1
|
Wu Y, Duan Z, Qu L, Liu Y, Ma X, Fan D. Ginsenoside Rk1 Ameliorates Non-Alcoholic Fatty Liver Disease by Targeting CD36 to Modulate the AMPK Signaling Pathway. Food Res Int 2025; 211:116426. [PMID: 40356178 DOI: 10.1016/j.foodres.2025.116426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 03/28/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent metabolic disorder associated with obesity and insulin resistance. Ginsenoside Rk1 (Rk1), a natural compound extracted from ginseng, exerts various pharmacological effects, including anti-inflammatory, hepatoprotective, hypolipidemic, and insulin-sensitizing properties. This study investigated the therapeutic effects of Rk1 on NAFLD induced by a Western diet in mice and explored the underlying molecular mechanisms. The results showed that Rk1 treatment significantly reduced weight gain, improved insulin sensitivity, and attenuated liver damage and lipid accumulation in the mice. RNA sequencing and molecular analyses showed that Rk1 downregulated the expression of the fatty acid translocase CD36, and restored AMP-activated protein kinase phosphorylation. Furthermore, Rk1 alleviated hepatic inflammation and oxidative stress in NAFLD mice by affecting the sirtuin 1 (SIRT1) and forkhead box O1 (FOXO1) pathways and regulated fatty acid synthesis by affecting the expression of the mechanistic target of rapamycin complex 1 (mTORc1) and sterol regulatory element-binding protein 1c (SREBP1). These findings highlight the potential of Rk1 as a natural therapeutic agent for NAFLD and contribute to research on the use of functional foods and bioactive compounds for metabolic disease management.
Collapse
Affiliation(s)
- Yuqing Wu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Linlin Qu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China; Xi'an Giant Biotechnology Co., Ltd., Xi'an, 710076, China.
| | - Yannan Liu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Xiaoxuan Ma
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an, 710127, China; Biotech. & Biomed. Research Institute, Northwest University, Xi'an, 710127, China.
| |
Collapse
|
2
|
Cho W, Choi SW, Lim DS, Gwon HJ, Abd El-Aty AM, Ahmet Aydemir H, Hong SA, Jeong JH, Jung TW. Donepezil alleviates hepatic steatosis by mitigating ER stress via the AMPK/autophagy pathway. Mol Cell Endocrinol 2025; 601:112523. [PMID: 40118333 DOI: 10.1016/j.mce.2025.112523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 03/23/2025]
Abstract
Donepezil (Do), a drug known for its ability to reduce neuronal inflammation and for its use in the treatment of Alzheimer's disease, has shown promise in combating hepatic lipid accumulation in hyperlipidemic conditions and endoplasmic reticulum (ER) stress, a factor associated with alterations in hepatic lipid metabolism. However, the mechanisms by which these problems are alleviated have not been fully elucidated. In this study, we investigated the effects of Do on hepatic lipid metabolism through both in vitro and in vivo studies. We examined the expression of proteins associated with lipogenesis and ER stress via immunoblot analysis, and hepatic lipid accumulation was assessed via oil red O staining. In addition, autophagosome formation was analyzed by counting MDC-positive cells. Our results demonstrated that Do treatment improved hepatic lipid metabolism and reduced the expression of ER stress markers, resulting in decreased lipogenic lipid deposition and apoptosis in the hepatocytes and livers of hyperlipidemic mice. Mechanistically, knocking down AMPK or inhibiting autophagy with 3-methyladenine (3 MA) attenuated the effects of Do on palmitate-exposed hepatocytes. These results suggest that Do alleviates hepatic ER stress via the AMPK/autophagy pathway and AMPK-mediated fatty acid oxidation, resulting in improved hepatic lipid metabolism and reduced hepatic steatosis and apoptosis. Our study provides evidence that Do may be a promising therapeutic approach for Alzheimer's disease patients with metabolic dysfunction-associated steatotic liver disease (MASLD).
Collapse
Affiliation(s)
- Wonjun Cho
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Sung Woo Choi
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Do Su Lim
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hyeon Ji Gwon
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt; Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey.
| | - Hacı Ahmet Aydemir
- Department of Family Medicine, Erzurum Regional Training and Research Hospital, Erzurum 25000, Turkey; Dr. Filiz Dolunay Family Health Center Unit Number:59, Yakutiye, Erzurum, Turkey
| | - Soon Auck Hong
- Department of Pathology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea; Department of Global Innovative Drugs, Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Tae Woo Jung
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Cao P, Jaeschke H, Ni HM, Ding WX. The Ways to Die: Cell Death in Liver Pathophysiology. Semin Liver Dis 2025. [PMID: 40199509 DOI: 10.1055/a-2576-4332] [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] [Indexed: 04/10/2025]
Abstract
Liver diseases are closely associated with various cell death mechanisms, including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis. Each process contributes uniquely to the pathophysiology of liver injury and repair. Importantly, these mechanisms are not limited to hepatocytes; they also significantly involve nonparenchymal cells. This review examines the molecular pathways and regulatory mechanisms underlying these forms of cell death in hepatocytes, emphasizing their roles in several liver diseases, such as ischemia-reperfusion injury, metabolic dysfunction-associated steatotic liver disease, drug-induced liver injury, and alcohol-associated liver disease. Recent insights into ferroptosis and pyroptosis may reveal novel therapeutic targets for managing liver diseases. This review aims to provide a comprehensive overview of these cell death mechanisms in the context of liver diseases, detailing their molecular signaling pathways and implications for potential treatment strategies.
Collapse
Affiliation(s)
- Peng Cao
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Hong-Min Ni
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas
- Division of Gastroenterology, Hepatology and Mobility, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
4
|
Palomer X, Wang JR, Escalona C, Wu S, Wahli W, Vázquez-Carrera M. Targeting AMPK as a potential treatment for hepatic fibrosis in MASLD. Trends Pharmacol Sci 2025:S0165-6147(25)00065-3. [PMID: 40300935 DOI: 10.1016/j.tips.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease, and often progresses to hepatic fibrosis, cirrhosis, and liver failure. Despite its increasing prevalence, effective pharmacological treatments for MASLD-related fibrosis remain limited. Recent research has highlighted AMP-activated protein kinase (AMPK) as a key regulator of the processes that promote fibrogenesis, and AMPK activation shows potential in mitigating fibrosis. Advances in AMPK activators and deeper insights into their role in fibrotic pathways have recently revitalized interest in targeting AMPK for fibrosis treatment. This review discusses the molecular mechanisms linking AMPK to hepatic fibrosis and evaluates emerging AMPK-directed therapies. Furthermore, it addresses challenges in clinical translation. Importantly, we combine the latest mechanistic discoveries with recent therapeutic developments to provide a comprehensive perspective on AMPK as a target for hepatic fibrosis treatment.
Collapse
Affiliation(s)
- Xavier Palomer
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Jue-Rui Wang
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Claudia Escalona
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Siyuan Wu
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, CH-1015 Lausanne, Switzerland; ToxAlim (Research Center in Food Toxicology), Institut National de Recherche pour l'Agriculture, l'Alimentation, et l'Environnement (INRAE), Unité Mixte de Recherche (UMR) 1331, F-31300 Toulouse Cedex, France
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology, and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, 08028 Barcelona, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain; Pediatric Research Institute, Hospital Sant Joan de Déu, 08950 Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
5
|
Wang Y, Wang H, Li Q, Zhang Y, Dai R, Wu J, Zhang Y, Zhang X, Zhao L, Liu J. Identification of Novel Cyclobutane-Based Derivatives as Potent Acetyl-CoA Carboxylase Allosteric Inhibitors for Nonalcoholic Steatohepatitis Drug Discovery. J Med Chem 2025; 68:8578-8599. [PMID: 40227434 DOI: 10.1021/acs.jmedchem.5c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Nonalcoholic steatohepatitis (NASH) has become a leading cause of liver fibrosis and hepatocellular carcinoma; however, there are no efficient drugs for NASH therapy. Acetyl-CoA carboxylase (ACC) is a crucial enzyme regulating lipid metabolism that is considered as a potential target for NASH treatment. Allosteric inhibitors target nonfunctional sites, which tend to be highly variable in protein families; thus, allosteric inhibitors are explored as an important source of drug candidates. Herein, several hotspot residues are initially identified by utilizing molecular dynamic simulation, MM-GBSA calculation, and alanine mutation. Then, focusing on the interaction with hotspot residues, several cyclobutane-based ACC allosteric inhibitors are designed, synthesized, and biologically evaluated. Among them, B1 demonstrates potent ACC inhibitory activity in vitro, a higher distribution in liver than in other tissues, and a potent therapeutic effect for NASH in vivo, making it a promising candidate for the treatment of NASH.
Collapse
Affiliation(s)
- Yazhou Wang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Hai Wang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Qingqing Li
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Rupeng Dai
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Wu
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Yanan Zhang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Xiaomeng Zhang
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Liwen Zhao
- R & D Center, Nanjing Sanhome Pharmaceutical Co. Ltd., Nanjing 210049, China
| | - Jian Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
- School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| |
Collapse
|
6
|
Holm E, Vermeulen I, Parween S, López-Pérez A, Cillero-Pastor B, Vandenbosch M, Remeseiro S, Hörnblad A. AMPK activator ATX-304 reduces oxidative stress and improves MASLD via metabolic switching. JCI Insight 2025; 10:e179990. [PMID: 40197369 PMCID: PMC11981618 DOI: 10.1172/jci.insight.179990] [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: 02/05/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most common chronic liver disease worldwide for which there is only one approved treatment. Adenosine monophosphate-activated protein kinase (AMPK) is an interesting therapeutic target since it acts as a central regulator of cellular metabolism. Despite efforts to target AMPK, no direct activators have yet been approved for treatment of this disease. This study investigated the effect of the AMPK activator ATX-304 in a preclinical mouse model of progressive fatty liver disease. The data demonstrated that ATX-304 diminishes body fat mass, lowers blood cholesterol levels, and mitigates general liver steatosis and the development of liver fibrosis, but with pronounced local heterogeneities. The beneficial effects of ATX-304 treatment were accompanied by a shift in the liver metabolic program, including increased fatty acid oxidation, reduced lipid synthesis, as well as remodeling of cholesterol and lipid transport. We also observed variations in lipid distribution among liver lobes in response to ATX-304, and a shift in the zonal distribution of lipid droplets upon treatment. Taken together, our data suggested that ATX-304 holds promise as a potential treatment for MASLD.
Collapse
Affiliation(s)
- Emanuel Holm
- Department of Medical and Translational Biology, Umeå University, Umeå Sweden
| | - Isabeau Vermeulen
- Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, Limburg, Netherlands
| | - Saba Parween
- Department of Medical and Translational Biology, Umeå University, Umeå Sweden
| | - Ana López-Pérez
- Department of Medical and Translational Biology, Umeå University, Umeå Sweden
| | - Berta Cillero-Pastor
- Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, Limburg, Netherlands
- The MERLN Institute for Technology-Inspired Regenerative Medicine, Department of Cell Biology-Inspired Tissue Engineering, Maastricht University, Maastricht, Limburg, Netherlands
| | - Michiel Vandenbosch
- Maastricht MultiModal Molecular Imaging Institute (M4i), Maastricht University, Maastricht, Limburg, Netherlands
| | - Silvia Remeseiro
- Department of Medical and Translational Biology, Umeå University, Umeå Sweden
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Andreas Hörnblad
- Department of Medical and Translational Biology, Umeå University, Umeå Sweden
| |
Collapse
|
7
|
Zhang N, Xu D. Controlling pyroptosis through post-translational modifications of gasdermin D. Dev Cell 2025; 60:994-1007. [PMID: 40199241 DOI: 10.1016/j.devcel.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/15/2024] [Accepted: 02/05/2025] [Indexed: 04/10/2025]
Abstract
Pyroptosis, a lytic and programmed cell death pathway, is mediated by gasdermins (GSDMs), with GSDMD playing an important role in innate immunity and pathology. Upon activation, GSDMD is cleaved to release the active N-terminal fragment that oligomerizes into membrane pores, which promote pyroptosis and cytokine secretion, leading to inflammation. Emerging evidence indicates that post-translational modification (PTM) is an important regulatory mechanism of GSDMD activity. This review explores how PTMs, aside from proteolytic cleavage, control GSDMD activity and link biological contexts to pyroptosis in innate immunity and inflammation, which could inform future studies and therapeutic solutions for treating inflammatory conditions.
Collapse
Affiliation(s)
- Na Zhang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 201210, China; State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; Shanghai Key Laboratory of Aging Studies, Shanghai 201210, China; Shanghai Academy of Natural Sciences (SANS), Fudan University, Shanghai 200031, China.
| |
Collapse
|
8
|
Wang D, Zhang M, Zhang Y, Yin Z, Zhang S, Zhao Z, Duan Y. Hepatoprotective effects of polysaccharide from Morchella esculenta are associated with activation of the AMPK/Sirt1 signaling pathway in mice with NAFLD. Int J Biol Macromol 2025; 301:140444. [PMID: 39884630 DOI: 10.1016/j.ijbiomac.2025.140444] [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: 01/26/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The functional food application of edible fungus polysaccharides has been widely studied based on their variety of potential pharmacological activities. However, the hepatoprotective effects and mechanisms of Morchella esculenta polysaccharide against nonalcoholic fatty liver disease (NAFLD) remain unknown. A high-fat diet (HFD) fed C57BL/6 J mice for 8 weeks was employed to establish NAFLD with simple steatosis, methionine choline deficiency (MCD) diet for 4 weeks induced hepatic steatohepatitis and fibrosis. The M. esculenta polysaccharide (MCP) or saline was administered intragastrically. MCP markedly reduced hepatic and serum triglyceride (TG) and cholesterol contents in HFD-fed mice. Moreover, treatment with MCP ameliorated nonalcoholic steatohepatitis (NASH) progression in MCD-fed mice, as evidenced by ameliorated hepatic steatosis, inflammatory response, and fibrosis. Mechanistically, MCP suppressed the expression of lipogenic genes and inflammatory cytokines and upregulated peroxisome proliferator-activated receptor (PPAR)-α expression to induce fatty acid β-oxidation. These beneficial effects were attributed to activating the AMP-activated kinase (AMPK)/Sirtuin 1 (Sirt1) signaling pathway. Therefore, we provided evidence that MCP might be an effective dietary supplement to ameliorate NAFLD.
Collapse
Affiliation(s)
- Dandan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China; College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Menglian Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Yaowen Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui 230011, China
| | - Zequn Yin
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Shuang Zhang
- College of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui 230601, China
| | - Zhiwei Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| | - Yajun Duan
- Department of Cardiology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China.
| |
Collapse
|
9
|
Zhang Q, Wang J, Hu X, Lu W, Cao Y, Niu C, Yue H. GLP-1RAs regulate lipid metabolism and induce autophagy through AMPK/SIRT1 pathway to improve NAFLD. Prostaglandins Other Lipid Mediat 2025; 178:106987. [PMID: 40180281 DOI: 10.1016/j.prostaglandins.2025.106987] [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: 12/01/2024] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a leading cause of cirrhosis and a major risk factor for hepatocellular carcinoma and liver-related death. Diabetes medications have been studied as potential treatments for NAFLD. Glucagon-like peptide-1 agonists (GLP-1RAs) have been rarely reported in the treatment of NAFLD alone as an anti-diabetic drug, and its specific mechanism of action is unknown. We investigated whether the therapeutic effect of liraglutide (LRG, a representative drug of GLP-1RAs) on hepatic steatosis is related to regulating lipid metabolism and enhancing autophagy in the hepatocytes. METHODS We examined the effect of LRG on fat accumulation in fatty hepatocytes, and discussed its effects on enzymes related to lipid metabolism and autophagy. Meanwhile, knockdown of SIRT1 in free fatty acids(FFA)-treated cells was used to detected the influence of LRG on lipid metabolism and autophagy by regulating of AMPK/SIRT1 signaling. RESULTS Our findings showed that free fatty acids (FFA) induced hepatocyte steatosis, which was significantly reversed by LRG. Meanwhile, LRG significantly regulated the expression of hepatocyte lipogenesis and cytosolic lipolysis-related proteins (FAS, ACC1, ATGL, HSL, LAL). Furthermore, LRG enhanced FFA-induced suppression of autophagy and SIRT1 expression, reducing intracellular lipid accumulation. It is evident that LRG regulates lipid metabolism and induces autophagy in an (AMPK)-dependent manner. Moreover, SIRT1 knockdown inhibited the autophagy-inducing and lipid-lowering effects of LRG. CONCLUSION GLP-1RAs may lower hepatic steatosis by regulating lipid metabolism and enhancing autophagy in an AMPK/SIRT1-dependent manner, providing a new target for the treatment of NAFLD.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Gastroenterology, Yancheng Third People's Hospital (The Yancheng School of Clinical Medicine of Nanjing Medical University), Yancheng, Jiangsu Province 224000, PR China
| | - Jingyuan Wang
- Department of Rhematology and Immunology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian Province 361000, PR China
| | - Xiaojin Hu
- Department of Radiation Oncology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian Province 361000, PR China
| | - Wei Lu
- Department of Gastroenterology, Yancheng Third People's Hospital (The Yancheng School of Clinical Medicine of Nanjing Medical University), Yancheng, Jiangsu Province 224000, PR China
| | - Yang Cao
- Department of Gastroenterology, Yancheng Third People's Hospital (The Yancheng School of Clinical Medicine of Nanjing Medical University), Yancheng, Jiangsu Province 224000, PR China
| | - Chunyan Niu
- Department of Gastroenterology, Nanjing Lishui People's Hospital (Zhongda Hospital Lishui Branch, Southeast University), Nanjing, Jiangsu Province 210000, PR China.
| | - Hongqin Yue
- Department of Gastroenterology, Yancheng Third People's Hospital (The Yancheng School of Clinical Medicine of Nanjing Medical University), Yancheng, Jiangsu Province 224000, PR China.
| |
Collapse
|
10
|
Deng J, Ding K, Liu S, Chen F, Huang R, Xu B, Zhang X, Xie W. SOX9 Overexpression Ameliorates Metabolic Dysfunction-associated Steatohepatitis Through Activation of the AMPK Pathway. J Clin Transl Hepatol 2025; 13:189-199. [PMID: 40078197 PMCID: PMC11894392 DOI: 10.14218/jcth.2024.00197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 12/03/2024] [Accepted: 12/04/2024] [Indexed: 03/14/2025] Open
Abstract
Background and Aims The transcription factor sex-determining region Y-related high-mobility group-box gene 9 (SOX9) plays a critical role in organ development. Although SOX9 has been implicated in regulating lipid metabolism in vitro, its specific role in metabolic dysfunction-associated steatohepatitis (MASH) remains poorly understood. This study aimed to investigate the role of SOX9 in MASH pathogenesis and explored the underlying mechanisms. Methods MASH models were established using mice fed either a methionine- and choline-deficient (MCD) diet or a high-fat, high-fructose diet. To evaluate the effects of SOX9, hepatocyte-specific SOX9 deletion or overexpression was performed. Lipidomic analyses were conducted to assess how SOX9 influences hepatic lipid metabolism. RNA sequencing was employed to identify pathways modulated by SOX9 during MASH progression. To elucidate the mechanism further, HepG2 cells were treated with an adenosine monophosphate-activated protein kinase (AMPK) inhibitor to test whether SOX9 acts via AMPK activation. Results SOX9 expression was significantly elevated in hepatocytes of MASH mice. Hepatocyte-specific SOX9 deletion exacerbated MCD-induced MASH, whereas overexpression of SOX9 mitigated high-fat, high-fructose-induced MASH. Lipidomic and RNA sequencing analyses revealed that SOX9 suppresses the expression of genes associated with lipid metabolism, inflammation, and fibrosis in MCD-fed mice. Furthermore, SOX9 deletion inhibited AMPK pathway activation, while SOX9 overexpression enhanced it. Notably, administration of an AMPK inhibitor negated the protective effects of SOX9 overexpression, leading to increased lipid accumulation in HepG2 cells. Conclusions Our findings demonstrate that SOX9 overexpression alleviates hepatic lipid accumulation in MASH by activating the AMPK pathway. These results highlight SOX9 as a promising therapeutic target for treating MASH.
Collapse
Affiliation(s)
- Juan Deng
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Shuqing Liu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Fei Chen
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ru Huang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bonan Xu
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xin Zhang
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Weifen Xie
- Department of Gastroenterology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Gastroenterology, Changzheng Hospital, Naval Medical University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, China
| |
Collapse
|
11
|
Wang X, Abu Bakar MH, Liqun S, Kassim MA, Shariff KA, Karunakaran T. Targeting metabolic diseases with celastrol: A comprehensive review of anti-inflammatory mechanisms and therapeutic potential. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119560. [PMID: 40015541 DOI: 10.1016/j.jep.2025.119560] [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: 12/10/2024] [Revised: 02/15/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tripterygium wilfordii is a traditional Chinese medicine used to treat rheumatic diseases, with properties such as clearing heat, detoxifying, dispelling wind, and relieving pain. In recent years, its active compound, celastrol, garnered significant attention for its potential therapeutic effects on metabolic diseases. Celastrol exhibits bioactivities such as regulating metabolic functions and anti-inflammatory effects, positioning it as a promising candidate for the treatment of obesity, diabetes, atherosclerosis (AS), and non-alcoholic fatty liver disease (NAFLD). AIM OF THE REVIEW This review aims to explore the pharmacological mechanisms of celastrol in metabolic diseases, focusing on its anti-inflammatory mechanisms and metabolic regulation effects, providing theoretical support for further investigation of its therapeutic potential in metabolic diseases. METHODS Literature was retrieved from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar. This review primarily focuses on anti-inflammatory mechanisms of celastrol, its metabolic regulation, and toxicity studies, by systematically analyzing its effects in obesity, diabetes, AS, and NAFLD, providing scientific evidence for its potential clinical applications. RESULTS Celastrol regulates multiple signaling pathways, particularly inhibiting NF-κB and activating AMPK, reducing the production of pro-inflammatory cytokines and improving insulin sensitivity, enhancing its therapeutic potential in metabolic diseases. Additionally, celastrol regulates adipogenesis and energy metabolism by influencing key transcription factors such as PPARγ and SREBP-1c. Numerous studies highlight its role in alleviating oxidative stress and improving mitochondrial function, further enhancing its metabolic benefits. CONCLUSION In summary, celastrol holds great promise as a multi-target therapeutic agent for metabolic diseases, offering anti-inflammatory, metabolic regulatory, and antioxidative benefits. Despite these, challenges remain for the clinical application of celastrol due to its poor bioavailability and potential toxicity. Advanced formulation strategies and targeted delivery systems are urgently needed to overcome challenges related to bioavailability and clinical translation.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia; Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohamad Hafizi Abu Bakar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| | - Song Liqun
- Department of Pharmacy, Taishan Vocational College of Nursing, 271099, Tai'an, Shandong, China
| | - Mohd Asyraf Kassim
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia
| | - Khairul Anuar Shariff
- School of Materials & Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, 14300, Penang, Malaysia
| | | |
Collapse
|
12
|
Zhang J, Wang Q, Zhou N, Liu J, Tao L, Peng Z, Hu G, Wang H, Fu L, Peng S. Fluorofenidone attenuates choline-deficient, l-amino acid-defined, high-fat diet-induced metabolic dysfunction-associated steatohepatitis in mice. Sci Rep 2025; 15:9863. [PMID: 40118958 PMCID: PMC11928590 DOI: 10.1038/s41598-025-94401-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/13/2025] [Indexed: 03/24/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH), a severe form of metabolic dysfunction-associated steatotic liver disease (MASLD), involves hepatic lipid accumulation, inflammation, and fibrosis. It can progress to cirrhosis or hepatocellular carcinoma without timely treatment. Current treatment options for MASH are limited. This study explores the therapeutic effects of fluorofenidone (AKF-PD), a novel small-molecule compound with antifibrotic and anti-inflammatory properties, on MASH in mouse model. Mice fed a choline-deficient, l-amino acid-defined, high-fat diet (CDAHFD) were treated with AKF-PD, resulting in reduced serum ALT, AST, hepatic lipid accumulation, liver inflammation, and fibrosis. Network pharmacology and RNA-sequencing analyses suggested that AKF-PD influenced multiple metabolic, inflammatory, and fibrosis-related pathways. Further experiments verified that AKF-PD activated hepatic AMPK signaling, leading to the inhibition of the downstream SREBF1/SCD1 pathway and the activation of autophagy. Additionally, AKF-PD suppressed the expression of various inflammatory factors, reduced macrophage infiltration, and inhibited NLRP3 inflammasome activation. Moreover, AKF-PD attenuated liver fibrosis by inhibiting TGFβ1/SMAD signaling. In conclusion, this study reveals that AKF-PD effectively decreases hepatic lipid accumulation, liver inflammation and fibrosis in a CDAHFD-induced MASH model, positioning AKF-PD as a promising candidate for the treatment of MASH.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Qianbing Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Nianqi Zhou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinqing Liu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Gaoyun Hu
- Faculty of Pharmaceutical Sciences, Central South University, Changsha, 410008, Hunan, China
| | - Huiwen Wang
- Department of Infection Control Center, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Lei Fu
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Shifang Peng
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
13
|
Wan T, Chen Z, Li J, Yuan X, Zheng M, Qin L, Zhang L, Hou T, Liu C, Li R. AMPK agonist AICAR ameliorates maternal hepatic lipid metabolism disorder, inflammation, and fibrosis caused by PM 2.5 exposure during pregnancy. Sci Rep 2025; 15:8689. [PMID: 40082541 PMCID: PMC11906884 DOI: 10.1038/s41598-025-93395-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Liver is an important target organ of ambient fine particulate matter (PM2.5). Numerous studies have shown that PM2.5 exposure can cause liver lipid metabolism disorders and other liver damage in mammals. However, the impact of PM2.5 on liver health during pregnancy, a sensitive life stage, remains understudied, and the underlying mechanisms are also unknown. Given the critical role of adenosine 5'-monophosphate activated protein kinase (AMPK) in regulating lipid metabolism and inflammation, we hypothesize that AMPK activation may mitigate maternal hepatic lipid metabolism disorders, reduce inflammation, and attenuate fibrosis induced by PM2.5 exposure during pregnancy. To test this hypothesis, pregnant C57BL/6 mice were randomly assigned to 4 groups: filtered air (FA) + NS (normal saline), PM2.5+NS, FA + AICAR (acadesine, an AMPK activator), and PM2.5+AICAR. PM2.5+NS and PM2.5+AICAR groups were continuously exposed to PM2.5 with a whole-body PM2.5 exposure chamber, while the other two groups were exposed to filtered air in the FA chamber. Simultaneously, the FA + AICAR and PM2.5+AICAR groups received intraperitoneal injections of the AMPK agonist AICAR (200 mg/kg∙bw per day) from gestational day 13 (GD13) to GD17, while mice in the FA + NS and PM2.5+NS groups were administered normal saline injection. We found that gestational PM2.5 exposure induced dyslipidemia in pregnant mice, which was alleviated by AICAR treatment. Histopathological analysis showed that the exposure to PM2.5 during pregnancy induced hepatic lipid deposition and fibrosis in pregnant mice, and biochemical assays revealed that hepatic triglyceride and cholesterol levels were also significantly increased in pregnant mice after exposure to PM2.5, whereas the AICAR treatment ameliorated hepatic lipid deposition and fibrosis induced by the exposure to PM2.5 during pregnancy. Furthermore, PM2.5 exposure during pregnancy disrupted the expression of key genes and proteins associated with hepatic lipid synthesis, cholesterol synthesis, inflammation, and fibrosis, while treatment with AICAR mitigated these effects. These findings demonstrated that AMPK activation ameliorates hepatic lipid metabolism disorders, reduces inflammation, and attenuates fibrosis caused by PM2.5 exposure in mice during pregnancy. AMPK may be a target of action for maternal liver injury induced by PM2.5 exposure during pregnancy.
Collapse
Affiliation(s)
- Teng Wan
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuan Chen
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangyi Yuan
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingmeng Zheng
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Qin
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tong Hou
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ran Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
14
|
Chowdhury K, Das D, Huang M. Advancing the Metabolic Dysfunction-Associated Steatotic Liver Disease Proteome: A Post-Translational Outlook. Genes (Basel) 2025; 16:334. [PMID: 40149485 PMCID: PMC11941888 DOI: 10.3390/genes16030334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver disorder with limited treatment options. This review explores the role of post-translational modifications (PTMs) in MASLD pathogenesis, highlighting their potential as therapeutic targets. We discuss the impact of PTMs, including their phosphorylation, ubiquitylation, acetylation, and glycosylation, on key proteins involved in MASLD, drawing on studies that use both human subjects and animal models. These modifications influence various cellular processes, such as lipid metabolism, inflammation, and fibrosis, contributing to disease progression. Understanding the intricate PTM network in MASLD offers the potential for developing novel therapeutic strategies that target specific PTMs to modulate protein function and alleviate disease pathology. Further research is needed to fully elucidate the complexity of PTMs in MASLD and translate these findings into effective clinical applications.
Collapse
Affiliation(s)
- Kushan Chowdhury
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA; (K.C.); (D.D.)
| | - Debajyoti Das
- Department of Medicine, Vatche and Tamar Manoukian Division of Digestive Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA; (K.C.); (D.D.)
| | - Menghao Huang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indiana University, Indianapolis, IN 46202, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Melvin & Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
15
|
Feng J, MengHuan L, TingTing Y, XueJie Y, HaiNing G. Research progress on AMPK in the pathogenesis and treatment of MASLD. Front Immunol 2025; 16:1558041. [PMID: 40134423 PMCID: PMC11932893 DOI: 10.3389/fimmu.2025.1558041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD; formerly known as non-alcoholic fatty liver disease, NAFLD) has become one of the most prevalent chronic liver diseases worldwide, with its incidence continuously rising alongside the epidemic of metabolic disorders. AMP-activated protein kinase (AMPK), as a key regulator of cellular energy metabolism, influences multiple pathological processes associated with MASLD. This review systematically summarizes the regulatory roles of AMPK in lipid metabolism, inflammatory response, cell apoptosis, and fibrosis. Additionally, it discusses the latest developments of AMPK activators from preclinical to clinical studies, while analyzing the major challenges currently faced and potential strategies for resolution. A deeper understanding of AMPK regulatory mechanisms will contribute to the development of more effective therapeutic approaches for MASLD.
Collapse
Affiliation(s)
- Jiang Feng
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Li MengHuan
- School of Physical Education, Liaoning Normal University, Dalian, China
| | - Yao TingTing
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Yi XueJie
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Gao HaiNing
- School of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
16
|
Berdowska I, Matusiewicz M, Fecka I. A Comprehensive Review of Metabolic Dysfunction-Associated Steatotic Liver Disease: Its Mechanistic Development Focusing on Methylglyoxal and Counterbalancing Treatment Strategies. Int J Mol Sci 2025; 26:2394. [PMID: 40141037 PMCID: PMC11942149 DOI: 10.3390/ijms26062394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/21/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a multifactorial disorder characterized by excessive lipid accumulation in the liver which dysregulates the organ's function. The key contributor to MASLD development is insulin resistance (IR) which affects many organs (including adipose tissue, skeletal muscles, and the liver), whereas the molecular background is associated with oxidative, nitrosative, and carbonyl stress. Among molecules responsible for carbonyl stress effects, methylglyoxal (MGO) seems to play a major pathological function. MGO-a by-product of glycolysis, fructolysis, and lipolysis (from glycerol and fatty acids-derived ketone bodies)-is implicated in hyperglycemia, hyperlipidemia, obesity, type 2 diabetes, hypertension, and cardiovascular diseases. Its causative effect in the stimulation of prooxidative and proinflammatory pathways has been well documented. Since metabolic dysregulation leading to these pathologies promotes MASLD, the role of MGO in MASLD is addressed in this review. Potential MGO participation in the mechanism of MASLD development is discussed in regard to its role in different signaling routes leading to pathological events accelerating the disorder. Moreover, treatment strategies including approved and potential therapies in MASLD are overviewed and discussed in this review. Among them, medications aimed at attenuating MGO-induced pathological processes are addressed.
Collapse
Affiliation(s)
- Izabela Berdowska
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Małgorzata Matusiewicz
- Department of Medical Biochemistry, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland;
| | - Izabela Fecka
- Department of Pharmacognosy and Herbal Medicines, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wrocław, Poland
| |
Collapse
|
17
|
Irshad I, Alqahtani SA, Ikejima K, Yu ML, Romero-Gomez M, Eslam M. Energy metabolism: An emerging therapeutic frontier in liver fibrosis. Ann Hepatol 2025; 30:101896. [PMID: 40057035 DOI: 10.1016/j.aohep.2025.101896] [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: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/18/2025]
Abstract
Liver fibrosis is a progressive response to chronic liver diseases characterized by a wound-healing process that leads to the accumulation of fibrillary extracellular matrix (ECM) proteins in and around the liver tissue. If left untreated, liver fibrosis can advance to cirrhosis and ultimately result in liver failure. Although there have been significant advancements in understanding the molecular mechanisms involved in liver fibrosis, effective therapeutic strategies to reverse or halt the condition remain limited. Recent research has underscored the critical role of energy metabolism in the initiation and progression of liver fibrosis. In response to liver injury, hepatic cells undergo metabolic reprogramming to meet the energy demands of myofibroblasts. This reprogramming involves various metabolic changes, including mitochondrial dysfunction, alterations in cellular bioenergetics, shifts in glycolysis and oxidative phosphorylation, as well as changes in lipid metabolism. These modifications can disrupt cellular energy homeostasis and increase energy release, activating hepatic cells, primarily hepatic stellate cells (HSCs). Activated HSCs then stimulate fibrogenic pathways, leading to the accumulation of ECM proteins in the liver, which exacerbates the progression of fibrosis. This review aims to explore the emerging connection between energy metabolism and liver fibrosis, focusing on the metabolic alterations and molecular mechanisms that drive this condition. We also examine the therapeutic implications of modulating energy metabolism to reduce energy release and mitigate liver fibrosis. Altering energy metabolism to decrease energy release may represent a promising approach for treating liver fibrosis and chronic liver diseases.
Collapse
Affiliation(s)
- Iram Irshad
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia
| | - Saleh A Alqahtani
- Liver, Digestive, & Lifestyle Health Research Section, and Organ Transplant Center of Excellence, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia; Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Kenichi Ikejima
- Department of Gastroenterology, Juntendo University School of Medicine, Japan
| | - Ming-Lung Yu
- School of Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan; Hepatobiliary Division, Department of Internal Medicine, Kaohsiung Medical University Hospital; College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Manuel Romero-Gomez
- Digestive Diseases Department and Ciberehd, Virgen del Rocío University Hospital, Institute of Biomedicine of Seville (HUVR/CSIC/US), University of Seville, Seville, Spain
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, NSW, Australia.
| |
Collapse
|
18
|
Karimzadeh K, Unniappan S, Zahmatkesh A. Spirulina platensis Peptide-Loaded Nanoliposomes Alleviate Hepatic Lipid Accumulation in Male Wistar Rats by Influencing Redox Homeostasis and Lipid Metabolism via the AMPK Signaling Pathway. Appl Biochem Biotechnol 2025; 197:1696-1725. [PMID: 39601973 DOI: 10.1007/s12010-024-05089-w] [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] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Spirulina platensis low-molecular-weight peptides (SP) have been reported to exhibit antioxidant and hepatoprotective properties. However, the limited bioavailability and solubility of SPs limit their potential applications. In this study, to examine the potential anti-obesity effects and underlying mechanisms of SPs, high-fat diet-induced non-alcoholic fatty liver disease (NAFLD) model rats were treated with SPs and SP-loaded nanoliposomes. Furthermore, hepatic biochemical parameters, inflammatory markers, histopathological changes, and genes involved in AMPK signaling were analyzed. SP-loaded nanoliposomes demonstrated a spherical shape with slower and sustained SP release. SP and SP-loaded nanoliposomes mitigated hepatic damage by lowering serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and increasing hepatic antioxidant enzymes, which are manifested in improving histopathological findings. In addition, notably, SP-loaded nanoliposomes downregulated lipogenic fatty acid synthase (FAS) and sterol regulatory element-binding protein-1c (SREBP-1c) in the liver. Meanwhile, an upregulation of phosphorylated AMP-activated protein kinase (P-AMPK), lipid acid oxidation-related genes carnitine palmitoyltransferase-1 (CPT-1), and peroxisome proliferator-activated receptor alpha (PPAR-α) was found in the rat liver. This data implies that SP and SP-loaded nanoliposomes exhibit protective potential in rats against the HFD-induced NAFLD, which is mediated through the activation of the AMPK signaling pathway.
Collapse
Affiliation(s)
- Katayoon Karimzadeh
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada.
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.
| | - Suraj Unniappan
- Laboratory of Integrative Neuroendocrinology, Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Asgar Zahmatkesh
- Aquaculture Department, Gilan Agricultural and Natural Resources Research and Education Center, AREEO, Rasht, Iran
| |
Collapse
|
19
|
Guo Y, Huang H, Yang L, Shen Q, Liu Z, Wang Q, Chen S, Pan J, Zhai H, Li Y, Xu L, Yu C, Xu C. Amyloid precursor protein promotes MASH progression by upregulating death receptor 6-mediated hepatocyte apoptosis. J Biol Chem 2025; 301:108285. [PMID: 39938799 PMCID: PMC11923821 DOI: 10.1016/j.jbc.2025.108285] [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: 11/07/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a complicated process that contributes to end-stage liver disease and, eventually, hepatocellular carcinoma. Hepatocyte apoptosis, a well-defined form of cell death in MASH, is considered the primary cause of liver inflammation and fibrosis. However, the mechanisms underlying the regulation of hepatocyte apoptosis in MASH remain largely unclear. We explored the proapoptotic effect of hepatocyte amyloid precursor protein (APP) in MASH. C57BL/6J mice were fed a Western diet plus sugar water, a high-fat high-fructose diet, or a methionine and choline deficiency diet to induce MASH. APP expression was analyzed in murine MASH specimens. App-/- mice and mice with adeno-associated virus-mediated APP overexpression were established to study the role of APP in MASH. Palmitic acid was used to mimic lipotoxicity-induced MASH in AML12 cells. We identified a dramatic increase in APP expression in hepatocytes of patients with MASH and three different mouse models. Suppression of APP attenuated hepatic steatosis, inflammation, and fibrosis in MASH mice, whereas its restoration activated MASH pathogenesis. Furthermore, increased death receptor 6 (DR6) was observed in MASH mouse livers. Mechanistically, APP interacted with DR6, a tumor necrosis factor receptor, to facilitate DR6 expression and activation. Activated DR6 increased apoptosis in hepatocytes, which was associated with an increase in proapoptotic effectors (cleaved-caspase 3/7). Our results highlight the role of the APP-DR6 axis in hepatocyte apoptosis, inflammation activation, and fibrosis formation in murine MASH model, providing new insights into therapeutic strategies for MASH.
Collapse
Affiliation(s)
- Yanjun Guo
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hangkai Huang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling Yang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qien Shen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhening Liu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qinqiu Wang
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenghui Chen
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaqi Pan
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoliang Zhai
- Department of Gastroenterology, Haining Branch, The First Affiliated Hospital, Zhejiang University School of Medicine, Haining, China
| | - Youming Li
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Gastroenterology, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Chengfu Xu
- Department of Gastroenterology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
20
|
Raju C, Sankaranarayanan K. Insights on post-translational modifications in fatty liver and fibrosis progression. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167659. [PMID: 39788217 DOI: 10.1016/j.bbadis.2025.167659] [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: 09/06/2024] [Revised: 12/20/2024] [Accepted: 01/02/2025] [Indexed: 01/12/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease [MASLD] is a pervasive multifactorial health burden. Post-translational modifications [PTMs] of amino acid residues in protein domains demonstrate pivotal roles for imparting dynamic alterations in the cellular micro milieu. The crux of identifying novel druggable targets relies on comprehensively studying the etiology of metabolic disorders. This review article presents how different chemical moieties of various PTMs like phosphorylation, methylation, ubiquitination, glutathionylation, neddylation, acetylation, SUMOylation, lactylation, crotonylation, hydroxylation, glycosylation, citrullination, S-sulfhydration and succinylation presents the cause-effect contribution towards the MASLD spectra. Additionally, the therapeutic prospects in the management of liver steatosis and hepatic fibrosis via targeting PTMs and regulatory enzymes are also encapsulated. This review seeks to understand the function of protein modifications in progression and promote the markers discovery of diagnostic, prognostic and drug targets towards MASLD management which could also halt the progression of a catalogue of related diseases.
Collapse
Affiliation(s)
- Chithra Raju
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, Madras Institute of Technology Campus, Anna University, Chrompet, Chennai 600 044, Tamil Nadu, India.
| |
Collapse
|
21
|
Kudo S, Hikita H, Saito Y, Murai K, Kodama T, Tatsumi T, Takehara T. Collaborative orchestration of BH3-only proteins governs Bak/Bax-dependent hepatocyte apoptosis under antiapoptotic protein-deficiency in mice. Cell Death Differ 2025:10.1038/s41418-025-01458-y. [PMID: 39994353 DOI: 10.1038/s41418-025-01458-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 01/10/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
The fine-tuned balance between anti-apoptotic Bcl-2 family proteins, such as Bcl-xL and Mcl-1, and pro-apoptotic Bcl-2 family proteins, like Bak and Bax, is crucial for maintaining hepatocyte integrity. BH3-only proteins, including Bid, Bim, Puma, Noxa, Bad, Bmf, Bik and Hrk, serve as apoptosis initiators. They are activated by various stimuli, which leads to Bak/Bax activation. We previously reported that Bid and Bim contributed to hepatocyte apoptosis through Bak/Bax activation in the absence of anti-apoptotic proteins Bcl-xL and/or Mcl-1. However, the comprehensive involvement of all eight BH3-only proteins in Bak/Bax-dependent hepatocyte apoptosis remains unclear. Puma disruption suppressed hepatocyte apoptosis in hepatocyte-specific Bcl-xL or Mcl-1 knockout (Bcl-xLΔHep/ΔHep or Mcl-1ΔHep/ΔHep) mice. Disruption of Bid and Bim partially prevented lethality in Mcl-1ΔHep/+ Bcl-xLΔHep/ΔHep mice, although severe hepatocyte apoptosis persisted, which was suppressed by additional Puma disruption. However, hepatocyte apoptosis was still induced compared to that in Mcl-1ΔHep/+ Bcl-xLΔHep/ΔHep BaxΔHep/ΔHep Bak-/- mice. Triple disruption of Bid, Bim and Puma did not prevent induction of hepatocyte apoptosis in tamoxifen-induced Mcl-1iΔHep/iΔHep Bcl-xLiΔHep/iΔHep mice. Primary hepatocytes, isolated from Mcl-1fl/fl Bcl-xLfl/fl Bid-/- Bim-/- Puma-/- mice and immortalized, underwent apoptosis with doxycycline-dependent Cre recombination. Among the remaining five BH3-only proteins, Bik and Hrk were not expressed in these cells, and Noxa knockdown, but not Bad or Bmf knockdown, reduced apoptosis. Noxa disruption alleviated hepatocyte apoptosis in Mcl-1ΔHep/ΔHep mice and tamoxifen-induced Mcl-1iΔHep/iΔHep Bcl-xLiΔHep/iΔHep Bid-/- Bim-/- Puma-/- mice, prolonging survival. Apoptosis persisted in immortalized primary hepatocytes isolated from Mcl-1fl/fl Bcl-xLfl/fl Bid-/- Bim-/- Puma-/- Noxa-/- mice where doxycycline-dependent Cre recombination was induced, but was completely suppressed by Bak/Bax knockdown, while Bad or Bmf knockdown had no effect. In conclusion, among the eight BH3-only proteins, Puma and Noxa, alongside Bid and Bim, contributed to Bak/Bax-dependent hepatocyte apoptosis, but not indispensably, in the absence of Mcl-1 and Bcl-xL.
Collapse
Affiliation(s)
- Shinnosuke Kudo
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshinobu Saito
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuhiro Murai
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
22
|
Geng W, Liao W, Cao X, Yang Y. Therapeutic Targets and Approaches to Manage Inflammation of NAFLD. Biomedicines 2025; 13:393. [PMID: 40002806 PMCID: PMC11853636 DOI: 10.3390/biomedicines13020393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its advanced form, non-alcoholic steatohepatitis (NASH), are the leading causes of chronic liver disease globally. They are driven by complex mechanisms where inflammation plays a pivotal role in disease progression. Current therapies, including lifestyle changes and pharmacological agents, are limited in efficacy, particularly in addressing the advanced stages of the disease. Emerging approaches targeting inflammation, metabolic dysfunction, and fibrosis offer promising new directions, though challenges such as treatment complexity and heterogeneity persist. This review concludes the main therapeutic targets and approaches to manage inflammation currently and emphasizes the critical need for future drug development and combination therapy for NAFLD/NASH management.
Collapse
Affiliation(s)
- Wanying Geng
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China;
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Wanying Liao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Xinyuan Cao
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| | - Yingyun Yang
- Department of Gastroenterology, Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (W.L.); (X.C.)
| |
Collapse
|
23
|
Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther 2025; 10:33. [PMID: 39904973 PMCID: PMC11794951 DOI: 10.1038/s41392-024-02072-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 10/06/2024] [Accepted: 11/12/2024] [Indexed: 02/06/2025] Open
Abstract
As a highly complex organ with digestive, endocrine, and immune-regulatory functions, the liver is pivotal in maintaining physiological homeostasis through its roles in metabolism, detoxification, and immune response. Various factors including viruses, alcohol, metabolites, toxins, and other pathogenic agents can compromise liver function, leading to acute or chronic injury that may progress to end-stage liver diseases. While sharing common features, liver diseases exhibit distinct pathophysiological, clinical, and therapeutic profiles. Currently, liver diseases contribute to approximately 2 million deaths globally each year, imposing significant economic and social burdens worldwide. However, there is no cure for many kinds of liver diseases, partly due to a lack of thorough understanding of the development of these liver diseases. Therefore, this review provides a comprehensive examination of the epidemiology and characteristics of liver diseases, covering a spectrum from acute and chronic conditions to end-stage manifestations. We also highlight the multifaceted mechanisms underlying the initiation and progression of liver diseases, spanning molecular and cellular levels to organ networks. Additionally, this review offers updates on innovative diagnostic techniques, current treatments, and potential therapeutic targets presently under clinical evaluation. Recent advances in understanding the pathogenesis of liver diseases hold critical implications and translational value for the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Can Gan
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuan Yuan
- Aier Institute of Ophthalmology, Central South University, Changsha, China
| | - Haiyuan Shen
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Jinhang Gao
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiangxin Kong
- Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Zhaodi Che
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yangkun Guo
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China.
| | - Erdan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital, School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Jia Xiao
- Clinical Medicine Research Institute and Department of Anesthesiology, The First Affiliated Hospital of Jinan University, Guangzhou, China.
- Department of Gastroenterology, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
24
|
Wan Z, Yuan M, Liu Z, Cai Y, He H, Hao K. Impact of Dapagliflozin on Hepatic Lipid Metabolism and a Dynamic Model of Ketone Body Levels. AAPS J 2025; 27:38. [PMID: 39900889 DOI: 10.1208/s12248-025-01024-x] [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: 11/20/2024] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
Abstract
The rising prevalence of metabolic-associated steatotic liver disease emphasizes the need to understand its lipid metabolism. Dapagliflozin may improve hepatic steatosis but could also increase the risk of ketoacidosis by elevating β-hydroxybutyrate (KB) levels. This study investigates dapagliflozin's effects on hepatic lipid metabolism and quantifies KB levels in vivo. Male Sprague-Dawley rats were fed either a normal diet or a high-fat diet (HFD) for 12 weeks. The HFD rats were then divided into four subgroups to receive vehicle, 0.5 mg/kg, 1 mg/kg, and 3 mg/kg of dapagliflozin for four weeks. Free fatty acids (FFA) and KB levels were monitored, while protein and gene expression were analyzed. And a dynamic model of KB was developed for humans based on preclinical data. Dapagliflozin decreased body weight and visceral fat in HFD rats, increasing KB by upregulating CPT1a, HMGCS2, and HMGCL, and downregulating ACC. These changes correlated with reduced liver/fat index, liver pathology score, and oil-red staining area. A pharmacokinetic/pharmacodynamic (PK/PD) model was created from preclinical data to quantify KB levels in rats and validated in humans. Dapagliflozin reduces hepatic steatosis by enhancing fatty acid β-oxidation and ketogenesis and inhibiting fat synthesis. A dynamic model accurately predicts ketone body levels in treated individuals.
Collapse
Affiliation(s)
- Zhijie Wan
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Ming Yuan
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziao Liu
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuan Cai
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua He
- Center of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| | - Kun Hao
- State Key Laboratory of Natural Medicine, Jiangsu Province Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
25
|
Qin W, Ding Y, Zhang W, Sun L, Weng J, Zheng X, Luo S. Small molecule-driven LKB1 deacetylation is responsible for the inhibition of hepatic lipid response in NAFLD. J Lipid Res 2025; 66:100740. [PMID: 39755206 PMCID: PMC11808498 DOI: 10.1016/j.jlr.2024.100740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a progressive condition characterized by ectopic fat accumulation in the liver, for which no FAD-approved drugs currently exist. Emerging evidence highlights the role of liver kinase B1 (LKB1), a key metabolic regulator, has been proposed in NAFLD, particularly in response to excessive nutrient levels. However, few agents have been identified that can prevent the progression of nonalcoholic steatohepatitis (NASH) by targeting LKB1 deacetylation. Through comprehensive screening of our in-house chemical library, we identified tranilast, a small molecule with remarkable inhibitory efficacy against lipid deposition induced by palmitic acid/oleic acid (PO). In this study, we investigated the novel biological function and mechanism of tranilast in regulating hepatic lipid response in NAFLD, focusing on its role in LKB1 deacetylation within hepatocytes. Our findings demonstrate that tranilast effectively reduced hepatic steatosis, inflammation, and fibrosis in NASH models induced by high-fat and high-cholesterol (HFHC) and methionine choline-deficient (MCD) diets. Mechanistic analysis using RNA sequencing revealed that tranilast mitigated hepatic lipid response by promoting LKB1 deacetylation and activating AMPK. Notably, in vivo experiments showed that the beneficial effects of tranilast in MCD diet-induced NASH model were reversed by the compound C (C-C), a known AMPK inhibitor, confirming that tranilast's effects on hepatic lipid response are mediated through the AMPK pathway. In summary, tranilast inhibits hepatic lipid response in NAFLD through LKB1 deacetylation, providing robust experimental evidence for the role of LKB1 in NAFLD. These findings position tranilast as a promising therapeutic candidate for the pharmacological management of metabolic diseases.
Collapse
Affiliation(s)
- Weiwei Qin
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China; Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yu Ding
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wenhao Zhang
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lu Sun
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jianping Weng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xueying Zheng
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Sihui Luo
- Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
26
|
Kim S, Han HJ, Rho H, Kang S, Mukherjee S, Kim J, Kim D, Ko HW, Lim SM, Im SS, Chung JY, Song J. Ebastine-mediated destabilization of E3 ligase MKRN1 protects against metabolic dysfunction-associated steatohepatitis. Cell Mol Life Sci 2025; 82:66. [PMID: 39888429 PMCID: PMC11785899 DOI: 10.1007/s00018-024-05535-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/08/2024] [Accepted: 11/29/2024] [Indexed: 02/01/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic condition encompassing metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), which can progress to fibrosis, cirrhosis, or hepatocellular carcinoma (HCC). The heterogeneous and complex nature of MASLD complicates optimal drug development. Ebastine, an antihistamine, exhibits antitumor activity in various types of cancer. However, its effects on MASH remain unexplored. In the present study, we identified ebastine as a potential treatment for MASH. Our results indicated that ebastine acts as a novel MKRN1 inhibitor by promoting MKRN1 destabilization through self-ubiquitination, leading to AMP-activated protein kinase (AMPK) activation. Ebastine appeared to bind to the C-terminal domain of MKRN1, particularly at residues R298 and K360. Notably, Mkrn1 knockout (KO) mice demonstrated resistance to MASH, including obesity, steatosis, inflammation, and fibrosis under high-fat-high-fructose diet (HFHFD) conditions. Additionally, liver-specific Mkrn1 knockdown using AAV8 alleviated MASH symptoms in HFHFD-fed mice, implicating MKRN1 as a potential therapeutic target. Consistent with these findings, treatment with ebastine significantly reduced the risk of MASH in HFHFD-fed mice, with a decrease in MKRN1 expression and an increase in AMPK activity. Our study suggests that ebastine binds to MKRN1, promoting its destabilization and subsequent degradation by stimulating its ubiquitination. This enhances AMPK stability and activity, suppressing lipid accumulation, inflammation, and fibrosis. Moreover, the knockout of Mkrn1 mice decreased the risk of MASH, suggesting that ebastine could be a promising therapeutic agent for the treatment of MASH.
Collapse
Affiliation(s)
- Seungyeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyun-Ji Han
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Hyunjin Rho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Subin Kang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Sulagna Mukherjee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jiwoo Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Doyoun Kim
- Therapeutics & Biotechnology Division, Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
- Medicinal Chemistry and Pharmacology, Korea University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea
| | - Sang Min Lim
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, 07292, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Joon-Yong Chung
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jaewhan Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
- Department of Biochemistry, College of Life Science and Technology, Institute for Bio-medical Convergence Science and Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Yang W, Zeng S, Shao R, Jin R, Huang J, Wang X, Liu E, Zhou T, Li F, Chen Y, Chen D. Sulforaphane regulation autophagy-mediated pyroptosis in autoimmune hepatitis via AMPK/mTOR pathway. Int Immunopharmacol 2025; 146:113826. [PMID: 39673998 DOI: 10.1016/j.intimp.2024.113826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/07/2024] [Indexed: 12/16/2024]
Abstract
Autoimmune hepatitis (AIH) is a liver disease marked by inflammation of unknown origin. If untreated, it can progress to cirrhosis or liver failure, posing a significant health risk. Currently, effective drug therapies are lacking in clinical practice. Sulforaphane (SFN), a natural anti-inflammatory and antioxidant compound found in various cruciferous vegetables, alleviate pyroptosis and improve impaired autophagic flux, both of which contribute to AIH progression. However, whether SFN modulates autophagic flux and pyroptosis in S100-induced EAH through the AMPK/mTOR pathway remains unclear. Therefore, this study aims to investigate whether SFN can regulate AIH and elucidate its potential mechanisms of action. In this study, experimental AIH (EAH) was induced in male C57BL/6 J mice through intraperitoneal (i.p.) injection of S100. SFN was administered intraperitoneally every other day. After 28 days, the mice were euthanized, and their livers and serum were collected for histological and biochemical analyses. AML12 cells were used for the in vitro studies. The results showed that SFN mitigated pyroptosis by inhibiting the NLRP3 inflammasome and improving autophagic flux, which alleviates S100-induced EAH. Conversely, the autophagy inhibitor 3-MA negated the protective effects of SFN against inflammasome-mediated pyroptosis. Furthermore, SFN activated the AMPK/mTOR signaling pathway, offering protection against S100-induced EAH. Selective inhibition of AMPK suppressed the improvement in autophagic flux and protected against SFN-induced pyroptosis. Overall, SFN significantly ameliorates S100-induced EAH by enhancing autophagic flux and mitigating pyroptosis through activation of the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Weijian Yang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Shiyi Zeng
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Rongrong Shao
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Ru Jin
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Jiayin Huang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Xinyu Wang
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Enqian Liu
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Tenghui Zhou
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Fengfan Li
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China
| | - Yongping Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China.
| | - Dazhi Chen
- Hepatology Diagnosis and Treatment Center, The First Affiliated Hospital of Wenzhou Medical University and Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou 325035, China; School of Clinical Medicine, The First People's Hospital of Lin'an District, Hangzhou, Lin'an People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou 311300, China.
| |
Collapse
|
28
|
An H, Jang Y, Choi J, Hur J, Kim S, Kwon Y. New Insights into AMPK, as a Potential Therapeutic Target in Metabolic Dysfunction-Associated Steatotic Liver Disease and Hepatic Fibrosis. Biomol Ther (Seoul) 2025; 33:18-38. [PMID: 39702310 PMCID: PMC11704404 DOI: 10.4062/biomolther.2024.188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/08/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024] Open
Abstract
AMP-activated protein kinase (AMPK) activators have garnered significant attention for their potential to prevent the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) into liver fibrosis and to fundamentally improve liver function. The broad spectrum of pathways regulated by AMPK activators makes them promising alternatives to conventional liver replacement therapies and the limited pharmacological treatments currently available. In this study, we aim to illustrate the newly detailed multiple mechanisms of MASLD progression based on the multiple-hit hypothesis. This model posits that impaired lipid metabolism, combined with insulin resistance and metabolic imbalance, initiates inflammatory cascades, gut dysbiosis, and the accumulation of toxic metabolites, ultimately promoting fibrosis and accelerating MASLD progression to irreversible hepatocellular carcinoma (HCC). AMPK plays a multifaceted protective role against these pathological conditions by regulating several key downstream signaling pathways. It regulates biological effectors critical to metabolic and inflammatory responses, such as SIRT1, Nrf2, mTOR, and TGF-β, through complex and interrelated mechanisms. Due to these intricate connections, AMPK's role is pivotal in managing metabolic and inflammatory disorders. In this review, we demonstrate the specific roles of AMPK and its related pathways. Several agents directly activate AMPK by binding as agonists, while some others indirectly activate AMPK by modulating upstream molecules, including adiponectin, LKB1, and the AMP: ATP ratio. As AMPK activators can target each stage of MASLD progression, the development of AMPK activators offers immense potential to expand therapeutic strategies for liver diseases such as MASH, MASLD, and liver fibrosis.
Collapse
Affiliation(s)
- Haeun An
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yerin Jang
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jungin Choi
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Juhee Hur
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seojeong Kim
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
29
|
Tang M, Wang X, Wang S, Xing C, Xu Q, Mu Y, Wu X, Zhao ZA, Li F. 10-Hydroxy-2-decenoic acid attenuates nonalcoholic fatty liver disease by activating AMPK-α signaling pathway. Biochem Pharmacol 2025; 231:116648. [PMID: 39581533 DOI: 10.1016/j.bcp.2024.116648] [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: 06/23/2024] [Revised: 11/01/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) originates from metabolic dysfunctions, is one of the most commonly encountered liver disorders worldwide, characterized by ectopic lipid deposition within hepatocytes, accompanied by hepatocellular injury and necroinflammation. Currently, NAFLD has very few treatment options. Purified from royal jelly, 10-hydroxy-2-decenoic acid (10-HDA) is the primary bioactive ingredient with a series of beneficial effects against various metabolic diseases. Herein, we investigated the effects of 10-HDA in methionine and choline deficiency (MCD) diet induced NAFLD model and free fatty acids (FFAs) induced lipid-laden hepatocyte model and explored the underlying mechanisms. In the mice fed with MCD diet, 10-HDA treatment significantly reduced hepatic steatosis, hepatocellular injury, apoptosis, inflammatory response and fibrosis. In vitro, 10-HDA treatment reduced lipid accumulation and apoptosis in hepatocytes induced by FFAs. Mechanistically, 10-HDA therapy restored AMPK-α phosphorylation, leading to the phosphorylation and inactivation acetyl-CoA carboxylase (ACC). Consequently, this increased the expression of carnitine palmitoyl transferase 1α(CPT1α), and peroxisome proliferators-activated receptors α (PPARα), and lowered the expression of cleavage forms of sterol regulatory element binding protein-1 (SREBP-1) and fatty acid synthetase (FASN). Furthermore, pretreating the cells with the AMPK-α inhibitor, compound C, greatly eliminated these beneficial effects of 10-HDA. Additionally, molecular docking analysis indicated that 10-HDA bound the domain of AMPK-α1 subunit. Based on these findings, 10-HDA suppresses hepatic lipogenesis via AMPK-α-dependent suppression of the ACC pathway, thus inhibiting hepatocellular injury, apoptosis, inflammatory response and fibrosis. 10-HDA may represent a promising candidate drug for the treatment of NAFLD.
Collapse
Affiliation(s)
- Minyi Tang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xinzi Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shuai Wang
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Chaofeng Xing
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Southern Medical University, Foshan 528308, China
| | - Qihua Xu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511400, China
| | - Yunping Mu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaoli Wu
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Zijian Allan Zhao
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Southern Medical University, Foshan 528308, China.
| | - Fanghong Li
- The School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
30
|
Song K, Wu Y, Tan S. Caspases in PANoptosis. Curr Res Transl Med 2025; 73:103502. [PMID: 39985853 DOI: 10.1016/j.retram.2025.103502] [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: 06/19/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Recent studies prove that the three well-established cell death pathways-pyroptosis, apoptosis, and necroptosis-are not isolated but rather engage in extensive crosstalk. PANoptosis, a newly identified pathway of inflammatory regulated cell death (RCD), integrates characteristics of apoptosis, pyroptosis, and necroptosis. Caspases are a family of conserved cysteine proteases that play critical roles in pyroptosis, apoptosis, and necroptosis. Similarly, caspases also play a role in PANoptosis. In this paper, we review the molecular mechanisms of these three RCDs and the crosstalk between them. We also delineate the discovery of PANoptosis and its association with disease. Furthermore, we discuss the caspase function in PANoptosis, mainly focusing on caspase-6 and caspase-8 molecules. This review describes the key molecules, especially caspases, in the context of PANoptosis research, aiming to provide a foundation for targeted interventions in PANoptosis-associated diseases.
Collapse
Affiliation(s)
- Kaiyuan Song
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Yongbin Wu
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China
| | - Sipin Tan
- Department of Pathophysiology, Xiangya School of Basic Medicine Science, Central South University, Changsha, PR China; Sepsis Translational Medicine Key Laboratory of Hunan Province, Central South University, Changsha, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, PR China.
| |
Collapse
|
31
|
McAloon LM, Muller AG, Nay K, Lu EL, Smeuninx B, Means AR, Febbraio MA, Scott JW. CaMKK2: bridging the gap between Ca2+ signaling and energy-sensing. Essays Biochem 2024; 68:309-320. [PMID: 39268917 DOI: 10.1042/ebc20240011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Calcium (Ca2+) ions are ubiquitous and indispensable signaling messengers that regulate virtually every cell function. The unique ability of Ca2+ to regulate so many different processes yet cause stimulus specific changes in cell function requires sensing and decoding of Ca2+ signals. Ca2+-sensing proteins, such as calmodulin, decode Ca2+ signals by binding and modifying the function of a diverse range of effector proteins. These effectors include the Ca2+-calmodulin dependent protein kinase kinase-2 (CaMKK2) enzyme, which is the core component of a signaling cascade that plays a key role in important physiological and pathophysiological processes, including brain function and cancer. In addition to its role as a Ca2+ signal decoder, CaMKK2 also serves as an important junction point that connects Ca2+ signaling with energy metabolism. By activating the metabolic regulator AMP-activated protein kinase (AMPK), CaMKK2 integrates Ca2+ signals with cellular energy status, enabling the synchronization of cellular activities regulated by Ca2+ with energy availability. Here, we review the structure, regulation, and function of CaMKK2 and discuss its potential as a treatment target for neurological disorders, metabolic disease, and cancer.
Collapse
Affiliation(s)
- Luke M McAloon
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Abbey G Muller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Kevin Nay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Eudora L Lu
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Benoit Smeuninx
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, U.S.A
| | - Mark A Febbraio
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
| | - John W Scott
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria 3052, Australia
- St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
32
|
Li S, Chen F, Liu M, Zhang Y, Xu J, Li X, Shang Z, Huang S, Song S, Tu C. Knockdown of hepatic mitochondrial calcium uniporter mitigates MASH and fibrosis in mice. Cell Biosci 2024; 14:135. [PMID: 39523398 PMCID: PMC11550531 DOI: 10.1186/s13578-024-01315-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Mitochondrial calcium uniporter (MCU) plays pleiotropic roles in cellular physiology and pathology that contributes to a variety of diseases, but the role and potential mechanism of MCU in the pathogenesis of metabolic dysfunction-associated steatohepatitis (MASH) remain poorly understood. METHODS AND RESULTS Here, hepatic knockdown of MCU in C57BL/6J mice was achieved by tail vein injection of AAV8-mediated the CRISPR/Cas9. Mice were fed a Choline-deficient, L-amino acid-defined high-fat diet (CDAHFD) for 8 weeks to induce MASH and fibrosis. We find that expression of MCU enhanced in MASH livers of humans and mice. MCU knockdown robustly limits lipid droplet accumulation, steatosis, inflammation, and hepatocyte apoptotic death during MASH development both in vivo in mice and in vitro in cellular models. MCU-deficient mice strikingly mitigate MASH-related fibrosis. Moreover, the protective effects of MCU knockdown against MASH progression are accompanied by a reduced level of mitochondrial calcium, limiting hepatic oxidative stress, and attenuating mitochondrial dysfunction. Mechanically, RNA sequencing analysis and protein immunoblotting indicate that knockdown MCU inhibited the Hippo/YAP pathway activation and restored the AMP-activated protein kinase (AMPK) activity during MASH development both in vitro and in vivo. CONCLUSIONS MCU is up-regulated in MASH livers in humans and mice; and hepatic MCU knockdown protects against diet-induced MASH and fibrosis in mice. Thus, targeting MCU may represent a novel therapeutic strategy for MASH and fibrosis.
Collapse
Affiliation(s)
- Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fangyuan Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Min Liu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Yajun Zhang
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jingjing Xu
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Xi Li
- Department of Geriatrics, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhiyin Shang
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Shaoping Huang
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Shu Song
- Department of Pathology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| | - Chuantao Tu
- Department of Gastroenterology, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
33
|
Wang C, Du M, Jiang Z, Cong R, Wang W, Zhang T, Chen J, Zhang G, Li L. PI3K-AKT-mediated phosphorylation of Thr260 in CgCaspase-3/6/7 regulates heat-induced activation in oysters. Commun Biol 2024; 7:1459. [PMID: 39511363 PMCID: PMC11543851 DOI: 10.1038/s42003-024-07184-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024] Open
Abstract
Cysteine-aspartic proteases (caspases) are critical drivers of apoptosis, exhibiting expansion and domain shuffling in mollusks. However, the functions and regulatory mechanisms of these caspases remain unclear. In this study, we identified a group of Caspase-3/6/7 in Bivalvia and Gastropoda with a long inter-subunit linker (IL) that inhibits cleavage activation. Within this region, we found that conserved phosphorylation at Thr260 in oysters, mediated by the PI3K-AKT pathway, suppresses heat-induced activation. This mechanism is involved in divergent temperature adaptation between two allopatric congeneric oyster species, the relatively cold-adapted Crassostrea gigas and warm-adapted Crassostrea angulata. Our study elucidates the role of these effector caspase members and their long IL in bivalves, revealing that the PI3K-AKT pathway phosphorylates Thr260 on CgCASP3/6/7's linker to inhibit heat-induced activation. These findings provide insights into the evolution and function of apoptotic regulatory mechanisms in bivalves.
Collapse
Affiliation(s)
- Chaogang Wang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
| | - Mingyang Du
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuxiang Jiang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rihao Cong
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Wei Wang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Taiping Zhang
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jincheng Chen
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guofan Zhang
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China
| | - Li Li
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture(CAS), Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.
- National and Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China.
- Shandong Technology Innovation Center of Oyster Seed Industry, Qingdao, China.
| |
Collapse
|
34
|
Wu H, Han Y, Liu J, Zhao R, Dai S, Guo Y, Li N, Yang F, Zeng S. The assembly and activation of the PANoptosome promote porcine granulosa cell programmed cell death during follicular atresia. J Anim Sci Biotechnol 2024; 15:147. [PMID: 39497227 PMCID: PMC11536665 DOI: 10.1186/s40104-024-01107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/18/2024] [Indexed: 11/07/2024] Open
Abstract
BACKGROUND Follicular atresia significantly impairs female fertility and hastens reproductive senescence. Apoptosis of granulosa cells is the primary cause of follicular atresia. Pyroptosis and necroptosis, as additional forms of programmed cell death, have been reported in mammalian cells. However, the understanding of pyroptosis and necroptosis pathways in granulosa cells during follicular atresia remains unclear. This study explored the effects of programmed cell death in granulosa cells on follicular atresia and the underlying mechanisms. RESULTS The results revealed that granulosa cells undergo programmed cell death including apoptosis, pyroptosis, and necroptosis during follicular atresia. For the first time, we identified the formation of a PANoptosome complex in porcine granulosa cells. This complex was initially identified as being composed of ZBP1, RIPK3, and RIPK1, and is recruited through the RHIM domain. Additionally, we demonstrated that caspase-6 is activated and cleaved, interacting with RIPK3 as a component of the PANoptosome. Heat stress may exacerbate the activation of the PANoptosome, leading to programmed cell death in granulosa cells. CONCLUSIONS Our data identified the formation of a PANoptosome complex that promoted programmed cell death in granulosa cells during the process of follicular atresia. These findings provide new insights into the molecular mechanisms underlying follicular atresia.
Collapse
Affiliation(s)
- Hao Wu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yingxue Han
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jikang Liu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Rong Zhao
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shizhen Dai
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yajun Guo
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Nan Li
- Department of Clinical Sciences, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Feng Yang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450046, China.
| | - Shenming Zeng
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
35
|
Liu JY, Liu JX, Li R, Zhang ZQ, Zhang XH, Xing SJ, Sui BD, Jin F, Ma B, Zheng CX. AMPK, a hub for the microenvironmental regulation of bone homeostasis and diseases. J Cell Physiol 2024; 239:e31393. [PMID: 39210747 DOI: 10.1002/jcp.31393] [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: 03/19/2024] [Revised: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024]
Abstract
AMP-activated protein kinase (AMPK), a crucial regulatory kinase, monitors energy levels, conserving ATP and boosting synthesis in low-nutrition, low-energy states. Its sensitivity links microenvironmental changes to cellular responses. As the primary support structure and endocrine organ, the maintenance, and repair of bones are closely associated with the microenvironment. While a series of studies have explored the effects of specific microenvironments on bone, there is lack of angles to comprehensively evaluate the interactions between microenvironment and bone cells, especially for bone marrow mesenchymal stem cells (BMMSCs) which mediate the differentiation of osteogenic lineage. It is noteworthy that accumulating evidence has indicated that AMPK may serve as a hub between BMMSCs and microenvironment factors, thus providing a new perspective for us to understand the biology and pathophysiology of stem cells and bone. In this review, we emphasize AMPK's pivotal role in bone microenvironment modulation via ATP, inflammation, reactive oxygen species (ROS), calcium, and glucose, particularly in BMMSCs. We further explore the use of AMPK-activating drugs in the context of osteoarthritis and osteoporosis. Moreover, building upon the foundation of AMPK, we elucidate a viewpoint that facilitates a comprehensive understanding of the dynamic relationship between the microenvironment and bone homeostasis, offering valuable insights for prospective investigations into stem cell biology and the treatment of bone diseases.
Collapse
Affiliation(s)
- Jin-Yu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Jie-Xi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Rang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Zi-Qi Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Xiao-Hui Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Shu-Juan Xing
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
- College of Life Science, Northwest University, Xi'an, China
| | - Bing-Dong Sui
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Fang Jin
- Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, China
| | - Bo Ma
- State Key Laboratory of National Security Specially Needed Medicines, Academy of Military Medical Sciences, Beijing, China
| | - Chen-Xi Zheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
36
|
Yanagi T, Kikuchi H, Takeuchi K, Susa K, Mori T, Chiga M, Yamamoto K, Furukawa A, Kanazawa T, Kato Y, Takahashi N, Suzuki T, Mori Y, Carter BC, Mori M, Nakano Y, Fujiki T, Hara Y, Suzuki S, Ando F, Mandai S, Honda S, Torii S, Shimizu S, Tanaka H, Fujii Y, Rai T, Uchida S, Sohara E. ULK1-regulated AMP sensing by AMPK and its application for the treatment of chronic kidney disease. Kidney Int 2024; 106:887-906. [PMID: 39428173 DOI: 10.1016/j.kint.2024.08.024] [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: 08/17/2023] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 10/22/2024]
Abstract
Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a central kinase involved in energy homeostasis. Increased intracellular AMP levels result in AMPK activation through the binding of AMP to the γ-subunit of AMPK. Recently, we reported that AMP-induced AMPK activation is impaired in the kidneys in chronic kidney disease (CKD) despite an increase in the AMP/ATP ratio. However, the mechanisms by which AMP sensing is disrupted in CKD are unclear. Here, we identified mechanisms of energy homeostasis in which Unc-51-like kinase 1 (ULK1)-dependent phosphorylation of AMPKγ1 at Ser260/Thr262 promoting AMP sensitivity of AMPK. AMPK activation by AMP was impaired in Ulk1 knockout mice despite an increased AMP/ATP ratio. ULK1 expression is markedly downregulated in CKD kidneys, leading to AMP sensing failure. Additionally, MK8722, an allosteric AMPK activator, stimulated AMPK in the kidneys of a CKD mouse model (5/6th nephrectomy) via a pathway that is independent of AMP sensing. Thus, our study shows that MK8722 treatment significantly attenuates the deterioration of kidney function in CKD and may be a potential therapeutic option in CKD therapeutics.
Collapse
Affiliation(s)
- Tomoki Yanagi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroaki Kikuchi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| | - Koh Takeuchi
- Graduate School of Pharmaceutical Sciences, the University of Tokyo, Tokyo, Japan
| | - Koichiro Susa
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takayasu Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Motoko Chiga
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Asuka Furukawa
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takumi Kanazawa
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuki Kato
- Department of Human Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Naohiro Takahashi
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takefumi Suzuki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yutaro Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Benjamin C Carter
- Department of Biochemistry, Purdue University, West Lafayette, Indiana, USA
| | - Makiko Mori
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuta Nakano
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tamami Fujiki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yu Hara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Soichiro Suzuki
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Fumiaki Ando
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shintaro Mandai
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shinya Honda
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Satoru Torii
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shigeomi Shimizu
- Department of Pathological Cell Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hajime Tanaka
- Department of Urology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yasuhisa Fujii
- Department of Urology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tatemitsu Rai
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan; Department of Nephrology and Hypertension, Dokkyo Medical University, Tochigi, Japan
| | - Shinichi Uchida
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.
| |
Collapse
|
37
|
Wang P, Li J, Ji M, Pan J, Cao Y, Kong Y, Zhu L, Li J, Li B, Chang L, Zhang Z. Vitamin D receptor attenuates carbon tetrachloride-induced liver fibrosis via downregulation of YAP. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135480. [PMID: 39146589 DOI: 10.1016/j.jhazmat.2024.135480] [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: 05/13/2024] [Revised: 08/02/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024]
Abstract
Liver fibrosis is characterized by the excessive accumulation of extracellular matrix proteins, which can lead to cirrhosis and liver cancer. Metabolic dysfunction-associated steatosis liver diseases are common causes of liver fibrosis, sharing a similar pathogenesis with carbon tetrachloride (CCl₄) exposure. This process involves the activation of hepatic stellate cells (HSCs) into myofibroblasts. However, the detailed mechanism and effective treatment strategies require further investigation. In this study, we uncovered a negative correlation between VDR expression and YAP within HSCs. Subsequently, we demonstrated that VDR exerted a downregulatory influence on YAP transcriptional activity in HSCs. Intriguingly, activation VDR effectively inhibited the culture induced activation of primary HSCs by suppressing the transcriptional activity of early YAP. Furthermore, in vivo results manifested that hepatic-specific deletion of YAP/TAZ ameliorates CCl4-induced liver fibrosis, and nullified the antifibrotic efficacy of VDR. Importantly, a YAP inhibitor rescued the exacerbation of liver fibrosis induced by hepatic-specific VDR knockout. Moreover, the combined pharmacological of VDR agonist and YAP inhibitor demonstrated a synergistic effect in diminishing CCl4-induced liver fibrosis, primary HSCs activation and hepatic injury in vivo. These effects were underpinned by their collective ability to inhibit HSC activation through AMPK activation, consequently curbing ATP synthesis and HSCs proliferation. In conclusion, our results not only revealed the inhibition of VDR on YAP-activated liver stellate cells but also identified a synergistic effect of VDR agonist and YAP inhibitor in an AMPKα-dependent manner, providing a practical foundation for integration of multi-targeted drugs in the therapy of CCl4-induced hepatic fibrosis.
Collapse
Affiliation(s)
- Ping Wang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jie Li
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Mintao Ji
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity. The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Jinjing Pan
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Yanmei Cao
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou 215007, China
| | - Yulin Kong
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou 215007, China
| | - Li Zhu
- Department of Infectious Diseases, The Affiliated Infectious Diseases Hospital of Soochow University, Suzhou 215007, China
| | - Jiafu Li
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China
| | - Bingyan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| | - Lei Chang
- State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Jiangsu Key Laboratory of Infection and Immunity. The Fourth Affiliated Hospital of Soochow University, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou 215123, China; Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200433, China.
| | - Zengli Zhang
- Department of Occupational and Environmental Health, School of Public Health, Suzhou Medical College of Soochow University, Suzhou 215123, China.
| |
Collapse
|
38
|
Chu X, Liu S, Qu B, Xin Y, Lu L. Salidroside may target PPARα to exert preventive and therapeutic activities on NASH. Front Pharmacol 2024; 15:1433076. [PMID: 39415834 PMCID: PMC11479876 DOI: 10.3389/fphar.2024.1433076] [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: 05/15/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
Background Salidroside (SDS), a phenylpropanoid glycoside, is an antioxidant component isolated from the traditional Chinese medicine Rhodiola rosea and has multifunctional bioactivities, particularly possessing potent hepatoprotective function. Non-alcoholic steatohepatitis (NASH) is one of the most prevalent chronic liver diseases worldwide, but it still lacks efficient drugs. This study aimed to assess the preventive and therapeutic effects of SDS on NASH and its underlying mechanisms in a mouse model subjected to a methionine- and choline-deficient (MCD) diet. Methods C57BL/6J mice were fed an MCD diet to induce NASH. During or after the formation of the MCD-induced NASH model, SDS (24 mg/kg/day) was supplied as a form of diet for 4 weeks. The histopathological changes were evaluated by H&E staining. Oil Red O staining and Sirius Red staining were used to quantitatively determine the lipid accumulation and collagen fibers in the liver. Serum lipid and liver enzyme levels were measured. The morphology of autophagic vesicles and autophagosomes was observed by transmission electron microscopy (TEM), and qRT-PCR and Western blotting were used to detect autophagy-related factor levels. Immunohistochemistry and TUNEL staining were used to evaluate the apoptosis of liver tissues. Flow cytometry was used to detect the composition of immune cells. ELISA was used to evaluate the expression of serum inflammatory factors. Transcript-proteome sequencing, molecular docking, qRT-PCR, and Western blotting were performed to explore the mechanism and target of SDS in NASH. Results The oral administration of SDS demonstrated comprehensive efficacy in NASH. SDS showed both promising preventive and therapeutic effects on NASH in vivo. SDS could upregulate autophagy, downregulate apoptosis, rebalance immunity, and alleviate inflammation to exert anti-NASH properties. Finally, the results of transcript-proteome sequencing, molecular docking evaluation, and experimental validation showed that SDS might exert its multiple effects through targeting PPARα. Conclusion Our findings revealed that SDS could regulate liver autophagy and apoptosis, regulating both innate immunity and adaptive immunity and alleviating inflammation in NASH prevention and therapy via the PPAR pathway, suggesting that SDS could be a potential anti-NASH drug in the future.
Collapse
Affiliation(s)
- Xueru Chu
- Department of Infectious Disease, Qingdao Municipal Hospital, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, China
| | - Shousheng Liu
- Clinical Research Center, Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Baozhen Qu
- Qingdao Cancer Prevention and Treatment Research Institute, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Yongning Xin
- Department of Infectious Disease, Qingdao Municipal Hospital, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Department of Infectious Disease, Qingdao Municipal Hospital, Qingdao, China
| | - Linlin Lu
- Qingdao Cancer Prevention and Treatment Research Institute, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, China
| |
Collapse
|
39
|
Zhang M, Barroso E, Peña L, Rada P, Valverde ÁM, Wahli W, Palomer X, Vázquez-Carrera M. PPARβ/δ attenuates hepatic fibrosis by reducing SMAD3 phosphorylation and p300 levels via AMPK in hepatic stellate cells. Biomed Pharmacother 2024; 179:117303. [PMID: 39153437 DOI: 10.1016/j.biopha.2024.117303] [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: 06/05/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
The role of peroxisome proliferator-activated receptor (PPAR)β/δ in hepatic fibrosis remains a subject of debate. Here, we examined the effects of a PPARβ/δ agonist on the pathogenesis of liver fibrosis and the activation of hepatic stellate cells (HSCs), the main effector cells in liver fibrosis, in response to the pro-fibrotic stimulus transforming growth factor-β (TGF-β). The PPARβ/δ agonist GW501516 completely prevented glucose intolerance and peripheral insulin resistance, blocked the accumulation of collagen in the liver, and attenuated the expression of inflammatory and fibrogenic genes in mice fed a choline-deficient high-fat diet (CD-HFD). The antifibrogenic effect of GW501516 observed in the livers CD-HFD-fed mice could occur through an action on HSCs since primary HSCs isolated from Ppard-/- mice showed increased mRNA levels of the profibrotic gene Col1a1. Moreover, PPARβ/δ activation abrogated TGF-β1-mediated cell migration (an indicator of cell activation) in LX-2 cells (immortalized activated human HSCs). Likewise, GW501516 attenuated the phosphorylation of the main downstream intracellular protein target of TGF-β1, suppressor of mothers against decapentaplegic (SMAD)3, as well as the levels of the SMAD3 co-activator p300 via the activation of AMP-activated protein kinase (AMPK) and the subsequent inhibition of extracellular signal-regulated kinase-1/2 (ERK1/2) in LX-2 cells. Overall, these findings uncover a new mechanism by which the activation of AMPK by a PPARβ/δ agonist reduces TGF-β1-mediated activation of HSCs and fibrosis via the reduction of both SMAD3 phosphorylation and p300 levels.
Collapse
Affiliation(s)
- Meijian Zhang
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Emma Barroso
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| | - Lucía Peña
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Patricia Rada
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Ángela M Valverde
- Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC/UAM), Madrid, Spain
| | - Walter Wahli
- Center for Integrative Genomics, University of Lausanne, Lausanne CH-1015, Switzerland; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; ToxAlim (Research Center in Food Toxicology), INRAE, UMR1331, Toulouse Cedex F-31300, France
| | - Xavier Palomer
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain
| | - Manuel Vázquez-Carrera
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), University of Barcelona, Barcelona 08028, Spain; Spanish Biomedical Research Center in Diabetes and Associated Metabolic Diseases (CIBERDEM)-Instituto de Salud Carlos III, Madrid 28029, Spain; Pediatric Research Institute-Hospital Sant Joan de Déu, Esplugues de Llobregat 08950, Spain.
| |
Collapse
|
40
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
41
|
Zhou B, Luo Y, Bi H, Zhang N, Ma M, Dong Z, Ji N, Zhang S, Wang X, Liu Y, Guo X, Wei W, Xie C, Wu L, Wan X, Zheng MH, Zhao B, Li Y, Hu C, Lu Y. Amelioration of nonalcoholic fatty liver disease by inhibiting the deubiquitylating enzyme RPN11. Cell Metab 2024; 36:2228-2244.e7. [PMID: 39146936 DOI: 10.1016/j.cmet.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 05/16/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD), including its more severe manifestation nonalcoholic steatohepatitis (NASH), is a global public health challenge. Here, we explore the role of deubiquitinating enzyme RPN11 in NAFLD and NASH. Hepatocyte-specific RPN11 knockout mice are protected from diet-induced liver steatosis, insulin resistance, and steatohepatitis. Mechanistically, RPN11 deubiquitinates and stabilizes METTL3 to enhance the m6A modification and expression of acyl-coenzyme A (CoA) synthetase short-chain family member 3 (ACSS3), which generates propionyl-CoA to upregulate lipid metabolism genes via histone propionylation. The RPN11-METTL3-ACSS3-histone propionylation pathway is activated in the livers of patients with NAFLD. Pharmacological inhibition of RPN11 by Capzimin ameliorated NAFLD, NASH, and related metabolic disorders in mice and reduced lipid contents in human hepatocytes cultured in 2D and 3D. These results demonstrate that RPN11 is a novel regulator of NAFLD/NASH and that suppressing RPN11 has therapeutic potential for the treatment.
Collapse
Affiliation(s)
- Bing Zhou
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunchen Luo
- Department of Endocrinology and Metabolism, Shanghai General Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Hanqi Bi
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ni Zhang
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mingyue Ma
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhixia Dong
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nana Ji
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuo Zhang
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoye Wang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yuejun Liu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaozhen Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Wei Wei
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Cen Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ling Wu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinjian Wan
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Hua Zheng
- MAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China.
| | - Yao Li
- Department of Laboratory Animal Science, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai, China.
| | - Yan Lu
- Institute of Metabolism and Regenerative Medicine, Digestive Endoscopic Center, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
42
|
Liu X, Liang Q, Wang Y, Xiong S, Yue R. Advances in the pharmacological mechanisms of berberine in the treatment of fibrosis. Front Pharmacol 2024; 15:1455058. [PMID: 39372209 PMCID: PMC11450235 DOI: 10.3389/fphar.2024.1455058] [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: 06/26/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
The rising incidence of fibrosis poses a major threat to global public health, and the continuous exploration of natural products for the effective treatment of fibrotic diseases is crucial. Berberine (BBR), an isoquinoline alkaloid, is widely used clinically for its anti-inflammatory, anti-tumor and anti-fibrotic pharmacological effects. Until now, researchers have worked to explore the mechanisms of BBR for the treatment of fibrosis, and multiple studies have found that BBR attenuates fibrosis through different pathways such as TGF-β/Smad, AMPK, Nrf2, PPAR-γ, NF-κB, and Notch/snail axis. This review describes the anti-fibrotic mechanism of BBR and its derivatives, and the safety evaluation and toxicity studies of BBR. This provides important therapeutic clues and strategies for exploring new drugs for the treatment of fibrosis. Nevertheless, more studies, especially clinical studies, are still needed. We believe that with the continuous implementation of high-quality studies, significant progress will be made in the treatment of fibrosis.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qingzhi Liang
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | | | - Shuai Xiong
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Rensong Yue
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Clinical Medical School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
43
|
Li B, Xiao Q, Zhao H, Zhang J, Yang C, Zou Y, Zhang B, Liu J, Sun H, Liu H. Schisanhenol ameliorates non-alcoholic fatty liver disease via inhibiting miR-802 activation of AMPK-mediated modulation of hepatic lipid metabolism. Acta Pharm Sin B 2024; 14:3949-3963. [PMID: 39309511 PMCID: PMC11413670 DOI: 10.1016/j.apsb.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 09/25/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), characterized by hepatic steatosis, is a common metabolic liver disease worldwide. Currently, satisfactory drugs for NAFLD treatment remain lacking. Obesity and diabetes are the leading causes of NAFLD, and compounds with anti-obesity and anti-diabetic activities are considered suitable candidates for treating NAFLD. In this study, biochemical and histological assays revealed that a natural lignan schisanhenol (SAL) effectively decreased lipid accumulation and improved hepatic steatosis in free fatty acid (FFA)-treated HepG2 cells and high-fat diet (HFD)-induced NAFLD mice. Further, molecular analyses, microRNA (miRNA)-seq, and bioinformatics analyses revealed that SAL may improve NAFLD by targeting the miR-802/adenosine monophosphate-activated protein kinase (AMPK) pathway. Liver-specific overexpression of miR-802 in NAFLD mice significantly impaired SAL-mediated liver protection and decreased the protein levels of phosphorylated (p)-AMPK and PRKAB1. Dual-luciferase assay analysis further confirmed that miR-802 inhibits hepatic AMPK expression by binding to the 3' untranslated region of mouse Prkab1 or human PRKAA1. Additionally, genetic silencing of PRKAA1 blocked SAL-induced AMPK pathway activation in FFA-treated HepG2 cells. The results demonstrate that SAL is an effective drug candidate for treating NAFLD through regulating miR-802/AMPK-mediated lipid metabolism.
Collapse
Affiliation(s)
- Bin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Qi Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Hongmei Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing 100005, China
| | - Jianuo Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Chunyan Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Yucen Zou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Bengang Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Jiushi Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Haitao Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Haitao Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| |
Collapse
|
44
|
Fernández-Tussy P, Cardelo MP, Zhang H, Sun J, Price NL, Boutagy NE, Goedeke L, Cadena-Sandoval M, Xirouchaki CE, Brown W, Yang X, Pastor-Rojo O, Haeusler RA, Bennett AM, Tiganis T, Suárez Y, Fernández-Hernando C. miR-33 deletion in hepatocytes attenuates MASLD-MASH-HCC progression. JCI Insight 2024; 9:e168476. [PMID: 39190492 PMCID: PMC11466198 DOI: 10.1172/jci.insight.168476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 08/21/2024] [Indexed: 08/29/2024] Open
Abstract
The complexity of the mechanisms underlying metabolic dysfunction-associated steatotic liver disease (MASLD) progression remains a significant challenge for the development of effective therapeutics. miRNAs have shown great promise as regulators of biological processes and as therapeutic targets for complex diseases. Here, we study the role of hepatic miR-33, an important regulator of lipid metabolism, during the progression of MASLD and the development of hepatocellular carcinoma (HCC). We report that miR-33 was elevated in the livers of humans and mice with MASLD and that its deletion in hepatocytes (miR-33 HKO) improved multiple aspects of the disease, including steatosis and inflammation, limiting the progression to metabolic dysfunction-associated steatotic hepatitis (MASH), fibrosis, and HCC. Mechanistically, hepatic miR-33 deletion reduced lipid synthesis and promoted mitochondrial fatty acid oxidation, reducing lipid burden. Additionally, absence of miR-33 altered the expression of several known miR-33 target genes involved in metabolism and resulted in improved mitochondrial function and reduced oxidative stress. The reduction in lipid accumulation and liver injury resulted in decreased YAP/TAZ pathway activation, which may be involved in the reduced HCC progression in HKO livers. Together, these results suggest suppressing hepatic miR-33 may be an effective therapeutic approach to temper the development of MASLD, MASH, and HCC in obesity.
Collapse
Affiliation(s)
- Pablo Fernández-Tussy
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Magdalena P. Cardelo
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Hanming Zhang
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
| | - Jonathan Sun
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Nathan L. Price
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Experimental Gerontology Section, Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, Maryland, USA
| | - Nabil E. Boutagy
- Vascular Biology and Therapeutics Program
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Leigh Goedeke
- Cardiovascular Research Institute and Division of Cardiology, Department of Medicine; and
- Diabetes, Obesity and Metabolism Institute and Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Martí Cadena-Sandoval
- Department of Pathology & Cell Biology and Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Chrysovalantou E. Xirouchaki
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Surgery, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Wendy Brown
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Xiaoyong Yang
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Molecular and Cellular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Oscar Pastor-Rojo
- Vascular Biology and Therapeutics Program
- Servicio de Bioquímica Clínica, Hospital Universitario Ramón y Cajal IRYCIS, Madrid, Spain
- Departamento de Biología de Sistemas, Universidad de Alcalá de Henares, Madrid, Spain
| | - Rebecca A. Haeusler
- Department of Pathology & Cell Biology and Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Anton M. Bennett
- Yale Center for Molecular and System Metabolism, and
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Department of Surgery, Alfred Hospital and Monash University, Melbourne, Victoria, Australia
| | - Yajaira Suárez
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Carlos Fernández-Hernando
- Vascular Biology and Therapeutics Program
- Department of Comparative Medicine
- Yale Center for Molecular and System Metabolism, and
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
45
|
Xue J, Liu Y, Liu B, Jia X, Fang X, Qin S, Zhang Y. Celastrus orbiculatus Thunb. extracts and celastrol alleviate NAFLD by preserving mitochondrial function through activating the FGF21/AMPK/PGC-1α pathway. Front Pharmacol 2024; 15:1444117. [PMID: 39161898 PMCID: PMC11330833 DOI: 10.3389/fphar.2024.1444117] [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: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease globally, characterized by the accumulation of lipids, oxidative stress, and mitochondrial dysfunction in the liver. Celastrus orbiculatus Thunb. (COT) and its active compound celastrol (CEL) have demonstrated antioxidant and anti-inflammatory properties. Our prior research has shown the beneficial effects of COT in mitigating NAFLD induced by a high-fat diet (HFD) in guinea pigs by reducing hepatic lipid levels and inhibiting oxidative stress. This study further assessed the effects of COT on NAFLD and explored its underlying mitochondria-related mechanisms. Methods COT extract or CEL was administered as an intervention in C57BL/6J mice fed a HFD or in HepG2 cells treated with sodium oleate. Oral glucose tolerance test, biochemical parameters including liver enzymes, blood lipid, and pro-inflammatory factors, and steatosis were evaluated. Meanwhile, mitochondrial ultrastructure and indicators related to oxidative stress were tested. Furthermore, regulators of mitochondrial function were measured using RT-qPCR and Western blot. Results The findings demonstrated significant reductions in hepatic steatosis, oxidative stress, and inflammation associated with NAFLD in both experimental models following treatment with COT extract or CEL. Additionally, improvements were observed in mitochondrial structure, ATP content, and ATPase activity. This improvement can be attributed to the significant upregulation of mRNA and protein expression levels of key regulators including FGF21, AMPK, PGC-1α, PPARγ, and SIRT3. Conclusion These findings suggest that COT may enhance mitochondrial function by activating the FGF21/AMPK/PGC-1α signaling pathway to mitigate NAFLD, which indicated that COT has the potential to target mitochondria and serve as a novel therapeutic option for NAFLD.
Collapse
Affiliation(s)
- Junli Xue
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Yunchao Liu
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Boyan Liu
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Xiubin Jia
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| | - Xinsheng Fang
- College of Agronomy, Shandong Agricultural University, Tai’an, Shandong, China
| | - Shucun Qin
- Taishan Institute for Hydrogen Biomedicine, The Second Affiliated Hospital of Shandong First Medical University and Shandong Academy of Medical Sciences, Tai’an, Shandong, China
| | - Ying Zhang
- School of Pharmaceutical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji’nan, Shandong, China
| |
Collapse
|
46
|
Chakraborty S, Mishra A, Choudhuri A, Bhaumik T, Sengupta R. Leveraging the redundancy of S-denitrosylases in response to S-nitrosylation of caspases: Experimental strategies and beyond. Nitric Oxide 2024; 149:18-31. [PMID: 38823434 DOI: 10.1016/j.niox.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Redox-based protein posttranslational modifications, such as S-nitrosylation of critical, active site cysteine thiols have garnered significant clinical attention and research interest, reasoning for one of the crucial biological implications of reactive messenger molecule, nitric oxide in the cellular repertoire. The stringency of the S-(de)nitrosylation-based redox switch governs the activity and contribution of several susceptible enzymes in signal transduction processes and diverse pathophysiological settings, thus establishing it as a transient yet reasonable, and regulated mechanism of NO adduction and release. Notably, endogenous proteases like cytosolic and mitochondrial caspases with a molecular weight ranging from 33 to 55 kDa are susceptible to performing this biochemistry in the presence of major oxidoreductases, which further unveils the enormous redox-mediated regulational control of caspases in the etiology of diseases. In addition to advancing the progress of the current state of understanding of 'redox biochemistry' in the field of medicine and enriching the existing dynamic S-nitrosoproteome, this review stands as a testament to an unprecedented shift in the underpinnings for redundancy and redox relay between the major redoxin/antioxidant systems, fine-tuning of which can command the apoptotic control of caspases at the face of nitro-oxidative stress. These intricate functional overlaps and cellular backups, supported rationally by kinetically favorable reaction mechanisms suggest the physiological relevance of identifying and involving such cognate substrates for cellular S-denitrosylases that can shed light on the bigger picture of extensively proposing targeted therapies and redox-based drug designing to potentially alleviate the side effects of NOx/ROS in disease pathogenesis.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Tamal Bhaumik
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
47
|
Wang S, Chen B, Du R, Zhong M, Zhang C, Jin X, Cui X, Zhou Y, Kang Q, Xu H, Li Y, Wu Q, Tong G, Luo L. An herbal formulation "Shugan Xiaozhi decoction" ameliorates methionine/choline deficiency-induced nonalcoholic steatohepatitis through regulating inflammation and apoptosis-related pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118127. [PMID: 38583728 DOI: 10.1016/j.jep.2024.118127] [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: 12/23/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shugan Xiaozhi (SGXZ) decoction is a traditional Chinese medicine used for treating nonalcoholic steatohepatitis (NASH). It has been used clinically for over 20 years and proved to be effective; however, the molecular mechanism underlying the effects of SGXZ decoction remains unclear. AIM OF THE STUDY We analyzed the chemical components, core targets, and molecular mechanisms of SGXZ decoction to improve NASH through network pharmacology and in vivo experiments. MATERIALS AND METHODS The chemical components, core targets, and related signaling pathways of SGXZ decoction intervention in NASH were predicted using network pharmacology. Molecular docking was performed to verify chemical components and their core targets. The results were validated in the NASH model treated with SGXZ decoction. Mouse liver function was assessed by measuring ALT and AST levels. TC and TG levels were determined to evaluate lipid metabolism, and lipid deposition was assessed via oil red O staining. Mouse liver damage was determined via microscopy following hematoxylin and eosin staining. Liver fibrosis was assessed via Masson staining. Western blot (WB) and immunohistochemical (IHC) analyses were performed to detect inflammation and the expression of apoptosis-related proteins, including IL-1β, IL-6, IL-18, TNF-α, MCP1, p53, FAS, Caspase-8, Caspase-3, Caspase-9, Bax, Bid, Cytochrome c, Bcl-2, and Bcl-XL. In addition, WB and IHC were used to assess protein expression associated with the TLR4/MyD88/NF-κB pathway. RESULTS Quercetin, luteolin, kaempferol, naringenin, and nobiletin in SGXZ decoction were effective chemical components in improving NASH, and TNF-α, IL-6, and IL-1β were the major core targets. Molecular docking indicated that these chemical components and major core targets might interact. KEGG pathway analysis showed that the pathways affected by SGXZ decoction, primarily including apoptosis and TLR4/NF-κB signaling pathways, interfere with NASH. In vivo experiments indicated that SGXZ decoction considerably ameliorated liver damage, fibrosis, and lipid metabolism disorder in MCD-induced NASH mouse models. In addition, WB and IHC verified the underlying molecular mechanisms of SGXZ decoction as predicted via network pharmacology. SGXZ decoction inhibited the activation of apoptosis-related pathways in MCD-induced NASH mice. Moreover, SGXZ decoction suppressed the activation of TLR4/MyD88/NF-κB pathway in MCD-induced NASH mice. CONCLUSION SGXZ decoction can treat NASH through multiple targets and pathways. These findings provide new insights into the effective treatment of NASH using SGXZ decoction.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Bohao Chen
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Ruili Du
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Mei Zhong
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Chunmei Zhang
- School of Basic Medical Science of Luoyang Polytechnic, No. 6 Keji Avenue, Yibin District, Henan, 471099, China
| | - Xiaoming Jin
- Department of Nephrology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Xiang Cui
- Ankang Traditional Chinese Medicine Hospital, Ankang, 725000, Shaanxi, China
| | - Yuhang Zhou
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Qinyang Kang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Hang Xu
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Yuting Li
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China.
| | - Guangdong Tong
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau, 999078, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China.
| | - Lidan Luo
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China; Shenzhen Key Laboratory of Liver Diseases of Traditional Chinese Medicine, No.15, Yingchun Road, Luohu District, Guangdong, 518033, China.
| |
Collapse
|
48
|
Cheng Z, Chu H, Seki E, Lin R, Yang L. Hepatocyte programmed cell death: the trigger for inflammation and fibrosis in metabolic dysfunction-associated steatohepatitis. Front Cell Dev Biol 2024; 12:1431921. [PMID: 39071804 PMCID: PMC11272544 DOI: 10.3389/fcell.2024.1431921] [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: 05/13/2024] [Accepted: 06/28/2024] [Indexed: 07/30/2024] Open
Abstract
By replacing and removing defective or infected cells, programmed cell death (PCD) contributes to homeostasis maintenance and body development, which is ubiquitously present in mammals and can occur at any time. Besides apoptosis, more novel modalities of PCD have been described recently, such as necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death. PCD not only regulates multiple physiological processes, but also participates in the pathogenesis of diverse disorders, including metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD is mainly classified into metabolic dysfunction-associated steatotic liver (MASL) and metabolic dysfunction-associated steatohepatitis (MASH), and the latter putatively progresses to cirrhosis and hepatocellular carcinoma. Owing to increased incidence and obscure etiology of MASH, its management still remains a tremendous challenge. Recently, hepatocyte PCD has been attracted much attention as a potent driver of the pathological progression from MASL to MASH, and some pharmacological agents have been proved to exert their salutary effects on MASH partly via the regulation of the activity of hepatocyte PCD. The current review recapitulates the pathogenesis of different modalities of PCD, clarifies the mechanisms underlying how metabolic disorders in MASLD induce hepatocyte PCD and how hepatocyte PCD contributes to inflammatory and fibrotic progression of MASH, discusses several signaling pathways in hepatocytes governing the execution of PCD, and summarizes some potential pharmacological agents for MASH treatment which exert their therapeutic effects partly via the regulation of hepatocyte PCD. These findings indicate that hepatocyte PCD putatively represents a new therapeutic point of intervention for MASH.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ekihiro Seki
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rong Lin
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
49
|
Lv M, Chen S, Shan M, Si Y, Huang C, Chen J, Gong L. Arctigenin induces activated HSCs quiescence via AMPK-PPARγ pathway to ameliorate liver fibrosis in mice. Eur J Pharmacol 2024; 974:176629. [PMID: 38679116 DOI: 10.1016/j.ejphar.2024.176629] [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: 01/17/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/01/2024]
Abstract
Arctigenin (ATG), a traditional Chinese herbal medicine, is a natural lignan compound extracted from the seeds of burdock (Arctium lappa L, Asteraceae). As a natural product with multiple biological activities, the effect and mechanism of ATG against liver fibrosis are not fully elucidated yet. In current work, we first discovered that ATG could improve CCl4-induced liver injury reflected by lower plasma ALT and AST levels, liver coefficient and pathological scoring of ballooning. Furthermore, it also could reduce the positive areas of Masson, Sirius red and α-SMA staining, inhibit the expression of fibrosis-related genes (Col1a1, Col3a1, Acta2), and decrease the content of hydroxyproline, indicated ATG treatment had benefits in alleviating CCl4-induced liver fibrosis. In vitro, we observed that ATG can inhibit collagen production stimulated by TGF-β1 in LX2 cells. By analysis of the information obtained from SymMap and GeneCards databases and in vitro validation experiments, ATG was proven to be an indirect PPARγ agonist and its effect on collagen production was dependent on PPARγ. Subsequently, we confirmed that ATG activating AMPK was the contributor of its effect on PPARγ and collagen production. Finally, the transformation of activated hepatic stellate cells was determined after treated with ATG, in which ATG treatment could return activated LX2 cells to quiescence because of the elevated quiescent markers and lipid droplets. Our work has highlighted the potential of ATG in the treatment of liver fibrosis and clarified that ATG can activate AMPK/PPARγ pathway to restore the activated hepatic stellate cell to quiescence thereby improving liver fibrosis.
Collapse
Affiliation(s)
- Mengjia Lv
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Shiyi Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Mengwen Shan
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Yuan Si
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China
| | - Chenggang Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China.
| | - Jing Chen
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| | - Likun Gong
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, Shanghai, 201203, China; University of Chinese Academy of Sciences, No.19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
50
|
Zhou Z, Jiang X, Yi L, Li C, Wang H, Xiong W, Li Z, Shen J. Mitochondria Energy Metabolism Depression as Novel Adjuvant to Sensitize Radiotherapy and Inhibit Radiation Induced-Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401394. [PMID: 38715382 PMCID: PMC11234447 DOI: 10.1002/advs.202401394] [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: 02/07/2024] [Indexed: 07/11/2024]
Abstract
Currently, the typical combination therapy of programmed death ligand-1 (PD-L1) antibodies with radiotherapy (RT) still exhibits impaired immunogenic antitumor response in clinical due to lessened DNA damage and acquired immune tolerance via the upregulation of some other immune checkpoint inhibitors. Apart from this, such combination therapy may raise the occurrence rate of radiation-induced lung fibrosis (RIPF) due to enhanced systemic inflammation, leading to the ultimate death of cancer patients (average survival time of about 3 years). Therefore, it is newly revealed that mitochondria energy metabolism regulation can be used as a novel effective PD-L1 and transforming growth factor-β (TGF-β) dual-downregulation method. Following this, IR-TAM is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with oxidative phosphorylation (OXPHOS) inhibitor Tamoxifen (TAM), which then self-assembled with albumin (Alb) to form IR-TAM@Alb nanoparticles. By doing this, tumor-targeting IR-TAM@Alb nanoparticle effectively reversed tumor hypoxia and depressed PD-L1 and TGF-β expression to sensitize RT. Meanwhile, due to the capacity of heptamethine cyanine dye in targeting RIPF and the function of TAM in depressing TGF-β, IR-TAM@Alb also ameliorated fibrosis development induced by RT.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Yi
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Cheng Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Xiong
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Zhipeng Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|