1
|
Li Z, Chen Y, Shi T, Cao H, Chen G, Yu L. Potential of queen bee larvae as a dietary supplement for obesity management: modulating the gut microbiota and promoting liver lipid metabolism. Food Funct 2025; 16:3848-3861. [PMID: 40131738 DOI: 10.1039/d5fo00166h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Queen bee larvae (QBL) have been consumed as both a traditional food and medicine in China for thousands of years; however, their specific benefits for human health, particularly their potential anti-obesity property, remain underexplored. This study investigated the anti-obesity effect of QBL freeze-dried powder (QBLF) on high-fat diet (HFD) induced obesity in mice and explored the underlying mechanisms. Our findings showed that QBLF effectively reduced body weight, fasting blood glucose levels, lipid accumulation, and inflammation in HFD mice. 16S rRNA sequencing revealed that QBLF significantly modulated the gut microbiota disrupted by an HFD, notably increasing the relative abundance of beneficial microbes such as Ileibacterium, Clostridium sensu stricto 1, Incertae sedis, Streptococcus, Lactococcus, Clostridia UCG-014, and Lachnospiraceae UCG-006, which were inversely associated with obesity-related phenotypes in the mice. RNA sequencing analysis further demonstrated that QBLF intervention upregulated the expression of genes involved in liver lipid metabolism, including Pck1, Cyp4a10, Cyp4a14, and G6pc, while downregulating genes associated with the inflammatory response, such as Cxcl10, Ccl2, Traf1, Mapk15, Lcn2, and Fosb. These results suggested that QBLF can ameliorate HFD-induced obesity through regulating the gut microbiota, promoting liver lipid metabolism, and reducing inflammatory response.
Collapse
Affiliation(s)
- Zhuang Li
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Yiang Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Tengfei Shi
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Haiqun Cao
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| | - Guijie Chen
- National Key Laboratory for Tea Plant Germplasm Innovation and Resource Utilization, School of Tea Science, Anhui Agricultural University, Hefei, 230036, China.
| | - Linsheng Yu
- School of Plant Protection, Anhui Province Key Laboratory of Crop Integrated Pest Management, Hefei 230031, China.
- Apiculture Research Institute, Anhui Agricultural University, Hefei 230031, China
- Biotechnology Center of Anhui Agriculture University, Hefei 230031, China
| |
Collapse
|
2
|
Gu X, Zhang Z, Zhao L, Lu L, Lu X, Li Y, Gu T, Huang X, Huang G, Liang Y, Meng H, Li B, Zhang X, Zhang J, Wang X, Du Y. Exposure to polyethylene terephthalate micro(nano)plastics exacerbates inflammation and fibrosis after myocardial infarction by reprogramming the gut and lung microbiota and metabolome. JOURNAL OF HAZARDOUS MATERIALS 2025; 488:137410. [PMID: 39919626 DOI: 10.1016/j.jhazmat.2025.137410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/06/2025] [Accepted: 01/25/2025] [Indexed: 02/09/2025]
Abstract
Micro(nano)plastics (MNPs), a ubiquitous environmental pollutant, have received increasing attention for their impacts on human health. We conducted an in-depth study on the role of polyethylene terephthalate (PET) MNPs in myocardial infarction (MI). Blood from the coronary circulation of MI patients was collected to detect microplastics (MPs). Peripheral monocytes (PBMCs) and AC16 cells were used to assess inflammation, cell proliferation and apoptosis after PET nanoplastics (NPs) stimulation. The mouse MI model was established after PET NPs respiratory or oral exposure. The results showed that various types of MPs, including high levels of PET MPs, were detected in the coronary circulation. PET NPs promoted inflammatory factors secretion by PBMCs, inhibited AC16 cell proliferation and promoted hypoxia-induced AC16 cell apoptosis. PET NPs exacerbated post-MI inflammation and fibrosis through activating the NLRP3 inflammasome pathway. Through macrogenetic sequencing and metabolomics analyses, we observed that PET NPs reprogrammed the intestinal and lung microbiota and metabolome in MI mice, leading to chronic inflammation. In conclusion, PET MPs were widely present in the coronary circulation of MI patients. PET MNPs can activate the NLRP3 inflammasome pathway to exacerbate post-MI ventricular remodelling, which may be related to the reprogramming of the gut and lung microbiota and metabolome.
Collapse
Affiliation(s)
- Xin Gu
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Zhixuan Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Lin Zhao
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Lijie Lu
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China
| | - Xin Lu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210004, China
| | - Yafei Li
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China
| | - Tianya Gu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin Huang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Guangyi Huang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Yan Liang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Haoyu Meng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Baihong Li
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Xiaodong Zhang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China
| | - Jun Zhang
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China.
| | - Xiaoyan Wang
- Department of Cardiology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, China.
| | - Yingqiang Du
- Department of Cardiology, Gusu School, Nanjing Medical University, Suzhou, Jiangsu 215008, China; Department of Cardiology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215008, China; Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, China.
| |
Collapse
|
3
|
Ren G, Lin Y, Fu Y, Liu F, Wang R, Zhang C, Qiu J, Chen L, Dou X. Multi-omics joint analysis: Pachymic acid ameliorated non-alcoholic fatty liver disease by regulating gut microbiota. Food Res Int 2025; 209:116178. [PMID: 40253122 DOI: 10.1016/j.foodres.2025.116178] [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: 10/04/2024] [Revised: 01/12/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Poria cocos a traditional Chinese medicinal material with both culinary and therapeutic applications, contains pachymic acid (Pac) as one of its main active compounds, which has demonstrated anti-lipid accumulation and hypoglycemic effects. However, its impact on the biochemical changes in the enterohepatic axis induced by a high-fat diet remains poorly understood. This study investigated the protective mechanism of Pac using a high-fat diet-induced non-alcoholic fatty liver disease (NAFLD) mouse model. 16S rRNA sequencing of gut microbiota revealed that Pac administration reduced the Firmicutes to Bacteroidetes ratio, restored Akkermansia abundance, decreased Desulfovibrio and Streptococcus population, and ameliorated gut dysbiosis. Concurrently, Pac treatment reduced the expression of hepatic inflammatory factors by mainly adjusted LPS/TLR4/MYD88/NFκB pathway. Liver transcriptome analysis indicated that Pac primarily affects genes involved in lipid metabolism, apoptosis, and inflammatory responses. Specifically, Pac inhibited FASN, SREBP1c, and SCD1 expression while upregulating PPARα and CPT1α, thereby improving high-fat diet-induced hepatic steatosis in mice. Additionally, Pac treatment reduced hepatocellular apoptosis. Non-targeted liver metabolomics analysis following Pac intervention revealed increased levels of acylcarnitine and oleic acid. Collectively, these findings suggest that Pac alleviates high-fat diet-induced hepatic lipid accumulation and damage by modulating gut microbiota, lipid metabolism, inflammation, and apoptosis. This comprehensive study provides valuable insights into the therapeutic potential of Pac and offers a reference for the development and utilization of Poria cocos resources in addressing NAFLD.
Collapse
Affiliation(s)
- Guilin Ren
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yiyou Lin
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yilong Fu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Fucai Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Ruiqi Wang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Congcong Zhang
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
4
|
Qiu J, Xu F, Wei H, Gao Y, Liu N, Zhao J, Yu Z, Chen L, Dou X. Metabolic restoration: Rhubarb polysaccharides as a shield against non-alcoholic fatty liver disease. Int J Biol Macromol 2025; 305:141151. [PMID: 39965694 DOI: 10.1016/j.ijbiomac.2025.141151] [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: 10/10/2024] [Revised: 02/09/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) accounts for remarkable burden of death and costs worldwide with no recommended pharmacological intervention for the clinical management. This study aimed to investigate the efficacy and underlying mechanisms of rhubarb-derived polysaccharides (RP) in mitigating high-fat diet (HFD)-induced NAFLD and to analyze the primary monosaccharide components of RP. Forty male C57BL/6 mice were subjected to a dietary intervention consisting of either a high fat or chow diet for a duration of 12 weeks. RP (270, 540 mg·kg-1·d-1) was administered to the mice for 4 consecutive weeks from the 9th week. Various assessments were conducted, including histopathological examination, liver transcriptome analysis, non-targeted metabolomics analysis, and evaluation of protein expressions related to lipid and bile acid metabolism. This study found RP demonstrate a protective effect on the livers of NAFLD mice by inhibiting lipid accumulation and reducing hepatocyte inflammatory damage. The metabolomics analysis of multi-tissues revealed that the RP exert a hepatoprotective effect against NAFLD by restoring the altered bile acids (BAs) and fatty acids (FFAs) metabolism through the improvement of BA transporter, nucleus hormone receptor, lipogenesis protein, FFA transporter, and lipolysis proteins. Hence, RP may serve as a potential therapeutic agent for NAFLD.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Fangying Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huaxin Wei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanyan Gao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Nian Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jinghua Zhao
- The First Affiliated Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Shang DF, Xu WQ, Zhao Q, Zhao CL, Wang SY, Han YL, Li HG, Liu MH, Zhao WX. Molecular mechanisms of pyroptosis in non-alcoholic steatohepatitis and feasible diagnosis and treatment strategies. Pharmacol Res 2025; 216:107754. [PMID: 40306603 DOI: 10.1016/j.phrs.2025.107754] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/11/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a distinct form of cell death that plays a critical role in intensifying inflammatory responses. It primarily occurs via the classical pathway, non-classical pathway, caspase-3/6/7/8/9-mediated pathways, and granzyme-mediated pathways. Key effector proteins involved in the pyroptosis process include gasdermin family proteins and pannexin-1 protein. Pyroptosis is intricately linked to the onset and progression of non-alcoholic steatohepatitis (NASH). During the development of NASH, factors such as pyroptosis, innate immunity, lipotoxicity, endoplasmic reticulum stress, and gut microbiota imbalance interact and interweave, collectively driving disease progression. This review analyzes the molecular mechanisms of pyroptosis and its role in the pathogenesis of NASH. Furthermore, it explores potential diagnostic and therapeutic strategies targeting pyroptosis, offering new avenues for improving the diagnosis and treatment of NASH.
Collapse
Affiliation(s)
- Dong-Fang Shang
- Henan University of CM, Zhengzhou 450000, China; The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Wen-Qian Xu
- Henan University of CM, Zhengzhou 450000, China
| | - Qing Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Chen-Lu Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Si-Ying Wang
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - Yong-Li Han
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China
| | - He-Guo Li
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Ming-Hao Liu
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| | - Wen-Xia Zhao
- The First Affiliated Hospital of Henan University of CM, Zhengzhou 450003, China.
| |
Collapse
|
6
|
Meybodi SM, Rezazadeh Khabaz MJ, Vojdani A, Nasiri Z, Mazhari SA, Tabar FA, Javazm SA, Owrang M, Noori Z, Pishva MS, Badameh P, Maleki MH, Nadimi E. Bifidobacterium adolescentis prevents diabetes-induced liver injury via pyroptosis attenuation. Exp Cell Res 2025; 447:114518. [PMID: 40097086 DOI: 10.1016/j.yexcr.2025.114518] [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: 06/02/2024] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD), along with non-alcoholic steatohepatitis (NASH), lacks definitive therapy and typically remains asymptomatic until reaching advanced stages. Lipid metabolism and inflammation management using probiotics such as Bifidobacterium adolescentis is suggested to alleviate or suppress NAFLD development. Hence, this study aims to investigate the effects of Bifidobacterium adolescentis treatment on mitigating pyroptosis, an inflammatory cell death pathway, in the liver of rats with NAFLD induced by high-fat diet (HFD) and streptozotocin (STZ) administration. METHODS Forty 8-week adult male Sprague Dawley rats were divided into four groups. Bifidobacterium adolescentis was administered for 8 and 16 weeks at 4 × 1010 CFU/day to rats fed a high-fat diet (HFD). Subsequently, the mRNA expression levels of pyroptotic-related genes including Cas1, Cas3, Cas11, NLRP3, GSDMD, IL-1β, and NF-κB were quantified in liver tissue using quantitative polymerase chain reaction (qPCR). Histopathological alterations and stereological changes in liver structure, as well as lipid profile (FBG, TG, TC, HDL, LDL), and liver indices (ALT, AST, ALP, LDH), were also evaluated across the different groups. RESULTS Bifidobacterium adolescentis administration significantly reduced the expression levels of NF-κB and pyroptotic-related genes. Additionally, this probiotic effectively reversed the adverse effects of the high-fat diet (HFD) on liver volume, Kupffer cell numbers, and hepatocyte nuclei. Furthermore, it improved the lipid profile and liver indices of rats fed with the HFD. CONCLUSION This study demonstrates that B. adolescentis supplementation prevents diabetes-induced liver injury by attenuating pyroptosis. These findings suggest that Bifidobacterium adolescentis may be a promising therapeutic approach for managing NAFLD and its associated complications, primarily by modulating key genes associated with pyroptosis and inflammation in rats fed with a high-fat diet.
Collapse
Affiliation(s)
- Seyed Mohammadmahdi Meybodi
- Faculty of Veterinary Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | | | - Andia Vojdani
- Department of Microbiology, School of Biology, University of Tehran, Tehran, Iran.
| | - Zahra Nasiri
- Department of Cellular and Molecular Biology, Faculty of Materials, Najafabad Branch, Islamic Azad University, Isfahan, Iran.
| | | | - Farideh Akhlaghi Tabar
- Department of Genetics, Faculty of Basic Science, Qom Branch, Islamic Azad University, Qom, Iran.
| | - Sara Abdizadeh Javazm
- Department of Microbiology, Faculty of Sciences, Karaj Branch, Islamic Azad University, Karaj, Iran.
| | - Marzieh Owrang
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Zahra Noori
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Maryam Sadat Pishva
- University of Tehran, Kish International Campus, School of Biology, Kish Island, Iran.
| | - Parisa Badameh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mohammad Hasan Maleki
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Elham Nadimi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Biotechnology Department, School of Advanced, Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
7
|
Lin Q, Huang K, Ge X, Ma M, Wang W, Yang L, Chen C, Han B, Liu D. LC-HRMS profiling of Dendrobium huoshanense aqueous extract and its therapeutic effects on nonalcoholic fatty liver disease in mice through the TLR2-NF-κB and AMPK-SREBP1-SIRT1 signaling pathways. J Chromatogr B Analyt Technol Biomed Life Sci 2025; 1256:124563. [PMID: 40107184 DOI: 10.1016/j.jchromb.2025.124563] [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: 09/23/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Dendrobium huoshanense (DH) belongs to the Dendrobium genus of the Orchidaceae family and is a herbaceous plant that protects the liver and nourishes the Yin according to traditional Chinese Medicine (TCM) theory. This research aimed to determine the therapeutic effect and mechanisms of DH on a nonalcoholic fatty liver disease (NAFLD) mouse model and its chemical composition. For pharmacological research, the pathological damage and lipid accumulation in liver tissues were evaluated using HE and oil red staining, respectively. The differential proteins between the model and DHH groups were screened using 4D label-free quantitative proteomics, and the proteomic results were verified using Western blot. The potential mechanism was validated by metabolomic analysis. The main active ingredients in a DH aqueous extract were identified using UHPLC-Q Exactive HF HRMS. Pathological staining results showed that DH can reverse liver pathological damage and lipid accumulation in the NAFLD model. Quantitative proteomics revealed that the differential proteins were mainly associated with liver lipid deposition (LAL, AMPK, TM7SF2, SBCAD, and SIRT1), insulin resistance (GYS1, GYS2, PYGL, FoxO1, and PPAR-γ), and inflammation (TLR2 and MAPKAPK). Western blot verified the above-mentioned results. Metabolomic analysis also indicated that the DH aqueous extract ameliorated NAFLD in mice by affecting cholesterol metabolism and AMPK signaling pathway, proving its significant therapeutic effects on the NAFLD model. Sixty-five compounds were identified from DH aqueous extract by analyzing the precise molecular weight and MS/MS fragmentation pathway. The pharmacological mechanism of DH in treating NAFLD mainly involved the TLR2-NF-κB and AMPK-SREBP1-SIRT1 signaling pathways.
Collapse
Affiliation(s)
- Qiyan Lin
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Ke Huang
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Xiyu Ge
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Menghua Ma
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Wei Wang
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Cunwu Chen
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China
| | - Bangxing Han
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China.
| | - Dong Liu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Traditional Chinese Medicine Institute of Anhui Dabie Mountain, Anhui Engineering Research Center for Eco-agriculture of Traditional Chinese Medicine, Generic Technology Research Center for Anhui Traditional Chinese Medicine Industry, Anhui Key Laboratory of Protection and Utilization of Characteristic Biological Resources in Dabie Mountains, Lu'an, 237012, Anhui, PR China.
| |
Collapse
|
8
|
Liu X, Guan B, Hu Z, Hu X, Liu S, Yang K, Zhou L, Yu L, Yang J, Chen S, Chen Q, Liu D, Liu G, Pan H. Combined traditional Chinese medicine and probiotics (TCMP) alleviates lipid accumulation and improves metabolism in high-fat diet mice via the microbiota-gut-liver axis. Food Res Int 2025; 207:116064. [PMID: 40086971 DOI: 10.1016/j.foodres.2025.116064] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/09/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
Lipid accumulation and metabolic disorders caused by a high-fat diet (HFD) pose significant threats to human health, and place a substantial burden on individuals and society. In this study, a novel combination comprising three traditional Chinese herbs (lotus leaf, hawthorn, and leaf of Chinese holly) and a probiotic (Bifidobacterium lactis BPL-1) (TCMP) was prepared. Then, its effects on growth performance, fat accumulation, hepatic function and gut microbiota in mice fed a high-fat diet were investigated. According to the results, TCMP significantly reduced adipose tissue fat accumulation, improved hepatic lipid metabolism, and ameliorated glucose homeostasis in HFD-fed mice. Notably, TCMP not only improved the abundance and diversity of gut microbiota and increased the content of beneficial intestinal bacteria related to lipid metabolism (especially Bifidobacterium animalis), but also increased the production of short-chain fatty acids, including2-methylbutyrate, isovaleric acid and isobutyric acid. Additionally, multi-omics (transcriptome and metabolome) analysis revealed that TCMP significantly inhibited the expression of genes involved in the lipid biosynthesis process and modulated the purine and glycerophospholipid metabolism caused by a high-fat diet, thereby achieving the purpose of reducing fat accumulation and regulating lipid metabolism. Taken together, our finding demonstrates the potential of TCMP as a promising therapeutic candidate for combatting obesity and lipid metabolism disorders induced by a high-fat diet.
Collapse
Affiliation(s)
- Xiayu Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Boyuan Guan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Ziyi Hu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Xiaoyan Hu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Shuaixing Liu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Ke Yang
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Liping Zhou
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Longli Yu
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Jinyan Yang
- Guangdong Qingyunshan Chinese Medicine Innovation Co., Ltd., Shaoguan 512000, China
| | - Shiguo Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Qihe Chen
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Guanchen Liu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| | - Haibo Pan
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| |
Collapse
|
9
|
Zhu J, Song N, Wang J, Wang Q, Cao Y, Zhang M, Sun X, Jia L. Mechanism of action of Danlou tablets affecting MAFLD via KEAP1-mediated oxeiptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 344:119521. [PMID: 39978447 DOI: 10.1016/j.jep.2025.119521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Metabolic fatty liver disease (MAFLD) is a significant risk factor for atherosclerotic cardiovascular disease. Several preliminary studies on MAFLD animal models have indicated the therapeutic potential of Danlou tablets (DLT), a primary Chinese medicine used for managing coronary artery disease. However, the underlying mechanism of DLT in the treatment of MAFLD remains elusive. AIM OF THE STUDY Clarify the potential effective components of DLT in the treatment of MAFLD, and preliminarily verify the molecular mechanism against MAFLD in vivo and in vitro experiments. MATERIALS AND METHODS The composition of DLT and their content in DLT-treated rat serum were analyzed using UPLC/ESI- Q TRAP-MS/MS. Mice were given a high-fat diet to establish the MAFLD model. Then, the MAFLD mice were treated with DLT. Liver sections were taken for histopathological assessment. Furthermore, in vivo and in vitro alterations in the oxeiptosis pathway, de novo fatty acid synthesis, and Triglyceride catabolism were verified by qRT-PCR, Western Blot, and Immunofluorescence experiments. Moreover, how DLT modulated the oxeiptosis pathway was further investigated by rescue experimental strategies. RESULTS We isolated and detected a total of 1003 compounds from DLT, 109 of which were found in rat plasma, and hypothesized that 11 active ingredients represented by Tanshinone IIA might play a major role in anti-MAFLD. Furthermore, we found that DLT increased Triglyceride catabolism and suppressed de novo fatty acid synthesis in vivo and in vitro, thereby significantly attenuating hepatic lipid deposition. Mechanistically, DLT restored the phosphorylation of Protein Kinase B, promoted Triglyceride catabolism and inhibited the de novo fatty acid synthesis through the oxeiptosis pathway (KEAP1/PGAM5/AIFM1). CONCLUSIONS Our findings suggest that DLT promotes Triglyceride catabolism and inhibit de novo fatty acid synthesis by affecting the activation of the oxeiptosis pathway, suggesting a potential therapeutic strategy for ameliorating NAFLD.
Collapse
Affiliation(s)
- Jingxuan Zhu
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Nan Song
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaxin Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Qun Wang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuan Cao
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Meiling Zhang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xiaofei Sun
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Lianqun Jia
- Liaoning University of Traditional Chinese Medicine, Shenyang, China.
| |
Collapse
|
10
|
Yang G, Tan X, Zhai Q, Wang Y, Zhang X, Zhao P, Liang F, Lu J, Bao L. Plasma Lipidomics, Gut Microbiota Profile, and Phenotype of Adipose Tissue in an ApoE-/- Mouse Model of Plaque Instability. FRONT BIOSCI-LANDMRK 2025; 30:27236. [PMID: 40152393 DOI: 10.31083/fbl27236] [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: 10/30/2024] [Revised: 01/12/2025] [Accepted: 02/08/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND An appropriate animal model that can simulate the pathological process of atherosclerosis is urgently needed to improve treatment strategies. This study aimed to develop a new atherosclerosis model using ApoE-/- mice and to characterize lipidomics, gut microbiota profiles, and phenotypic alterations in adipose tissue using this model. METHODS After a 14- or 18-week high-fat diet (HFD), male ApoE-/- mice were randomly divided into four groups and treated separately with or without short-term and strong co-stimulation, including ice water bath and intraperitoneal injection of lipopolysaccharide and phenylephrine. As a control group, C57BL/6 mice were fed with conventional chow. The serum lipid levels, aortic arch pathology, adipose tissue phenotypic changes, plasma lipidomics, and 16S rDNA gene sequencing of colon feces were investigated. RESULTS The serum lipid levels were significantly lowered following extended HFD feeding for four weeks. However, co-stimulation increased serum interleukin (IL)-1β levels but did not affect serum lipid profiles. Co-stimulation revealed typical vulnerable atherosclerotic plaque characteristics and defective adipose hypertrophy associated with peroxisome proliferator-activated receptor γ (PPARγ) regulation in adipose tissue and a reduction in mitochondrial uncoupling protein 1 (UCP1) within brown adipose tissue. Plasma lipidomic analysis showed that sphingomyelin (SM), ceramide (Cer), and monohexosylceramide (HexCer) levels in plasma were significantly elevated by HFD feeding, whereas co-stimulation further elevated HexCer levels. Additionally, glycerophosphocholines (16:0/16:0, 18:2/20:4, 18:1/18:1) and HexCer (C12:1, C16:0), Cer (d18:1/16:0), and SM (C16:0) were the most sensitive to co-stimulation. Combined co-stimulation and HFD-fed increased the abundance of Firmicutes, the abundance of f_Erysipelotrichaceae, and the Firmicutes/Bacteroidota ratio but decreased the abundance of microflora promoting bile acid metabolism and short-chain fatty acids (SCFAs) in mouse feces. The results were consistent with the findings of epidemiologic atherosclerotic cardiovascular disease studies. CONCLUSIONS This study established an ApoE-/- mouse atherosclerotic vulnerable plaque model using a multi-index evaluation method. Adipogenic disorders, dysregulation of lipid metabolism at the molecular level, and increasing harmful gut microbiota are significant risk factors for vulnerable plaques, with sphingolipid metabolism receiving the most attention.
Collapse
Affiliation(s)
- Guanlin Yang
- Laboratory of Pharmacology, Zaozhuang Thoracic Hospital, 277500 Zaozhuang, Shandong, China
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xin Tan
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Qiong Zhai
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Yuewu Wang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Xuan Zhang
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Pengwei Zhao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Fangyuan Liang
- School of Pharmacy, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - Jingkun Lu
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| | - LiLi Bao
- School of Basic Medicine, Inner Mongolia Medical University, 010107 Hohhot, Inner Mongolia, China
| |
Collapse
|
11
|
Mao Y, Xie Z, Zhang X, Fu Y, Yu X, Deng L, Zhang X, Hou B, Wang X, Ma M, Ren F. Ergothioneine Ameliorates Liver Fibrosis by Inhibiting Glycerophospholipids Metabolism and TGF-β/Smads Signaling Pathway: Based on Metabonomics and Network Pharmacology. J Appl Toxicol 2025; 45:514-530. [PMID: 39579000 DOI: 10.1002/jat.4728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024]
Abstract
Ergothioneine (EGT) is a diet-derived natural sulfur-containing amino acid that exhibits strong anti-oxidant and anti-inflammation activities. Oxidative stress and chronic inflammatory injury are predominant pro-fibrogenic factors. Therefore, EGT may have therapeutic potential against liver fibrosis; however, its underlying mechanism is incompletely understood. This study aimed at investigating the protective effects of EGT on liver fibrosis based on metabonomics and network pharmacology. A mouse model of liver fibrosis was established by intraperitoneal injection with 40% CCl4 solution (2 mL/kg, twice a week) and intragastric administration with EGT (5, 10 mg/kg/d) for six weeks. Results showed that EGT improved liver function by reducing serum levels of ALT (alanine aminotransferase), AST (aspartate aminotransferase), and TBIL (total bilirubin), and alleviated liver fibrosis by reducing LN (laminin) and HyP (hydroxyproline) levels, decreasing expressions of α-SMA (α-smooth muscle actin), Col-I (collagen type I), and Col-III (collagen type III), and improving pathological changes. EGT also significantly inhibited CCl4-induced hepatic inflammation and TGF-β/Smads signaling pathway. Metabolomics identified six key metabolic pathways, such as purine metabolism, glycerophospholipid metabolism, and sphingolipid metabolism, and eight key metabolites, such as xanthine, guanine, ATP, phosphatidylcholine, and sphingosine. Network pharmacology analysis showed that IL-17, cAMP and NF-κB signaling pathways were potential key mechanisms. Integrated analysis revealed that PLA2G2A might be a potential target of EGT against liver fibrosis. EGT may inhibit the glycerophospholipid metabolism through PLA2G2A to inhibit the TGF-β/Smads signaling pathway, thereby alleviating fibrosis. The present study indicates that EGT may be considered a valid therapeutic strategy to regress liver fibrosis, and provides novel insights into the pharmacological mechanism of EGT against liver fibrosis.
Collapse
Affiliation(s)
- Yaping Mao
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Zhenghui Xie
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiangxia Zhang
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Department of Morphology, School of Nursing and Health, Qingdao Huanghai University, Qingdao, China
| | - Yu Fu
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiaotong Yu
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Lili Deng
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Xiu Zhang
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
| | - Bo Hou
- Department of Morphology, School of Nursing and Health, Qingdao Huanghai University, Qingdao, China
| | - Xiao Wang
- Department of Gastroenterology, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Mingyue Ma
- Department of Toxicology, School of Public Health, Shenyang Medical College, Shenyang, China
| | - Fu Ren
- Department of Anatomy, School of Basic Medicine, Shenyang Medical College, Shenyang, China
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang, China
| |
Collapse
|
12
|
Yin J, Chen L, Lin Y, Qiu J, Liu F, Wang Y, Dou X. Bifidobacterium bifidum reduces oxidative stress and alters gut flora to mitigate acute liver injury caused by N-acetyl-p-aminophenol. BMC Microbiol 2025; 25:87. [PMID: 40000948 PMCID: PMC11853282 DOI: 10.1186/s12866-025-03775-1] [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: 10/29/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Pharmacologically-induced liver injury from N-acetyl-p-aminophenol (APAP) overdose has become a leading cause of acute liver failure. Extensive research has elucidated the relationship between the intestinal microbiota and the pathophysiology of liver diseases. The growing body of evidence supporting the beneficial effects of probiotics, coupled with their established safety profile, has led to their widespread adoption in clinical practice. Among these, Bifidobacterium bifidum has garnered substantial attention due to its potential hepatoprotective properties, particularly in APAP-induced acute liver injury (AILI). However, the precise therapeutic effects and underlying mechanisms of its potential to alleviate drug-induced liver toxicity remain largely unexplored. To address this knowledge gap, the present study aimed to investigate the role of a new Bifidobacterium bifidum strain CGMCC No. 29,545 isolated from faeces on AILI. A mouse model was constructed through the administration of heat-killed or active B. bifidum CGMCC No. 29,545 preparations via gavage, followed by an intraperitoneal overdose of APAP. The results showed that the active B. bifidum could significantly reverse the increase in plasma transaminase levels and reduce the necrotic area of liver cells in AILI mice. A reduction in oxidative stress accompanied a reduction in this effect. Furthermore, B. bifidum attenuated plasma endotoxin levels and improved colonic inflammation, reducing hepatocyte apoptosis. The 16 S rRNA diversity of intestinal contents suggests that the involvement of B. bifidum in the regulation of the intestinal microbiota also plays a crucial role in the protection against AILI. The above results suggest that the amelioration of multiple injuries due to APAP overprocessing is closely related to active B. bifidum, which was confirmed by heat-killed B. bifidum preparations. Heat-killed B. bifidum preparations did not attenuate the degree of liver injury and oxidative stress caused by APAP treatment. The effects of two different active B. bifidum preparations provide new insights into the protective strategies of active B. bifidum as a probiotic against AILI.
Collapse
Affiliation(s)
- Juan Yin
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
- Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Zhejiang, 310053, PR China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Yiyou Lin
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Fucai Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China
| | - Yuhao Wang
- School of Medicine, Zhejiang University, Hangzhou, 310020, Zhejiang, PR China.
- , 268 Kaixuan Road, Shangcheng District, Hangzhou, 310029, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, PR China.
- Zhejiang-Hong Kong Joint Laboratory of Liver and Spleen Simultaneous Treatment in Traditional Chinese Medicine, Zhejiang, 310053, PR China.
| |
Collapse
|
13
|
Wang J, Wang Z, Yu Y, Cheng S, Wu J. Advances in research on metabolic dysfunction-associated steatotic liver disease. Life Sci 2025; 362:123362. [PMID: 39761743 DOI: 10.1016/j.lfs.2024.123362] [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: 09/14/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
The global increase in obesity-related metabolic disorders has led to metabolic dysfunction-associated steatotic liver disease (MASLD) emerging as one of the most prevalent chronic liver disease worldwide. Despite growing concerns, the exact pathogenesis of MASLD remains unclear and no definitive treatments have been made available. Consequently, the need for comprehensive research on MASLD is more critical than ever. Gaining insight into the mechanisms of the disease can lay the groundwork for identifying new therapeutic targets and can facilitate the development of diagnostic tools that enable the early detection and intervention of MASLD. Research has discovered a multifactorial etiology for MASLD, suggesting that potential therapeutic strategies should be considered from a variety of perspectives. This review delves into the pathogenesis of MASLD, current diagnostic approaches, potential therapeutic targets, the status of clinical trials for emerging drugs, and the most promising treatment methods available today. With a focus on therapeutic targets, the aim is to offer fresh insights and guide for future research in the treatment of MASLD.
Collapse
Affiliation(s)
- Jiawang Wang
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Zhongyu Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning 530021, China
| | - Yao Yu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Si Cheng
- Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| | - Jianping Wu
- Hubei Key Laboratory of Nanomedicine for Neurodegenerative Diseases, School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China; Department of Pharmacology, Hubei University of Medicine, Shiyan 440070, China; Beijing Tiantan Hospital, Capital Medical University, Beijing 10070, China; China National Clinical Research Center for Neurological Diseases, Beijing 10070, China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 10070, China.
| |
Collapse
|
14
|
Han J, Miao Y, Song L, Zhou X, Liu Y, Wang L, Zhu K, Ma H, Ma Y, Li Q, Han D. Xuefu Zhuyu Decoction improves hyperlipidemia through the MAPK/NF-κB and MAPK/PPARα/CPT-1A signaling pathway. FASEB J 2025; 39:e70363. [PMID: 39878687 DOI: 10.1096/fj.202402688r] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/29/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Xuefu Zhuyu Decoction (XZD) is widely used in the treatment of cardiovascular diseases. The purpose of this study was to explore the pharmacological effects and molecular mechanisms of XZD in improving hyperlipidemia and to provide a theoretical framework for clinical application. In this study, the signaling pathways regulated by XZD in improving hyperlipidemia were predicted by network pharmacology. Molecular docking was used to verify the affinity between the components in XZD and the target. Furthermore, a hyperlipidemic model in rats was constructed through feeding a high-fat diet. The effect of XZD on hyperlipidemia was verified by histopathological staining, Elisa, and western blot. The results found that the XZD improved dyslipidemia and inflammatory factor disorders, and inhibited liver function damage, pathological damage, and oxidative stress damage in hyperlipidemic rats. The findings from molecular docking and network pharmacology suggested that the mechanism of XZD improving hyperlipidemia may be closely related to the MAPK, NF-κB, and PPAR pathways. This study demonstrated that the XZD inhibited liver lipid metabolism disorder and inflammatory response by regulating the MAPK/NF-κB and MAPK/PPARα/CPT-1A pathway, significantly improved liver histopathological damage and oxidative stress injury, and played a protective role in hyperlipidemic rats.
Collapse
Affiliation(s)
- Jiajun Han
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yuyang Miao
- Department of Endocrinology and Metabolism, Jilin Province People's Hospital, Jilin, China
| | - Linze Song
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Xianfeng Zhou
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yan Liu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Lin Wang
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Kai Zhu
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - He Ma
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| | - Yan Ma
- Department of Endocrinology and Metabolism, Jilin Province People's Hospital, Jilin, China
| | - Qingjie Li
- Research Center of Traditional Chinese Medicine, Affiliated Hospital to Changchun University of Chinese Medicine, Jilin, China
| | - Dong Han
- College of Pharmacy, Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
15
|
Wu D, Yang S, Yuan C, Zhang K, Tan J, Guan K, Zeng H, Huang C. Targeting purine metabolism-related enzymes for therapeutic intervention: A review from molecular mechanism to therapeutic breakthrough. Int J Biol Macromol 2024; 282:136828. [PMID: 39447802 DOI: 10.1016/j.ijbiomac.2024.136828] [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: 04/23/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Purines are ancient metabolites with established and emerging metabolic and non-metabolic signaling attributes. The expression of purine metabolism-related genes is frequently activated in human malignancies, correlating with increased cancer aggressiveness and chemoresistance. Importantly, under certain stimulating conditions, the purine biosynthetic enzymes can assemble into a metabolon called "purinosomes" to enhance purine flux. Current evidence suggests that purine flux is regulated by a complex circuit that encompasses transcriptional, post-translational, metabolic, and association-dependent regulatory mechanisms. Furthermore, purines within the tumor microenvironment modulate cancer immunity through signaling mediated by purinergic receptors. The deregulation of purine metabolism has significant metabolic consequences, particularly hyperuricemia. Herbal-based therapeutics have emerged as valuable pharmacological interventions for the treatment of hyperuricemia by inhibiting the activity of hepatic XOD, modulating the expression of renal urate transporters, and suppressing inflammatory responses. This review summarizes recent advancements in the understanding of purine metabolism in clinically relevant malignancies and metabolic disorders. Additionally, we discuss the role of herbal interventions and the interaction between the host and gut microbiota in the regulation of purine homeostasis. This information will fuel the innovation of therapeutic strategies that target the disease-associated rewiring of purine metabolism for therapeutic applications.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Shengqiang Yang
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China
| | - Chenyang Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Jiachen Tan
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China.
| | - Hong Zeng
- School of Basic Medicine, Youjiang Medical University for Nationalities, Baise 533000, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
16
|
Zhang Y, Wang H, Sang Y, Liu M, Wang Q, Yang H, Li X. Gut microbiota in health and disease: advances and future prospects. MedComm (Beijing) 2024; 5:e70012. [PMID: 39568773 PMCID: PMC11577303 DOI: 10.1002/mco2.70012] [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: 06/28/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
The gut microbiota plays a critical role in maintaining human health, influencing a wide range of physiological processes, including immune regulation, metabolism, and neurological function. Recent studies have shown that imbalances in gut microbiota composition can contribute to the onset and progression of various diseases, such as metabolic disorders (e.g., obesity and diabetes) and neurodegenerative conditions (e.g., Alzheimer's and Parkinson's). These conditions are often accompanied by chronic inflammation and dysregulated immune responses, which are closely linked to specific forms of cell death, including pyroptosis and ferroptosis. Pathogenic bacteria in the gut can trigger these cell death pathways through toxin release, while probiotics have been found to mitigate these effects by modulating immune responses. Despite these insights, the precise mechanisms through which the gut microbiota influences these diseases remain insufficiently understood. This review consolidates recent findings on the impact of gut microbiota in these immune-mediated and inflammation-associated conditions. It also identifies gaps in current research and explores the potential of advanced technologies, such as organ-on-chip models and the microbiome-gut-organ axis, for deepening our understanding. Emerging tools, including single-bacterium omics and spatial metabolomics, are discussed for their promise in elucidating the microbiota's role in disease development.
Collapse
Affiliation(s)
- Yusheng Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Hong Wang
- School of Traditional Chinese Medicine Southern Medical University Guangzhou China
| | - Yiwei Sang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Mei Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| | - Qing Wang
- School of Life Sciences Beijing University of Chinese Medicine Beijing China
| | - Hongjun Yang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs China Academy of Chinese Medical Sciences Beijing China
| | - Xianyu Li
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases Experimental Research Center China Academy of Chinese Medical Sciences Beijing China
| |
Collapse
|
17
|
Mao D, Guo J, Yang K, Yang F, Peng J, Jia X, Luo Z, Liu L, Yang E, Tang R, Lan H, Zheng Q. Mechanism of epigallocatechin gallate in treating non-alcoholic fatty liver disease: Insights from network pharmacology and experimental validation. Biochem Biophys Res Commun 2024; 734:150424. [PMID: 39083974 DOI: 10.1016/j.bbrc.2024.150424] [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: 04/26/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024]
Abstract
To explore the therapeutic effects along with the molecular mechanisms of epigallocatechin gallate (EGCG) in non-alcoholic fatty liver disease (NAFLD) treatment using network pharmacology as well as animal experiments. Firstly, the Traditional Chinese Medicine (TCM) Systems Pharmacology Database was searched to identify the potential targets of EGCG. The DisGeNET Database was used to screen the potential targets of NAFLD. The GeneCards Database was searched to identify related genes involved in pyroptosis. Subsequently, the intersecting genes of EGCG targeting pyroptosis to regulate NAFLD were obtained using a Venn diagram. Simultaneously, the aforementioned intersecting genes were used to construct a drug-disease target protein-protein interaction (PPI) network. The DAVID database was adopted for Gene Ontology (GO) as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. The main pathway-target network was determined. Next, the potential mechanism of EGCG targeting pyroptosis to regulate NAFLD was investigated and validated through in vivo experiments. 626 potential targets of EGCG, 447 target genes of NAFLD, and 568 potential targets of pyroptosis were identified. The number of common targets between EGCG, NAFLD, and pyroptosis was 266. GO biological process items and 92 KEGG pathways were determined based on the analysis results. Animal experiments demonstrated that EGCG could ameliorate body weight, glucolipid metabolism, steatosis, and liver injury, enhance insulin sensitivity, and improve glucose tolerance in NAFLD mice through the classical pathway of pyroptosis. EGCG could effectively treat NAFLD through multiple targets and pathways. It was concluded that EGCG ameliorates hepatocyte steatosis, pyroptosis, dyslipidemia, and inflammation in NAFLD mice fed a high-fat diet (HFD), and the protective mechanism could be associated with the NLRP3-Caspase-1-GSDMD classical pyroptosis pathway.
Collapse
Affiliation(s)
- Danting Mao
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Jianwei Guo
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Kunli Yang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Fan Yang
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China.
| | - Jiaojiao Peng
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Xu Jia
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, China.
| | - Ziren Luo
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Lu Liu
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Enjie Yang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Rui Tang
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Haitao Lan
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| | - Qian Zheng
- Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637000, China.
| |
Collapse
|
18
|
Zhang H, Zhao X, Zhang L, Sun D, Ma Y, Bai Y, Bai X, Liang X, Liang H. Nicotinamide Riboside Ameliorates Fructose-Induced Lipid Metabolism Disorders in Mice by Activating Browning of WAT, and May Be Also Related to the Regulation of Gut Microbiota. Nutrients 2024; 16:3920. [PMID: 39599706 PMCID: PMC11597130 DOI: 10.3390/nu16223920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
OBJECTIVES This study aims to observe the preventive effect of nicotinamide riboside (NR) on fructose-induced lipid metabolism disorders and explore its mechanism. METHODS Male C57BL/6J mice were fed a 20% fructose solution and given 400 mg/kg NR daily by gavage for 10 weeks. RESULTS The results indicated that NR supplementation significantly reduced the body weight, liver weight, white adipose tissue (WAT) weight, serum, and hepatic lipid levels. NR upregulated the protein expression levels of sirtuin-1 (SIRT1), AMP-activated protein kinase (AMPK), PR domain containing 16 (PRDM16), uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor-gamma coactiva-tor-1-alpha (PGC-1α), nuclear respiratory factor 1-encoding gene (NRF1), mitochondrial transcription factor A (TFAM), cluster of differentiation 137 (CD137), transmembrane protein 26 (TMEM26), and T-box 1 (TBX1). Moreover, NR enhanced the Actinobacteria and Enterorhabdus abundance. Spearman's correlation analysis revealed that significant correlations exist between Firmicutes, Bacteroidetes, and Erysipelotrichaceae with browning-related indicators. CONCLUSIONS In conclusion, NR could alleviate lipid metabolic abnormalities induced by fructose through activating SIRT1/AMPK-mediated browning of WAT. The mechanism by which NR improves fructose-induced lipid metabolism disorders may also be associated with the modulation of intestinal flora.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hui Liang
- Department of Nutrition and Food Hygiene, School of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China; (H.Z.); (X.Z.); (L.Z.); (D.S.); (Y.M.); (Y.B.); (X.B.); (X.L.)
| |
Collapse
|
19
|
Qiu J, Le Y, Liu N, Chen L, Jiang Y, Wang Y, Fan X, Rong X, Yu Z, Li S, Dou X. Nuciferine Alleviates High-Fat Diet- and ApoE -/--Induced Hepatic Steatosis and Ferroptosis in NAFLD Mice via the PPARα Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24417-24431. [PMID: 39445611 DOI: 10.1021/acs.jafc.4c04929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) causes significant global mortality and healthcare costs with no recommended pharmacological intervention for clinical management. Nuciferine (Nuc) is an alkaloid with aromatic rings, abundantly found in Nelumbo nucifera Gaertn. In this study, we explored the protective mechanisms of Nuc against hepatic steatosis and ferroptosis in NAFLD. High-fat diet (HFD) and healthy male ApoE-/- mice were used to induce NAFLD and a hypercholesterolemia model. Nuc was administered to the mice for four consecutive weeks from the ninth week. Various assessments, including histopathology, RNA sequencing, lipid metabolism, and ferroptosis-related protein expression, showed that Nuc alleviated hepatic steatosis and ferroptosis. We further showed that Nuc improves fatty acid accumulation and ferroptosis through the PPARα signaling pathway in mice and RSL3-treated AML-12 cells. The PPARα inhibitor GW6471 blocked Nuc's protective effects, leading to excess accumulation of iron ions. Thus, Nuc may be a potential therapeutic agent for NAFLD.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
- School of Public Health, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Nian Liu
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yuhao Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Shangcheng District, Hangzhou, Zhejiang 310020, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 38 Zheda Road, Hangzhou, Zhejiang 310027, China
| | - Xianglu Rong
- Chinese Medicine Institute, Guangdong Pharmaceutical University, No. 280 Outer Ring East Road, University Town, Guangzhou, Guangdong 510006, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong 999077, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| |
Collapse
|
20
|
Wang Y, Gu W, Xu Z, Lv L, Wang D, Jin Y, Wang X. Comprehensive multi-omics investigation of sub-chronic toxicity induced by Cadmium and Triazophos Co-exposure in hook snout carps (Opsariichthys bidens). JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135104. [PMID: 38970972 DOI: 10.1016/j.jhazmat.2024.135104] [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: 05/07/2024] [Revised: 06/18/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
The coexistence of heavy metals and pesticides poses a critical challenge in agricultural ecosystems. Traditional toxicity assessments often focus only on the individual impacts of either pesticides or heavy metals. Here, the untargeted metabolomics and 16 S rRNA sequencing were used to assess the individual and combined effects of cadmium (Cd) and triazophos (TRI) on hook snout carps (Opsariichthys bidens). Cd caused much more serious impacts on hepatic metabolism and gut microbiota than those in TRI. Combined Cd and TRI exposure synergistically affected hepatic metabolism, causing mitochondrial dysfunction and even oxidative damage. Simultaneously, 16 S rRNA sequencing highlighted significant variations in the composition and abundance of gut microbiota. A noteworthy connection emerged between these distinct microbiota profiles and disruptions in energy metabolism, ultimately leading to disorders in metabolites. These findings enhanced the understanding of risks posed by heavy metals and pesticides, providing insights for better environmental risk assessments of aquatic organisms.
Collapse
Affiliation(s)
- Yanhua Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Weijie Gu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China
| | - Zhenlan Xu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Lu Lv
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Dou Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, Zhejiang, PR China.
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, Zhejiang, PR China.
| |
Collapse
|
21
|
Qiu J, Lin C, Ren G, Xu F, Hu T, Le Y, Fan X, Yu Z, Liu Q, Wang X, Dou X. Geniposide dosage and administration time: Balancing therapeutic benefits and adverse reactions in liver disease treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155799. [PMID: 38968789 DOI: 10.1016/j.phymed.2024.155799] [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: 02/22/2024] [Revised: 05/05/2024] [Accepted: 06/02/2024] [Indexed: 07/07/2024]
Abstract
Gardenia jasminoides Ellis, a staple in herbal medicine, has long been esteemed for its purported hepatoprotective properties. Its primary bioactive constituent, geniposide, has attracted considerable scientific interest owing to its multifaceted therapeutic benefits across various health conditions. However, recent investigations have unveiled potential adverse effects associated with its metabolite, genipin, particularly at higher doses and prolonged durations of administration, leading to hepatic injury. Determining the optimal dosage and duration of geniposide administration while elucidating its pharmacological and toxicological mechanisms is imperative for safe and effective clinical application. This study aimed to evaluate the safe dosage and administration duration of geniposide in mice and investigate its toxicological mechanisms within a comprehensive dosage-duration-efficacy/toxicity model. Four distinct mouse models were employed, including wild-type mice, cholestasis-induced mice, globally farnesoid X-activated receptor (FXR) knock out mice, and high-fat diet-induced (HFD) NAFLD mice. Various administration protocols, spanning one or four weeks and comprising two or three oral doses, were tailored to each model's requirements. Geniposide has positive effects on bile acid and lipid metabolism at doses below 220 mg/kg/day without causing liver injury in normal mice. However, in mice with NAFLD, this dosage is less effective in improving liver function, lipid profiles, and bile acid metabolism compared to lower doses. In cholestasis-induced mice, prolonged use of geniposide at 220 mg/kg/day worsened liver damage. Additionally, in NAFLD mice, this dosage of geniposide for four weeks led to intestinal pyroptosis and liver inflammation. These results highlight the lipid-lowering and bile acid regulatory effects of geniposide, but also warn of potential negative impacts on intestinal epithelial cells, particularly with higher doses and longer treatment durations. Therefore, achieving optimal therapeutic results requires a decrease in treatment duration as the dosage increases, in order to maintain a balanced approach to the use of geniposide in clinical settings.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China; E-institute of Shanghai Municipal Education Committee, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Chen Lin
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Guilin Ren
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Fangying Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Tianxiao Hu
- Department of Ultrasonography, Chinese PLA 903rd Hospital, Hangzhou 310013, China
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Qingsheng Liu
- Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang 310000, China
| | - Xiaoning Wang
- E-institute of Shanghai Municipal Education Committee, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China.
| |
Collapse
|
22
|
Dong X, Xiong YT, He T, Zheng C, Li J, Zhuang Y, Xu Y, Xiu Y, Wu Z, Zhao X, Xiao X, Bai Z, Gao L. Protective effects of Nogo-B deficiency in NAFLD mice and its multiomics analysis of gut microbiology and metabolism. GENES & NUTRITION 2024; 19:17. [PMID: 39182019 PMCID: PMC11344411 DOI: 10.1186/s12263-024-00754-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 08/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver ailment that can lead to serious conditions such as cirrhosis and hepatocellular carcinoma. Hepatic Nogo-B regulates glucose and lipid metabolism, and its inhibition has been shown to be protective against metabolic syndrome. Increasing evidence suggests that imbalances in the gut microbiota (GM) and lipid metabolism disorders are significant contributors to NAFLD progression. Nevertheless, it is not yet known whether Nogo-B can affect NAFLD by influencing the gut microbiota and metabolites. Hence, the aim of the present study was to characterize this process and explore its possible underlying mechanisms. METHODS A NAFLD model was constructed by administering a high-fat diet (HFD) to Nogo-B-/- and WT mice from the same litter, and body weight was measured weekly in each group. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed to assess blood glucose levels. At the end of the 12-week period, samples of serum, liver, and intestinal contents were collected and used for serum biochemical marker and inflammatory factor detection; pathology evaluation; and gut microbiome and metabolomics analysis. Spearman's correlation analysis was performed to determine possible correlations between differential gut microbiota and differential serum metabolites between groups. RESULTS Nogo-B deficiency attenuated the effects of the HFD, including weight gain, liver weight gain, impaired glucose tolerance, hepatic steatosis, elevated serum lipid biochemicals levels, and liver function. Nogo-B deficiency suppressed M1 polarization and promoted M2 polarization, thus inhibiting inflammatory responses. Furthermore, Nogo-B-/--HFD-fed mice presented increased gut microbiota richness and diversity, decreased Firmicutes/Bacteroidota (F/B) ratios, and altered serum metabolites compared with those of WT-HFD-fed mice. During analysis, several differential gut microbiota, including Lachnoclostridium, Harryflintia, Odoribacter, UCG-009, and unclassified_f_Butyricoccaceae, were screened between groups. These microbiota were found to be positively correlated with upregulated purine metabolism and bile acid metabolites in Nogo-B deficiency, while they were negatively correlated with downregulated corticosterone and tricarboxylic acid cyclic metabolites in Nogo-B deficiency. CONCLUSION Nogo-B deficiency delayed NAFLD progression, as demonstrated by reduced hepatocellular lipid accumulation, attenuated inflammation and liver injury, and ameliorated gut microbiota dysbiosis and metabolic disorders. Importantly, Odoribacter was strongly positively correlated with ALB and taurodeoxycholic acid, suggesting that it played a considerable role in the influence of Nogo-B on the progression of NAFLD, a specific feature of NAFLD in Nogo-B-/- mice. The regulation of bile acid metabolism by the gut microbiota may be a potential target for Nogo-B deficiency to ameliorate NAFLD.
Collapse
Affiliation(s)
- Xu Dong
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yu-Ting Xiong
- 307 Clinical Medical College of PLA, Anhui Medical University, Beijing, China
| | - Tingting He
- Department of Hepatology Medicine of Traditional Chinese Medicine, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Congyang Zheng
- Medical School of Chinese PLA, Beijing, China
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junjie Li
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
- School of Chengde Medical University, Chengdeshi, China
| | - Yingjie Zhuang
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yingjie Xu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ye Xiu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhixin Wu
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaomei Zhao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Zhaofang Bai
- Department of Hepatology, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
- China Military Institute of Chinese Materia, The Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.
| | - Lili Gao
- Medical School of Chinese PLA, Beijing, China.
- Department of Gastroenterology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
23
|
Tian L, Chen J, Yang M, Chen L, Qiu J, Jiang Y, Tan X, Qian Q, Liang X, Dou X. Xiezhuo Tiaozhi formula inhibits macrophage pyroptosis in the non-alcoholic fatty liver disease by targeting the SIRT1 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155776. [PMID: 38851104 DOI: 10.1016/j.phymed.2024.155776] [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: 02/07/2024] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is a challenging disease to interfere with and represents a potential long-term risk factor for hepatic fibrosis and liver cancer. The Xiezhuo Tiaozhi (XZTZ) formula, a water extract from crude herbs, has been widely used as an anti-NAFLD agent through clinical observation. However, the underlying pharmacological mechanisms of the XZTZ formula and its impact on the potential pathways against NAFLD have not been elucidated. PURPOSE Our study aims to investigate the pharmacological effects and underlying regulatory mechanisms of the XZTZ formula to treat NAFLD. METHODS The possible active components and pharmacological mechanisms of the XZTZ formula against NAFLD were identified using ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS) and molecular docking. To further explore the potential mechanisms, forty-eight 6-week-old male C57BL/6 J mice were given individual attention with high-fat and high-sugar diet (HFHSD) or relevant control (Ctrl) diets for 16 weeks to successfully construct a NAFLD mouse model. Subsequently, the levels of serum biochemicals, pathological changes in the liver, and pyroptosis levels were assessed in mice to investigate the therapeutic effects of the XZTZ formula. Further, LPS-induced RAW264.7 cells and Immortalized Mouse Kupffer cells (ImKC) were used to verify the potential mechanisms of the XZTZ formula against NAFLD in vitro. RESULTS We identified 7 chemical compounds and 2 potential therapeutic targets as plausible therapeutic points for the treatment of NAFLD using the XZTZ formula. Subsequent histopathological analysis revealed marked hepatic steatosis and lipid accumulation in the HFHSD mice liver, while conditions were effectively ameliorated by administration of the XZTZ formula. Additionally, our work demonstrated that the XZTZ formula could attenuate M1 polarization, promote M2 polarization, and suppress pyroptosis via the SIRT1 pathway in tissue samples. Moreover, validation performed through LPS-induced RAW264.7 and ImKC cells by showing that silencing SIRT1 weaken the effects of the XZTZ formula on relative pyroptosis affirmed that its role was associated with the SIRT1 pathway in macrophage. CONCLUSION These findings suggest that the XZTZ formula alleviated hepatic steatosis and lipid accumulation in NAFLD mice. These ameliorations are associated with mechanisms involving the attenuation of M1 polarization, promotion of M2 polarization, and anti-pyroptosis effects through the SIRT1 pathway.
Collapse
Affiliation(s)
- Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Meiqi Yang
- Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, Liaoning, China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiaolong Tan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qianyu Qian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiao Liang
- Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
24
|
Xue Y, Lu J, Liu Y, Gao Y, Gong Y, Yang Y, Xiong Y, Shi X. Dihydroartemisinin modulated arachidonic acid metabolism and mitigated liver inflammation by inhibiting the activation of 5-LOX and COX-2. Heliyon 2024; 10:e33370. [PMID: 39027511 PMCID: PMC11255665 DOI: 10.1016/j.heliyon.2024.e33370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
Background Dihydroartemisinin (DHA), a derivative of Artemisia annua, has been shown to possess anti-inflammatory properties. Besides, Yes-associated protein 1 (YAP1) plays a crucial role in maintaining liver homeostasis. Methods This study used Yap1 Flox/Flox, Albumin-Cre mice with hepatocyte-specific Yap1 knockout (referred to as Yap1 LKO) and their control mice (Yap1 Flox/Flox, referred to as Yap1 Flox). The effect of Yap1 on lipid metabolism homeostasis was investigated through non-targeted metabolomic analysis of mouse liver. Subsequently, DHA was administered to Yap1 LKO mice to assess its potential as a treatment. Liver pathology was evaluated via H&E staining, and the levels of AST, ALT, and TG were quantified using biochemical assays. The contents of arachidonic acid (AA), prostaglandin E1 (PGE1), and leukotrienes (LT) in the liver were measured using ELISA, while the protein expressions of PLIN2, 5-lipoxygenase (5-LOX), and cyclooxygenase-2 (COX-2) were analyzed through IHC staining. Results Hepatocyte-specific Yap1 knockout activated the AA metabolic pathway, resulting in increased elevated levels of AA, PGE1, and LT levels, along with inflammatory cytokine infiltration. DHA mitigated the elevation of metabolites such as PGE1 and LT caused by the AA metabolic pathway activation by down-regulating the levels of COX-2 and 5-LOX in the liver of Yap1 LKO mice. Moreover, it alleviated the accumulation of lipid vacuoles and reduced triglyceride (TG) and perilipin-2 (PLIN2) levels in the liver of Yap1 LKO mice. Conclusions Excessively low YAP1 expression induces liver inflammation and disturbances in lipid metabolism, whereas DHA modulated AA metabolism and mitigated liver inflammation by inhibiting the activation of 5-LOX and COX-2.
Collapse
Affiliation(s)
- Yu Xue
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Junlan Lu
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yiwei Liu
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yuting Gao
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yi Gong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yanguang Yang
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Yajun Xiong
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
| | - Xinli Shi
- Laboratory of Integrated Medicine Tumor Immunology, Shanxi University of Chinese Medicine, Taiyuan, 030000, China
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| |
Collapse
|
25
|
Wang T, Liao H, Lin J, Zhang M, Chen B, Yin R, Sun J, Dai H, Liu H. Antidiabetic action of the Chinese formula Shouhuitongbian and the underlying mechanism associated with alteration of gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155575. [PMID: 38636179 DOI: 10.1016/j.phymed.2024.155575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND The prevalence and incidence of type 2 diabetes mellitus (T2DM) have dramatically increased. The intestinal flora and its derived metabolites are demonstrated to play vital roles in the etiology and onset of T2DM. Shouhuitongbian (SHTB) is a traditional Chinese formula to treat constipation. SHTB is composed of seven herbs and components of Colla corii asini (CCA) that are obtained from the hide of Equus asinus L.. Some of herbs in SHTB such as Aloe vera (L.) Burm.f., Cassia obtusifolia L., fruits of Lycium barbarum L., and Citrus aurantium L. have shown to improve insulin resistance (IR) and T2DM in early reports. We hypothesized that SHTB composed of these herbs has antidiabetic effects. The antidiabetic efficacy and mechanism of action of SHTB have not been previously reported. HYPOTHESIS/PURPOSE To demonstrate the antidiabetic effect and elucidate the underlying mechanisms of SHTB from the perspective of gut microbiota. STUDY DESIGN The main compounds were identified and quantified by high-performance liquid chromatography (HPLC)-mass spectrometry analysis. High fat diet (HFD)-fed mice and db/db mice were used to assess the antidiabetic effects and the mechanism of SHTB. The underlying mechanisms were evaluated by enzyme-linked immunosorbent assay (ELISA), western blot analysis, quantitative real time polymerase chain reaction (qPCR) analysis, 16S rRNA high-throughput sequencing, and targeted metabolome analysis. METHODS HFD-fed mice and db/db mice were orally treated with the standard positive drug metformin (100 mg/kg/d) and with SHTB (200 and 100 mg/kg/d), which was chemically characterized according to the European Medicine Agency (EMA) guidelines. The beneficial effects of SHTB were studied by homeostasis model assessment of insulin resistance (HOMA-IR) index, oral glucose tolerance test (OGTT), insulin tolerance test (ITT), total cholesterol (T-CHO), triglyceride (TG), and inflammation. Subsequently, 16S rDNA-based high-throughput pyrosequencing and GC-MS-based targeted metabolomics profiling were performed to analyze the gut microbiota composition and metabolites profile in the gut, respectively. Moreover, the mammalian target of rapamycin complex 1 (mTORC1) / insulin receptor substrate 1 (IRS-1) / phosphoinositide 3-kinase (PI3K) / protein kinase B (AKT) pathway was evaluated via qPCR and western blot. RESULTS Chemically characterized SHTB, in which six markers were quantified, effectively alleviated glucose intolerance and IR, ameliorated lipid metabolism dysfunction, and reduced inflammation. In addition, 16S rDNA sequencing found that SHTB reshaped the composition of intestinal flora, as indicated by the enrichment of Akkermansia and Parabacteroides in both HFD-fed and db/db mice. Moreover, SHTB enhanced the intestinal production of short-chain fatty acids (SCFAs) and branched short-chain fatty acids (BSCFAs), and reduced the levels of the fecal and circulating branched-chain amino acids (BCAAs). The IRS-1/PI3K/AKT signaling pathway was upregulated after treatment with SHTB. CONCLUSION Orally administration of SHTB effectively improved IR and reduced hyperglycemia in mice. Treatment with SHTB regulated the gut BCAAs-mTORC1/IRS-1/PI3K/AKT axis by enhancing the BCAAs catabolism in the gut, which attenuated the deleterious effect of BCAAs on the IRS-1 signaling pathway.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Huan Liao
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jinghan Lin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Mingkai Zhang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Key Laboratory of Structure-Based Drug Design & Discovery of Education, College of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Baosong Chen
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Ruopeng Yin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, No. 1 Beichenxi Road, Chaoyang District, Beijing, 100101, PR China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| |
Collapse
|
26
|
Zhang L, Li Y, Fan CD, Jiang YH, Sheng LS, Song XY, Lin YX, Xue YT, Sun R. Chinese medicinal formula Fu Xin decoction against chronic heart failure by inhibiting the NLRP3/caspase-1/GSDMD pyroptotic pathway. Biomed Pharmacother 2024; 174:116548. [PMID: 38599064 DOI: 10.1016/j.biopha.2024.116548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/28/2024] [Accepted: 04/04/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Various heart diseases ultimately lead to chronic heart failure (CHF). In CHF, the inflammatory response is associated with pyroptosis, which is mediated by the NOD-like receptor protein 3 (NLRP3) inflammasome. Fu Xin decoction (FXD) is commonly used in clinical practice to treat CHF and improve inflammatory conditions. However, the specific pharmacological mechanisms of action for FXD in these processes have yet to be fully understood. PURPOSE The objective of this study was to examine the protective mechanism of FXT against CHF, both in H9c2 cells and mice. METHOD A CHF mouse model was established, and the effect of FXD was observed via gavage. Cardiac function was evaluated using echocardiography, while serum BNP and LDH levels were analyzed to assess the severity of CHF. Hematoxylin and eosin staining (H&E) and Masson staining were performed to evaluate myocardial pathological changes, and TdT-mediated dUTP Nick-End Labeling staining was used to detect DNA damage. Additionally, doxorubicin was utilized to induce myocardial cell injury in H9c2 cells, establishing a relevant model. CCK8 was used to observe cell viability and detect LDH levels in the cell supernatant. Subsequently, the expression of pyroptosis-related proteins was detected using immunohistochemistry, immunofluorescence, and western blotting. Finally, the pharmacological mechanism of FXD against CHF was further validated by treating H9c2 cells with an NLRP3 activator and inducing NLRP3 overexpression. RESULT According to current research findings, echocardiography demonstrated a significant improvement of cardiac function by FXD, accompanied by reduced levels of BNP and LDH, indicating the amelioration of cardiac injury in CHF mice. FXD exhibited the ability to diminish serum CRP and MCP inflammatory markers in CHF mice. The results of HE and Masson staining analyses revealed a significant reduction in pathological damage of the heart tissue following FXD treatment. The CCK8 assay demonstrated the ability of FXD to enhance H9c2 cell viability, improve cell morphology, decrease LDH levels in the cell supernatant, and alleviate cell damage. Immunohistochemistry, Western blotting, and immunofluorescence staining substantiated the inhibitory effect of FXD on the NLRP3/caspase-1/GSDMD pyroptosis signaling pathway in both CHF and H9c2 cell injury models. Ultimately, the administration of the NLRP3 activator (Nigericin) and the overexpression of NLRP3 counteract the effects of FXD on cardiac protection and pyroptosis inhibition in vitro. CONCLUSION FXD exhibits a cardioprotective effect, improving CHF and alleviating pyroptosis by inhibiting the NLRP3/caspase-1/GSDMD pathway.
Collapse
Affiliation(s)
- Lei Zhang
- The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Cun-Dong Fan
- Department of Neurology, Key Lab of Cerebral Microcirculation in Universities of Shandong, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong, China
| | - Yong-Hao Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Li-Song Sheng
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China; Shandong Academy of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xu-Yu Song
- The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yu-Xing Lin
- The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Yi-Tao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Rong Sun
- The Second Hospital of Shandong University, Jinan, Shandong, China; Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
27
|
Chang K, Guo R, Hu W, Wang X, Cao F, Qiu J, Li J, Han Q, Du Z, Dou X, Li S. Xie Zhuo Tiao Zhi formula ameliorates chronic alcohol-induced liver injury in mice. Front Pharmacol 2024; 15:1363131. [PMID: 38681193 PMCID: PMC11045942 DOI: 10.3389/fphar.2024.1363131] [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: 12/29/2023] [Accepted: 04/03/2024] [Indexed: 05/01/2024] Open
Abstract
This study aimed to evaluate the protective role and potential mechanisms of Xie Zhuo Tiao Zhi decoction (XZTZ) on alcohol-associated liver disease (ALD). XZTZ significantly alleviated alcohol-induced liver dysfunction, based on histological examinations and biochemical parameters after 4-week administration. Mechanically, alcohol-stimulated hepatic oxidative stress was ameliorated by XZTZ, accompanied by the improvement of Nrf2/Keap1 expression and alcohol-activated phosphorylation of pro-inflammatory transcription factors, including JNK, P38, P65, and IκBα, were rescued by XZTZ. In conclusion, XZTZ demonstrates potential in alleviating alcohol-induced liver injury, oxidative stress, and inflammation possibly through modulation of Nrf2/Keap1 and MAPKs/NF-κB signaling pathways, suggesting its potential as a therapeutic option for patients with alcoholic liver disease.
Collapse
Affiliation(s)
- Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rui Guo
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenbo Hu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuezhu Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Feiwei Cao
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaomei Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiang Han
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhongyan Du
- Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Zhejiang Engineering Research Center for ‘Preventive Treatment’ Smart Health of Traditional Chinese Medicine, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
28
|
Wang M, Li H, Liu C, Zhang Y, Wu Q, Yang Y. Lingguizhugan Decoction Improved Obesity by Modulating the Gut Microbiota and its Metabolites in Mice. Curr Drug Metab 2024; 25:276-287. [PMID: 38982915 DOI: 10.2174/0113892002289388240705113755] [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: 02/25/2024] [Revised: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND The global obese population is rapidly increasing, urgently requiring the development of effective and safe weight-loss medications. The classic Chinese medicine formulation Lingguizhugan Decoction has exerted a significant anti-obesity effect. However, the underlying mechanism is still unclear. OBJECTIVE This study aimed to explore the mechanism of LGZGD in the treatment of obesity based on the gut microbiota and its metabolites. METHODS Three different dosages of LGZGD were gavaged to ob/ob mice for 8 weeks. Body mass and visceral fat mass were evaluated. Additionally, the changes in gut microbiota, fecal and plasma metabolites in mice after LGZGD treatment were analyzed by metagenomics and non-targeted metabolomics. RESULTS The results demonstrated a significant anti-obesity effect of LGZGD treatment in ob/ob mice. Furthermore, the metagenomic analysis revealed that LGZGD reduced the ratio of Firmicutes / Bacteroidetes (F to B) in the gut, restored gut microbiota diversity, and identified 3 enriched KEGG pathways, including energy metabolism, lipid metabolism, and energy production and conversion pathways. Based on non-targeted metabolomics analysis, 20 key metabolites in the feces and 30 key metabolites in the plasma responding to LGZGD treatment were identified, and the levels of Eicosapentaenoic acid (EPA) and Myristoleic acid (MA) might be the metabolites related to gut microbiota after LGZGD treatment. Their biological functions were mainly related to the metabolism pathway. CONCLUSIONS These findings suggested that LGZGD had therapeutic potential for obesity. The mechanism of LGZGD alleviating obesity was associated with improving dysbiosis of the gut microbiota. LDZGD affected gut microbiota-derived metabolites of EPA and MA and may act on energy metabolism pathways.
Collapse
Affiliation(s)
- Meiling Wang
- Traditional Chinese Medicine Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
- Guangdong Pharmaceutical University, Xiaoguwei Street, Panyu District, Guangzhou, China
| | - Hairong Li
- Guangdong Pharmaceutical University, Xiaoguwei Street, Panyu District, Guangzhou, China
| | - Chunmei Liu
- Guangdong Pharmaceutical University, Xiaoguwei Street, Panyu District, Guangzhou, China
| | - Yuanyuan Zhang
- Guangdong Pharmaceutical University, Xiaoguwei Street, Panyu District, Guangzhou, China
| | - Qian Wu
- Guangdong Pharmaceutical University, Xiaoguwei Street, Panyu District, Guangzhou, China
| | - Yubin Yang
- Traditional Chinese Medicine Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, China
| |
Collapse
|
29
|
Liu YC, Wei G, Liao ZQ, Wang FX, Zong C, Qiu J, Le Y, Yu ZL, Yang SY, Wang HS, Dou XB, Wang CY. Design and Synthesis of Novel Indole Ethylamine Derivatives as a Lipid Metabolism Regulator Targeting PPARα/CPT1 in AML12 Cells. Molecules 2023; 29:12. [PMID: 38202597 PMCID: PMC10779794 DOI: 10.3390/molecules29010012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/13/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Peroxisome proliferator-activated receptor alpha (PPARα) and carnitine palmitoyltransferase 1 (CPT1) are important targets of lipid metabolism regulation for nonalcoholic fatty liver disease (NAFLD) therapy. In the present study, a set of novel indole ethylamine derivatives (4, 5, 8, 9) were designed and synthesized. The target product (compound 9) can effectively activate PPARα and CPT1a. Consistently, in vitro assays demonstrated its impact on the lipid accumulation of oleic acid (OA)-induced AML12 cells. Compared with AML12 cells treated only with OA, supplementation with 5, 10, and 20 μM of compound 9 reduced the levels of intracellular triglyceride (by 28.07%, 37.55%, and 51.33%) with greater inhibitory activity relative to the commercial PPARα agonist fenofibrate. Moreover, the compound 9 supplementations upregulated the expression of hormone-sensitive triglyceride lipase (HSL) and adipose triglyceride lipase (ATGL) and upregulated the phosphorylation of acetyl-CoA carboxylase (ACC) related to fatty acid oxidation and lipogenesis. This dual-target compound with lipid metabolism regulatory efficacy may represent a promising type of drug lead for NAFLD therapy.
Collapse
Affiliation(s)
- Yu-Chen Liu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| | - Gang Wei
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, China; (G.W.); (Z.-Q.L.); (F.-X.W.); (H.-S.W.)
| | - Zhi-Qiang Liao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, China; (G.W.); (Z.-Q.L.); (F.-X.W.); (H.-S.W.)
| | - Fang-Xin Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, China; (G.W.); (Z.-Q.L.); (F.-X.W.); (H.-S.W.)
| | - Chunxiao Zong
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| | - Jiannan Qiu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| | - Yifei Le
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| | - Zhi-Ling Yu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China;
| | - Seo Young Yang
- Department of Biology Education, Teachers College and Institute for Phylogenomics and Evolution, Kyungpook National University, Daegu 41566, Republic of Korea;
| | - Heng-Shan Wang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin 541004, China; (G.W.); (Z.-Q.L.); (F.-X.W.); (H.-S.W.)
| | - Xiao-Bing Dou
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| | - Cai-Yi Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou 310053, China; (Y.-C.L.); (C.Z.); (J.Q.); (Y.L.)
| |
Collapse
|