signaling pathway. This investigation holds significance in delineating the beneficial effects of ELP3 and ELE in ALD alleviation.
Collapse
Affiliation(s)
- Xiao-Na Le
- College of Mechanical and Electronic Engineering, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China
| | - Da-Ping Long
- College of Forestry, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China
| | - Shuang-Shuang Yin
- College of Mechanical and Electronic Engineering, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China
| | - Ren-Yan Qing
- College of Chemistry and Pharmacy, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China
| | - Zhi-Zheng Chi
- College of Forestry, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China
| | - Ming-Qing Gao
- School of Medicine, Northwest University, Xi'an 710000, China.
| | - Ming-Qiang Zhu
- College of Mechanical and Electronic Engineering, Northwest A&F University, Yangling 712100, China; College of Forestry, Northwest A&F University, Yangling 712100, China; Western Scientific Observation and Experiment Station for Development and Utilization of Rural Renewable Energy, M.O.A, Northwest Agriculture & Forestry University, Yangling 712100, China.
| |
Collapse
|
49
|
Wang L, Dong W, Fan L, Kong H, Liang S, Huang Z, Chen J, Zhi S, Xu S, Qiu Q, Yang M, Hou Y, Hu Y, Pan T, Zheng M, Li X, Huang Z, Song L. Repression of the ERRγ-CYP2E1 pathway by FGF4 mitigates alcohol-associated liver injury. Hepatology 2025:01515467-990000000-01184. [PMID: 40009617 DOI: 10.1097/hep.0000000000001282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/25/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND AND AIMS Alcohol-associated liver disease (ALD) represents a critical global health challenge characterized by liver damage resulting from excessive alcohol consumption. Early detection and timely intervention are essential for optimizing patient outcomes. However, the mechanisms underlying alcohol-induced liver injury have not been fully elucidated. Fibroblast growth factor 4 (FGF4) has been implicated in the progression of various liver diseases. This study aims to elucidate the role of FGF4 in the pathogenesis of ALD. APPROACH AND RESULTS We analyzed human liver specimens and observed significant upregulation of FGF4 mRNA and protein levels in patients with ALD. Consistent findings were noted in mouse models subjected to a Lieber-DeCarli liquid diet. Importantly, hepatic FGF4 expression exhibited a positive correlation with ALD severity in both human subjects and murine models. Hepatocyte-specific deletion of Fgf4 ( Fgf4 -LKO) exacerbated alcohol-induced liver injury through increased oxidative stress, inflammation, and apoptosis. Specifically, Fgf4 -LKO mice demonstrated heightened susceptibility to ethanol plus CCl 4 -induced fibrosis and liver injury. However, treatment with the ERRγ inverse agonist GSK5182 and CYP2E1 inhibitor chlormethiazole (CMZ) mitigated the exacerbated liver injury associated with Fgf4 deficiency. Mechanistic investigations revealed that FGFR4 phosphorylates ERRγ, promoting its ubiquitination and degradation in hepatocytes. Hepatic-specific knockout of Fgfr4 intensified alcohol-induced liver injury and nullified the protective conferred of recombinant FGF4 △NT . CONCLUSIONS Our study identifies FGF4 as a stress-responsive regulator in liver pathophysiology, operating through an FGFR4-mediated ERRγ-CYP2E1 signaling pathway. These results underscore the potential of FGF4 and its downstream pathways as therapeutic targets for ALD treatment.
Collapse
Affiliation(s)
- Luyao Wang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenliya Dong
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lei Fan
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongru Kong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyu Liang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhuobing Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Chen
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Zhi
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Siyan Xu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qiaoling Qiu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Miaomiao Yang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yushu Hou
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yue Hu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tongtong Pan
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Minghua Zheng
- Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaokun Li
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhifeng Huang
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lintao Song
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
50
|
Wang J, Chen B, Cheng C, Wang Q, Yang L, Li Z, Lv X. Timosaponin B II as a novel KEAP1-NRF2 inhibitor to alleviate alcoholic liver disease:Receptor structure-based virtual screening and biological evaluation. Chem Biol Interact 2025; 408:111390. [PMID: 39862944 DOI: 10.1016/j.cbi.2025.111390] [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: 11/24/2024] [Revised: 01/02/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Oxidative stress induced by excess ethanol is an important factor in the progression of alcoholic liver disease (ALD). In recent years, inhibiting Kelch-like ECH-associated protein 1 (KEAP1) to activate the antioxidant regulator Nuclear factor erythroid 2-related factor 2 (NRF2) has been considered an effective strategy for treating oxidative stress-related diseases, but its application in ALD remains insufficiently explored. This study aims to discover high-affinity inhibitors targeting the KEAP1 receptor. We conducted virtual screening of a compound library based on a structure-based pharmacophore model, ultimately identifying the candidate compound Timosaponin B II (TBII). Subsequently, we established ALD models in AML-12 cells and C57BL/6 mice, and evaluated the therapeutic effects and mechanisms of TBII on ALD using methods including Immunofluorescence, Western blotting, RT-qPCR, Biochemical assays, and histological staining. Results indicate that TBII significantly improved ethanol-induced liver injury, inhibited the elevation of serum Alanine Aminotransferase (ALT), Aspartate Aminotransferase (AST), Total Cholesterol (T-CHO), and Triglycerides (TG) levels, and reduced lipid droplet accumulation in liver tissues. Furthermore, TBII treatment enhanced the antioxidant capacity of AML-12 cells and mouse liver, increasing Glutathione (GSH) and Superoxide Dismutase (SOD) levels while reducing Malondialdehyde (MDA) and Reactive Oxygen Species (ROS) levels. Mechanistic studies indicated that TBII inhibited the ethanol-induced increase in KEAP1 and reversed the ethanol-induced changes in NRF2 and its downstream targets. In conclusion, this study suggests that TBII may become a potential therapeutic agent for ALD by modulating the KEAP1-NRF2 pathway to alleviate oxidative stress and lipid metabolism abnormalities.
Collapse
Affiliation(s)
- Junjie Wang
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China
| | - Baoyi Chen
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China
| | - Chaofan Cheng
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China
| | - Qingqing Wang
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China
| | - Lili Yang
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China
| | - Zeng Li
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China.
| | - Xiongwen Lv
- Anhui Prevention and Control Engineering Research Center for Fatty Liver Disease, Hefei, Anhui, 230032, PR China; The Key Laboratory of Anti-inflammatory and Immune Medicines, Ministry of Education, Hefei, PR China; Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, PR China.
| |
Collapse
|