1
|
Yin J, Song Z, Zhang L, Cong J. Methylophiopogonanone A alleviates diabetic cardiomyopathy via inhibiting JNK1 signaling. Cell Signal 2025; 131:111762. [PMID: 40139620 DOI: 10.1016/j.cellsig.2025.111762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/13/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DCM) is a common complication of type 2 diabetes mellitus (T2DM). The effects of methylophiopogonanone A (MO-A), a natural homoisoflavonoid with anti-inflammatory effects, on DCM and its underlying mechanisms were investigated in this study. METHODS The T2DM mouse model was induced by intraperitoneal injection of 30 mg/kg streptozotocin for 7 consecutive days and fed with a high-fat diet for 12 weeks. T2DM mice received MO-A (2.5, 5, or 10 mg/kg) treatment for two weeks. Cardiac function, hypertrophy, fibrosis, and inflammation were evaluated. The binding energy between MO-A and JNK1 was analyzed using molecular docking. The underlying mechanism was further investigated in high glucose (HG)-induced H9C2 cells. The cytotoxic effects, cardiomyocyte hypertrophy, fibrosis, inflammation, and relevant signaling proteins were assessed. RESULTS MO-A treatment alleviated cardiac function and histopathological changes in DCM mice. Moreover, MO-A treatment significantly decreased COLI, TGF-β1, MYH7, and ANP expression levels in DCM mice. Furthermore, TNF-α, IL-6, and IL-1β expression levels were notably downregulated after treatment with MO-A in DCM mice. Similar results were also observed in vitro. Mechanistically, MO-A targets JNK1 and downregulates its phosphorylation levels in DCM mice. The protective properties of MO-A were reversed by JNK1 overexpression in HG-induced H9C2 cells. CONCLUSION Our results revealed that MO-A could alleviate cardiac function, hypertrophy, fibrosis, and inflammation in DCM via inhibiting JNK1 signaling.
Collapse
Affiliation(s)
- Jing Yin
- Department of Traditional Chinese Medicine, Yantaishan Hospital, Yantai 264003, Shandong, China
| | - Zhicheng Song
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China
| | - Lijun Zhang
- Department of Endocrinology, Longkou Traditional Chinese Medicine Hospital, Yantai 265701, Shandong, China
| | - Jialin Cong
- Department of Integrated Chinese and Western Medicine, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai 264000, Shandong, China.
| |
Collapse
|
2
|
He C, Mao Y, Wan H. In-depth understanding of the structure-based reactive metabolite formation of organic functional groups. Drug Metab Rev 2025:1-43. [PMID: 40008940 DOI: 10.1080/03602532.2025.2472076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/19/2025] [Indexed: 02/27/2025]
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a leading cause of drug attrition and/or withdrawal. The formation of reactive metabolites is widely accepted as a key factor contributing to idiosyncratic DILI. Therefore, identifying reactive metabolites has become a critical focus during lead optimization, and a combination of GSH-/cyano-trapping and cytochrome P450 inactivation studies is recommended to identify compounds with the potential to generate reactive metabolites. Daily dose, clinical indication, detoxication pathways, administration route, and treatment duration are the most considerations when deprioritizing candidates that generate reactive metabolites. Removing the structural alerts is considered a pragmatic strategy for mitigating the risk associated with reactive metabolites, although this approach may sometimes exclude otherwise potent molecules. In this context, an in-depth insight into the structure-based reactive metabolite formation of organic functional groups can significantly aid in the rational design of drug candidates with improved safety profiles. The primary goal of this review is to delve into an analysis of the bioactivation mechanisms of organic functional groups and their potential detrimental effects with recent examples to assist medicinal chemists and metabolism scientists in designing safer drug candidates with a higher likelihood of success.
Collapse
Affiliation(s)
- Chunyong He
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Yuchang Mao
- Department of DMPK/Tox, Shanghai Hengrui Pharmaceutical Co. Ltd., Shanghai, China
| | - Hong Wan
- WHDex Consulting AB, Mölndal, Sweden
| |
Collapse
|
3
|
Xie T, Hao Y, Xie Y. A Validated Liquid Chromatography-Tandem Mass Spectrometry Method for the Determination of Methylophiopogonanone A in Rat Plasma and Its Application to Pharmacokinetic Study. Biomed Chromatogr 2024:e6050. [PMID: 39562503 DOI: 10.1002/bmc.6050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 10/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024]
Abstract
Methylophiopogonanone A (MOA) is one of the homoisoflavonoids isolated from Ophiopogon japonicus, which has been demonstrated to have extensive pharmacological activities. The aim of this study was to develop and validate a liquid chromatography-tandem mass spectrometry method for the measurement of MOA in rat plasma. Methylophiopogonanone B (MOB) was used as internal standard. After precipitation with acetonitrile, the samples were separated using a Waters ACQUITY HSS T3 column with 0.1% formic acid solution and acetonitrile as mobile phase. Mass detection was monitored using multiple reactions monitoring mode with precursor-to-product ion transition of m/z 343.2 > 135.1 for MOA and m/z 329.2 > 121.1 for internal standard. The assay showed good linearity over the concentration range of 1-1000 ng/mL, with correlation coefficient > 0.997. The lower limit of quantification (LLOQ) was 1 ng/mL. Acetonitrile-mediated precipitation showed high extraction efficiency (> 80%). The accuracy and precision were within the acceptable limits. MOA was demonstrated to be stable in rat plasma under the tested storage conditions. After validation, the proposed method was applied to the pharmacokinetic study of MOA in rat plasma, and the data revealed that MOA showed rapid absorption and elimination from rat plasma. The oral bioavailability of MOA in rat was 24.5%.
Collapse
Affiliation(s)
- Tiantian Xie
- School of Traditional Chinese Medicine, Jiangsu Medical College, Yancheng, Jiangsu, China
| | - Yanling Hao
- National Demonstration Center for Experimental Basic Medical Science Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanzheng Xie
- School of Traditional Chinese Medicine, Jiangsu Medical College, Yancheng, Jiangsu, China
| |
Collapse
|
4
|
Liu S, Zhao Y, Tang X, Yang J, Pan C, Liu C, Han J, Li C, Yi Y, Li Y, Cheng J, Zhang Y, Wang L, Tian J, Wang Y, Wang L, Liang A. In vitro inhibition of six active sesquiterpenoids in zedoary turmeric oil on human liver cytochrome P450 enzymes. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117588. [PMID: 38104879 DOI: 10.1016/j.jep.2023.117588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 11/29/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Affiliation(s)
- Suyan Liu
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yong Zhao
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Xuan Tang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Junling Yang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Chen Pan
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Chenyue Liu
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jiayin Han
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Chunying Li
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yan Yi
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yingfei Li
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jintang Cheng
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yushi Zhang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Lianmei Wang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Jingzhuo Tian
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Yuan Wang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Liping Wang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China
| | - Aihua Liang
- State Key Laboratory of Innovative Natural Medicine and TCM Injections, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, PR China.
| |
Collapse
|
5
|
Guo X, Wang L, Zhang J, Liu Q, Wang B, Liu D, Gao F, Lanzi G, Zhao Y, Shi Y. Thwarting resistance: MgrA inhibition with methylophiopogonanone a unveils a new battlefront against S. aureus. NPJ Biofilms Microbiomes 2024; 10:15. [PMID: 38413623 PMCID: PMC10899606 DOI: 10.1038/s41522-024-00485-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Limitations in the clinical treatment of Staphylococcus aureus (S. aureus) infections have arisen due to the advent of antibiotic-resistant strains. Given the immense potential of therapeutic strategies targeting bacterial virulence, the role of MgrA as a pivotal virulence determinant in S. aureus-orchestrating resistance, adherence, and hundreds of virulence targets-becomes indispensable. In this investigation, leveraging advanced virtual screening and fluorescence anisotropy assays, we discerned methylophiopogonanone A (Mo-A), a flavonoid derivative, as a potent disruptor of the MgrA-DNA interaction nexus. Subsequent analysis revealed that Mo-A effectively inhibits the expression of virulence factors such as Hla and Pvl in S. aureus and markedly reduces its adhesion capability to fibrinogen. On a cellular landscape, Mo-A exerts a mitigating influence on the deleterious effects inflicted by S. aureus USA300 on A549 cells. Furthermore, our data indicate that Mo-A downregulates the transcription of genes associated with immune evasion, such as nucleases (nuc), Staphylococcal Chemotaxis Inhibitory Protein (chips), and Staphylococcal Complement Inhibitor (scin), thereby undermining immune escape and amplifying neutrophil chemotaxis. Upon application in an in vivo setting, Mo-A assumes a protective persona in a murine model of S. aureus USA300-induced pneumonia and demonstrates efficacy in the Galleria mellonella infection model. Of note, S. aureus displayed no swift acquisition of resistance to Mo-A, and the effect was synergistically enhanced when used in combination with vancomycin. Our findings add substantive weight to the expanding field of virulence-targeted therapeutic strategies and set the stage for more comprehensive exploration of Mo-A potential in combating antibiotic-resistant S. aureus.
Collapse
Affiliation(s)
- Xuerui Guo
- School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Li Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Jinlong Zhang
- School of Pharmaceutical Science, Jilin University, Changchun, China
| | - Quan Liu
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Bingmei Wang
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun, Jilin, China
| | | | - Yicheng Zhao
- Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China.
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China.
| | - Yan Shi
- School of Pharmaceutical Science, Jilin University, Changchun, China.
| |
Collapse
|
6
|
Li C, Li X, Fan A, He N, Wu D, Yu H, Wang K, Jiao W, Zhao X. Evidence for cytochrome P450 3A4-mediated metabolic activation of SCO-267. Biopharm Drug Dispos 2024; 45:30-42. [PMID: 38236698 DOI: 10.1002/bdd.2381] [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: 10/28/2023] [Revised: 12/04/2023] [Accepted: 12/24/2023] [Indexed: 02/15/2024]
Abstract
SCO-267 is a potent G-protein-coupled receptor 40 agonist that is undergoing clinical development for the treatment of type 2 diabetes mellitus. The current work was undertaken to investigate the bioactivation potential of SCO-267 in vitro and in vivo. Three SCO-267-derived glutathione (GSH) conjugates (M1-M3) were found both in rat and human liver microsomal incubations supplemented with GSH and nicotinamide adenine dinucleotide phosphate. Two GSH conjugates (M1-M2) together with two N-acetyl-cysteine conjugates (M4-M5) were detected in the bile of rats receiving SCO-267 at 10 mg/kg. The identified conjugates suggested the generation of quinone-imine and ortho-quinone intermediates. CYP3A4 was demonstrated to primarily catalyze the bioactivation of SCO-267. In addition, SCO-267 concentration-, time-, and NADPH-dependently inactivated CYP3A in human liver microsomes using testosterone as a probe substrate, along with KI and kinact values of 4.91 μM and 0.036 min-1 , respectively. Ketoconazole (a competitive inhibitor of CYP3A) displayed no significant protective effect on SCO-267-induced CYP3A inactivation. However, inclusion of GSH showed significant protection. These findings revealed that SCO-267 undergoes a facile CYP3A4-catalyzed bioactivation with the generation of quinone-imine and ortho-quinone intermediates, which were assumed to be involved in SCO-267 induced CYP3A inactivation. These findings provide further insight into the bioactivation pathways involved in the generation of reactive, potentially toxic metabolites of SCO-267. Further studies are needed to evaluate the influence of SCO-267 metabolism on the safety of this drug in vivo.
Collapse
Affiliation(s)
- Cui Li
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Xiaokun Li
- Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Ali Fan
- TriApex Laboratories Co. Ltd, Nanjing, China
| | - Ning He
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Dongmei Wu
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Hongyan Yu
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Kun Wang
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Weijie Jiao
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| | - Xu Zhao
- Department of Pharmacy, Henan Province Hospital of Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan Province, China
| |
Collapse
|
7
|
Yumoto Y, Endo T, Harada H, Kobayashi K, Nakabayashi T, Abe Y. High-throughput assay to simultaneously evaluate activation of CYP3A and the direct and time-dependent inhibition of CYP3A, CYP2C9, and CYP2D6 using liquid chromatography-tandem mass spectrometry. Xenobiotica 2024; 54:45-56. [PMID: 38265764 DOI: 10.1080/00498254.2024.2308818] [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: 10/25/2023] [Accepted: 01/18/2024] [Indexed: 01/25/2024]
Abstract
In the early stages of drug discovery, adequate evaluation of the potential drug-drug interactions (DDIs) of drug candidates is important. Several CYP3A activators are known to lead to underestimation of DDIs. These compounds affect midazolam 1'-hydroxylation but not midazolam 4-hydroxylation.We used both metabolic reactions of midazolam to evaluate the activation and inhibition of CYP3A activators simultaneously. For our CYP inhibition assay using cocktail probe substrates, simultaneous liquid chromatography-tandem mass spectrometry monitoring of 1'-hydroxymidazolam and 4-hydroxymidazolam for CYP3A was established in addition to monitoring of 4-hydroxydiclofenac and 1'-hydroxybufuralol for CYP2C9 and CYP2D6.The results of our cocktail inhibition assay were well correlated with those of a single inhibition assay, as were the estimated inhibition parameters for typical CYP3A inhibitors. In our assay, a proprietary compound that activated midazolam 1'-hydroxylation and tended to inhibit 4-hydroxylation was evaluated along with known CYP3A activators. All compounds were well characterised by comparison of the results of midazolam 1'- and 4-hydroxylation.In conclusion, our CYP cocktail inhibition assay can detect CYP3A activation and assess the direct and time-dependent inhibition potentials for CYP3A, CYP2C9, and CYP2D6. This method is expected to be very efficient in the early stages of drug discovery.
Collapse
Affiliation(s)
- Yu Yumoto
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takuro Endo
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Hiroshi Harada
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Kaoru Kobayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Takeshi Nakabayashi
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| | - Yoshikazu Abe
- Central Research Laboratories, Kissei Pharmaceutical Co., Ltd, Azumino, Nagano, Japan
| |
Collapse
|
8
|
Dai Z, Wu Y, Xiong Y, Wu J, Wang M, Sun X, Ding X, Yang L, Sun X, Ge G. CYP1A inhibitors: Recent progress, current challenges, and future perspectives. Med Res Rev 2024; 44:169-234. [PMID: 37337403 DOI: 10.1002/med.21982] [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: 12/09/2022] [Revised: 03/28/2023] [Accepted: 05/23/2023] [Indexed: 06/21/2023]
Abstract
Mammalian cytochrome P450 1A (CYP1A) are key phase I xenobiotic-metabolizing enzymes that play a distinctive role in metabolic activation or metabolic clearance of a variety of procarcinogens, drugs, and endogenous substances. Human CYP1A subfamily contains two members (hCYP1A1 and hCYP1A2), which are known to catalyze the oxidative activation of some environmental procarcinogens into carcinogenic species. Increasing evidence has demonstrated that CYP1A inhibitor therapies are promising strategies for cancer chemoprevention or overcoming CYP1A-associated drug toxicity and resistance. Herein, we reviewed recent advances in the discovery and characterization of hCYP1A inhibitors, from the discovery approaches to structural features and biomedical applications of hCYP1A inhibitors. The inhibition potentials, inhibition modes, and inhibition constants of all reported hCYP1A inhibitors are comprehensively summarized. Meanwhile, the structural features and structure-activity relationships of different classes of hCYP1A1 and hCYP1A2 inhibitors are analyzed and discussed in depth. Furthermore, the major challenges and future directions for this field are presented and highlighted. Collectively, the information and knowledge presented here will strongly facilitate the researchers to discover and develop more efficacious CYP1A inhibitors for specific purposes, such as chemo-preventive agents or as tool molecules in hCYP1A-related fundamental studies.
Collapse
Affiliation(s)
- Ziru Dai
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Wu
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Xiong
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Min Wang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, America
| | - Ling Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xiaobo Sun
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center for TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Huang H, Zhao Y, Huang C, Lv N, Zhao J, Sun S, Guo C, Zhao D, Chen X, Zhang Y. Unraveling a Combined Inactivation Mechanism of Cytochrome P450s by Genipin, the Major Reactive Aglycone Derived from Gardeniae Fructus. Chem Res Toxicol 2023; 36:1483-1494. [PMID: 37622730 DOI: 10.1021/acs.chemrestox.3c00102] [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: 08/26/2023]
Abstract
Genipin (GP) is the reactive aglycone of geniposide, the main component of traditional Chinese medicine Gardeniae Fructus (GF). The covalent binding of GP to cellular proteins is suspected to be responsible for GF-induced hepatotoxicity and inhibits drug-metabolizing enzyme activity, although the mechanisms remain to be clarified. In this study, the mechanisms of GP-induced human hepatic P450 inactivation were systemically investigated. Results showed that GP inhibited all tested P450 isoforms via distinct mechanisms. CYP2C19 was directly and irreversibly inactivated without time dependency. CYP1A2, CYP2C9, CYP2D6, and CYP3A4 T (testosterone as substrate) showed time-dependent and mixed-type inactivation, while CYP2B6, CYP2C8, and CYP3A4 M (midazolam as substrate) showed time-dependent and irreversible inactivation. For CYP3A4 inactivation, the kinact/KI values in the presence or absence of NADPH were 0.26 or 0.16 min-1 mM-1 for the M site and 0.62 or 0.27 min-1 mM-1 for the T site. Ketoconazole and glutathione (GSH) both attenuated CYP3A4 inactivation, suggesting an active site occupation- and reactive metabolite-mediated inactivation mechanism. Moreover, the in vitro and in vivo formation of a P450-dependent GP-S-GSH conjugate indicated the involvement of metabolic activation and thiol residues binding in GP-induced enzyme inactivation. Lastly, molecular docking analysis simulated potential binding sites and modes of GP association with CYP2C19 and CYP3A4. We propose that direct covalent binding and metabolic activation mediate GP-induced P450 inactivation and alert readers to potential risk factors for GP-related clinical drug-drug interactions.
Collapse
Affiliation(s)
- Haoyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yulin Zhao
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chunyan Huang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ning Lv
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Zhao
- Pharmaceutical Animal Experimental Center, China Pharmaceutical University, Nanjing 210009, China
| | - Shanliang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Chaorui Guo
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Di Zhao
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xijing Chen
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yongjie Zhang
- Clinical Pharmacology Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
10
|
Chen X, Han L, Zhao Y, Huang H, Pan H, Zhang C, Chen H, Sun S, Yao S, Chen X, Zhang Y. Mechanistic Study of Icaritin-Induced Inactivation of Cytochrome P450 2C9. Drug Metab Dispos 2023; 51:771-781. [PMID: 36863865 DOI: 10.1124/dmd.122.001245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 02/28/2023] [Indexed: 03/04/2023] Open
Abstract
Icaritin (ICT) is a prenylflavonoid derivative that has been approved by National Medical Products Administration for the treatment of hepatocellular carcinoma. This study aims to evaluate the potential inhibitory effect of ICT against cytochrome P450 (CYP) enzymes and to elucidate the inactivation mechanisms. Results showed that ICT inactivated CYP2C9 in a time-, concentration-, and NADPH-dependent manner with Ki = 1.896 μM, Kinact = 0.02298 minutes-1, and Kinact/Ki = 12 minutes-1 mM-1, whereas the activities of other CYP isozymes was minimally affected. Additionally, the presence of CYP2C9 competitive inhibitor, sulfaphenazole, superoxide dismutase/catalase system, and GSH all protected CYP2C9 from ICT-induced activity loss. Moreover, the activity loss was neither recovered by washing the ICT-CYP2C9 preincubation mixture nor the addition of potassium ferricyanide. These results, collectively, implied the underlying inactivation mechanism involved the covalent binding of ICT to the apoprotein and/or the prosthetic heme of CYP2C9. Furthermore, an ICT-quinone methide (QM)-derived GSH adduct was identified, and human glutathione S-transferases (GST) isozymes GSTA1-1, GSTM1-1, and GSTP1-1 were shown to be substantially involved in the detoxification of ICT-QM. Interestingly, our systematic molecular modeling work predicted that ICT-QM was covalently bound to C216, a cysteine residue located in the F-G loop downstream of substrate recognition site (SRS) 2 in CYP2C9. The sequential molecular dynamics simulation confirmed the binding to C216 induced a conformational change in the active catalytic center of CYP2C9. Lastly, the potential risks of clinical drug-drug interactions triggered by ICT as a perpetrator were extrapolated. In summary, this work confirmed that ICT was an inactivator of CYP2C9. SIGNIFICANCE STATEMENT: This study is the first to report the time-dependent inhibition of CYP2C9 by icaritin (ICT) and the intrinsic molecular mechanism behind it. Experimental data indicated that the inactivation was via irreversible covalent binding of ICT-quinone methide to CYP2C9, while molecular modeling analysis provided additional evidence by predicting C216 as the key binding site which influenced the structural confirmation of CYP2C9's catalytic center. These findings suggest the potential of drug-drug interactions when ICT is co-administered with CYP2C9 substrates clinically.
Collapse
Affiliation(s)
- Xiang Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Luyao Han
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Yulin Zhao
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Haoyan Huang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - He Pan
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Chenmeng Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Huili Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Shanliang Sun
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Sihui Yao
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Xijing Chen
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| | - Yongjie Zhang
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China (Xiang Chen, L.H., Y. Zhao, H.H., H.P., C.Z., Xijing Chen, Y. Zhang); Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida (H.C.); National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, China (S.S.); and Editorial Department of Progress in Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China (S.Y.)
| |
Collapse
|
11
|
He RJ, Tian ZH, Huang J, Sun MR, Wei F, Li CY, Zeng HR, Zhang F, Guan XQ, Feng Y, Meng XM, Yang H, Ge GB. Rationally Engineered CYP3A4 Fluorogenic Substrates for Functional Imaging Analysis and Drug-Drug Interaction Studies. J Med Chem 2023; 66:6743-6755. [PMID: 37145039 DOI: 10.1021/acs.jmedchem.3c00101] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Cytochrome P450 3A4 (CYP3A4) is a key xenobiotic-metabolizing enzyme-mediated drug metabolism and drug-drug interaction (DDI). Herein, an effective strategy was used to rationally construct a practical two-photon fluorogenic substrate for hCYP3A4. Following two-round structure-based substrate discovery and optimization, we have successfully constructed a hCYP3A4 fluorogenic substrate (F8) with desirable features, including high binding affinity, rapid response, excellent isoform specificity, and low cytotoxicity. Under physiological conditions, F8 is readily metabolized by hCYP3A4 to form a brightly fluorescent product (4-OH F8) that can be easily detected by various fluorescence devices. The practicality of F8 for real-time sensing and functional imaging of hCYP3A4 has been examined in tissue preparations, living cells, and organ slices. F8 also demonstrates good performance for high-throughput screening of hCYP3A4 inhibitors and assessing DDI potentials in vivo. Collectively, this study develops an advanced molecular tool for sensing CYP3A4 activities in biological systems, which strongly facilitates CYP3A4-associated fundamental and applied research studies.
Collapse
Affiliation(s)
- Rong-Jing He
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhen-Hao Tian
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jian Huang
- Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai 201203, China
| | - Meng-Ru Sun
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Feng Wei
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
| | - Chun-Yu Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hai-Rong Zeng
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao-Qing Guan
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan Feng
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
| | - Xiang-Ming Meng
- School of Chemistry and Chemical Engineering & Center for Atomic Engineering of Advanced Materials & Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei 230601, China
- Institute of Physical Science and Information Technology, Anhui University, Hefei 230601, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
12
|
Wu D, Wang C, Zhang X, Xie T. A high-resolution mass spectrometry-based methodology for characterization and identification of methylophiopogonanone B metabolites from cryopreserved hepatocytes and liver microsomes. Biomed Chromatogr 2023; 37:e5574. [PMID: 36527223 DOI: 10.1002/bmc.5574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/04/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Methylophiopogonanone B (MOB), one of the homoisoflavonoids isolated from Ophiopogon japonicus, has been demonstrated to possess antioxidative and antitumor activities. The aim of this work was to investigate the metabolism of MOB using liver microsomes and hepatocytes. MOB was individually incubated with rat, monkey, and human hepatocytes to generate the metabolites. To investigate the bioactivation pathways, MOB was incubated with liver microsomes in the presence of glutathione (GSH). All the metabolites were detected and identified using LC with a quadrupole Orbitrap mass spectrometer. Under the current conditions, nine metabolites were identified in hepatocyte incubations. Of these metabolites, M7 derived from hydroxylation was identified as the most abundant metabolite in hepatocyte incubation. MOB was metabolized via demethylation, hydroxylation, and glucuronidation. In liver microsomes, five GSH conjugates were detected and identified. MOB was subjected to bioactivation through demethylation yielding M9, which further formed quinone-methide and ortho-quinone intermediates, followed by GSH conjugation. This work is the first to study the metabolism of MOB, which will help us understand its disposition and efficacy.
Collapse
Affiliation(s)
- Dawei Wu
- Jiangsu Vocational College of Medicine, Yancheng, China
| | | | - Xiaolong Zhang
- Jiangsu Food & Pharmaceutical Science College, Huai'an, China
| | - Tiantian Xie
- Jiangsu Vocational College of Medicine, Yancheng, China
| |
Collapse
|
13
|
Lee SG, Cho KH, Nguyen TTL, Vo DK, Chae YJ, Maeng HJ. Inhibitory effect of 20(S)-protopanaxadiol on cytochrome P450: Potential of its pharmacokinetic interactions in vivo. Biomed Pharmacother 2022; 153:113514. [DOI: 10.1016/j.biopha.2022.113514] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/24/2022] [Accepted: 08/02/2022] [Indexed: 12/28/2022] Open
|
14
|
Zhang F, Zhang T, Gong J, Fang Q, Qi S, Li M, Han Y, Liu W, Ge G. The Chinese herb Styrax triggers pharmacokinetic herb-drug interactions via inhibiting intestinal CYP3A. Front Pharmacol 2022; 13:974578. [PMID: 36110541 PMCID: PMC9469097 DOI: 10.3389/fphar.2022.974578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/28/2022] [Indexed: 12/04/2022] Open
Abstract
Human cytochrome P450 3A4 (hCYP3A4) is a predominant enzyme to trigger clinically relevant drug/herb-drug interactions (DDIs or HDIs). Although a number of herbal medicines have been found with strong anti-hCYP3A4 effects in vitro, the in vivo modulatory effects of herbal medicines on hCYP3A4 and their potential risks to trigger HDIs are rarely investigated. Herein, we demonstrate a case study to efficiently find the herbal medicine(s) with potent hCYP3A4 inhibition in vitro and to accurately assess the potential HDIs risk in vivo. Following screening over 100 herbal medicines, the Chinese herb Styrax was found with the most potent hCYP3A4 inhibition in HLMs. In vitro assays demonstrated that Styrax could potently inhibit mammalian CYP3A in liver and intestinal microsomes from both humans and rats. In vivo pharmacokinetic assays showed that Styrax (i.g., 100 mg/kg) significantly elevated the plasma exposure of two CYP3A-substrate drugs (midazolam and felodipine) when midazolam or felodipine was administered orally. By contrast, the plasma exposure of either midazolam or felodipine was hardly affected by Styrax (i.g.) when the victim drug was administered intravenously. Further investigations demonstrated that seven pentacyclic triterpenoid acids (PTAs) in Styrax were key substances responsible for CYP3A inhibition, while these PTAs could be exposed to intestinal tract at relatively high exposure levels but their exposure levels in rat plasma and liver were extremely low. These findings well explained why Styrax (i.g.) could elevate the plasma exposure of victim drugs only when these agents were orally administrated. Collectively, our findings demonstrate that Styrax can modulate the pharmacokinetic behavior of CYP3A-substrate drugs via inhibiting intestinal CYP3A, which is very helpful for the clinical pharmacologists to better assess the HDIs triggered by Styrax or Styrax-related herbal products.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tiantian Zhang
- School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jiahao Gong
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qinqin Fang
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shenglan Qi
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengting Li
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Han
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
15
|
Li X, Yin D, Sun Y. Identification of the metabolite of ophiopogonanone A by liquid chromatography/quadrupole time-of-flight mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9311. [PMID: 35557016 DOI: 10.1002/rcm.9311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 06/15/2023]
Abstract
RATIONALE Ophiopogonanone A (OPA) is one of the representative homoisoflavonoids isolated from Ophiopogonis Radix. The aim of this study was to identify and characterize the metabolites of OPA generated in the liver microsomes and hepatocytes of rats and humans. METHODS The metabolites were generated by incubating OPA (5 μM) with liver microsomes or hepatocytes at 37°C. To trap the reactive metabolites, glutathione (GSH, 5mM) was added into microsomal incubations. The metabolite identification and profiling were performed using ultra-high-performance liquid chromatography combined with photo-diode array detector and quadrupole time-of-flight tandem mass spectrometry (LC-Q/TOF-MS). The acquired mass data were processed by MetaboLynx software. The structures of the metabolites were tentatively characterized in terms of their accurate masses, product ions, and retention times. RESULTS Under the present conditions, a total of nine metabolites were detected and their structures were tentatively identified. Among these metabolites, M8 (OPA catechol) was the most abundant metabolite both in rat and human liver microsomes. M7 (glucuronidation product of M8) was the major metabolite both in rat and human hepatocytes. The metabolic pathways of OPA include demethylenation, dehydrogenation, hydroxylation, methylation and glucuronidation and GSH conjugation. CONCLUSION Our results provided valuable information regarding the in vitro metabolism of OPA, which would help us understand the mechanism of the elimination of OPA and in turn the effectiveness and potential toxicity.
Collapse
Affiliation(s)
- Xiao Li
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Dengyang Yin
- Department of Clinical Pharmacy, Jingjiang People's Hospital, The Seventh Affiliated Hospital of Yangzhou University, Jingjiang, Jiangsu Province, China
| | - Ying Sun
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| |
Collapse
|
16
|
Wu X, Sun S, Wu X, Sun Z. Identification of the metabolites of methylophiopogonanone A by ultra-high-performance liquid chromatography combined with high-resolution mass spectrometry. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2022; 36:e9304. [PMID: 35347765 DOI: 10.1002/rcm.9304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 06/14/2023]
Abstract
RATIONALE Methylophiopogonanone A (MOA) is a naturally occurring homoisoflavonoid from the Chinese herb Ophiopogon japonicus, which has been demonstrated to attenuate myocardial apoptosis. However, the metabolism of MOA remains unknown. The goal of the present work was to investigate the in vitro metabolism of MOA using liver microsomes and hepatocytes. METHODS The metabolites were generated by incubating MOA with rat, monkey and human liver microsomes or hepatocytes. The resulting samples were analyzed by using a quadrupole-orbitrap high-resolution mass spectrometer. The metabolites were identified through the measurements of the exact mass, elemental composition and product ions. RESULTS A total of 15 metabolites were detected and identified. Among these metabolites, M7 (demethylenation) was the most abundant metabolite in liver microsomes, while M6 (hydroxylation) was the predominant metabolite in hepatocytes, and glucuronidation metabolites (M9 and M10) were also the main metabolites in hepatocytes. The metabolic pathways of MOA included hydroxylation, demethylenation, glucuronidation, methylation, sulfation and glutathione conjugation. CONCLUSIONS This study for the first time provides valuable data on the metabolites of MOA, which will be of great importance for a better understanding of its disposition and to predict human pharmacokinetics.
Collapse
Affiliation(s)
- Xiaowen Wu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Shuai Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Xiaoyi Wu
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| | - Zengxian Sun
- Department of Pharmacy, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu Province, China
| |
Collapse
|
17
|
Wang YS, Zheng W, Jiang N, Jin YX, Meng ZK, Sun MX, Zong YL, Xu T, Zhu J, Tan RX. Alteration of the Catalytic Reaction Trajectory of a Vicinal Oxygen Chelate Enzyme by Directed Evolution. Angew Chem Int Ed Engl 2022; 61:e202201321. [DOI: 10.1002/anie.202201321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Indexed: 11/09/2022]
Affiliation(s)
- Yi Shuang Wang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Wan Zheng
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Nan Jiang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing 210023 China
| | - Yun Xia Jin
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zi Kang Meng
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Meng Xin Sun
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Yu Liang Zong
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Tong Xu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing 210023 China
| | - Jiapeng Zhu
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Ren Xiang Tan
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
- State Key Laboratory of Pharmaceutical Biotechnology Institute of Functional Biomolecules School of Life Sciences Nanjing University Nanjing 210023 China
| |
Collapse
|
18
|
Cheng C, Zhao S, Gu YL, Pang J, Zhao Y. Characterization and identification of the metabolites of dihydromethysticin by ultra-high-performance liquid chromatography orbitrap high-resolution mass spectrometry. J Sep Sci 2022; 45:2914-2923. [PMID: 35689602 DOI: 10.1002/jssc.202200250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/27/2022] [Accepted: 06/06/2022] [Indexed: 11/06/2022]
Abstract
Dihydromethysticin, a natural component from Piper methysticum Forst, has been reported to display pharmacological effects in mental disorders and some malignant tumors. However, the metabolism of this component remained unknown. The goal of this work was conducted to discover the metabolic profiles of dihydromethysticin. The in vitro incubation was performed by incubating dihydromethysticin with rat, monkey, and human liver microsomes and hepatocytes. An analytical assay of ultra-high performance liquid chromatography combined with Orbitrap high-resolution mass spectrometry was utilized to detect and identify the metabolites. With high resolution mass spectrometric determination, the accurate mass, elemental composition, and product ions of the metabolites were determined, which enabled structural characterization to become easy. Under the present conditions, four phase-I metabolites, as well as six phase-II metabolites, were detected and their tentative structures were characterized by mass spectra. M4 was found as the most abundant metabolite both in liver microsomes and hepatocytes. Cytochrome P450 1A2, 2C9, and 3A4 contributed to the formation of this metabolite by using human recombinant P450 enzymes. M4 can be oxidized into reactive ortho-quinone intermediate followed by conjugating with glutathione. M4 was also subject to glucuronidation (M1 and M2) and methylation (M5). Demethylenation, oxidation, hydroxylation, glucuronidation, glutathionylation, and methylation were the primary metabolic pathways of dihydromethysticin. This study provides in vitro metabolism data of dihydromethysticin, which is indispensable for understanding the disposition of this compound.
Collapse
Affiliation(s)
- Cong Cheng
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang, P. R. China
| | - Shanshan Zhao
- Jiangsu Wanbang Pharmaceutical Technology Co. Ltd, P. R. China
| | - Yong-Li Gu
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang, P. R. China
| | - Jie Pang
- Department of Pharmacy, The First Affiliated Hospital of Kangda College of Nanjing Medical University/The First People's Hospital of Lianyungang, Lianyungang, P. R. China
| | - Yanyun Zhao
- Department of Pharmacy, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang, P. R. China
| |
Collapse
|
19
|
Zhang Q, Liu H, Wu D, Yu H, Wang K, Jiao W, Zhao X. Methysticin Acts as a Mechanism-Based Inactivator of Cytochrome P450 2C9. Chem Res Toxicol 2022; 35:1117-1124. [PMID: 35583123 DOI: 10.1021/acs.chemrestox.2c00098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Methysticin is one of the naturally occurring bioactive constituents extracted from Piper methysticum Forst. In the present study, we intended to investigate the inhibitory effect of methysticin on cytochrome P450 (P450) enzymes. Methysticin exhibited time-, concentration-, and NADPH-dependent inhibition on CYP2C9 using diclofenac as a probe substrate. Approximately 85% of CYP2C9 activity was inhibited by methysticin at 50 μM after a 30 min preincubation with human liver microsomes in the presence of NADPH. The kinetic parameters KI, kinact, and t1/2,inact were 13.32 ± 1.35 μM, 0.054 ± 0.005 min-1, and 12.83 ± 3.23 min, respectively. Sulfaphenazole (competitive inhibitor of CYP2C9) displayed a significant protective effect on methysticin-induced CYP2C9 inactivation. However, the inclusion of catalase/superoxide dismutase or glutathione (GSH) showed no such protection. A carbene intermediate was postulated to be involved in methysticin-induced CYP2C9 inactivation as K3Fe(CN)6 recovered 14.96% of CYP2C9 activity. A methysticin-derived ortho-quinone intermediate dependent on NADPH was trapped by GSH, and this intermediate was believed to be involved in CYP2C9 inactivation. CYP1A2, 2C9, and 3A4 were the major enzymes responsible for methysticin bioactivation. Taken together, the present work demonstrated that methysticin was a mechanism-based inactivator of CYP2C9. Both ortho-quinone and carbene intermediates appeared to be involved in the inactivation of CYP2C9 induced by methysticin.
Collapse
Affiliation(s)
- Qiuying Zhang
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| | - Hui Liu
- Department of Applied Chemistry, North China University of Water Resources and Electric Power, Zhengzhou 450046, China
| | - Dongmei Wu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| | - Hongyan Yu
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| | - Kun Wang
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| | - Weijie Jiao
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| | - Xu Zhao
- Department of Pharmacy, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, No. Dongfeng Road, Jinshui District, Zhengzhou 450002, Henan, China
| |
Collapse
|
20
|
Wang YS, Zheng W, Jiang N, Jin YX, Meng ZK, Sun MX, Zong YL, Xu T, Zhu J, Tan RX. Alteration of the Catalytic Reaction Trajectory of a Vicinal Oxygen Chelate Enzyme by Directed Evolution. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202201321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Yi Shuang Wang
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Wan Zheng
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Nan Jiang
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing 210023 China
| | - Yun Xia Jin
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Zi Kang Meng
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Meng Xin Sun
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Yu Liang Zong
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Tong Xu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine School of Pharmacy Nanjing Medical University Nanjing 210023 China
| | - Jiapeng Zhu
- School of Medicine and Holistic Integrative Medicine Nanjing University of Chinese Medicine Nanjing 210023 China
| | - Ren Xiang Tan
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy Nanjing University of Chinese Medicine Nanjing 210023 China
- State Key Laboratory of Pharmaceutical Biotechnology Institute of Functional Biomolecules School of Life Sciences Nanjing University Nanjing 210023 China
| |
Collapse
|
21
|
Zhou QH, Zhu GH, Song YQ, Que YF, He QQ, Tu DZ, Zeng HR, Qin WW, Ai CZ, Ge GB. Methylophiopogonanone A is a naturally occurring broad-spectrum inhibitor against human UDP-glucuronosyltransferases: Inhibition behaviours and implication in herb-drug interactions. Basic Clin Pharmacol Toxicol 2021; 129:437-449. [PMID: 34478607 DOI: 10.1111/bcpt.13651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Methylophiopogonanone A (MOA) is an abundant homoisoflavonoid in the Chinese herb Ophiopogonis Radix. Recent investigations revealed that MOA inhibited several human cytochrome P450 enzymes (CYPs) and stimulated OATP1B1. However, the inhibitory effects of MOA on phase II drug-metabolizing enzymes, such as human UDP-glucuronosyltransferases (hUGTs), have not been well investigated. Herein, the inhibition potentials of MOA on hUGTs were assessed. The results clearly demonstrated that MOA dose-dependently inhibited all tested hUGTs including UGT1A1 (IC50 = 1.23 μM), one of the most important detoxification enzymes in humans. Further investigations showed that MOA strongly inhibited UGT1A1-catalysed NHPH-O-glucuronidation in a range of biological settings including hUGT1A1, human liver microsomes (HLM) and HeLa cells overexpressing UGT1A1. Inhibition kinetic analyses demonstrated that MOA competitively inhibited UGT1A1-catalysed NHPH-O-glucuronidation in both hUGT1A1 and HLM, with Ki values of 0.52 and 1.22 μM, respectively. Collectively, our findings expanded knowledge of the interactions between MOA and human drug-metabolizing enzymes, which would be very helpful for guiding the use of MOA-related herbal products in clinical settings.
Collapse
Affiliation(s)
- Qi-Hang Zhou
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang-Hao Zhu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yun-Qing Song
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan-Fang Que
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qing-Qing He
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong-Zhu Tu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hai-Rong Zeng
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei-Wei Qin
- Department of Pharmacy and Worldwide Medical Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chun-Zhi Ai
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmacy, Guangxi Normal University, Guilin, China
| | - Guang-Bo Ge
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|