1
|
Liu X, Chen G, Yang Y, Liu F, Wu G, An L, Tang T, Zhang J. Comprehensive multi-omics analysis reveals the mechanism of hepatotoxicity induced by Emilia sonchifolia (L.) DC. JOURNAL OF ETHNOPHARMACOLOGY 2025:119371. [PMID: 39826791 DOI: 10.1016/j.jep.2025.119371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Emilia sonchifolia is a very widely used traditional Chinese medicine, with the efficacy of heat-clearing, detoxicating, dissipating blood stasis, reducing swelling and relieving pain. As a widely used traditional miao herb, Emilia sonchifolia is often used to treat upper respiratory tract infections, oral ulcer, pneumonia, mastitis, enteritis, bacillum, urinary tract infection, sores, eczema, falls and injuries, etc. In fact, many cases of liver injury caused by Emilia sonchifolia have been reported clinically. However, the mechanisms underlying hepatotoxicity induced by Emilia sonchifolia remain poorly understood. AIM OF THE STUDY This study aimed to systematically evaluate the acute and chronic hepatotoxicity of water extract from Emilia sonchifolia, identify its hepatotoxic metabolites, and elucidate the potential mechanisms underlying Emilia sonchifolia-induced hepatotoxicity. MATERIAL AND METHOD The chemical components in the water extract of Emilia sonchifolia were identified using mass spectrometry. The acute toxicity study was conducted by orally administering a gradient dose of water extract of Emilia sonchifolia ranging from 0 to 37.6 g/kg. Mice were orally administered a water extract of Emilia sonchifolia at a dose of 13.72 g/kg/d for 14 days to induce liver injury. The hepatotoxicity was evaluated using hematoxylin and eosin staining as well as enzyme-linked immunosorbent assay (ELISA). The mechanisms of hepatotoxicity were explored through transcriptomics, proteomics, and metabolomics analysis. Meanwhile, the core pathways related to the hepatotoxicity of Emilia sonchifolia were analyzed and validated using quantitative reverse transcription polymerase chain reaction (qRT-PCR) and ELISA. RESULT The present study demonstrates that the water extract of Emilia sonchifolia can induce hepatotoxicity in mice. We found that the water extract of Emilia sonchifolia contained hepatotoxic pyrrolizidine alkaloids, such as seneciphyllin, senecionine, rinderine, echimidine, retrorsine and echimidine N-oxide. A dose of 19.20 g/kg or higher of the water extract of Emilia sonchifolia caused acute liver failure and death in mice. A dose of 13.72 g/kg or lower of the water extract of Emilia sonchifolia produced dose-dependent acute hepatotoxicity. Meanwhile, a dose of 13.72 g/kg of the water extract from Emilia sonchifolia induced chronic hepatotoxicity in mice. Furthermore, the results of liver transcriptomics, proteomics, and metabolomics indicate that the mechanism of hepatotoxicity induced by the water extract of Emilia sonchifolia is associated with ferroptosis caused by abnormalities in bile acid accumulation, lipid and bilirubin accumulation, and glutathione metabolism. The validation experiment results demonstrate that in mice treated with the water extract of Emilia sonchifolia, the gene levels of Cyp2c29, Cyp3a41a and Ugt2b1 decreased while the gene level of Hsd3b3 increased. In mice treated with a water extract of Emilia sonchifolia, the levels of total bilirubin, direct bilirubin, total bile acids, alkaline phosphatase, and γ-glutamyl transferase were significantly elevated. Additionally, in mice treated with a water extract of Emilia sonchifolia, the levels of malondialdehyde increased while the levels of catalase and superoxide dismutase decreased. CONCLUSION In conclusion, our results suggest that the water extract of Emilia sonchifolia can cause hepatotoxicity in mice. The chronic hepatotoxicity of Emilia sonchifolia is associated with Cyp2c29, Cyp3a41a, Ugt2b1, and Hsd3b3-mediated cholestasis, oxidative stress, and ferroptosis.
Collapse
Affiliation(s)
- Xin Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Gongzhen Chen
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China.
| | - Yuqi Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Feng Liu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Guangzhou Wu
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Lili An
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| | - Ting Tang
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China.
| | - Jinqiang Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China.
| |
Collapse
|
2
|
Xia N, Chen QH, Meng ZJ, Ma SY, Huang JL, Shen R, Dong YT, Du HW, Zhou K. Isobavachin induces autophagy-mediated cytotoxicity in AML12 cells via AMPK and PI3K/Akt/mTOR pathways. Toxicol In Vitro 2024; 100:105919. [PMID: 39154867 DOI: 10.1016/j.tiv.2024.105919] [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: 04/22/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
Isobavachin (IBA) is a dihydroflavonoid compound with various pharmacological effects. However, further investigation into the hepatotoxicity of IBA is necessary. This study aims to identify the hepatotoxic effects of IBA and explore its potential mechanisms. The study assessed the impact of IBA on the viability of AML12, HepG2, LO2, rat, and mouse primary hepatocytes using MTT and LDH assays. Autophagy was detected in AML12 cells after IBA treatment using electron microscopy, MDC, and Ad-mCherry-GFP-LC3B fluorescence. The effect of IBA on autophagy-related proteins was examined using Western blot. The results showed that IBA had dose-dependent inhibitory effects on five cells, induced autophagy in AML12 cells, and promoted autophagic flux. The study found that IBA treatment inhibited phosphorylation of PI3K, Akt, and mTOR, while increasing phosphorylation levels of AMPK and ULK1. Treatment with both AMPK and PI3K inhibitors reversed the expression of AMPK and PI3K-Akt-mTOR signaling pathway proteins. These results suggest that IBA may have hepatocytotoxic effects but can also prevent IBA hepatotoxicity by inhibiting the AMPK and PI3K/Akt/mTOR signaling pathways. This provides a theoretical basis for preventing and treating IBA hepatotoxicity in clinical settings.
Collapse
Affiliation(s)
- Ning Xia
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing-Hai Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhao-Jun Meng
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shu-Yue Ma
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia-Li Huang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong Shen
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu-Tong Dong
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hai-Wei Du
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Kun Zhou
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
3
|
Rao SW, Liu CJ, Liang D, Duan YY, Chen ZH, Li JJ, Pang HQ, Zhang FX, Shi W. Multi-omics and chemical profiling approaches to understand the material foundation and pharmacological mechanism of sophorae tonkinensis radix et rhizome-induced liver injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118224. [PMID: 38642623 DOI: 10.1016/j.jep.2024.118224] [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: 11/06/2023] [Revised: 03/31/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sophorae tonkinensis Radix et Rhizoma (STR) is an extensively applied traditional Chinese medicine (TCM) in southwest China. However, its clinical application is relatively limited due to its hepatotoxicity effects. AIM OF THE STUDY To understand the material foundation and liver injury mechanism of STR. MATERIALS AND METHODS Chemical compositions in STR and its prototypes in mice were profiled by ultra-performance liquid chromatography coupled quadrupole-time of flight mass spectrometry (UPLC-Q/TOF MS). STR-induced liver injury (SILI) was comprehensively evaluated by STR-treated mice mode. The histopathologic and biochemical analyses were performed to evaluate liver injury levels. Subsequently, network pharmacology and multi-omics were used to analyze the potential mechanism of SILI in vivo. And the target genes were further verified by Western blot. RESULTS A total of 152 compounds were identified or tentatively characterized in STR, including 29 alkaloids, 21 organic acids, 75 flavonoids, 1 quinone, and 26 other types. Among them, 19 components were presented in STR-medicated serum. The histopathologic and biochemical analysis revealed that hepatic injury occurred after 4 weeks of intragastric administration of STR. Network pharmacology analysis revealed that IL6, TNF, STAT3, etc. were the main core targets, and the bile secretion might play a key role in SILI. The metabolic pathways such as taurine and hypotaurine metabolism, purine metabolism, and vitamin B6 metabolism were identified in the STR exposed groups. Among them, taurine, hypotaurine, hypoxanthine, pyridoxal, and 4-pyridoxate were selected based on their high impact value and potential biological function in the process of liver injury post STR treatment. CONCLUSIONS The mechanism and material foundation of SILI were revealed and profiled by a multi-omics strategy combined with network pharmacology and chemical profiling. Meanwhile, new insights were taken into understand the pathological mechanism of SILI.
Collapse
Affiliation(s)
- Si-Wei Rao
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, PR China
| | - Cheng-Jun Liu
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China
| | - Dong Liang
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China
| | - Yuan-Yuan Duan
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China
| | - Zi-Hao Chen
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China
| | - Jin-Jin Li
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China
| | - Han-Qing Pang
- Institute of Translational Medicine, Medical College, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, PR China
| | - Feng-Xiang Zhang
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| | - Wei Shi
- 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), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin, 541004, PR China.
| |
Collapse
|
4
|
Meng D, Dong Y, Shang Q, Sun Z. Anti-tumor effect and hepatotoxicity mechanisms of psoralen. Front Pharmacol 2024; 15:1442700. [PMID: 39161897 PMCID: PMC11331265 DOI: 10.3389/fphar.2024.1442700] [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: 06/02/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
In recent years, natural products have gradually become an important source for new drug development due to their advantages of multi-components, multi-targets, and good safety profiles. Psoralen, a furanocoumarin compound extracted from the traditional Chinese medicine psoralea corylifolia, is widely distributed among various plants. It has attracted widespread attention in the research community due to its pharmacological activities, including antitumor, anti-inflammatory, antioxidant, and neuroprotective effects. Studies have shown that psoralen has broad spectrum anti-tumor activities, offering resistance to malignant tumors such as breast cancer, liver cancer, glioma, and osteosarcoma, making it a natural, novel potential antitumor drug. Psoralen mainly exerts its antitumor effects by inhibiting tumor cell proliferation, inducing apoptosis, inhibiting tumor cell migration, and reversing multidrug resistance, presenting a wide application prospect in the field of antitumor therapy. With the deepening research on psoralea corylifolia, its safety has attracted attention, and reports on the hepatotoxicity of psoralen have gradually increased. Therefore, this article reviews recent studies on the mechanism of antitumor effects of psoralen and focuses on the molecular mechanisms of its hepatotoxicity, providing insights for the clinical development of low-toxicity, high-efficiency antitumor drugs and the safety of clinical medication.
Collapse
Affiliation(s)
- Dandan Meng
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yanling Dong
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qingxin Shang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Ziyuan Sun
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
5
|
Yang H, Yao Z, Yang K, Wang C, Li M, Zhang Y, Yan J, Lv R, Wang Y, Huang A, Zhang D, Li W, Wu Y, Miao Z. Synthesis and Antibacterial Evaluation of Novel Psoralen Derivatives against Methicillin-Resistant Staphylococcus aureus (MRSA). Chem Biodivers 2024; 21:e202302048. [PMID: 38263380 DOI: 10.1002/cbdv.202302048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/25/2024]
Abstract
Today, the bacterial infections caused by multidrug-resistant pathogens seriously threaten human health. Thereby, there is an urgent need to discover antibacterial drugs with novel mechanism. Here, novel psoralen derivatives had been designed and synthesized by a scaffold hopping strategy. Among these targeted twenty-five compounds, compound ZM631 showed the best antibacterial activity against methicillin-resistant S. aureus (MRSA) with the low MIC of 1 μg/mL which is 2-fold more active than that of the positive drug gepotidacin. Molecular docking study revealed that compound ZM631 fitted well in the active pockets of bacterial S. aureus DNA gyrase and formed a key hydrogen bond binding with the residue ASP-1083. These findings demonstrated that the psoralen scaffold could serve as an antibacterial lead compound for further drug development against multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Hang Yang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Zheng Yao
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Keli Yang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Chuanhao Wang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, 200 Xiaolingwei Road, Nanjing, 210094, the People's Republic of China
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Mochenxuan Li
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Yanming Zhang
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Jianyu Yan
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Rongxue Lv
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Yongchuang Wang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| | - Anhua Huang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Daozuan Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Wei Li
- School of Pharmacy, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, the People's Republic of China
| | - Yuelin Wu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai, 201418, The People's Republic of China
| | - Zhenyuan Miao
- School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai, 200433, the People's Republic of China
| |
Collapse
|
6
|
Su L, Song G, Zhou T, Tian H, Xin H, Zou X, Xu Y, Jin X, Gui S, Lu X. Colon-targeted oral nanoliposomes loaded with psoralen alleviate DSS-induced ulcerative colitis. Biomater Sci 2024; 12:3212-3228. [PMID: 38757193 DOI: 10.1039/d4bm00321g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Oral administration, while convenient, but complex often faces challenges due to the complexity of the digestive environment. In this study, we developed a nanoliposome (NLP) encapsulating psoralen (P) and coated it with chitosan (CH) and pectin (PT) to formulate PT/CH-P-NLPs. PT/CH-P-NLPs exhibit good biocompatibility, superior to liposomes loaded with psoralen and free psoralen alone. After oral administration, PT/CH-P-NLPs remain stable in the stomach and small intestine, followed by a burst release of psoralen after reaching the slightly alkaline and gut microbiota-rich colon segment. In the DSS-induced ulcerative colitis of mice, PT/CH-P-NLPs showed significant effects on reducing inflammation, mitigating oxidative stress, protecting the integrity of the colon mucosal barrier, and modulating the gut microbiota. In conclusion, the designed nanoliposomes demonstrated the effective application of psoralen in treating ulcerative colitis.
Collapse
Affiliation(s)
- Liqian Su
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
| | - Gaoqing Song
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People's Republic of China
| | - Tao Zhou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People's Republic of China
| | - Hongmei Tian
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People's Republic of China
| | - Hui Xin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People's Republic of China
| | - Xuan Zou
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
| | - Yinghua Xu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotechnology Products, National Institutes for Food and Drug Control, Beijing 102629, People's Republic of China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
| | - Shuiqing Gui
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, People's Republic of China
| | - Xuemei Lu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of BasicMedical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, People's Republic of China.
- Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, People's Republic of China
| |
Collapse
|
7
|
Chen LM, Qian ST, Li ZQ, He MF, Li HJ. Psoralen and Isopsoralen, Two Estrogen -Like Natural Products from Psoraleae Fructus, Induced Cholestasis via Activation of ERK1/2. Chem Res Toxicol 2024; 37:804-813. [PMID: 38646980 DOI: 10.1021/acs.chemrestox.4c00054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
With the increasing use of oral contraceptives and estrogen replacement therapy, the incidence of estrogen-induced cholestasis (EC) has tended to rise. Psoralen (P) and isopsoralen (IP) are the major bioactive components in Psoraleae Fructus, and their estrogen-like activities have already been recognized. Recent studies have also reported that ERK1/2 plays a critical role in EC in mice. This study aimed to investigate whether P and IP induce EC and reveal specific mechanisms. It was found that P and IP increased the expression of esr1, cyp19a1b and the levels of E2 and VTG at 80 μM in zebrafish larvae. Exemestane (Exe), an aromatase antagonist, blocked estrogen-like activities of P and IP. At the same time, P and IP induced cholestatic hepatotoxicity in zebrafish larvae with increasing liver fluorescence areas and bile flow inhibition rates. Further mechanistic analysis revealed that P and IP significantly decreased the expression of bile acids (BAs) synthesis genes cyp7a1 and cyp8b1, BAs transport genes abcb11b and slc10a1, and BAs receptor genes nr1h4 and nr0b2a. In addition, P and IP caused EC by increasing the level of phosphorylation of ERK1/2. The ERK1/2 antagonists GDC0994 and Exe both showed significant rescue effects in terms of cholestatic liver injury. In conclusion, we comprehensively studied the specific mechanisms of P- and IP-induced EC and speculated that ERK1/2 may represent an important therapeutic target for EC induced by phytoestrogens.
Collapse
Affiliation(s)
- Liang-Min Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Si-Tong Qian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Zhuo-Qing Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ming-Fang He
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing 211816, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
8
|
Bishnoi A, Parsad D. Phototherapy for vitiligo: A narrative review on the clinical and molecular aspects, and recent literature. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2024; 40:e12968. [PMID: 38632705 DOI: 10.1111/phpp.12968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Vitiligo is characterized by depigmented patches resulting from loss of melanocytes. Phototherapy has emerged as a prominent treatment option for vitiligo, utilizing various light modalities to induce disease stability and repigmentation. AIMS AND METHODS This narrative review aims to explore the clinical applications and molecular mechanisms of phototherapy in vitiligo. RESULTS AND DISCUSSION The review evaluates existing literature on phototherapy for vitiligo, analyzing studies on hospital-based and home-based phototherapy, as well as outcomes related to stabilization and repigmentation. Narrowband ultra-violet B, that is, NBUVB remains the most commonly employed, studied and effective phototherapy modality for vitiligo. Special attention is given to assessing different types of lamps, dosimetry, published guidelines, and the utilization of targeted phototherapy modalities. Additionally, the integration of phototherapy with other treatment modalities, including its use as a depigmenting therapy in generalized/universal vitiligo, is discussed. Screening for anti-nuclear antibodies and tailoring approaches for non-photo-adapters are also examined. CONCLUSION In conclusion, this review provides a comprehensive overview of phototherapy for vitiligo treatment. It underscores the evolving landscape of phototherapy and offers insights into optimizing therapeutic outcomes and addressing the challenges ahead. By integrating clinical evidence with molecular understanding, phototherapy emerges as a valuable therapeutic option for managing vitiligo, with potential for further advancements in the field.
Collapse
Affiliation(s)
- Anuradha Bishnoi
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Davinder Parsad
- Department of Dermatology, Venereology and Leprology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
9
|
Zhang ML, Li WX, Wang XY, Zhang H, Wu YL, Yang LQ, Chen XF, Zhang SQ, Chen YL, Feng KR, Tang JF. A gene expression profile-based approach to screen the occurrence and predisposed host characteristics of drug-induced liver injury: a case study of Psoralea corylifolia Linn. Front Chem 2023; 11:1259569. [PMID: 37867998 PMCID: PMC10588485 DOI: 10.3389/fchem.2023.1259569] [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: 07/16/2023] [Accepted: 09/12/2023] [Indexed: 10/24/2023] Open
Abstract
Drug-induced liver injury (DILI) is one of the most common causes of a drug being withdrawn, and identifying the culprit drugs and the host factors at risk of causing DILI has become a current challenge. Recent studies have found that immune status plays a considerable role in the development of DILI. In this study, DILI-related differentially expressed genes mediated by immunoinflammatory cytokines were obtained from the Gene Expression Omnibus (GEO) database to predict the occurrence of DILI (named the DILI predictive gene set, DILI_PGS), and the predictability of the DILI_PGS was verified using the Connectivity Map (CMap) and LiverTox platforms. The results obtained DILI_PGS from the GEO database could predict 81.25% of liver injury drugs. In addition, the Coexpedia platform was used to predict the DILI_PGS-related characteristics of common host diseases and found that the DILI_PGS mainly involved immune-related diseases and tumor-related diseases. Then, animal models of immune stress (IS) and immunosuppressive (IP) were selected to simulate the immune status of the above diseases. Meanwhile, psoralen, a main component derived from Psoralea corylifolia Linn. with definite hepatotoxicity, was selected as an experimental drug with highly similar molecular fingerprints to three idiosyncratic hepatotoxic drugs (nefazodone, trovafloxacin, and nimesulide) from the same DILI_PGS dataset. The animal experiment results found a single administration of psoralen could significantly induce liver injury in IS mice, while there was no obvious liver function change in IP mice by repeatedly administering the same dose of psoralen, and the potential mechanism of psoralen-induced liver injury in IS mice may be related to regulating the expression of the TNF-related pathway. In conclusion, this study constructed the DILI_PGS with high accuracy to predict the occurrence of DILI and preliminarily identified the characteristics of host factors inducing DILI.
Collapse
Affiliation(s)
- Ming-Liang Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Wei-Xia Li
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Yan Wang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Hui Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Ya-Li Wu
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Liu-Qing Yang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Xiao-Fei Chen
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Shu-Qi Zhang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Yu-Long Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Ke-Ran Feng
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
| | - Jin-Fa Tang
- The Department of Pharmacy, First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center for Clinical Application, Evaluation and Transformation of Traditional Chinese Medicine, Zhengzhou, China
- Henan Provincial Key Laboratory for Clinical Pharmacy of Traditional Chinese Medicine, Zhengzhou, China
- Henan Province Engineering Research Center of Safety Evaluation and Risk Management of Traditional Chinese Medicine, Zhengzhou, China
- Henan University of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
10
|
Lu Y, Zhang M, Zhang J, Jiang M, Bai G. Psoralen prevents the inactivation of estradiol and treats osteoporosis via covalently targeting HSD17B2. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116426. [PMID: 36997132 DOI: 10.1016/j.jep.2023.116426] [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: 11/04/2022] [Revised: 02/22/2023] [Accepted: 03/21/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Psoralea corylifolia L. seeds (P. corylifolia), popularly known as Buguzhi in traditional Chinese medicine, are often used to treat osteoporosis in China. Psoralen (Pso) is the key anti-osteoporosis constituent in P. corylifolia, however, its targets and mechanism of action are still unclear. AIM OF THE STUDY The purpose of this study was to explore the interaction between Pso and 17-β hydroxysteroid dehydrogenase type 2 (HSD17B2), an estrogen synthesis-related protein that inhibits the inactivation of estradiol (E2) to treat osteoporosis. MATERIALS AND METHODS Tissue distribution of Pso was analyzed by in-gel imaging after oral administration of an alkynyl-modified Pso probe (aPso) in mice. The target of Pso in the liver was identified and analyzed using chemical proteomics. Co-localization and cellular thermal shift assays (CETSA) were used to verify the key action targets. To detect the key pharmacophore of Pso, the interaction of Pso and its structural analogs with HSD17B2 was investigated by CETSA, HSD17B2 activity assay, and in-gel imaging determination. Target competitive test, virtual docking, mutated HSD17B2 activity, and CETSA assay were used to identify the binding site of Pso with HSD17B2. A mouse model of osteoporosis was established by ovariectomies, and the efficacy of Pso in vivo was confirmed by micro-CT, H&E staining, HSD17B2 activity, and bone-related biochemical assays. RESULTS Pso regulated estrogen metabolism by targeting HSD17B2 in the liver, with the α, β-unsaturated ester in Pso being the key pharmacophore. Pso significantly suppressed HSD17B2 activity by irreversibly binding to Lys236 of HSD17B2 and preventing NAD+ from entering the binding pocket. In vivo studies in ovariectomized mice revealed that Pso could inhibit HSD17B2 activity, prevent the inactivation of E2, increase levels of endogenous estrogen, improve bone metabolism-related indices, and play a role in anti-osteoporosis. CONCLUSIONS Pso covalently binds to Lys236 of HSD17B2 in hepatocytes to prevent the inactivation of E2, thereby aiding in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Yujie Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Man Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Jin Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China.
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 300353, China.
| |
Collapse
|
11
|
Di Stasi LC. Natural Coumarin Derivatives Activating Nrf2 Signaling Pathway as Lead Compounds for the Design and Synthesis of Intestinal Anti-Inflammatory Drugs. Pharmaceuticals (Basel) 2023; 16:ph16040511. [PMID: 37111267 PMCID: PMC10142712 DOI: 10.3390/ph16040511] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/24/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Nrf2 (nuclear factor erythroid 2-related factor 2) is a transcription factor related to stress response and cellular homeostasis that plays a key role in maintaining the redox system. The imbalance of the redox system is a triggering factor for the initiation and progression of non-communicable diseases (NCDs), including Inflammatory Bowel Disease (IBD). Nrf2 and its inhibitor Kelch-like ECH-associated protein 1 (Keap1) are the main regulators of oxidative stress and their activation has been recognized as a promising strategy for the treatment or prevention of several acute and chronic diseases. Moreover, activation of Nrf2/keap signaling pathway promotes inhibition of NF-κB, a transcriptional factor related to pro-inflammatory cytokines expression, synchronically promoting an anti-inflammatory response. Several natural coumarins have been reported as potent antioxidant and intestinal anti-inflammatory compounds, acting by different mechanisms, mainly as a modulator of Nrf2/keap signaling pathway. Based on in vivo and in vitro studies, this review focuses on the natural coumarins obtained from both plant products and fermentative processes of food plants by gut microbiota, which activate Nrf2/keap signaling pathway and produce intestinal anti-inflammatory activity. Although gut metabolites urolithin A and urolithin B as well as other plant-derived coumarins display intestinal anti-inflammatory activity modulating Nrf2 signaling pathway, in vitro and in vivo studies are necessary for better pharmacological characterization and evaluation of their potential as lead compounds. Esculetin, 4-methylesculetin, daphnetin, osthole, and imperatorin are the most promising coumarin derivatives as lead compounds for the design and synthesis of Nrf2 activators with intestinal anti-inflammatory activity. However, further structure-activity relationships studies with coumarin derivatives in experimental models of intestinal inflammation and subsequent clinical trials in health and disease volunteers are essential to determine the efficacy and safety in IBD patients.
Collapse
Affiliation(s)
- Luiz C Di Stasi
- Laboratory of Phytomedicines, Pharmacology and Biotechnology (PhytoPharmaTech), Department of Biophysics and Pharmacology, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| |
Collapse
|
12
|
Ni B, Wang W, Liu M, Xu Y, Zhao J. Paris saponin Ⅰ induce toxicity in zebrafish by up-regulation of p53 pathway and down-regulation of wnt pathway. Toxicon 2023; 228:107094. [PMID: 37003302 DOI: 10.1016/j.toxicon.2023.107094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 03/06/2023] [Accepted: 03/22/2023] [Indexed: 04/03/2023]
Abstract
Paris saponin I, II, and VII are three important components in Paris polyphylla, which have been widely studied as tumor cytotoxic drugs, but their safety in vivo has not been reported. Therefore, this study evaluated the safety of these three drugs based on the zebrafish model. Firstly, the lethality curves and half lethal rates of the three saponins were determined and the results showed the values of LC50 of Paris saponin I, II, and VII were 122.2, 210.7, 566.2 ng/ml, respectively. And then our data revealed that Paris saponin I, II and VII had definite hepatotoxicity, as shown by their significant reduction in the liver area and fluorescence intensity of zebrafish. Besides, Paris saponin Ⅰ affected the heart rate of zebrafish obviously, suggesting its cardiovascular toxicity. Afterwards, we found Paris saponin Ⅰ and Ⅶ reduced the area and fluorescence intensity of kidney in zebrafish, and had mild nephrotoxicity. And when treated with Paris saponin I, the pathological section of liver tissue in zebrafish showed vacuoles, severe necrosis of hepatocytes, and then the apoptosis of hepatocytes could be observed by TUNEL staining. Eventually, we found that the genes expression of p53, Bax and β-catenin changed significantly in the administration group of Paris saponin I. In general, our study proved Paris saponin Ⅰ was the most toxic of the three saponins, and the most definite toxic target sites were liver and cardiovascular. And it was further inferred that the hepatotoxicity of Paris saponin Ⅰ may be related to the regulation of p53 pathway and Wnt pathway. These results above showed the toxicity of the three saponins in zebrafish, suggesting their safety should be paid more attention in the future.
Collapse
Affiliation(s)
- Boran Ni
- Department of Endocrinology, Guang' Anmen Hospital of China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Wenping Wang
- Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Manting Liu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yuchen Xu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jinxi Zhao
- Section II of Endocrinology & Nephropathy, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
13
|
Liu J, Yang G, Zhang H. Glyphosate-triggered hepatocyte ferroptosis via suppressing Nrf2/GSH/GPX4 axis exacerbates hepatotoxicity. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 862:160839. [PMID: 36521597 DOI: 10.1016/j.scitotenv.2022.160839] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 11/16/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
Glyphosate (GLY) exposure has been reported to damage organs in animals, in particular the liver, due to increased reactive oxygen species (ROS). Ferroptosis is defined as a new type of cell death that is characterized by the increase of ROS. The purpose of this study was to elucidate whether the relationship between ferroptosis and GLY-induced hepatotoxicity is of significance to enlarge the knowledge about GLY toxicity and consequences for human and animal health. To this end, in this study, we investigated the role of ferroptosis in GLY-induced hepatotoxicity both in vivo and in vitro. The results showed that GLY exposure triggered ferroptosis in L02 cells, but pretreatment with ferroptosis inhibitor ferrostatin (Fer-1) rescued ferroptosis-induced injury, thereby indicating that ferroptosis plays a key role in GLY-induced hepatotoxicity. Moreover, N-acetylcysteine, a glutathione (GSH) synthesis precursor, reversed GLY-triggered ferroptosis damage, thus indicating that GSH exhaustion may be a prerequisite for GLY-triggered hepatotoxicity. Mechanistically, GLY inhibited GSH biosynthesis via blocking the phosphorylation and nuclear translocation of Nrf2, which resulted in GSH depletion-induced hepatocyte ferroptosis. In a mouse model, GLY exposure triggered ferroptosis-induced liver damage, which can be rescued by pretreatment with Fer-1 or tBHQ (a specific agonist of Nrf2). To our knowledge, this is the first study to reveal that GLY-triggered hepatocyte ferroptosis via suppressing Nrf2/GSH/GPX4 axis exacerbates hepatotoxicity, which expands our knowledge about GLY toxicity in animal and human health.
Collapse
Affiliation(s)
- Jingbo Liu
- College of Biological and Brewing Engineering, Taishan University, No. 525 Dongyue Street, 271000 Tai'an City, Shandong Province, China.
| | - Guangcheng Yang
- College of Biological and Brewing Engineering, Taishan University, No. 525 Dongyue Street, 271000 Tai'an City, Shandong Province, China
| | - Hongna Zhang
- College of Bioscience and Engineering, Hebei University of Economics and Business, No. 47 Xuefu Road, 050061 Shijiazhuang City, Hebei Province, China.
| |
Collapse
|