1
|
Arafa SS, Elnoury HA, Badr El-Din S, Sakr MA, Hendawi FF, Masoud RAE, Barghash SS, Elbehairy DS, Hemeda AA, Farrag IM, Abdelrahman DS, Elsadek AM, Ghanem SK, AboShabaan HS, Atwa AM, Nour El Din M, Radwan AF, Al-Zahrani M, Alhomodi AF, Abdulfattah AM, Abdelkader A. Acetamiprid-induced pulmonary toxicity via oxidative stress, epithelial-mesenchymal transition, apoptosis, and extracellular matrix accumulation in human lung epithelial cells and fibroblasts: Protective role of heat-killed Lactobacilli. Food Chem Toxicol 2025; 198:115322. [PMID: 39961414 DOI: 10.1016/j.fct.2025.115322] [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: 10/08/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Acetamiprid (ACE) is a neonicotinoid insecticide with widespread global application, resulting in persistent human exposure. The current research examined the toxicological implications of ACE exposure on human lung fibroblasts (MRC-5 cells) and bronchial epithelial cells (BEAS-2B cells). The following implications were explored: oxidative stress, epithelial-mesenchymal transition, apoptosis, cellular proliferation, and extracellular matrix accumulation. The prospective protective properties of heat-killed Lactobacillus fermentum and Lactobacillus delbrueckii (HKL) were further studied. The 14-day exposure to ACE at 4 μM triggered oxidative stress and inflammation. ACE promoted epithelial-mesenchymal transition, as evidenced by the decline of protein and mRNA abundances of E-cadherin alongside increased protein and mRNA quantities of α-SMA and N-cadherin in BEAS-2B cells. Additionally, it elicited apoptosis in BEAS-2B cells and stimulated the cellular growth of MRC-5 cells. The TGF-β1/Smad pathway was activated upon ACE exposure, leading to the accumulation of extracellular matrix. HKL demonstrated antioxidant, anti-apoptotic, anti-proliferative, and anti-fibrotic properties, mitigating ACE-induced toxicity. Our findings delineate the molecular mechanisms underlying epithelial-mesenchymal transition, inflammation, oxidative stress, and extracellular matrix accumulation in ACE-induced pulmonary fibrosis, which provides new insights into pulmonary injury. Additionally, this investigation would offer us an approach to mitigate lung deterioration induced by ACE through utilizing heat-killed probiotic supplementation.
Collapse
Affiliation(s)
- Samah S Arafa
- Department of Pesticides, Faculty of Agriculture, Menoufia University, Shibin Elkom, Egypt.
| | - Heba A Elnoury
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Sahar Badr El-Din
- Department of Pharmacology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed A Sakr
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Suez University, Suez, Egypt
| | - Fatma Fawzi Hendawi
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Rehab Ali Elsayed Masoud
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Samia Soliman Barghash
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt; Department of Pharmacology and Toxicology, Pharmacy College, Qassim University, Saudi Arabia
| | - Doaa Sabry Elbehairy
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Ayat Abdelaty Hemeda
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Islam Mostafa Farrag
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Doaa Sayed Abdelrahman
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Amira Mohammad Elsadek
- Department of Chest Diseases, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - Sahar K Ghanem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sohag University, Sohag, Egypt
| | - Hind S AboShabaan
- Department of Clinical Pathology, National Liver Institute Hospital, Menoufia University, Shibin Elkom, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt; Department of Pharmacology and Toxicology, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Mahmoud Nour El Din
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, University of Sadat City, Cairo, Egypt
| | - Abdullah F Radwan
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt; Department of Pharmacy, Kut University College, Al Kut, Wasit, Iraq
| | - Majid Al-Zahrani
- Department of Biological Sciences, College of Sciences and Art, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Ahmad F Alhomodi
- Department of Biology, College of Science and Arts, Najran University, Saudi Arabia
| | - Ahmed M Abdulfattah
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Embryonic Stem Cell Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Afaf Abdelkader
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
2
|
Xie S, Meng Q, Wang L. The effect of gut microbiome and plasma metabolome on systemic sclerosis: a bidirectional two-sample Mendelian randomization study. Front Microbiol 2024; 15:1427195. [PMID: 39086645 PMCID: PMC11288946 DOI: 10.3389/fmicb.2024.1427195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Background Cellular and molecular biology, combined with research on the human microbiome and metabolome, have provided new insights into the pathogenesis of systemic sclerosis (SSc). However, most studies on gut microbiota (GM) and metabolome in SSc are observational studies. The impact of confounding factors and reverse causation leads to different insights. To shed light on this matter, we utilized Mendelian randomization (MR) to determine the causal effect of GM/metabolites on SSc. Methods Based on summary-level data from genome-wide association studies, bidirectional Two-sample MR was conducted involving 196 GM, 1400 plasma metabolism, and 9,095 SSc. Inverse Variance Weighting (IVW) was mainly used for effect estimation. Results Forward MR analysis found that three GM and two plasma metabolites are causally related to SSc. IVW results showed Victivallaceae (family) (OR, 1.469; 95%CI, 1.099-1.963; p = 0.009) and LachnospiraceaeUCG004 (genus) (OR, 1.548; 95%CI, 1.020-2.349; p = 0.04) were risk factor of SSc. Conversely, Prevotella7 (genus) (OR, 0.759; 95%CI, 0.578-0.997; p = 0.048)was a protective factor of SSc. The results on plasma metabolites indicated that Pregnenediol disulfate (C21H34O8S2) levels (OR, 1.164; 95%CI, 1.006-1.347; p = 0.041)was a risk factor of SSc, while Sphingomyelin (d18:1/19:0, d19:1/18:0) levels (OR, 0.821; 95%CI, 0.677-0.996; p = 0.045)was a protective factor of SSc. Reverse MR analysis did not find causally relationship between SSc and the above GM/plasma metabolites. Conclusion Our results revealed the causally effect between GM/plasma metabolites and SSc. These findings provided new insights into the mechanism of SSc. In particular, we demonstrated Prevotella7 was a protective factor of SSc despite its controversial role in SSc in previous researches.
Collapse
Affiliation(s)
- Shasha Xie
- Department of Rheumatology and Nephrology, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qiming Meng
- Department of Rheumatology and Nephrology, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Wang
- Department of Rheumatology and Nephrology, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Gao Y, Hao Z, Zhang H, Liu J, Zhou G, Wen H, Su Q, Tong C, Huang S, Wang X. Forsythiaside A attenuates lipopolysaccharide-induced mouse mastitis by activating autophagy and regulating gut microbiota and metabolism. Chem Biol Interact 2024; 396:111044. [PMID: 38729284 DOI: 10.1016/j.cbi.2024.111044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/12/2024]
Abstract
Mastitis is an inflammatory disease of the mammary gland with a high incidence in lactating animals, significantly impacting their health and breastfeeding. Moreover, mastitis adversely affects milk quality and yield, resulting in substantial economic losses for the dairy farming industry. Forsythiaside A (FTA), a phenylethanol glycoside analog extracted from Forsythia, exhibits notable anti-inflammatory and antioxidant properties. However, its protective effects and specific mechanisms against mastitis remain unclear. In this study, a lipopolysaccharide (LPS)-induced mouse mastitis model was used to investigate the protective effect of FTA on LPS-induced mastitis and its potential mechanism using histological assays, Western blot, qRT-PCR, FITC-albumin permeability test, 16s rRNA gene sequencing analysis and non-targeted metabolomics assays to investigate the protective effect of FTA on LPS-induced mastitis model and its potential mechanism. The results demonstrated that FTA significantly mitigated LPS-induced mouse mastitis by reducing inflammation and apoptosis levels, modulating the PI3K/AKT/mTOR signaling pathways, inducing autophagy, and enhancing antioxidant capacity and the expression of tight junction proteins. Furthermore, FTA increased the abundance of beneficial microbiota while decreasing the levels of harmful microbiota in mice, thus counteracting the gut microbiota disruption induced by LPS stimulation. Intestinal metabolomics analysis revealed that FTA primarily regulated LPS-induced metabolite alterations through key metabolic pathways, such as tryptophan metabolism. This study confirms the anti-inflammatory and antioxidant effects of FTA on mouse mastitis, which are associated with key metabolic pathways, including the restoration of gut microbiota balance and the regulation of tryptophan metabolism. These findings provide a novel foundation for the treatment and prevention of mammalian mastitis using FTA.
Collapse
Affiliation(s)
- Yingkui Gao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Zhonghua Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Huaqiang Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Jingjing Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Guangwei Zhou
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Haojie Wen
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Qing Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China.
| | - Chao Tong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China; Zhengzhou Key Laboratory of Research and Evaluation of Traditional Chinese Veterinary Medicine, Zhengzhou, 450046, PR China.
| | - Shucheng Huang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Zhengzhou Key Laboratory of Research and Evaluation of Traditional Chinese Veterinary Medicine, Zhengzhou, 450046, PR China.
| | - Xuebing Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, PR China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, PR China; Zhengzhou Key Laboratory of Research and Evaluation of Traditional Chinese Veterinary Medicine, Zhengzhou, 450046, PR China.
| |
Collapse
|
4
|
Chuandong Z, Hu J, Li J, Wu Y, Wu C, Lai G, Shen H, Wu F, Tao C, Liu S, Zhang W, Shao H. Distribution and roles of Ligilactobacillus murinus in hosts. Microbiol Res 2024; 282:127648. [PMID: 38367479 DOI: 10.1016/j.micres.2024.127648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/26/2023] [Accepted: 02/10/2024] [Indexed: 02/19/2024]
Abstract
Ligilactobacillus murinus, a member of the Ligilactobacillus genus, holds significant potential as a probiotic. While research on Ligilactobacillus murinus has been relatively limited compared to well-studied probiotic lactic acid bacteria such as Limosilactobacillus reuteri and Lactobacillus gasseri, a mounting body of evidence highlights its extensive involvement in host intestinal metabolism and immune activities. Moreover, its abundance exhibits a close correlation with intestinal health. Notably, beyond the intestinal context, Ligilactobacillus murinus is gaining recognition for its contributions to metabolism and regulation in the oral cavity, lungs, and vagina. As such, Ligilactobacillus murinus emerges as a potential probiotic candidate with a pivotal role in supporting host well-being. This review delves into studies elucidating the multifaceted roles of Ligilactobacillus murinus. It also examines its medicinal potential and associated challenges, underscoring the imperative to delve deeper into unraveling the mechanisms of its actions and exploring its health applications.
Collapse
Affiliation(s)
- Zhou Chuandong
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jicong Hu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jiawen Li
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Yuting Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Chan Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Guanxi Lai
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Han Shen
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Fenglin Wu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Changli Tao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Song Liu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Zhang
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| | - Hongwei Shao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
5
|
Hua H, Zhao QQ, Kalagbor MN, Yu GZ, Liu M, Bian ZR, Zhang BB, Yu Q, Xu YH, Tang RX, Zheng KY, Yan C. Recombinant adeno-associated virus 8-mediated inhibition of microRNA let-7a ameliorates sclerosing cholangitis in a clinically relevant mouse model. World J Gastroenterol 2024; 30:471-484. [PMID: 38414587 PMCID: PMC10895596 DOI: 10.3748/wjg.v30.i5.471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/17/2023] [Accepted: 01/12/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Primary sclerosing cholangitis (PSC) is characterized by chronic inflammation and it predisposes to cholangiocarcinoma due to lack of effective treatment options. Recombinant adeno-associated virus (rAAV) provides a promising platform for gene therapy on such kinds of diseases. A microRNA (miRNA) let-7a has been reported to be associated with the progress of PSC but the potential therapeutic implication of inhibition of let-7a on PSC has not been evaluated. AIM To investigate the therapeutic effects of inhibition of a miRNA let-7a transferred by recombinant adeno-associated virus 8 (rAAV8) on a xenobiotic-induced mouse model of sclerosing cholangitis. METHODS A xenobiotic-induced mouse model of sclerosing cholangitis was induced by 0.1% 3,5-Diethoxycarbonyl-1,4-Dihydrocollidine (DDC) feeding for 2 wk or 6 wk. A single dose of rAAV8-mediated anti-let-7a-5p sponges or scramble control was injected in vivo into mice onset of DDC feeding. Upon sacrifice, the liver and the serum were collected from each mouse. The hepatobiliary injuries, hepatic inflammation and fibrosis were evaluated. The targets of let-7a-5p and downstream molecule NF-κB were detected using Western blot. RESULTS rAAV8-mediated anti-let-7a-5p sponges can depress the expression of let-7a-5p in mice after DDC feeding for 2 wk or 6 wk. The reduced expression of let-7a-5p can alleviate hepato-biliary injuries indicated by serum markers, and prevent the proliferation of cholangiocytes and biliary fibrosis. Furthermore, inhibition of let-7a mediated by rAAV8 can increase the expression of potential target molecules such as suppressor of cytokine signaling 1 and Dectin1, which consequently inhibit of NF-κB-mediated hepatic inflammation. CONCLUSION Our study demonstrates that a rAAV8 vector designed for liver-specific inhibition of let-7a-5p can potently ameliorate symptoms in a xenobiotic-induced mouse model of sclerosing cholangitis, which provides a possible clinical translation of PSC of human.
Collapse
Affiliation(s)
- Hui Hua
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Qian-Qian Zhao
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Miriam Nkesichi Kalagbor
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Guo-Zhi Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Man Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Zheng-Rui Bian
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Bei-Bei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Qian Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Yin-Hai Xu
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu Province, China
| | - Ren-Xian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| |
Collapse
|
6
|
Li Y, Peng X, Wang G, Zan B, Wang Y, Zou J, Tian T, Meng Q, Shi R, Wang T, Wu J, Ma Y. Identifying hepatoprotective mechanism and effective components of Yinchenzhufu decoction in chronic cholestatic liver injury using a comprehensive strategy based on metabolomics, molecular biology, pharmacokinetics, and cytology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117060. [PMID: 37598769 DOI: 10.1016/j.jep.2023.117060] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/22/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Traditional Chinese Medicine (TCM), cholestasis liver disease belongs to jaundice. Yinchenzhufu decoction (YCZFD) is a classic formula used for treating jaundice. AIM OF THE STUDY This study was aimed to investigate the potential mechanism and effective components of YCZFD in chronic cholestatic liver injury (CCLI). MATERIALS AND METHODS A chronic cholestatic mouse model induced by 3, 5-diethoxycarbonyl-1, 4-dihydroxychollidine was used to investigate the effect of YCZFD. Then, metabolomics was used to investigate the metabolites influenced by YCZFD. Serum and liver bile acid (BA) levels were measured using liquid chromatography coupled with triple quadruple mass spectrometry (LC-MS/MS), and the gene and protein expressions of BA transporters and metabolic enzymes were detected. Additionally, the pharmacokinetics of multiple components of YCZFD was explored to clarify the potential effective components. The effects of absorbed components of YCZFD on BA metabolism and transporter function, inflammation, and farnesoid X receptor (FXR) and pregnane X receptor (PXR) activation were analyzed using sandwich cultured rat hepatocytes, AML12 cells, and dual-luciferase receptor systems, respectively. RESULTS YCZFD decreased the liver damage in chronic cholestatic mice. Serum metabolomics results indicated that the main pathways influenced by YCZFD involved primary BA biosynthesis and arachidonic acid metabolism. YCZFD upregulated the expression of FXR, PXR, and BA efflux transporters and the metabolic enzymes of liver tissues, promoting BA excretion and metabolism in cholestatic mice. Additionally, YCZFD downregulated the expression of genes and proteins of the toll-like receptor 4 (TLR4)/nuclear factor kappa-B (NF-κB) pathway and decreased liver inflammation. The pharmacokinetic study indicated that multiple components showed different pharmacokinetic properties. Among the absorbed components of YCZFD, multiple components activated the transcription of FXR and PXR, regulated BA transporters and metabolic enzyme function, and reduced the gene expression of TLR4 and NF-κB1. CONCLUSION YCZFD can ameliorate CCLI by promoting the excretion and metabolism of BAs and inhibiting inflammation via the TLR4/NF-κB signaling pathway. The multiple components of YCZFD could act on BA homeostasis regulation and anti-inflammation, exhibiting a combined effect against CCLI.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Xiaotian Peng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Guofeng Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Bin Zan
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Yahang Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Juan Zou
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Tian Tian
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Qian Meng
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Rong Shi
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Tianming Wang
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Jiasheng Wu
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Yueming Ma
- Department of Pharmacology, School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China; Shanghai Key Laboratory of Compound Chinese Medicines, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|