1
|
Sun J, Li R, Zhang J, Huang Y, Lu Y, Liu C, Li Y, Liu T. Analysis of compatibility mechanism of shenxiong glucose injection after multiple dosing based on differences of PK-PD correlation and cytochrome P450 enzyme. J Pharm Biomed Anal 2024; 239:115899. [PMID: 38103414 DOI: 10.1016/j.jpba.2023.115899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Shenxiong glucose injection (SGI) containing a water extract from the roots of Danshen and Ligustrazine hydrochloride, is the main drug used for the prevention and treatment of acute myocardial ischemia (AMI) in China. Based on the characteristics of drug clinical applications, this study aims to uncover the compatibility mechanism of SGI by investigating pharmacokinetic (PK) and pharmacodynamic (PD) differences between Danshen glucose injection (DGI), Ligustrazine glucose injection (LGI) and SGI groups after multiple dosing during the pathological state from the perspective of metabolic enzymes. Compared to the LGI group, the absorption (Cmax) and exposure (AUC) of ligustrazine increased significantly, and the protein expression of CYP1A2, CYP2C11 and CYP3A2 in the SGI group decreased significantly. Furthermore, the PK and PD experimental data for Danshen and ligustrazine in AMI rats were fitted to obtain a PK-PD binding model with three components. PK-PD parameter analysis showed that in the SGI group the IC50 values of ligustrazine and danshensu on AST, CK-MB, cTn-I and the IC50 values of rosmarinic acid on AST and CK-MB were lower than the DGI or LGI group. It is speculated that Danshen inhibited CYP1A2, CYP2C11 and CYP3A2 mediating the metabolism of ligustrazine and decreased the expression of these three isozymes, which further affected the in vivo process of ligustrazine. Moreover, the combination of Danshen and ligustrazine could have better regulating effect on AST, CK-MB and cTn-I. This preliminary study has provided a scientific basis for understanding the compatibility mechanism of SGI from the viewpoint of the regulation of CYP enzymes in the PK-PD model.
Collapse
Affiliation(s)
- Jia Sun
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China; National Engineering Research Center of Miao's Medicines, Guiyang 550004, China
| | - Rong Li
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Jingya Zhang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China; School of Pharmacy, Guizhou Medical University, Guiyang 550004, China
| | - Yong Huang
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China; National Engineering Research Center of Miao's Medicines, Guiyang 550004, China
| | - Yuan Lu
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China
| | - Chunhua Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and Traditional Chinese Medicine Development and Application, Guizhou Medical University, Guiyang 550004, China
| | - Yongjun Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Research Center for the Development and Application of Ethnic Medicine and Traditional Chinese Medicine Development and Application, Guizhou Medical University, Guiyang 550004, China.
| | - Ting Liu
- Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
2
|
Zhao Y, Yang C, Liu Y, Qin M, Sun J, Liu G. Effects of sodium ferulate for injection on anticoagulation of warfarin in rats in vivo. BMC Complement Med Ther 2024; 24:87. [PMID: 38355450 PMCID: PMC10865636 DOI: 10.1186/s12906-024-04389-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/03/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Herb-drug interactions may result in increased adverse drug reactions or diminished drug efficacy, especially for drugs with a narrow therapeutic index such as warfarin. The current study investigates the effects of sodium ferulate for injection (SFI) on anticoagulation of warfarin from aspects of pharmacodynamics and pharmacokinetics in rats and predicts the risk of the combination use. METHODS Rats were randomly divided into different groups and administered single- or multiple-dose of warfarin (0.2 mg/kg) with or without SFI of low dose (8.93 mg/kg) or high dose (26.79 mg/kg). Prothrombin time (PT) and activated partial thromboplastin time (APTT) were detected by a blood coagulation analyzer, and international normalized ratio (INR) values were calculated. UPLC-MS/MS was conducted to measure concentrations of warfarin enantiomers and pharmacokinetic parameters were calculated by DAS2.0 software. RESULTS The single-dose study demonstrated that SFI alone had no effect on coagulation indices, but significantly decreased PT and INR values of warfarin when the two drugs were co-administered (P < 0.05 or P < 0.01), while APTT values unaffected (P > 0.05). Cmax and AUC of R/S-warfarin decreased but CL increased significantly in presence of SFI (P < 0.01). The multiple-dose study showed that PT, APTT, INR, and concentrations of R/S-warfarin decreased significantly when SFI was co-administered with warfarin (P < 0.01). Warfarin plasma protein binding rate was not significantly changed by SFI (P > 0.05). CONCLUSIONS The present study implied that SFI could accelerate warfarin metabolism and weaken its anticoagulation intensity in rats.
Collapse
Affiliation(s)
- Yue Zhao
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, The Heilongjiang Key Laboratory of Drug Research, Harbin, 150086, P.R. China
| | - Chunjuan Yang
- College of Pharmacy, Harbin Medical University, Harbin, 150086, P.R. China
| | - Yan Liu
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, The Heilongjiang Key Laboratory of Drug Research, Harbin, 150086, P.R. China
| | - Mengnan Qin
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, The Heilongjiang Key Laboratory of Drug Research, Harbin, 150086, P.R. China
| | - Jiahui Sun
- College of Pharmacy, Harbin Medical University, Harbin, 150086, P.R. China
| | - Gaofeng Liu
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, The Heilongjiang Key Laboratory of Drug Research, Harbin, 150086, P.R. China.
| |
Collapse
|
3
|
Wu ZX, Chen SS, Lu DY, Xue WN, Sun J, Zheng L, Wang YL, Li C, Li YJ, Liu T. Shenxiong glucose injection inhibits oxidative stress and apoptosis to ameliorate isoproterenol-induced myocardial ischemia in rats and improve the function of HUVECs exposed to CoCl 2. Front Pharmacol 2023; 13:931811. [PMID: 36686658 PMCID: PMC9849394 DOI: 10.3389/fphar.2022.931811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
Background: Shenxiong Glucose Injection (SGI) is a traditional Chinese medicine formula composed of ligustrazine hydrochloride and Danshen (Radix et rhizoma Salviae miltiorrhizae; Salvia miltiorrhiza Bunge, Lamiaceae). Our previous studies and others have shown that SGI has excellent therapeutic effects on myocardial ischemia (MI). However, the potential mechanisms of action have yet to be elucidated. This study aimed to explore the molecular mechanism of SGI in MI treatment. Methods: Sprague-Dawley rats were treated with isoproterenol (ISO) to establish the MI model. Electrocardiograms, hemodynamic parameters, echocardiograms, reactive oxygen species (ROS) levels, and serum concentrations of cardiac troponin I (cTnI) and cardiac troponin T (cTnT) were analyzed to explore the protective effect of SGI on MI. In addition, a model of oxidative damage and apoptosis in human umbilical vein endothelial cells (HUVECs) was established using CoCl2. Cell viability, Ca2+ concentration, mitochondrial membrane potential (MMP), apoptosis, intracellular ROS, and cell cycle parameters were detected in the HUVEC model. The expression of apoptosis-related proteins (Bcl-2, Caspase-3, PARP, cytoplasmic and mitochondrial Cyt-c and Bax, and p-ERK1/2) was determined by western blotting, and the expression of cleaved caspase-3 was analyzed by immunofluorescence. Results: SGI significantly reduced ROS production and serum concentrations of cTnI and cTnT, reversed ST-segment elevation, and attenuated the deterioration of left ventricular function in ISO-induced MI rats. In vitro, SGI treatment significantly inhibited intracellular ROS overexpression, Ca2+ influx, MMP disruption, and G2/M arrest in the cell cycle. Additionally, SGI treatment markedly upregulated the expression of anti-apoptotic protein Bcl-2 and downregulated the expression of pro-apoptotic proteins p-ERK1/2, mitochondrial Bax, cytoplasmic Cyt-c, cleaved caspase-3, and PARP. Conclusion: SGI could improve MI by inhibiting the oxidative stress and apoptosis signaling pathways. These findings provide evidence to explain the pharmacological action and underlying molecular mechanisms of SGI in the treatment of MI.
Collapse
Affiliation(s)
- Zhong-Xiu Wu
- State Key Laboratory of Functions and Applications of Medicinal Plants and Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China,School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Shuai-Shuai Chen
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education) and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Ding-Yan Lu
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education) and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Wei-Na Xue
- School of Medicine and Health Management, Guizhou Medical University, Guiyang, China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants and Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Lin Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants and Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Yong-Lin Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants and Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Chun Li
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yong-Jun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education) and State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China,*Correspondence: Yong-Jun Li, ; Ting Liu,
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants and Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China,*Correspondence: Yong-Jun Li, ; Ting Liu,
| |
Collapse
|
4
|
Guo J, Xu Y, Chen LJ, Zhang SX, Liou YL, Chen XP, Tan ZR, Zhou HH, Zhang W, Chen Y. Gut microbiota and host Cyp450s co-contribute to pharmacokinetic variability in mice with non-alcoholic steatohepatitis: Effects vary from drug to drug. J Adv Res 2022; 39:319-332. [PMID: 35777915 PMCID: PMC9263650 DOI: 10.1016/j.jare.2021.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/09/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Drugs’ pharmacokinetics were changed in NASH disease. A systematical research on cocktail drugs in NASH. Gut microbiota can bio-transform some drugs in vitro, and the metabolic rate was different in NASH. The gut microbiota and the host co-contributed the pharmacokinetic variability of drugs in NASH. The degree of influence on pharmacokinetic variability varies from drug to drug.
Introduction Pharmacokinetic variability in disease state is common in clinical practice, but its underlying mechanism remains unclear. Recently, gut microbiota has been considered to be pharmacokinetically equivalent to the host liver. Although some studies have explored the roles of gut microbiota and host Cyp450s in drug pharmacokinetics, few have explored their effects on pharmacokinetic variability, especially in disease states. Objectives In this study, we aim to investigate the effects of gut microbiota and host Cyp450s on pharmacokinetic variability in mice with non-alcoholic steatohepatitis (NASH), and to elucidate the contribution of gut microbiota and host Cyp450s to pharmacokinetic variability in this setting. Methods The pharmacokinetic variability of mice with NASH was explored under intragastric and intravenous administrations of a cocktail mixture of omeprazole, phenacetin, midazolam, tolbutamide, chlorzoxazone, and metoprolol, after which the results were compared with those obtained from the control group. Thereafter, the pharmacokinetic variabilities of all drugs and their relations to the changes in gut microbiota and host Cyp450s were compared and analyzed. Results The exposures of all drugs, except metoprolol, significantly increased in the NASH group under intragastric administration. However, no significant increase in the exposure of all drugs, except tolbutamide, was observed in the NASH group under intravenous administration. The pharmacokinetic variabilities of phenacetin, midazolam, omeprazole, and chlorzoxazone were mainly associated with decreased elimination activity in the gut microbiota. By contrast, the pharmacokinetic variability of tolbutamide was mainly related to the change in the host Cyp2c65. Notably, gut microbiota and host Cyp450s exerted minimal effects on the pharmacokinetic variability of metoprolol. Conclusion Gut microbiota and host Cyp450s co-contribute to the pharmacokinetic variability in mice with NASH, and the degree of contribution varies from drug to drug. The present findings provide new insights into the explanation of pharmacokinetic variability in disease states.
Collapse
Affiliation(s)
- Jing Guo
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Ying Xu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Li-Jie Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Song-Xia Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Yu-Ligh Liou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Zhi-Rong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, China.
| |
Collapse
|
5
|
Zuo HL, Huang HY, Lin YCD, Cai XX, Kong XJ, Luo DL, Zhou YH, Huang HD. Enzyme Activity of Natural Products on Cytochrome P450. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020515. [PMID: 35056827 PMCID: PMC8779343 DOI: 10.3390/molecules27020515] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/27/2022]
Abstract
Drug-metabolizing enzymes, particularly the cytochrome P450 (CYP450) monooxygenases, play a pivotal role in pharmacokinetics. CYP450 enzymes can be affected by various xenobiotic substrates, which will eventually be responsible for most metabolism-based herb–herb or herb–drug interactions, usually involving competition with another drug for the same enzyme binding site. Compounds from herbal or natural products are involved in many scenarios in the context of such interactions. These interactions are decisive both in drug discovery regarding the synergistic effects, and drug application regarding unwanted side effects. Herein, this review was conducted as a comprehensive compilation of the effects of herbal ingredients on CYP450 enzymes. Nearly 500 publications reporting botanicals’ effects on CYP450s were collected and analyzed. The countries focusing on this topic were summarized, the identified herbal ingredients affecting enzyme activity of CYP450s, as well as methods identifying the inhibitory/inducing effects were reviewed. Inhibitory effects of botanicals on CYP450 enzymes may contribute to synergistic effects, such as herbal formulae/prescriptions, or lead to therapeutic failure, or even increase concentrations of conventional medicines causing serious adverse events. Conducting this review may help in metabolism-based drug combination discovery, and in the evaluation of the safety profile of natural products used therapeutically.
Collapse
Affiliation(s)
- Hua-Li Zuo
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- School of Computer Science and Technology, University of Science and Technology of China, Hefei 230027, China
| | - Hsi-Yuan Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yang-Chi-Dung Lin
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Xiao-Xuan Cai
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
| | - Xiang-Jun Kong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China;
| | - Dai-Lin Luo
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
| | - Yu-Heng Zhou
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
| | - Hsien-Da Huang
- School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China; (H.-L.Z.); (H.-Y.H.); (Y.-C.-D.L.); (X.-X.C.); (D.-L.L.); (Y.-H.Z.)
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
- Correspondence: ; Tel.: +86-0755-2351-9601
| |
Collapse
|
6
|
Kwon HC, Sohn H, Kim DH, Jeong CH, Kim DW, Han SG. Effects of Flutriafol Fungicide on the Lipid Accumulation in Human Liver Cells and Rat Liver. Foods 2021; 10:foods10061346. [PMID: 34200939 PMCID: PMC8230498 DOI: 10.3390/foods10061346] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Flutriafol (FTF) is a triazole fungicide that can cause liver toxicity through the ingestion of its residues in food and water. However, little is known about the liver toxicity of FTF, particularly nonalcoholic fatty liver disease (NAFLD) in humans. Therefore, the purpose of this study was to investigate whether FTF induces NAFLD in human liver cells and animal liver. HepG2 cells and Sprague Dawley (SD) rats were treated with FTF at doses of 0–640 µM for 24 h and 0–150 mg/kg bw/day for 28 days, respectively. FTF (80, 160, and 320 µM) treatment to cells induced lipid accumulation. FTF (80 and 160 µM)-treated cells had higher levels of cytochrome P450 enzymes and reactive oxygen species and increased mitochondrial membrane potential loss than the control. FTF also increased the mRNA levels of antioxidant enzymes through oxidative stress and nuclear factor erythroid 2-related factor 2 pathways in HepG2 cells. However, a higher level of FTF (320 µM) induced apoptosis. The treatment of SD rats with FTF (2.5–150 mg/kg bw/day) induced fatty infiltration in the liver by impairing liver metabolism and inducing apoptosis. Therefore, our data suggest that human exposure to FTF residues may be a risk factor for liver diseases, such as NAFLD.
Collapse
Affiliation(s)
- Hyuk-Cheol Kwon
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea; (H.-C.K.); (H.S.); (D.-H.K.)
| | - Hyejin Sohn
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea; (H.-C.K.); (H.S.); (D.-H.K.)
| | - Do-Hyun Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea; (H.-C.K.); (H.S.); (D.-H.K.)
| | - Chang-Hee Jeong
- Microbiology and Functionality Research Group, World Institute of Kimchi, Gwangju 61755, Korea;
| | - Dong-Wook Kim
- Department of Poultry Science, Korea National College of Agriculture and Fisheries, Jeonju 54874, Korea;
| | - Sung-Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea; (H.-C.K.); (H.S.); (D.-H.K.)
- Correspondence:
| |
Collapse
|
7
|
Tu DZ, Mao X, Zhang F, He RJ, Wu JJ, Wu Y, Zhao XH, Zheng J, Ge GB. Reversible and Irreversible Inhibition of Cytochrome P450 Enzymes by Methylophiopogonanone A. Drug Metab Dispos 2021; 49:459-469. [PMID: 33811108 DOI: 10.1124/dmd.120.000325] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Methylophiopogonanone A (MOA), an abundant homoisoflavonoid bearing a methylenedioxyphenyl moiety, is one of the major constituents in the Chinese herb Ophiopogon japonicas This work aims to assess the inhibitory potentials of MOA against cytochrome P450 enzymes and to decipher the molecular mechanisms for P450 inhibition by MOA. The results showed that MOA concentration-dependently inhibited CYP1A, 2C8, 2C9, 2C19, and 3A in human liver microsomes (HLMs) in a reversible way, with IC50 values varying from 1.06 to 3.43 μM. By contrast, MOA time-, concentration-, and NADPH-dependently inhibited CYP2D6 and CYP2E1, along with KI and kinact values of 207 µM and 0.07 minute-1 for CYP2D6, as well as 20.9 µM and 0.03 minutes-1 for CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in an HLM incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate. Additionally, the potential risks of herb-drug interactions triggered by MOA or MOA-related products were also predicted. Collectively, our findings verify that MOA is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. SIGNIFICANCE STATEMENT: Methylophiopogonanone A (MOA), an abundant homoisoflavonoid isolated from the Chinese herb Ophiopogon japonicas, is a reversible inhibitor of CYP1A, 2C8, 2C9, 2C19, and 3A but acts as an inactivator of CYP2D6 and CYP2E1. Further investigations demonstrated that a quinone metabolite of MOA could be trapped by glutathione in a human liver microsome incubation system, and CYP2D6, 1A2, and 2E1 were the major contributors to catalyze the metabolic activation of MOA to the corresponding O-quinone intermediate.
Collapse
Affiliation(s)
- Dong-Zhu Tu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xu Mao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jing-Jing Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Yue Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Xiao-Hua Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Jiang Zheng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China (D.-Z.T., F.Z., R.-J.H., Y.W., X.-H.Z., G.-B.G.); Heilongjiang Key Laboratory of Tissue Damage and Repair, Mudanjiang Medical University, Heilongjiang, China (X.M.); Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China (X.M., J.Z.); Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China (J.-J.W.); and State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang, Guizhou, China (J.Z.)
| |
Collapse
|
8
|
Zhang F, Huang J, He RJ, Wang L, Huo PC, Guan XQ, Fang SQ, Xiang YW, Jia SN, Ge GB. Herb-drug interaction between Styrax and warfarin: Molecular basis and mechanism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 77:153287. [PMID: 32739573 DOI: 10.1016/j.phymed.2020.153287] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Styrax, one of the most famous folk medicines, has been frequently used for the treatment of cardiovascular diseases and skin problems in Asia and Africa. It is unclear whether Styrax or Styrax-related herbal medicines may trigger clinically relevant herb-drug interactions. PURPOSE This study was carried out to investigate the inhibitory effects of Styrax on human cytochrome P450 enzymes (CYPs) and to clarify whether this herb may modulate the pharmacokinetic behavior of the CYP-substrate drug warfarin when co-administered. STUDY DESIGN The inhibitory effects of Styrax on CYPs were assayed in human liver microsomes (HLM), while the pharmacokinetic interactions between Styrax and warfarin were investigated in rats. The bioactive constituents in Styrax with strong CYP3A inhibitory activity were identified and their inhibitory mechanisms were carefully investigated. METHODS The inhibitory effects of Styrax on human CYPs were assayed in vitro, while the pharmacokinetic interactions between Styrax and warfarin were studied in rats. Fingerprinting analysis of Styrax coupled with LC-TOF-MS/MS profiling and CYP inhibition assays were used to identify the constituents with strong CYP3A inhibitory activity. The inhibitory mechanism of oleanonic acid (the most potent CYP3A inhibitor occurring in Styrax) against CYP3A4 was investigated by a panel of inhibition kinetics analyses and in silico analysis. RESULTS In vitro assays demonstrated that Styrax extract strongly inhibited human CYP3A and moderately inhibited six other tested human CYPs, as well as potently inhibited warfarin 10-hydroxylation in liver microsomes from both humans and rats. In vivo assays demonstrated that compared with warfarin given individually in rats, Styrax (100 mg/kg) significantly prolonged the plasma half-life of warfarin by 2.3-fold and increased the AUC(0-inf) of warfarin by 2.7-fold when this herb was co-administrated with warfarin (2 mg/kg) in rats. Two LC fractions were found with strong CYP3A inhibitory activity and the major constituents in these fractions were characterized by LC-TOF-MS/MS. Five pentacyclic triterpenoid acids (including epibetulinic acid, betulinic acid, betulonic acid, oleanonic acid and maslinic acid) present in Styrax were potent CYP3A inhibitors, and oleanonic acid was a competitive inhibitor against CYP3A-mediated testosterone 6β-hydroxylation. CONCLUSION Styrax and the pentacyclic triterpenoid acids occurring in this herb strongly modulate the pharmacokinetic behavior of warfarin via inhibition of CYP3A.
Collapse
Affiliation(s)
- Feng Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jian Huang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Pharmacology and Toxicology Division, Shanghai Institute of Food and Drug Control, Shanghai, China
| | - Rong-Jing He
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peng-Chao Huo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qing Guan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sheng-Quan Fang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China
| | - Yan-Wei Xiang
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shou-Ning Jia
- Qinghai Hospital of Traditional Chinese Medicine, Xining, China
| | - Guang-Bo Ge
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200473, China.
| |
Collapse
|
9
|
Lu D, Zhang Y, Xue W, Sun J, Yang C, Lin C, Li Y, Liu T. Shenxiong Glucose Injection Protects H9c2 Cells From CoCl 2-Induced Oxidative Damage via Antioxidant and Antiapoptotic Pathways. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20920054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease has become one of the main diseases that endanger humans, and oxidative damage plays an important role in this. Shenxiong glucose injection (SGI) is a common clinical treatment in China for the treatment of this condition. To understand further the protective effects and related mechanisms of SGI on cardiovascular diseases, H9c2 cells were treated with SGI at different concentrations (0.5%, 1%, 2% [v/v]) before hypoxic damage was induced by treatment with CoCl2). In CoCl2-induced H9c2 cells, SGI treatment increased cell viability and the activity of superoxide dismutase, glutathione peroxidase, catalase, elevated mitochondrial membrane potential, and decreased the rate of cellular apoptosis, lactic dehydrogenase release, and the content of malondialdehyde and reactive oxygen species, while also upregulating Bcl-2 expression and downregulating Bax, Cyt-c, and cleaved caspase-3 expression. Together, these results suggested that SGI has a protective effect on CoCl2-induced damage, and its mechanism may be related to increased antioxidant and antiapoptosis capacity in H9c2 cells. This study provides the basis for further research and potential practical applications of SGI.
Collapse
Affiliation(s)
- Dingyan Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
- School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Yubin Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
- School of Pharmacy, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Weina Xue
- School of Medicine and Health Management, Guizhou Medical University, Guiyang, Guizhou, P.R. China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Chang Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Changhu Lin
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Yongjun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, Guizhou, P. R. China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants & Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, Guizhou, P. R. China
| |
Collapse
|
10
|
Chaihu-Shugan-San Reinforces CYP3A4 Expression via Pregnane X Receptor in Depressive Treatment of Liver-Qi Stagnation Syndrome. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:9781675. [PMID: 31781287 PMCID: PMC6875207 DOI: 10.1155/2019/9781675] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 07/24/2019] [Accepted: 08/13/2019] [Indexed: 01/11/2023]
Abstract
Backgrounds. Chaihu-Shugan-San (CSS) is a classic traditional Chinese herbal prescription for treating depression. However, the underlying mechanism of the Chinese syndrome-specific efficacy of CSS is poorly understood. Aim of the Study. From traditional Chinese medicine and pharmacogenetics perspectives, the present study aimed to investigate the antidepressant effects of CSS on a mouse model of Liver-Qi Stagnation (LQS) syndrome and its underlying mechanisms. Methods and Materials. We used two main mouse models of depressive syndromes in the study, including LQS and liver stagnation and spleen deficiency (LSSD) syndrome. Tail suspension and forced swimming tests were used to evaluate the effects of CSS on animal behaviour. The expression level of the CYP450 enzyme from liver microsomes was analysed by western blot (WB) analysis and quantitative real-time polymerase chain reaction (qRT-PCR). More specifically, we analysed the key compounds of CSS that are responsible for CYP450 regulation via bioinformatics. Ultimately, luciferase assays were employed to confirm the prediction in vitro. Results. CSS remarkably reduced the immobile time in LQS rather than in LSSD mice. Although CSS significantly upregulated CYP2C9 in mice with both syndromes, activated translation of CYP3A4 induced by CSS was only observed in the LQS group. Bioinformatics analysis revealed that the unique regulation of CYP3A4 was responsible for the effects of glycyrrhetinic acid (GA) from CSS. Further luciferase assays confirmed the enhancement of CYP3A4 expression via the pregnane X receptor (PXR) pathway in vitro. Conclusions. CSS specifically upregulates the translation of CYP3A4 via the PXR pathway in depressed LQS mice. GA, a bioactive compound that originates from CSS, contributes to this activation. This work provides novel insight into Chinese syndrome-based therapy for depression.
Collapse
|
11
|
Effect of Naoxintong Capsules on the Activities of CYP450 and Metabolism of Metoprolol Tartrate in Rats Evaluated by Probe Cocktail and Pharmacokinetic Methods. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:5242605. [PMID: 31662775 PMCID: PMC6778862 DOI: 10.1155/2019/5242605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/28/2019] [Accepted: 09/06/2019] [Indexed: 12/26/2022]
Abstract
Naoxintong capsule (NXT), a prescribed Chinese medicine, has been used clinically for more than 20 years and is widely received by patients. We determined five probe drugs, namely, omeprazole (CYP2C19), midazolam (CYP3A4), phenacetin (CYP1A2), tolbutamide (CYP2C9), and dextromethorphan (CYP2D6) to study the potential influences of NXT on the activities of CYP enzymes and assessed the pharmacokinetics effect of NXT on metoprolol tartrate in rat plasma. The study showed that AUC(0–24) and AUC(0–∞) of midazolam (CYP3A4) in NXT coadministration group (283.7 ± 65.2 h·ng·mL−1 and 292.0 ± 75.1 h·ng·mL−1 in group B; 295.7 ± 62.7 h·ng·mL−1 and 299.5 ± 60.0 h·ng·mL−1 in group C) were significantly decreased as compared to another group (416.8 ± 82.3 h·ng·mL−1 and 424.9 ± 77.9 h·ng·mL−1 in group A), while that of dextromethorphan (CYP2D6) showed an opposite tendency (540.7 ± 119.7 h·ng·mL−1 and 595.3 ± 122.2 h·ng·mL−1 in group A, 760.6 ± 184.9 h·ng·mL−1 and 788.7 ± 211.0 h·ng·mL−1 in group B, and 734.3 ± 118.5 h·ng·mL−1 and 757.2 ± 105.4 h·ng·mL−1 in group C). Moreover, NXT preadministration can enhance the metabolism of metoprolol tartrate and reduce the metabolism of O-demethylmetoprolol. The results indicated that NXT had potential effects in inducing CYP3A4 and inhibiting CYP2D6 in the metabolism of metoprolol tartrate. It suggests that patients who coadministered NXT and metoprolol tartrate should be advised of potential herb-drug interactions (HDIs) to reduce therapeutic failure or accelerated toxicity of conventional drug treatment.
Collapse
|
12
|
Shi Y, Zhang W, Jiang M, Huang L, Zhou Y, Chen J, Liu D, Liu G, Dong M. Effects of sulfotanshinone sodium injection on the pharmacokinetics and pharmacodynamics of warfarin in rats in vivo. Xenobiotica 2019; 50:705-712. [PMID: 31609652 DOI: 10.1080/00498254.2019.1681034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study was to explore the effects of sulfotanshinone sodium injection (SSI) on the pharmacokinetics and pharmacodynamics of warfarin in rats.The studies of single dose and multiple dose of warfarin were designed to assess the interaction between warfarin and SSI. Rats were divided into different groups randomly and administered with warfarin in the absence or presence of SSI. Prothrombin time (PT) and activated partial thromboplastin time (APTT) values were detected by blood coagulation analyzer, and international normalized ratio (INR) values were calculated. Plasma concentrations of warfarin enantiomers were determined by UPLC-MS/MS method, pharmacokinetic parameters were calculated.The single-dose study demonstrated that the repeated doses of SSI alone had no effect on PT, APTT and INR values, but had a significant effect on PT and INR values produced by a single dose of warfarin, APTT values were unaffected. The Cmax, AUC of R-warfarin and S-warfarin were reduced, t1/2 were shortened. The multiple-dose study showed that PT, APTT, INR values, and the Cmax and AUC of R-warfarin and S-warfarin decreased significantly after administration of SSI.The finding implied that SSI could accelerate warfarin metabolism and weaken its anticoagulation. However, human SSI-warfarin interaction studies need to be conducted to confirm this finding.
Collapse
Affiliation(s)
- Yuan Shi
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Wenlong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Meiting Jiang
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Lijun Huang
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yangxu Zhou
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jiayu Chen
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Duo Liu
- Department of Pharmacy, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Gaofeng Liu
- Department of Pharmacy, The Second Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Mei Dong
- Department of Pharmacy, The Third Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Wang F, Wu Y, Zhang J, Wang H, Xie X, Ye X, Peng D, Chen W. Induction of Cytochrome P450 Involved in the Accelerated Blood Clearance Phenomenon Induced by PEGylated Liposomes In Vivo. Drug Metab Dispos 2019; 47:364-376. [PMID: 30674617 DOI: 10.1124/dmd.118.085340] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
Polyethylene glycol (PEG) is recognized as an attractive excipient to modify liposomes due to its extended-circulation properties. Nevertheless, intravenous injection of polyethylene glycol-coated liposomes (PEG-L) usually triggers a rapid systemic clearance of the subsequent dose from blood circulation, which is referred to as an accelerated blood clearance (ABC) phenomenon. Therefore, since the induction of cytochrome P450 (P450) activity may lead to enhanced drug clearance, it motivated us to investigate the possibility of P450 involvement in the ABC phenomenon. In this study, polyethylene glycol-coated liposomal docetaxel was prepared and used to evaluate the magnitude of the ABC phenomenon in rats induced by repeated injection of PEG-modified liposomes. Notably, the ABC phenomenon was observed when the time interval between two doses was from 1 to 7 days, and its magnitude reached the maximum level at 3 days before gradually decreasing the time. Meanwhile, increased activity of CYP3A1, CYP2C6, and CYP1A2 was detected when PEG-L was repeatedly injected in male rats at a 3-day interval. Consistently, the expression levels of hepatic CYP3A1, CYP2C6, and CYP1A2 were also significantly increased in the repeated injection groups and their levels were highest in the 3-day interval group. P450 selective inhibitors confirmed the inhibition of hepatic CYP3A1 was accompanied by an attenuated magnitude of the ABC phenomenon, which strongly suggests that P450s may be induced by repeated injection of PEG-L, thus favoring metabolic clearance of the second dose. Collectively, herein, for the first time we demonstrate that the contribution of P450s should not be ignored in the ABC phenomenon.
Collapse
Affiliation(s)
- Fengling Wang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Yifan Wu
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Jiwen Zhang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Huihui Wang
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Xiaoting Xie
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Xi Ye
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Daiyin Peng
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| | - Weidong Chen
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, Anhui, China (F.W., Y.W., H.W., X.X., X.Y., D.P., W.C.); Department of Pharmacy, The Second People's Hospital of Hefei, Hefei, Anhui, China (F.W., X.Y.); and Center for Drug Delivery Systems, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China (J.Z.)
| |
Collapse
|
14
|
Effect of Gambogenic Acid on Cytochrome P450 1A2, 2B1 and 2E1, and Constitutive Androstane Receptor in Rats. Eur J Drug Metab Pharmacokinet 2018; 43:655-664. [DOI: 10.1007/s13318-018-0477-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|