1
|
Ran Q, Huang M, Wang L, Li Y, Wu W, Liu X, Chen J, Yang M, Han K, Guo X. Integrated bioinformatics and multi-omics to investigate the mechanism of Rhododendron molle Flos-induced hepatotoxicity. JOURNAL OF ETHNOPHARMACOLOGY 2024:119308. [PMID: 39746411 DOI: 10.1016/j.jep.2024.119308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Drug-induced liver injury (DILI) is an important and common adverse drug event. Rhododendron molle Flos (RMF), as one of toxic Traditional Chinese medicines (TCMs), holds a prominent position in clinical practice for treating rheumatoid arthritis. However, the toxicity of RMF limits its safe. Most of the concerns are about its rapid neurotoxicity and cardiotoxicity, with less attention paid to its hepatotoxicity, and the mechanism of which is still unclear. AIM OF THE STUDY To reveal the mechanism of RMF-induced hepatotoxicity by bioinformatics and multi-omics. MATERIALS AND METHODS Rats were intragastric administered RMF at doses of 0.8 g/kg, 0.4 g/kg, and 0.2 g/kg once daily for 2 weeks. Initially, hepatotoxicity was then evaluated using liver function enzymes, antioxidant enzymes, and histopathology. Subsequently, network toxicology, transcriptomics, and metabolomics were used to identify the genes and metabolites. In addition, molecular docking and Western blot were employed to verify toxic components and key targets. RESULTS RMF caused abnormal levels of ALT, γ-GT, TBIL, and TBA in the serum of rats, as well as abnormal levels of MDA, GSH-Px, and SOD in the liver, leading to inflammatory infiltration of liver cells, with a dose-dependent manner. RMF disordered the steroid hormone biosynthesis, pyruvate metabolism, fatty acid biosynthesis, and arachidonic acid metabolism. Six key targets were identified- UGT1A6, CYP2E1, ACOT1, ACSL5, CTH, and PKLR, along with their corresponding metabolites, namely 17β-estradiol, estriol, arachidonic acid, octadecanoic acid, and pyruvic acid. The hepatotoxicity could be attributed to five diterpenoid components, including grayanotoxin-III, rhodojaponin (RJ)-I, RJ-II, RJ-III, and RJ-V. CONCLUSIONS This study comprehensively identified the toxic components, upstream targets, and downstream metabolites of RMF-induced liver toxicity, providing a basis for evaluating and monitoring liver function in patients during clinical application.
Collapse
Affiliation(s)
- Qiang Ran
- Department of Orthopedics, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Mengjun Huang
- National-Local Joint Engineering Research Center for Innovative Targeted Drugs, Chongqing University of Arts and Sciences, Chongqing, 402160, China.
| | - Lijuan Wang
- Department of Pathology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Yanyan Li
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Wenhui Wu
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Xia Liu
- Department of Pharmacy, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Juan Chen
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Min Yang
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Keqing Han
- Department of Pharmacy, Shaanxi Provincial Tuberculosis Prevention and Control Hospital, Xi'an, Shanxi, 710100, China
| | - Xiaohong Guo
- Department of Preparation Center, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China
| |
Collapse
|
2
|
Qu T, Sun Q, Tan B, Wei H, Qiu X, Xu X, Gao H, Zhang S. Integration of network toxicology and transcriptomics reveals the novel neurotoxic mechanisms of 2, 2', 4, 4'-tetrabromodiphenyl ether. JOURNAL OF HAZARDOUS MATERIALS 2024; 486:136999. [PMID: 39740552 DOI: 10.1016/j.jhazmat.2024.136999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/30/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025]
Abstract
The brominated flame retardant 2, 2', 4, 4'-tetrabromodiphenyl ether (PBDE-47) is known as a developmental neurotoxicant, yet the underlying mechanisms remain unclear. This study aims to explore its neurotoxic mechanisms by integrating network toxicology with transcriptomics based on human neural precursor cells (hNPCs) and neuron-like PC12 cells. Network toxicology revealed that PBDE-47 crosses the blood-brain barrier more effectively than heavier PBDE congeners, and is associated with disruptions in 159 biological pathways, including cytosolic DNA-sensing pathway, ferroptosis, cellular senescence, and chemokine signaling pathway. Additionally, transcriptomic analyses of hNPCs and PC12 cells exposed to PBDE-47 uncovered substantial gene expression changes, with 855 and 702 genes up- and down-regulated in hNPCs, and 2844 and 2711 genes in PC12 cells, respectively. These differentially expressed genes were primarily implicated in crucial processes like neuroactive ligand-receptor interaction, nucleocytoplasmic transport, ferroptosis, p53 signaling, and cell cycle regulation. Integration of the results identified novel mechanisms of PBDE-47 neurotoxicity, such as neuroinflammation and cellular senescence, alongside established mechanisms like ferroptosis, apoptosis and cell cycle arrest. Overall, these findings provide critical insights into the mechanisms of PBDE-47 neurotoxicity, highlighting the integration of network toxicology and transcriptomics as a novel study approach to explore the modes of action of toxicants.
Collapse
Affiliation(s)
- Tengjiao Qu
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Sun
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Environmental health effects and risk assessment Key Laboratory of Luzhou, School of Public Health, Southwest Medical University, Luzhou, Sichuan, China
| | - Bo Tan
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hao Wei
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaoxuan Qiu
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojie Xu
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Gao
- Department of Clinical Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei, China.
| | - Shun Zhang
- Department of Occupational and Environmental Health, MOE Key Laboratory of Environment and Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Xiong R, Lei J, Wang L, Zhang S, Liu H, Wang H, Liu T, Lai X. Efficient analysis of adverse drug events and toxicological mechanisms of newly marketed drugs by integrating pharmacovigilance and network toxicology: selumetinib as an example. Front Pharmacol 2024; 15:1432759. [PMID: 39193326 PMCID: PMC11347787 DOI: 10.3389/fphar.2024.1432759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Objective To integrate pharmacovigilance and network toxicology methods to explore the potential adverse drug events (ADEs) and toxic mechanisms of selumetinib, and to provide a reference for quickly understanding the safety and toxicological mechanisms of newly marketed drugs. Methods Taking selumetinib as an example, this study integrated pharmacovigilance methods based on real-world data and network toxicology methods to analyze its ADE and its potential toxicological mechanism. First, the ADE reports of selumetinib were extracted from the US Food and Drug Administration (FDA) adverse event reporting system (FAERS), and the ADE signals were detected by reporting odds ratio (ROR) and UK medicines and healthcare products regulatory agency (MHRA) methods. The ADE signals were classified and described according to the preferred terms (PTs) and system organ class (SOC) derived from the Medical Dictionary for Regulatory Activities (MedDRA). The network toxicology method was used to analyze the toxicological mechanism of the interested SOCs. The specific steps included predicting the potential targets of selumetinib using TOXRIC, STITCH, ChEMBL, CTD, SwissTargetPreditcion, and Super-PRED databases, collecting the targets of SOC using GeneCards database, conducting protein-protein interaction (PPI) analysis through STRING database, conducting gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) analysis through DAVID database, and testing the molecular affinity using AutoDock software. Results A total of 1388 ADE reports related to selumetinib were extracted, and 53 positive signals were detected by ROR and MHRA methods, of which 20 signals were not mentioned in the package insert, including ingrowing nail, hyperphosphatemia, cardiac valve disease, hematuria, neutropenia, etc. Analysis of the toxicological mechanism of six SOCs involved in positive ADE signals revealed that the key targets included EGFR, STAT3, AKT1, IL6, BCL2, etc., and the key pathways included PI3K/Akt pathway, apoptosis, ErbB signaling pathway, and EGFR tyrosine kinase inhibitor resistance, etc. Molecular docking assays showed spontaneous binding of selumetinib to key targets in these pathways. Conclusion The pharmacovigilance analysis identified some new potential ADEs of selumetinib, and the network toxicology analysis showed that the toxic effects of selumetinib may be related to PI3K/Akt pathway, apoptosis, ErbB signaling pathway, EGFR tyrosine kinase inhibitor resistance and other pathways.
Collapse
Affiliation(s)
- Rui Xiong
- Department of Pharmacy, Jiangbei Campus of The First Affiliated Hospital of Army Medical University (The 958th Hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Jing Lei
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lu Wang
- Department of Pharmacy, Jiangbei Campus of The First Affiliated Hospital of Army Medical University (The 958th Hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Shipeng Zhang
- Department of Pharmacy, Jiangbei Campus of The First Affiliated Hospital of Army Medical University (The 958th Hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Hengxu Liu
- Department of Pharmacy, Jiangbei Campus of The First Affiliated Hospital of Army Medical University (The 958th Hospital of Chinese People’s Liberation Army), Chongqing, China
| | - Hongping Wang
- Department of Pharmacy, The First Affiliated Hospital of Army Medical University, Chongqing, China
| | - Tao Liu
- Department of Infectious Diseases, Navy No.971 Hospital, Qingdao, Shandong, China
| | - Xiaodan Lai
- Department of Pharmacy, Jiangbei Campus of The First Affiliated Hospital of Army Medical University (The 958th Hospital of Chinese People’s Liberation Army), Chongqing, China
| |
Collapse
|
4
|
Lin Z, Basili D, Chou WC. Preface to the special issue of Food and Chemical Toxicology on "New approach methodologies and machine learning in food safety and chemical risk assessment: Development of reproducible, open-source, and user-friendly tools for exposure, toxicokinetic, and toxicity assessments in the 21st century". Food Chem Toxicol 2024; 190:114809. [PMID: 38857761 DOI: 10.1016/j.fct.2024.114809] [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: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
This Special Issue contains articles on applications of various new approach methodologies (NAMs) in the field of toxicology and risk assessment. These NAMs include in vitro high-throughput screening, quantitative structure-activity relationship (QSAR) modeling, physiologically based pharmacokinetic (PBPK) modeling, network toxicology analysis, molecular docking simulation, omics, machine learning, deep learning, and "template-and-anchor" multiscale computational modeling. These in vitro and in silico approaches complement each other and can be integrated together to support different applications of toxicology, including food safety assessment, dietary exposure assessment, chemical toxicity potency screening and ranking, chemical toxicity prediction, chemical toxicokinetic simulation, and to investigate the potential mechanisms of toxicities, as introduced further in selected articles in this Special Issue.
Collapse
Affiliation(s)
- Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, FL 32608, United States; Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32608, United States.
| | - Danilo Basili
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Wei-Chun Chou
- Department of Environmental Sciences, College of Natural and Agricultural Sciences, University of California, Riverside, CA 92521, United States
| |
Collapse
|
5
|
Song L, Wang D, Zhai Y, Zhang X, Zhang Y, Yu Y, Sun L, Zhou K. Aqueous extract of Epimedium sagittatum (Sieb. et Zucc.) Maxim. induces liver injury in mice via pyroptosis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 329:118164. [PMID: 38593963 DOI: 10.1016/j.jep.2024.118164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 04/04/2024] [Accepted: 04/06/2024] [Indexed: 04/11/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Epimedium sagittatum (Sieb. et Zucc.) Maxim. has been used traditionally in Asia. It can dispel wind and cold, tonify the kidney, and strengthen bones and tendons. However, adverse effects of E. sagittatum have been reported, and the underlying mechanisms remain unclear. AIM OF THE STUDY This study aimed to investigate liver injury caused by an aqueous extract of E. sagittatum in Institute of Cancer Research (ICR) mice and explore its potential mechanisms. MATERIALS AND METHODS Dried E. sagittatum leaves were decocted in water to prepare aqueous extracts for ultra-high performance liquid chromatography analysis. Mice were administered an aqueous extract of E. sagittatum equivalent to either 3 g raw E. sagittatum/kg or 10 g raw E. sagittatum/kg once daily via intragastric injection for three months. The liver weights and levels of the serum biochemical parameters including alanine transaminase (ALT), aspartate aminotransferase (AST), lactate dehydrogenase (LDH), total bilirubin (TBIL), and alkaline phosphatase were measured. Hematoxylin-eosin staining was performed for histopathology. Apoptosis was detected using the TUNEL apoptosis assay kit. IL-1β was detected using ELISA kits. Proteomics was used to identify the differentially expressed proteins. Western blot analysis was performed to determine the levels of proteins significantly affected by the aqueous extract of E. sagittatum. RESULTS E. sagittatum treatment increased the liver weights and liver coefficients, and ALT and AST levels significantly increased (p < 0.05). A high dose of E. sagittatum significantly increased LDH and TBIL levels (p < 0.05). Ruptured cell membranes and multiple sites of inflammatory cell infiltration were also observed. No evidence of apoptosis was observed. IL-1β levels were significantly increased (p < 0.05). The expressions of PIK3R1, p-MAP2K4, p-Jun N-terminal kinase (JNK)/JNK, p-c-Jun, VDAC2, Bax, and CYC were upregulated, whereas that of Bcl-2 was inhibited by E. sagittatum. The expression of cleaved caspase-1 was significantly increased; however, its effects on GSDMD and GSDMD-N were significantly decreased. The expression levels of cleaved caspase-3 and its effector proteins GSDME and GSDME-N significantly increased. CONCLUSIONS Our results suggest that the aqueous extract of E. sagittatum induces liver injury in ICR mice after three months of intragastric injection via inflammatory pyroptosis.
Collapse
Affiliation(s)
- Lei Song
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin, 301617, China
| | - Dongyu Wang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuxia Zhai
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaoying Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yue Zhang
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin, 301617, China
| | - Yingli Yu
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin, 301617, China
| | - Likang Sun
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Kun Zhou
- Center of Drug Safety Evaluation, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-based Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
6
|
Zhao Z, Du JF, Wang QL, Qiu FN, Li P, Jiang Y, Li HJ. Natural Product Baohuoside I Impairs the Stability and Membrane Location of MRP2 Reciprocally Regulated by SUMOylation and Ubiquitination in Hepatocytes. Chem Res Toxicol 2024; 37:57-71. [PMID: 38177062 DOI: 10.1021/acs.chemrestox.3c00298] [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: 01/06/2024]
Abstract
Epimedii Folium (EF) is a botanical dietary supplement to benefit immunity. Baohuoside I (BI), a prenylated flavonoid derived from EF, has exhibited the cholestatic risk before. Here, the mechanism of BI on the stability and membrane localization of liver MRP2, a bile acid exporter in the canalicular membrane of hepatocytes, was investigated. The fluorescent substrate of MRP2, CMFDA was accumulated in sandwich-cultured primary mouse hepatocytes (SCH) under BI stimulation, followed by reduced membrane MRP2 expression. BI triggered MRP2 endocytosis associated with oxidative stress via inhibition of the NRF2 signaling pathway. Meanwhile, BI promoted the degradation of MRP2 by reducing its SUMOylation and enhancing its ubiquitination level. Co-IP and fluorescence colocalization experiments all proved that MRP2 was a substrate protein for SUMOylation for SUMO proteins. CHX assays showed that SUMO1 prolonged the half-life of MRP2 and further increased its membrane expression, which could be reversed by UBC9 knockdown. Correspondingly, MRP2 accumulated in the cytoplasm by GP78 knockdown or under MG132 treatment. Additionally, the SUMOylation sites of MRP2 were predicted by the algorithm, and a conversion of lysines to arginines at positions 940 and 953 of human MRP2 caused its decreased stability and membrane location. K940 was further identified as the essential ubiquitination site for MRP2 by an in vitro ubiquitination assay. Moreover, the decreased ubiquitination of MRP2 enhanced the SUMOylation MRP2 and vice versa, and the crosstalk of these two modifiers could be disrupted by BI. Collectively, our findings indicated the process of MRP2 turnover from the membrane to cytoplasm at the post-translational level and further elucidated the novel toxicological mechanism of BI.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Jin-Fa Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Qiao-Lei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Fang-Ning Qiu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing 210037, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing 210009, China
| |
Collapse
|
7
|
Sun J, Zhang K, Yin Y, Qi Y, Li S, Sun H, Luo M, Sun Y, Yu Z, Yang J, Wu J, Chen L, Xu W, Dong L. Arecoline-Induced Hepatotoxicity in Rats: Screening of Abnormal Metabolic Markers and Potential Mechanisms. TOXICS 2023; 11:984. [PMID: 38133385 PMCID: PMC10748282 DOI: 10.3390/toxics11120984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Arecoline is a pyridine alkaloid derived from areca nut in the Arecaceae family. It has extensive medicinal activity, such as analgesic, anti-inflammatory, and anti-allergic. However, the toxicity of Arecoline limits its application. Most current studies on its toxicity mainly focus on immunotoxicity, carcinogenesis, and cancer promotion. However, there are few systematic studies on its hepatotoxicity and mechanisms. Therefore, this research explored the mechanism of hepatotoxicity induced by Arecoline in rats and analyzed endogenous metabolite changes in rat plasma by combining network toxicology with metabolomics. The differential metabolites after Arecoline exposure, such as D-Lysine, N4-Acetylaminobutanal, and L-Arginine, were obtained by metabolomics study, and these differential metabolites were involved in the regulation of lipid metabolism, amino acid metabolism, and vitamin metabolism. Based on the strategy of network toxicology, Arecoline can affect the HIF-1 signaling pathway, MAPK signaling pathway, PI3K-Akt signaling pathway, and other concerning pathways by regulating critical targets, such as ALB, CASP3, EGFR, and MMP9. Integration of metabolomics and network toxicology results were further analyzed, and it was concluded that Arecoline may induce hepatotoxicity by mediating oxidative stress, inflammatory response, energy and lipid metabolism, and cell apoptosis.
Collapse
Affiliation(s)
- Jing Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (M.L.); (J.Y.); (J.W.)
| | - Kai Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Yihui Yin
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Yunpeng Qi
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Siyuan Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Haonan Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Min Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (M.L.); (J.Y.); (J.W.)
| | - Yixuan Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Zhiying Yu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Jie Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (M.L.); (J.Y.); (J.W.)
| | - Jingjing Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China; (M.L.); (J.Y.); (J.W.)
| | - Lijuan Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Wenjuan Xu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| | - Ling Dong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (J.S.); (K.Z.); (Y.Y.); (Y.Q.); (S.L.); (H.S.); (Y.S.); (Z.Y.); (L.C.)
| |
Collapse
|