1
|
Park JD. Metabolism and drug interactions of Korean ginseng based on the pharmacokinetic properties of ginsenosides: Current status and future perspectives. J Ginseng Res 2024; 48:253-265. [PMID: 38707645 PMCID: PMC11068998 DOI: 10.1016/j.jgr.2024.02.003] [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: 01/17/2024] [Revised: 02/21/2024] [Accepted: 02/24/2024] [Indexed: 05/07/2024] Open
Abstract
Orally administered ginsenosides, the major active components of ginseng, have been shown to be biotransformed into a number of metabolites by gastric juice, digestive and bacterial enzymes in the gastrointestinal tract and also in the liver. Attention is brought to pharmacokinetic studies of ginseng that need further clarification to better understand the safety and possible active mechanism for clinical application. Experimental results demonstrated that ginsenoside metabolites play an important role in the pharmacokinetic properties such as drug metabolizing enzymes and drug transporters, thereby can be applied as a metabolic modulator. Very few are known on the possibility of the consistency of detected ginsenosides with real active metabolites if taken the recommended dose of ginseng, but they have been found to act on the pharmacokinetic key factors in any clinical trial, affecting oral bioavailability. Since ginseng is increasingly being taken in a manner more often associated with prescription medicines, ginseng and drug interactions have been also reviewed. Considering the extensive oral administration of ginseng, the aim of this review is to provide a comprehensive overview and perspectives of recent studies on the pharmacokinetic properties of ginsenosides such as deglycosylation, absorption, metabolizing enzymes and transporters, together with ginsenoside and drug interactions.
Collapse
Affiliation(s)
- Jong Dae Park
- R&D Center, REBIO Co., Ltd., Seoul, Republic of Korea
| |
Collapse
|
2
|
Ben-Eltriki M, Shankar G, Tomlinson Guns ES, Deb S. Pharmacokinetics and pharmacodynamics of Rh2 and aPPD ginsenosides in prostate cancer: a drug interaction perspective. Cancer Chemother Pharmacol 2023; 92:419-437. [PMID: 37709921 DOI: 10.1007/s00280-023-04583-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Ginsenoside Rh2 and its aglycon (aPPD) are one of the major metabolites from Panax ginseng. Preclinical studies suggest that Rh2 and aPPD have antitumor effects in prostate cancer (PCa). Our aims in this review are (1) to describe the pharmacokinetic (PK) properties of Rh2 and aPPD ginsenosides; 2) to provide an overview of the preclinical findings on the use of Rh2 and aPPD in the treatment of PCa; and (3) to highlight the mechanisms of its PK and pharmacodynamic (PD) drug interactions. Increasing evidence points to the potential efficacy of Rh2 or aPPD for PCa treatment. Based on the laboratory studies, Rh2 or aPPD combinations revealed an additive or synergistic interaction or enhanced sensitivity of anticancer drugs toward PCa. This review reveals that enhanced anticancer activities were demonstrated in preclinical studies through interactions of Rh2 and/or aPPD with the proteins related to PK (e.g., cytochrome P450 enzymes, transporters) or PD of the other anticancer drugs or PCa signaling pathways. In conclusion, combining Rh2 or aPPD with anti-prostate cancer drugs leads to PK or PD interactions which could facilitate either therapeutically beneficial or toxic effects.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC, Canada.
- Community Pharmacist, Vancouver Area, BC, Canada.
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Gehana Shankar
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
3
|
Hu QR, Hong H, Zhang ZH, Feng H, Luo T, Li J, Deng ZY, Chen F. Methods on improvements of the poor oral bioavailability of ginsenosides: Pre-processing, structural modification, drug combination, and micro- or nano- delivery system. J Ginseng Res 2023; 47:694-705. [PMID: 38107396 PMCID: PMC10721471 DOI: 10.1016/j.jgr.2023.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 12/19/2023] Open
Abstract
Panax ginseng Meyer is a traditional Chinese medicine that is widely used as tonic in Asia. The main pharmacologically active components of ginseng are the dammarane-type ginsenosides, which have been shown to have anti-cancer, anti-inflammatory, immunoregulatory, neuroprotective, and metabolic regulatory activities. Moreover, some of ginsenosides (eg, Rh2 and Rg3) have been developed into nutraceuticals. However, the utilization of ginsenosides in clinic is restrictive due to poor permeability in cells and low bioavailability in human body. Obviously, the dammarane skeleton and glycosyls of ginsenosides are responsible for these limitations. Therefore, improving the oral bioavailability of ginsenosides has become a pressing issue. Here, based on the structures of ginsenosides, we summarized the understanding of the factors affecting the oral bioavailability of ginsenosides, introduced the methods to enhance the oral bioavailability and proposed the future perspectives on improving the oral bioavailability of ginsenosides.
Collapse
Affiliation(s)
- Qi-rui Hu
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Huan Hong
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Zhi-hong Zhang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Hua Feng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| | - Ting Luo
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Ze-yuan Deng
- State Key Laboratory of Food Science and Resources, College of Food Science, Nanchang University, Nanjing East Road, Nanchang, Jiangxi, China
| | - Fang Chen
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Bayi Avenue, Nanchang, Jiangxi, China
| |
Collapse
|
4
|
Xie Y, Wang C. Herb-drug interactions between Panax notoginseng or its biologically active compounds and therapeutic drugs: A comprehensive pharmacodynamic and pharmacokinetic review. JOURNAL OF ETHNOPHARMACOLOGY 2023; 307:116156. [PMID: 36754189 DOI: 10.1016/j.jep.2023.116156] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/24/2022] [Accepted: 01/06/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Herbs, along with the use of herb-drug interactions (HDIs) to combat diseases, are increasing in popularity worldwide. HDIs have two effects: favorable interactions that tend to improve therapeutic outcomes and/or minimize the toxic effects of drugs, and unfavorable interactions aggravating the condition of patients. Panax notoginseng (Burk.) F.H. Chen is a medicinal plant that has long been commonly used in traditional Chinese medicine to reduce swelling, relieve pain, clear blood stasis, and stop bleeding. Numerous studies have demonstrated the existence of intricate pharmacodynamic (PD) and pharmacokinetic (PK) interactions between P. notoginseng and conventional drugs. However, these HDIs have not been systematically summarized. AIM OF THE REVIEW To collect the available literature on the combined applications of P. notoginseng and drugs published from 2005 to 2022 and summarize the molecular mechanisms of interactions to circumvent the potential risks of combination therapy. MATERIALS AND METHODS This work was conducted by searching PubMed, Scopus, Web of Science, and CNKI databases. The search terms included "notoginseng", "Sanqi", "drug interaction," "synergy/synergistic", "combination/combine", "enzyme", "CYP", and "transporter". RESULTS P. notoginseng and its bioactive ingredients interact synergistically with numerous drugs, including anticancer, antiplatelet, and antimicrobial agents, to surmount drug resistance and side effects. This review elaborates on the molecular mechanisms of the PD processed involved. P. notoginseng shapes the PK processes of the absorption, distribution, metabolism, and excretion of other drugs by regulating metabolic enzymes and transporters, mainly cytochrome P450 enzymes and P-glycoprotein. This effect is a red flag for drugs with a narrow therapeutic window. Notably, amphipathic saponins in P. notoginseng act as auxiliary materials in drug delivery systems to enhance drug solubility and absorption and represent a new entry point for studying interactions. CONCLUSION This article provides a comprehensive overview of HDIs by analyzing the results of the in vivo and in vitro studies on P. notoginseng and its bioactive components. The knowledge presented here offers a scientific guideline for investigating the clinical importance of combination therapies. Physicians and patients need information on possible interactions between P. notoginseng and other drugs, and this review can help them make scientific predictions regarding the consequences of combination treatments.
Collapse
Affiliation(s)
- Yujuan Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
5
|
Hong X, Cai Z, Zhou F, Jin X, Wang G, Ouyang B, Zhang J. Improved pharmacokinetics of tenofovir ester prodrugs strengthened the inhibition of HBV replication and the rebalance of hepatocellular metabolism in preclinical models. Front Pharmacol 2022; 13:932934. [PMID: 36105197 PMCID: PMC9465247 DOI: 10.3389/fphar.2022.932934] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
Tenofovir (TFV) ester prodrugs, a class of nucleotide analogs (NAs), are the first-line clinical anti-hepatitis B virus (HBV) drugs with potent antiviral efficacy, low resistance rate and high safety. In this work, three marketed TFV ester drugs, tenofovir disoproxil fumarate (TDF), tenofovir alafenamide fumarate (TAF) and tenofovir amibufenamide fumarate (TMF), were used as probes to investigate the relationships among prodrug structures, pharmacokinetic characteristics, metabolic activations, pharmacological responses and to reveal the key factors of TFV ester prodrug design. The results indicated that TMF and TAF exhibited significantly stronger inhibition of HBV DNA replication than did TDF in HBV-positive HepG2.2.15 cells. The anti-HBV activity of TMF was slightly stronger than TAF after 9 days of treatment (EC50 7.29 ± 0.71 nM vs. 12.17 ± 0.56 nM). Similar results were observed in the HBV decline period post drug administration to the HBV transgenic mouse model, although these three TFV prodrugs finally achieved the same anti-HBV effect after 42 days treatments. Furthermore, TFV ester prodrugs showed a correcting effect on disordered host hepatic biochemical metabolism, including TCA cycle, glycolysis, pentose phosphate pathway, purine/pyrimidine metabolism, amino acid metabolism, ketone body metabolism and phospholipid metabolism. The callback effects of the three TFV ester prodrugs were ranked as TMF > TAF > TDF. These advantages of TMF were believed to be attributed to its greater bioavailability in preclinical animals (SD rats, C57BL/6 mice and beagle dogs) and better target loading, especially in terms of the higher hepatic level of the pharmacologically active metabolite TFV-DP, which was tightly related to anti-HBV efficacy. Further analysis indicated that stability in intestinal fluid determined the actual amount of TFV prodrug at the absorption site, and hepatic/intestinal stability determined the maintenance amount of prodrug in circulation, both of which influenced the oral bioavailability of TFV prodrugs. In conclusion, our research revealed that improved pharmacokinetics of TFV ester prodrugs (especially intestinal stability) strengthened the inhibition of HBV replication and the rebalance of hepatocellular metabolism, which provides new insights and a basis for the design, modification and evaluation of new TFV prodrugs in the future.
Collapse
Affiliation(s)
- Xiaodan Hong
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zuhuan Cai
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiaoliang Jin
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- *Correspondence: Guangji Wang, ; Bingchen Ouyang, ; Jingwei Zhang,
| | - Bingchen Ouyang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- *Correspondence: Guangji Wang, ; Bingchen Ouyang, ; Jingwei Zhang,
| | - Jingwei Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- *Correspondence: Guangji Wang, ; Bingchen Ouyang, ; Jingwei Zhang,
| |
Collapse
|
6
|
Ren HC, Sun JG, A JY, Gu SH, Shi J, Shao F, Ai H, Zhang JW, Peng Y, Yan B, Huang Q, Liu LS, Sai Y, Wang GJ, Yang CG. Mechanism-Based Pharmacokinetic Model for the Deglycosylation Kinetics of 20(S)-Ginsenosides Rh2. Front Pharmacol 2022; 13:804377. [PMID: 35694247 PMCID: PMC9175024 DOI: 10.3389/fphar.2022.804377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Aim: The 20(S)-ginsenoside Rh2 (Rh2) is being developed as a new antitumor drug. However, to date, little is known about the kinetics of its deglycosylation metabolite (protopanoxadiol) (PPD) following Rh2 administration. The aim of this work was to 1) simultaneously characterise the pharmacokinetics of Rh2 and PPD following intravenous and oral Rh2 administration, 2) develop and validate a mechanism-based pharmacokinetic model to describe the deglycosylation kinetics and 3) predict the percentage of Rh2 entering the systemic circulation in PPD form. Methods: Plasma samples were collected from rats after the I.V. or P.O. administration of Rh2. The plasma Rh2 and PPD concentrations were determined using HPLC-MS. The transformation from Rh2 to PPD, its absorption, and elimination were integrated into the mechanism based pharmacokinetic model to describe the pharmacokinetics of Rh2 and PPD simultaneously at 10 mg/kg. The concentration data collected following a 20 mg/kg dose of Rh2 was used for model validation. Results: Following Rh2 administration, PPD exhibited high exposure and atypical double peaks. The model described the abnormal kinetics well and was further validated using external data. A total of 11% of the administered Rh2 was predicted to be transformed into PPD and enter the systemic circulation after I.V. administration, and a total of 20% of Rh2 was predicted to be absorbed into the systemic circulation in PPD form after P.O. administration of Rh2. Conclusion: The developed model provides a useful tool to quantitatively study the deglycosylation kinetics of Rh2 and thus, provides a valuable resource for future pharmacokinetic studies of glycosides with similar deglycosylation metabolism.
Collapse
Affiliation(s)
- Hong-can Ren
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- DMPK and Clinical Pharmacology Group, Hutchison MediPharma Ltd., Shanghai, China
- Department of Biology, GenFleet Therapeutics, Shanghai, China
| | - Jian-guo Sun
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ji-ye A
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| | - Sheng-hua Gu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- School of Pharmacy, Shanghai University of Tranditional Chinese Medicine, Shanghai, China
| | - Jian Shi
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- College of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Feng Shao
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hua Ai
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jing-wei Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ying Peng
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Bei Yan
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Qing Huang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- NMPA Key Laboratory for Impurity Profile of Chemical Drugs, Jiangsu Institute for Food and Drug Control, Nanjing, China
| | - Lin-sheng Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Sai
- DMPK and Clinical Pharmacology Group, Hutchison MediPharma Ltd., Shanghai, China
| | - Guang-ji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| | - Cheng-guang Yang
- Department of General Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Ji-ye A, ; Guang-ji Wang, ; Cheng-guang Yang,
| |
Collapse
|
7
|
Probable Mechanisms of Doxorubicin Antitumor Activity Enhancement by Ginsenoside Rh2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030628. [PMID: 35163891 PMCID: PMC8838402 DOI: 10.3390/molecules27030628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 11/19/2022]
Abstract
Ginsenoside Rh2 increases the efficacy of doxorubicin (DOX) treatment in murine models of solid and ascites Ehrlich’s adenocarcinoma. In a solid tumor model (treatment commencing 7 days after inoculation), DOX + Rh2 co-treatment was significantly more efficacious than DOX alone. If treatment was started 24 h after inoculation, the inhibition of tumor growth of a solid tumor for the DOX + Rh2 co-treatment group was complete. Furthermore, survival in the ascites model was dramatically higher for the DOX + Rh2 co-treatment group than for DOX alone. Mechanisms underlying the combined DOX and Rh2 effects were studied in primary Ehrlich’s adenocarcinoma-derived cells and healthy mice’s splenocytes. Despite the previously established Rh2 pro-oxidant activity, DOX + Rh2 co-treatment revealed no increase in ROS compared to DOX treatment alone. However, DOX + Rh2 treatment was more effective in suppressing Ehrlich adenocarcinoma cell adhesion than either treatment alone. We hypothesize that the benefits of DOX + Rh2 combination treatment are due to the suppression of tumor cell attachment/invasion that might be effective in preventing metastatic spread of tumor cells. Ginsenoside Rh2 was found to be a modest activator in a Neh2-luc reporter assay, suggesting that Rh2 can activate the Nrf2-driven antioxidant program. Rh2-induced direct activation of Nrf2 might provide additional benefits by minimizing DOX toxicity towards non-cancerous cells.
Collapse
|
8
|
Liu Z, Wen X, Wang G, Zhou Y. Involvement of P-gp on Reversing Multidrug Resistance Effects of 23-Hydroxybetulinic Acid on Chemotherapeutic Agents. Front Pharmacol 2022; 12:796745. [PMID: 34975494 PMCID: PMC8714961 DOI: 10.3389/fphar.2021.796745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 11/22/2021] [Indexed: 01/22/2023] Open
Abstract
Betulinic acid (BA) and 23-Hydroxybetulinic acid (23-HBA) are natural products with similar structures, which show a range of biological effects including cytotoxicity activity. The aim of current research was to investigate and evaluate the combinational cytotoxicity of BA and 23-HBA with chemotherapeutic agents in vitro, and to clarify the potential interaction and related mechanism with P-gp. Instead of BA, 23-HBA could increase cytotoxicity of MCF-7/ADR cells to adriamaycin (ADR) and vincristine (VCR). The intracellular accumulation of ADR/VCR in MCF-7/ADR cells was obviously increased in the presence of 23-HBA. Furthermore, 23-HBA could show dose-dependent increase on the transport of VCR and digoxin, which are typical P-gp substrates, in both MDCK-MDR1 and Caco-2 cells. However, the transport of BA and 23-HBA was not influenced by P-gp inhibition in MDCK-MDR1 cells. MDR1 shift assay and molecular docking model suggested that both compounds showed interaction with P-gp, yet the binding affinity and sites are different. In conclusion, 23-HBA could strongly improve the efficacy of anti-tumor agents in multidrug resistance (MDR) cells, which was related to P-gp inhibition. The MDR1 shift assay and molecular docking study further revealed that 23-HBA and BA showed different interaction modes with P-gp.
Collapse
Affiliation(s)
- Zhihao Liu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiaozhou Wen
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China.,Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| | - Ying Zhou
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China.,Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
9
|
Xu JF, Wan Y, Tang F, Chen L, Yang Y, Xia J, Wu JJ, Ao H, Peng C. Emerging Significance of Ginsenosides as Potentially Reversal Agents of Chemoresistance in Cancer Therapy. Front Pharmacol 2022; 12:720474. [PMID: 34975466 PMCID: PMC8719627 DOI: 10.3389/fphar.2021.720474] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 12/03/2021] [Indexed: 12/16/2022] Open
Abstract
Chemoresistance has become a prevalent phenomenon in cancer therapy, which alleviates the effect of chemotherapy and makes it difficult to break the bottleneck of the survival rate of tumor patients. Current approaches for reversing chemoresistance are poorly effective and may cause numerous new problems. Therefore, it is urgent to develop novel and efficient drugs derived from natural non-toxic compounds for the reversal of chemoresistance. Researches in vivo and in vitro suggest that ginsenosides are undoubtedly low-toxic and effective options for the reversal of chemoresistance. The underlying mechanism of reversal of chemoresistance is correlated with inhibition of drug transporters, induction of apoptosis, and modulation of the tumor microenvironment(TME), as well as the modulation of signaling pathways, such as nuclear factor erythroid-2 related factor 2 (NRF2)/AKT, lncRNA cancer susceptibility candidate 2(CASC2)/ protein tyrosine phosphatase gene (PTEN), AKT/ sirtuin1(SIRT1), epidermal growth factor receptor (EGFR)/ phosphatidylinositol 3-kinase (PI3K)/AKT, PI3K/AKT/ mammalian target of rapamycin(mTOR) and nuclear factor-κB (NF-κB). Since the effects and the mechanisms of ginsenosides on chemoresistance reversal have not yet been reviewed, this review summarized comprehensively experimental data in vivo and in vitro to elucidate the functional roles of ginsenosides in chemoresistance reversal and shed light on the future research of ginsenosides.
Collapse
Affiliation(s)
- Jin-Feng Xu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yan Wan
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Tang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Yang
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia Xia
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiao-Jiao Wu
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Ao
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Characteristic Chinese Medicine Resources in Southwest China, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Ginsenoside 20(S)-Rh2 promotes cellular pharmacokinetics and intracellular antibacterial activity of levofloxacin against Staphylococcus aureus through drug efflux inhibition and subcellular stabilization. Acta Pharmacol Sin 2021; 42:1930-1941. [PMID: 34462563 PMCID: PMC8564512 DOI: 10.1038/s41401-021-00751-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/22/2021] [Indexed: 02/07/2023] Open
Abstract
Intracellular Staphylococcus aureus (S. aureus) often causes clinical failure and relapse after antibiotic treatment. We previously found that 20(S)-ginsenoside Rh2 [20(S)-Rh2] enhanced the therapeutic effect of quinolones in a mouse model of peritonitis, which we attributed to the increased concentrations of quinolones within bacteria. In this study, we investigated the enhancing effect of 20(S)-Rh2 on levofloxacin (LVF) from a perspective of intracellular bacteria. In S. aureus 25923-infected mice, coadministration of LVF (1.5 mg/kg, i.v.) and 20(S)-Rh2 (25, 50 mg/kg, i.g.) markedly increased the survival rate, and decreased intracellular bacteria counts accompanied by increased accumulation of LVF in peritoneal macrophages. In addition, 20(S)-Rh2 (1, 5, 10 μM) dose-dependently increased the uptake and accumulation of LVF in peritoneal macrophages from infected mice without drug treatment. In a model of S. aureus 25923-infected THP-1 macrophages, we showed that 20(S)-Rh2 (1, 5, 10 μM) dose-dependently enhanced the intracellular antibacterial activity of LVF. At the cellular level, 20(S)-Rh2 increased the intracellular accumulation of LVF by inhibiting P-gp and BCRP. PK-PD modeling revealed that 20(S)-Rh2 altered the properties of the cell but not LVF. At the subcellular level, 20(S)-Rh2 did not increase the distribution of LVF in lysosomes but exhibited a stronger sensitizing effect in acidic environments. Molecular dynamics (MD) simulations showed that 20(S)-Rh2 improved the stability of the DNA gyrase-LVF complex in lysosome-like acidic conditions. In conclusion, 20(S)-Rh2 promotes the cellular pharmacokinetics and intracellular antibacterial activities of LVF against S. aureus through efflux transporter inhibition and subcellular stabilization, which is beneficial for infection treatment.
Collapse
|
11
|
Truong VL, Jun M, Jeong WS. Phytochemical and Over-The-Counter Drug Interactions: Involvement of Phase I and II Drug-Metabolizing Enzymes and Phase III Transporters. J Med Food 2021; 24:786-805. [PMID: 34382862 DOI: 10.1089/jmf.2021.k.0003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Consumption of plant-derived natural products and over-the-counter (OTC) drugs is increasing on a global scale, and studies of phytochemical-OTC drug interactions are becoming more significant. The intake of dietary plants and herbs rich in phytochemicals may affect drug-metabolizing enzymes (DMEs) and transporters. These effects may lead to alterations in pharmacokinetics and pharmacodynamics of OTC drugs when concomitantly administered. Some phytochemical-drug interactions benefit patients through enhanced efficacy, but many interactions cause adverse effects. This review discusses possible mechanisms of phytochemical-OTC drug interactions mediated by phase I and II DMEs and phase III transporters. In addition, current information is summarized for interactions between phytochemicals derived from fruits, vegetables, and herbs and OTC drugs, and counseling is provided on appropriate and safe use of OTC drugs.
Collapse
Affiliation(s)
- Van-Long Truong
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Korea
| | - Mira Jun
- Brain Busan 21 Plus Program, Department of Food Science and Nutrition, Graduate School, Center for Silver-Targeted Biomaterials, Dong-A University, Busan, Korea
| | - Woo-Sik Jeong
- Food and Bio-Industry Research Institute, School of Food Science and Biotechnology, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Korea
| |
Collapse
|
12
|
Detoxification of toxic herbs in TCM prescription based on modulation of efflux transporters. DIGITAL CHINESE MEDICINE 2021. [DOI: 10.1016/j.dcmed.2021.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
13
|
Yang Y, Hu N, Gao XJ, Li T, Yan ZX, Wang PP, Wei B, Li S, Zhang ZJ, Li SL, Yan R. Dextran sulfate sodium-induced colitis and ginseng intervention altered oral pharmacokinetics of cyclosporine A in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113251. [PMID: 32810615 DOI: 10.1016/j.jep.2020.113251] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/04/2020] [Accepted: 08/04/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Application of cyclosporine A (CsA) as a rescue treatment in acute severe ulcerative colitis (UC) is limited by its narrow therapeutic window and great interpatient variability. As a substrate of cytochrome P450 3A enzyme (CYP3A) and P-glycoprotein (P-gp), the oral pharmacokinetics of CsA is susceptible to disease status and concomitant medications. Combined treatment with ginseng, a famous medicinal herb frequently prescribed for ameliorating abnormal immune response in many diseases including UC, showed immunologic safety in CsA-based immunosuppression. AIM OF THE STUDY Since the therapeutic levels of CsA can be achieved within 24 h, this study first assessed the impact of acute colitis and ginseng intervention on the single oral dose pharmacokinetics of CsA and explored the underlying mechanisms in dextran sulfate sodium (DSS)-induced colitis rats and Caco-2 cells. MATERIALS AND METHODS Rats received drinking water (normal group), 5% DSS (UC group), or 5% DSS plus daily oral ginseng extract (GS+UC group). On day 7, GS+UC group only received an oral dose of CsA (5 mg/kg), while animals of normal or UC group received an oral, intravenous (1.25 mg/kg), or intraperitoneal dose of CsA (1.25 mg/kg), respectively. Blood, liver/intestine tissues and fecal samples were collected for determining CsA and main hydroxylated metabolite HO-CsA or measuring hepatic/intestinal CYP3A activity. Caco-2 cells were incubated with gut microbial culture supernatant (CS) of different groups or ginseng (decoction or polysaccharides), and then CYP3A, P-gp and tight junction (TJ) proteins were determined. RESULTS Oral CsA exhibited enhanced absorption, systemic exposure and tissue accumulation, and lower fecal excretion, while intravenous or intraperitoneal CsA showed lower systemic exposure and enhanced distribution, in colitis rats. Diminished intestinal and hepatic P-gp expression well explained the changes with DSS-induced colitis. Moreover, blood exposures of HO-CsA in both normal and colitis after oral dosing were significantly higher than intravenous/intraperitoneal dosing, supporting the dominant role of intestinal first-pass metabolism. Interestingly, colitis reduced CYP3A expression in intestine and liver but only potentiated intestinal CYP3A activity, causing higher oral systemic exposure of HO-CsA. Oral ginseng mitigated colitis-induced down-regulation of CYP3A and P-gp expression, facilitated HO-CsA production, biliary excretion and colonic sequestration of CsA, while not affected CsA oral systemic exposure. In Caco-2 cells, gut microbial CS from both colitis and GS+UC group diminished P-gp function, while ginseng polysaccharides directly affected ZO-1 distribution and suppressed TJ proteins expression, explaining unaltered oral CsA systemic exposure. CONCLUSIONS DSS-induced colitis significantly altered oral CsA disposition through regulating intestinal and hepatic P-gp and CYP3A. One-week ginseng treatment enhanced colonic accumulation while not altered the systemic exposure of CsA after single oral dosing, indicating pharmacokinetic compatibility between the two medications.
Collapse
Affiliation(s)
- Ying Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Nan Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Xue-Jiao Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Zhi-Xiang Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Pan-Pan Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Bin Wei
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Sai Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China
| | - Zai-Jun Zhang
- Institute of New Drug Research and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Song-Lin Li
- Department of Pharmaceutical Analysis and Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, China
| | - Ru Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao, China; Zhuhai UM Science & Technology Research Institute, Zhuhai, 519080, China.
| |
Collapse
|
14
|
An UPLC-MS/MS Method for Determination of Osimertinib in Rat Plasma: Application to Investigating the Effect of Ginsenoside Rg3 on the Pharmacokinetics of Osimertinib. Int J Anal Chem 2021; 2020:8814214. [PMID: 33456471 PMCID: PMC7785372 DOI: 10.1155/2020/8814214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 11/17/2022] Open
Abstract
Osimertinib is a novel oral, potent, and irreversible epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) for treatment of advanced T790M mutation-positive advanced non-small cell lung cancer, which is commonly combined with ginsenoside Rg3 in clinic to enhance the efficacy and minimize adverse reactions. In the present study, a highly sensitive UPLC-MS/MS method was established and validated for analysis of osimertinib in rat plasma according to US FDA guideline. Separation was performed on a C18 (2.1 × 50 mm, 2.6 μm) column using a gradient elution of ammonium formate (10 mM) with 0.1% formic acid buffer (A) and ACN (B) at a flow rate of 0.2 mL/min. Detection was carried out on a triple quadrupole tandem mass spectrometer equipped with electrospray ionization in the MRM mode. The method was validated over a concentration range of 1-400 ng/mL for osimertinib. The intra- and interday accuracy and precision values were within ±15%. No significant degradation occurred under the experimental conditions in stability assays. There was a further investigation on the effects of multiple doses of ginsenoside Rg3 on the pharmacokinetics of osimertinib in rats for the first time. The results implied that osimertinib exhibited a slow absorption and moderate-rate elimination in rats following oral administration. Coadministeration with ginsenoside Rg3 (5 mg/kg, 7 days, i.g.) may have no effect on the pharmacokinetics of osimertinib in rats. The results provide a reference for the clinical concomitant medications of Rg3 and osimertinib.
Collapse
|
15
|
Yang L, Zhang C, Chen J, Zhang S, Pan G, Xin Y, Lin L, You Z. Shenmai injection suppresses multidrug resistance in MCF-7/ADR cells through the MAPK/NF-κB signalling pathway. PHARMACEUTICAL BIOLOGY 2020; 58:276-285. [PMID: 32251615 PMCID: PMC7170370 DOI: 10.1080/13880209.2020.1742167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Context: Shenmai Injection (SMI) is usually used to treat atherosclerotic coronary heart disease and viral myocarditis in China. However, the effect of SMI on multidrug resistance has not been reported.Objective: To investigate the reversal effect of SMI in adriamycin (ADR) resistant breast cancer cell line (MCF-7/ADR) and explore the related molecular mechanisms.Materials and methods: The effect of SMI (0.25, 0.5, 1 mg/mL) to reverse chemoresistance in MCF-7/ADR cells was elucidated by MTT, HPLC-FLD, DAPI staining, flow cytometric analysis, western blotting. At the same time, in vivo test was conducted to probe into the effect of SMI on reversing ADR resistance, and verapamil (10 μM) was used as a positive control.Results: The results showed that the toxicity of ADR to MCF-7/ADR cells was strengthened significantly after treated with SMI (0.25, 0.5, 1 mg/mL), the IC50 of ADR was decreased 54.4-fold. The intracellular concentrations of ADR were increased 2.2-fold (p < 0.05) and ADR accumulation was enhanced in the nuclei (p < 0.05). SMI could strongly enhance the ADR-induced apoptosis and increase intracellular rhodamine 123 accumulation in MCF-7/ADR cells. Additionally, a combination of ADR and SMI (5 mg/kg) could dramatically reduce the weight and volume of tumour (p < 0.05). Furthermore, the results revealed that SMI might reverse MDR via inhibiting ADR-induced activation of the mitogen-activated protein kinase/nuclear factor (NF)-κB pathway to down-regulated the expression of P-glycoprotein (P-gp).Discussion and conclusions: SMI could potentially be used to treat ADR-resistance. This suggests possibilities for future clinical research.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/metabolism
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Down-Regulation/drug effects
- Doxorubicin/metabolism
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Combinations
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Drugs, Chinese Herbal/administration & dosage
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Female
- Humans
- MAP Kinase Signaling System/drug effects
- MCF-7 Cells
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- NF-kappa B/metabolism
- Rhodamine 123/metabolism
- Signal Transduction/drug effects
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lin Yang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Chengda Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Jiaoting Chen
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
| | - Sheng Zhang
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Guixuan Pan
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Yanfei Xin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, China
- Yanfei Xin
| | - Lin Lin
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- Lin Lin Zhejiang Academy of Medical Sciences, 182 Tianmushan Road, Hangzhou310013, China
| | - Zhenqiang You
- Zhejiang Academy of Medical Sciences, Hangzhou Medical College, Hangzhou, China
- CONTACT Zhenqiang You
| |
Collapse
|
16
|
Domínguez CJ, Tocchetti GN, Rigalli JP, Mottino AD. Acute regulation of apical ABC transporters in the gut. Potential influence on drug bioavailability. Pharmacol Res 2020; 163:105251. [PMID: 33065282 DOI: 10.1016/j.phrs.2020.105251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/07/2020] [Accepted: 10/08/2020] [Indexed: 01/09/2023]
Abstract
The extensive intestinal surface offers an advantage regarding nutrient, ion and water absorptive capacity but also brings along a high exposition to xenobiotics, including drugs of therapeutic use and food contaminants. After absorption of these compounds by the enterocytes, apical ABC transporters play a key role in secreting them back to the intestinal lumen, hence acting as a transcellular barrier. Rapid and reversible modulation of their activity is a subject of increasing interest for pharmacologists. On the one hand, a decrease in transporter activity may result in increased absorption of therapeutic agents given orally. On the other hand, an increase in transporter activity would decrease their absorption and therapeutic efficacy. Although of less relevance, apical ABC transporters also contribute to disposition of drugs systemically administered. This review article summarizes the present knowledge on the mechanisms aimed to rapidly regulate the activity of the main apical ABC transporters of the gut: multidrug resistance protein 1 (MDR1), multidrug resistance-associated protein 2 (MRP2) and breast cancer resistance protein (BCRP). Regulation of these mechanisms by drugs, drug delivery systems, drug excipients and nutritional components are particularly considered. This information could provide the basis for controlled regulation of bioavailability of therapeutic agents and at the same time would help to prevent potential drug-drug interactions.
Collapse
Affiliation(s)
- Camila Juliana Domínguez
- Institute of Experimental Physiology, Faculty of Biochemical and Pharmaceutical Sciences, Rosario National University, Suipacha 570, 2000 Rosario, Argentina
| | - Guillermo Nicolás Tocchetti
- Institute of Experimental Physiology, Faculty of Biochemical and Pharmaceutical Sciences, Rosario National University, Suipacha 570, 2000 Rosario, Argentina; Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Juan Pablo Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany; Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, the Netherlands
| | - Aldo Domingo Mottino
- Institute of Experimental Physiology, Faculty of Biochemical and Pharmaceutical Sciences, Rosario National University, Suipacha 570, 2000 Rosario, Argentina.
| |
Collapse
|
17
|
Suroowan S, Mahomoodally MF. Herbal Medicine of the 21st Century: A Focus on the Chemistry, Pharmacokinetics and Toxicity of Five Widely Advocated Phytotherapies. Curr Top Med Chem 2020; 19:2718-2738. [PMID: 31721714 DOI: 10.2174/1568026619666191112121330] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/02/2019] [Accepted: 09/25/2019] [Indexed: 12/25/2022]
Abstract
Widely advocated for their health benefits worldwide, herbal medicines (HMs) have evolved into a billion dollar generating industry. Much is known regarding their wellness inducing properties, prophylactic and therapeutic benefits for the relief of both minor to chronic ailment conditions given their long-standing use among various cultures worldwide. On the other hand, their equally meaningful chemistry, pharmacokinetic profile in humans, interaction and toxicity profile have been poorly researched and documented. Consequently, this review is an attempt to highlight the health benefits, pharmacokinetics, interaction, and toxicity profile of five globally famous HMs. A systematic literature search was conducted by browsing major scientific databases such as Bentham Science, SciFinder, ScienceDirect, PubMed, Google Scholar and EBSCO to include 196 articles. In general, ginsenosides, glycyrrhizin and curcumin demonstrate low bioavailability when orally administered. Ginkgo biloba L. induces both CYP3A4 and CYP2C9 and alters the AUC and Cmax of conventional medications including midazolam, tolbutamide, lopinavir and nifedipine. Ginsenosides Re stimulates CYP2C9, decreasing the anticoagulant activity of warfarin. Camellia sinensis (L.) Kuntze increases the bioavailability of buspirone and is rich in vitamin K thereby inhibiting the activity of anticoagulant agents. Glycyrrhiza glabra L. displaces serum bound cardiovascular drugs such as diltiazem, nifedipine and verapamil. Herbal medicine can directly affect hepatocytes leading to hepatoxicity based on both intrinsic and extrinsic factors. The potentiation of the activity of concurrently administered conventional agents is potentially lethal especially if the drugs bear dangerous side effects and have a low therapeutic window.
Collapse
Affiliation(s)
- S Suroowan
- Department of Health Sciences, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - M F Mahomoodally
- Department of Health Sciences, Faculty of Science, University of Mauritius, Reduit, Mauritius.,Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
18
|
Liu J, Cai Q, Wang W, Lu M, Liu J, Zhou F, Sun M, Wang G, Zhang J. Ginsenoside Rh2 pretreatment and withdrawal reactivated the pentose phosphate pathway to ameliorate intracellular redox disturbance and promoted intratumoral penetration of adriamycin. Redox Biol 2020; 32:101452. [PMID: 32067911 PMCID: PMC7264470 DOI: 10.1016/j.redox.2020.101452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/19/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Improving the limited penetration, accumulation and therapeutic effects of antitumor drugs in the avascular region of the tumor mass is crucial during chemotherapy. P-gp inhibitors have achieved little success despite significant efforts. Excessive P-gp inhibition disturbed the kinetic balance between intracellular accumulation and intercellular penetration, thus resulting in a more inhomogeneous distribution of substrate drugs. Here, we found that ginsenoside Rh2 pretreatment mildly downregulated P-gp expression through reactivating the pentose phosphate pathway and rebalancing redox status. This mild P-gp inhibition not only significantly increased the growth inhibition effect and accumulation profile of adriamycin (ADR) throughout the multicellular tumor spheroid (MCTS) but also had unique advantages in improving drug penetration. Furthermore, we developed a novel individual-cell-based PK-PD integrated model and proved that metabolic reprogramming and redox rebalancing-based P-gp regulation was sufficient to increase the ADR effect in both central and peripheral cells of MCTS. Thus, a “ginsenoside Rh2-ADR” sequential regimen was proposed and exhibited a potent antitumor effect in vivo. This novel P-gp inhibition via metabolic reprogramming and redox rebalancing provided a new idea for achieving better antitumor effects in the tumor avascular region during chemotherapy. Rh2 pretreatment downregulated P-gp expression through metabolic reprogramming and redox rebalancing. Rh2-pretreatment improved ADR penetration into the core of MCTS and tumour mass. “Ginsenoside Rh2-ADR” sequential regimen exhibited potent antitumor effects in vivo.
Collapse
Affiliation(s)
- Jiali Liu
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qingyun Cai
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenjie Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meng Lu
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jianming Liu
- Clinical Pharmacology Institute, Nanchang University, Nanchang, China
| | - Fang Zhou
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Minjie Sun
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Jingwei Zhang
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Discovery and synthesis of 3- and 21-substituted fusidic acid derivatives as reversal agents of P-glycoprotein-mediated multidrug resistance. Eur J Med Chem 2019; 182:111668. [DOI: 10.1016/j.ejmech.2019.111668] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 08/30/2019] [Accepted: 08/30/2019] [Indexed: 12/30/2022]
|
20
|
Intravenous formulation of Panax notoginseng root extract: human pharmacokinetics of ginsenosides and potential for perpetrating drug interactions. Acta Pharmacol Sin 2019; 40:1351-1363. [PMID: 31358899 DOI: 10.1038/s41401-019-0273-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/11/2019] [Indexed: 12/12/2022] Open
Abstract
XueShuanTong, a lyophilized extract of Panax notoginseng roots (Sanqi) for intravenous administration, is extensively used as add-on therapy in the treatment of ischemic heart and cerebrovascular diseases and comprises therapeutically active ginsenosides. Potential for XueShuanTong-drug interactions was determined; the investigation focused on cytochrome P450 (CYP)3A induction and organic anion-transporting polypeptide (OATP)1B inhibition. Ginsenosides considerably bioavailable for drug interactions were identified by dosing XueShuanTong in human subjects and their interaction-related pharmacokinetics were determined. The CYP3A induction potential was determined by repeatedly dosing XueShuanTong for 15 days in human subjects and by treating cryopreserved human hepatocytes with circulating ginsenosides; midazolam served as a probe substrate. Joint inhibition of OATP1B by XueShuanTong ginsenosides was assessed in vitro, and the data were processed using the Chou-Talalay method. Samples were analyzed by liquid chromatography/mass spectrometry. Ginsenosides Rb1, Rd, and Rg1 and notoginsenoside R1 were the major circulating XueShuanTong compounds; their interaction-related pharmacokinetics comprised compound dose-dependent levels of systemic exposure and, for ginsenosides Rb1 and Rd, long terminal half-lives (32‒57 and 58‒307 h, respectively) and low unbound fractions in plasma (0.8%‒2.9% and 0.4%‒3.0%, respectively). Dosing XueShuanTong did not induce CYP3A. Based on the pharmacokinetics and inhibitory potency of the ginsenosides, XueShuanTong was predicted to have high potential for OATP1B3-mediated drug interactions (attributed chiefly to ginsenoside Rb1) suggesting the need for further model-based determination of the interaction potential for XueShuanTong and, if necessary, a clinical drug interaction study. Increased awareness of ginsenosides' pharmacokinetics and XueShuanTong-drug interaction potential will help ensure the safe use of XueShuanTong and coadministered synthetic drugs.
Collapse
|
21
|
Xia Y, Sun M, Li R, Liu Y. Simultaneous determination of eight bioactive components in rat plasma after oral administration of Yan-Ke-Ning-Tablet by liquid chromatography coupled with Q-Exactive Orbitrap tandem mass spectrometry. Biomed Chromatogr 2019; 33:e4519. [PMID: 30807650 DOI: 10.1002/bmc.4519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/21/2019] [Indexed: 11/10/2022]
Abstract
A rapid, sensitive and reliable quantitative method based on ultra-high performance liquid chromatography coupled with Q-Exactive Orbitrap tandem mass spectrometry was developed for simultaneous determination of berberine, berberrubine, palmatine, jatrorrhizine, columbamine, baicalin, baicalein and wogonin in rat plasma after oral administration with Yan-Ke-Ning-Tablet (YKNT). After precipitation with acetonitrile, the plasma samples were separated on a reverse-phase C18 column with 1 mm ammonium acetate containing 0.2% acetic acid-acetonitrile as mobile phase. Calibration curves showed good linearity (r > 0.9983) over the tested concentration ranges of 0.5-200 ng/mL for berberine, berberrubine, palmatine, jatrorrhizine and columbamine, and 1-300 ng/mL for baicalin, baicalein and wogonin. The precision (relative standard deviation) at three different concentration levels was <12.15% and the accuracy (relative error) ranged from -8.24 to 10.85%. No matrix effects were observed with matrix effect value ranging from 89.23 to 107.68%. The extraction recovery was in the range of 82.34-92.31%. The validated assay was further successfully applied to the pharmacokinetic study of these components after oral administration of YKNT. The present study provides the pharmacokinetic profiles of major bioactive components found in YKNT, and provides valuable information regarding the chemical components that were absorbed into plasma, which will be helpful for understanding the therapeutic effects of YKNT.
Collapse
Affiliation(s)
- Yu Xia
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| | - Mingyang Sun
- Department of Clinical Pharmacy, Hankun Hospital, Beijing, China
| | - Ren Li
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yue Liu
- Department of Pharmacy, Peking University People's Hospital, Beijing, China
| |
Collapse
|
22
|
Zhan T, Yao N, Wu L, Lu Y, Liu M, Liu F, Xiong Y, Xia C. The major effective components in Shengmai Formula interact with sodium taurocholate co-transporting polypeptide. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 59:152916. [PMID: 30978651 DOI: 10.1016/j.phymed.2019.152916] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/27/2019] [Accepted: 04/02/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Shengmai Formula (SMF) is widely used to treat cardiovascular disease such as chronic heart disease, coronary atherosclerotic heart disease, viral myocarditis, and others. Our previous studies have shown that OATP1B1/1B3 mediates the interactions between ophiopogon D and ginsenoside Rb1/Rd, which are the major active components in SMF. The herb-drug interactions that involve sodium taurocholate co-transporting polypeptide (NTCP) have been drawing increasing amounts of attention. PURPOSE The aim of the present study was to investigate the interactions of the major effective components in SMF mediated by NTCP. METHODS By using NTCP-overexpressing HEK293T cells and liquid chromatograph-mass spectrometer (LC-MS) analytical methods, we investigated the impact of the four main effective fractions and the 12 main effective components in SMF on NTCP-mediated sodium taurocholate (TCNa) uptake. The interactions of these effective components in SMF mediated by NTCP were further studied. RESULTS The main effective fractions, ginseng total saponins (GTS), ophiopogon total saponins (OTS), ophiopogon total flavonoids (OTF), and fructus schisandrae total lignans (STL), all exhibited a certain inhibitory effect on the uptake of TCNa. Among the 12 main effective components, only ginsenoside Rg1, ophiopogon D', and schizandrin A showed inhibition of TCNa uptake, with IC50 values of 50.49 ± 4.24 μM, 6.71 ± 0.70 μM, and 45.80 ± 3.10 μM, respectively. Additionally, we found that ginsenoside Re and schizandrin B could be transported by NTCP-overexpressing HEK293T cells, and that the uptake of ginsenoside Re was significantly inhibited by OTS, OTF, STL, ginsenoside Rg1, ophiopogon D', and schizandrin A. The uptake of schizandrin B was significantly inhibited by GTS, OTS, OTF, and ophiopogon D'. CONCLUSION Ginsenoside Rg1, ophiopogon D', and schizandrin A are potential inhibitors of NTCP and may interact with clinical drugs mediated by NTCP. Ginsenoside Re and schizandrin B are also potential substrates of NTCP, and their uptake mediated by NTCP was inhibited by the other components in SMF. The interaction of complex components based on NTCP may be one of the important compatibility mechanisms in SMF.
Collapse
Affiliation(s)
- Tao Zhan
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Na Yao
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Lingna Wu
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Yanli Lu
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Mingyi Liu
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Fanglan Liu
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Yuqing Xiong
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China
| | - Chunhua Xia
- Clinical Pharmacology Institute, Nanchang University, Bayi road 461#, Nanchang 330006, PR China.
| |
Collapse
|
23
|
Ma J, Gao G, Lu H, Fang D, Li L, Wei G, Chen A, Yang Y, Zhang H, Huo J. Reversal effect of ginsenoside Rh2 on oxaliplatin-resistant colon cancer cells and its mechanism. Exp Ther Med 2019; 18:630-636. [PMID: 31258699 PMCID: PMC6566025 DOI: 10.3892/etm.2019.7604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 04/18/2019] [Indexed: 01/28/2023] Open
Abstract
Chemotherapy is an important treatment modality for colon cancer, however, drug resistance is the main factor leading to treatment failure. Ginsenoside Rh2 (G-Rh2), the main bioactive metabolite of ginseng, is known to possess the ability to potently induce cell apoptosis, inhibit cell proliferation and reverse multidrug resistance in a variety of cancer cells. The present study examined the effect of G-Rh2 on oxaliplatin (L-OHP)-resistant colon cancer cells and its potential mechanism. L-OHP-resistant colon cancer cells (LoVo/L-OHP) and LoVo cells were used in the present study. The effect of G-Rh2 on LoVo/L-OHP and LoVo cell proliferation was measured using a 3-(4,5 dimethylthiazol-z-yl)-3,5-diphenyltetrazolium bromide assay. The effects of G-Rh2 on LoVo/L-OHP and LoVo cell apoptosis were detected by flow cytometry. The mRNA and protein expression of apoptosis-related genes Bax, Bcl-2 and caspase-3, drug resistance-related genes P-glycoprotein (P-gp) and Smad4, were determined in LoVo/L-OHP and LoVo cells treated with G-Rh2 by reverse transcription-quantitative polymerase chain reaction and western blot analyses. G-Rh2 treatment significantly inhibited the proliferation and induced the apoptosis of LoVo/L-OHP and LoVo cells. In addition, G-Rh2 treatment resulted in a significant increase in pro-apoptotic factors, Bax and caspase-3, and decrease in anti-apoptotic factor Bcl-2 in the LoVo/L-OHP and LoVo cells. Furthermore, G-Rh2 treatment significantly decreased the levels of P-gp and increased the levels of Smad4 in the LoVo/L-OHP and LoVo cells. It was found that L-OHP had no significant effects on LoVo/L-OHP cell proliferation or apoptosis, whereas G-Rh2 + L-OHP treatment significantly inhibited LoVo/L-OHP cell proliferation and induced apoptosis. L-OHP had no significant effects on the expression of P-gp, Smad4, Bcl-2, Bax or caspase-3 in LoVo/L-OHP cells. Treatment with G-Rh2 + L-OHP significantly reduced the expression of P-gp and Bcl-2, and enhanced the expression levels of Smad4, Bax and caspase-3. These findings demonstrated that G-Rh2 reversed the drug resistance of LoVo/L-OHP cells to L-OHP, and this may be mediated by inhibiting cell proliferation and promoting apoptosis and regulating the expression of drug resistance genes. These results suggest that G-Rh2 may function as a potent anticancer drug for drug resistance in colon cancer treatment.
Collapse
Affiliation(s)
- Jun Ma
- Department of Oncology, Huai'an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Guangyi Gao
- Department of Traditional Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Hong Lu
- Department of Oncology, Changshu No. 1 People's Hospital, Changshu, Jiangsu 215500, P.R. China
| | - Dong Fang
- Department of Oncology, Zhenjiang Hospital of Integrated Traditional and Western Medicine, Zhenjiang, Jiangsu 212000, P.R. China
| | - Lingchang Li
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| | - Guoli Wei
- Department of Oncology, Huai'an TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu 223001, P.R. China
| | - Aifei Chen
- Department of Oncology, Huai'an Hospital of Chinese Medicine, Huai'an, Jiangsu 223001, P.R. China
| | - Yong Yang
- Department of Oncology, Huai'an Hospital of Chinese Medicine, Huai'an, Jiangsu 223001, P.R. China
| | - Hongying Zhang
- Department of Oncology, Huai'an Hospital of Chinese Medicine, Huai'an, Jiangsu 223001, P.R. China
| | - Jiege Huo
- Department of Oncology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210028, P.R. China
| |
Collapse
|
24
|
Impact of Curcuma longa extract on the expression level of brain transporters in in vivo model. HERBA POLONICA 2019. [DOI: 10.2478/hepo-2019-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Summary
Introduction: Blood brain barrier and multidrug resistance phenomenon are subjects of many investigations. Mainly, because of their functions in protecting the central nervous system (CNS) by blocking the delivery of toxic substances to the brain. This special function has some disadvantages, like drug delivery to the brain in neurodegenerative diseases
Objective: The aim of this study was to examine how natural and synthetic substances affect the expression levels of genes (Mdr1a, Mdr1b, Mrp1, Mrp2, Oatp1a4, Oatp1a5 and Oatp1c1) that encode transporters in the blood-brain barrier.
Methods: cDNA was synthesized from total RNA isolated from rat hippocampus. The expression level of genes was determined using real-time PCR (RT-PCR) method.
Results: Our findings showed that verapamil, as a synthetic substance, caused the greatest reduction of mRNA level of genes studied. The standardized extract of Curcuma longa reduced the expression level for Mrp1 and Mrp2, whereas the increase of mRNA level was observed for Mdr1b, Oatp1a5 and Oatp1c1.
Conclusions: These results suggests that herbal extracts may play an important role in overcoming the blood brain barrier during pharmacotherapy.
Collapse
|
25
|
Natural products in licorice for the therapy of liver diseases: Progress and future opportunities. Pharmacol Res 2019; 144:210-226. [PMID: 31022523 DOI: 10.1016/j.phrs.2019.04.025] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/18/2019] [Accepted: 04/21/2019] [Indexed: 12/16/2022]
Abstract
Liver diseases related complications represent a significant source of morbidity and mortality worldwide, creating a substantial economic burden. Oxidative stress, excessive inflammation, and dysregulated energy metabolism significantly contributed to liver diseases. Therefore, discovery of novel therapeutic drugs for the treatment of liver diseases are urgently required. Licorice is one of the most commonly used herbal drugs in Traditional Chinese Medicine for the treatment of liver diseases and drug-induced liver injury (DILI). Various bioactive components have been isolated and identified from the licorice, including glycyrrhizin, glycyrrhetinic acid, liquiritigenin, Isoliquiritigenin, licochalcone A, and glycycoumarin. Emerging evidence suggested that these natural products relieved liver diseases and prevented DILI through multi-targeting therapeutic mechanisms, including anti-steatosis, anti-oxidative stress, anti-inflammation, immunoregulation, anti-fibrosis, anti-cancer, and drug-drug interactions. In the current review, we summarized the recent progress in the research of hepatoprotective and toxic effects of different licorice-derived bioactive ingredients and also highlighted the potency of these compounds as promising therapeutic options for the treatment of liver diseases and DILI. We also outlined the networks of underlying molecular signaling pathways. Further pharmacology and toxicology research will contribute to the development of natural products in licorice and their derivatives as medicines with alluring prospect in the clinical application.
Collapse
|
26
|
Bai J, Zhao S, Fan X, Chen Y, Zou X, Hu M, Wang B, Jin J, Wang X, Hu J, Zhang D, Li Y. Inhibitory effects of flavonoids on P-glycoprotein in vitro and in vivo: Food/herb-drug interactions and structure–activity relationships. Toxicol Appl Pharmacol 2019; 369:49-59. [DOI: 10.1016/j.taap.2019.02.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 12/24/2022]
|
27
|
Ma C, Guan H, Ju Z, Li S, Deng G, Zhang Y, Lin Q, Cheng X, Yang L, Wang Z, Wang C. Identification and characterization of forced degradation products and stability-indicating assay for notoginsenosidefc by using UHPLC-Q-TOF-MS and UHPLC-MS/MS: Insights into stability profile and degradation pathways. J Sep Sci 2019; 42:1550-1563. [DOI: 10.1002/jssc.201801295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 02/05/2019] [Accepted: 02/10/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Chao Ma
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Huida Guan
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Zhengcai Ju
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Shuping Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Gang Deng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Yunpeng Zhang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Qiyan Lin
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai P. R. China
| | - Li Yang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai P. R. China
| | - Zhengtao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai P. R. China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine; The MOE Key Laboratory for Standardization of Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine; Shanghai P. R. China
- Shanghai R&D Centre for Standardization of Chinese Medicines; Shanghai P. R. China
| |
Collapse
|
28
|
Bi X, Yuan Z, Qu B, Zhou H, Liu Z, Xie Y. Piperine enhances the bioavailability of silybin via inhibition of efflux transporters BCRP and MRP2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:98-108. [PMID: 30668388 DOI: 10.1016/j.phymed.2018.09.217] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/06/2018] [Accepted: 09/25/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Although silybin serves as a well-known hepatoprotective agent with prominent anti-inflammatory, anti-oxidant and anti-fibrotic activities, its low bioavailability limits its application in the treatment of chronic liver diseases. However, novel formulation products with increased solubility were not sufficient to achieve pharmacologically meaningful concentrations of silybin in the clinical studies even used at high dosage. HYPOTHESIS/PURPOSE We hypothesized that inhibiting efflux transporter(s) and/or glucuronidation by piperine might enhance the bioavailability and efficacy of silybin. METHODS Pharmacokinetics of silybin given alone or in-combination with piperine was determined by a validated LC-MS method. A CCl4 induced rat model of liver injury was prepared and verified for comparing the effects of silybin and combination treatment. To investigate the underlying mechanism, the inhibition effects of piperine on transportation of silybin were performed in Caco-2 and transfected MDCKII cell lines as well as sandwich-cultured rat hepatocytes (SCH). Human liver microsomes incubation was used for exploring the modulation effects of piperine on the phase-2 metabolism of silybin. RESULTS In the present study, we demonstrated for the first time that piperine as a bioenhancer increased the bioavailability of silybin (146%- 181%), contributing to a boosted therapeutic effect in CCl4-induced acute liver-injury rat model. The underlying mechanisms involved that piperine enhanced the absorption of silybin by inhibiting the efflux transporters including MRP2 and BCRP but not MDR1 in Caco-2 and transfected MDCKII cell lines. Moreover, piperine could inhibit the biliary excretion of silybin and conjugated metabolites in sandwich-cultured rat hepatocytes. Notably, we found that piperine did not affect the phase-2 metabolism of silybin. CONCLUSION Efflux transporters play an important role in the pharmacokinetic behavior of flavolignans, and modulating these transporters by bioenhancer such as piperine could enhance the in vivo absorption of silybin, leading to more effective treatments.
Collapse
Affiliation(s)
- Xiaoli Bi
- State Key Laboratory for Quality Research of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau (SAR), China; Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Zhongwen Yuan
- Department of Pharmacy, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong Province 510150, China
| | - Biao Qu
- State Key Laboratory for Quality Research of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau (SAR), China
| | - Hua Zhou
- State Key Laboratory for Quality Research of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau (SAR), China
| | - Zhongqiu Liu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, School of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Ying Xie
- State Key Laboratory for Quality Research of Chinese Medicines, Macau University of Science and Technology, Taipa, Macau (SAR), China.
| |
Collapse
|
29
|
Increase in P-glycoprotein levels in the blood-brain barrier of partial portal vein ligation /chronic hyperammonemia rats is medicated by ammonia/reactive oxygen species/ERK1/2 activation: In vitro and in vivo studies. Eur J Pharmacol 2019; 846:119-127. [PMID: 30639310 DOI: 10.1016/j.ejphar.2019.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
Liver failure altered P-glycoprotein (P-gp) function and expression at blood-brain barrier (BBB), partly owing to hyperammonemia. We aimed to examine the effects of partial portal vein ligation (PVL) plus chronic hyperammonemia (CHA) on P-gp function and expression at rat BBB. Experimental rats included sham-operation (SH), PVL, CHA and PVL+CHA. The PVL+CHA rats were developed by ammonia-containing diet for 2 weeks after operation. The brain-to-plasma concentration ratios (Kp) and apparent unidirectional influx constants (Kin) of rhodamine123 and sodium fluorescein were measured to assess function of P-gp and BBB integrity, respectively. Human cerebral microvascular endothelial cells (HCMEC/D3) were used to assess effects of ammonia on P-gp expression and function. It was found that PVL+CHA significantly decreased Kp and Kin of rhodamine123 without affecting brain distribution of fluorescein. The P-gp expressions in membrane protein in cortex and hippocampus were significantly increased in CHA and PVL +CHA rats, especially in PVL + CHA rats, while remarkably increased phosphorylated ERK1/2 was only found in PVL +CHA rats. Expressions of tight junction proteins claudin-5 and occluding in rat brain remained unchanged. In vitro data showed that NH4Cl increased reactive oxygen species, membrane expression and function of P-gp as well as phosphorylated ERK1/2 levels in HCMEC/D3. The NH4Cl-induced alterations were reversed by reactive oxygen species scavenger N-acetylcysteine and ERK1/2 inhibitor U0126. In conclusion, PVL+CHA increased function and membrane translocation of P-gp at rat BBB partly via ammonia. Reactive oxygen species/ERK1/2 pathway activation may be one of the reasons that ammonia upregulated P-gp expression and function at BBB.
Collapse
|
30
|
Dunkoksung W, Vardhanabhuti N, Jianmongkol S. Potential P-glycoprotein-mediated herb-drug interaction of phyllanthin at the intestinal absorptive barrier. J Pharm Pharmacol 2018; 71:213-219. [DOI: 10.1111/jphp.13019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 09/02/2018] [Indexed: 01/14/2023]
Abstract
Abstract
Objectives
This study investigated the absorptive potential of phyllanthin across the polarized Caco-2 monolayers and the potential role of phyllanthin in P-glycoprotein (P-gp)-mediated drug interaction.
Methods
The absorptive potential of phyllanthin was predicted from its apparent permeability (Papp) across the Caco-2 monolayers under the pH gradient condition (pH 6.5AP–7.4BL) at 37°C. Integrity of paracellular transport was assessed by monitoring transepithelial electrical resistance (TEER) and lucifer yellow (LY) leakage. P-gp-mediated interaction was evaluated by transport studies of phyllanthin and rhodamine-123.
Key findings
The absorptive Papp of phyllanthin (34.90 ± 1.18 × 10−6 cm/s) was in the same rank order as the high permeable theophylline and antipyrine. Phyllanthin transport in the absorptive and secretive directions was comparable (the efflux ratio (ER) of 1.19 ± 0.01). Phyllanthin caused no changes in TEER nor LY leakage in the monolayers. However, phyllanthin increased rhodamine-123 ER in a concentration-dependent manner, suggesting its inhibition on P-gp function. In addition, phyllanthin aqueous solubility was <5 μg/ml at 37°C.
Conclusions
Phyllanthin is a highly permeable compound that could passively diffuse through the absorptive barrier via transcellular pathway with little hindrance from P-gp. Phyllanthin could interfere with transport of P-gp drug substrates, when concomitantly administered. In addition, aqueous solubility could be a limiting factor in phyllanthin absorption.
Collapse
Affiliation(s)
- Wilasinee Dunkoksung
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Nontima Vardhanabhuti
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Suree Jianmongkol
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
31
|
Wu W, Jiao C, Li H, Ma Y, Jiao L, Liu S. LC-MS based metabolic and metabonomic studies of Panax ginseng. PHYTOCHEMICAL ANALYSIS : PCA 2018; 29:331-340. [PMID: 29460310 DOI: 10.1002/pca.2752] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/02/2018] [Accepted: 01/03/2018] [Indexed: 05/28/2023]
Abstract
INTRODUCTION Panax ginseng has received much attention as a valuable health supplement with medicinal potential. Its chemical diversity and multiple pharmacological properties call for comprehensive methods to better understand the effects of ginseng and ginsenosides. Liquid chromatography-mass spectrometry (LC-MS) based metabonomic approaches just fit the purpose. OBJECTIVE Aims to give a review of recent progress on LC-MS based pharmacokinetic, metabolic, and phytochemical metabolomic studies of ginseng, and metabonomic studies of ginseng intervention effects. METHODS The review has four sections: the first section discusses metabolic studies of ginsenosides based on LC-MS, the second focuses on ginsenoside-drug interactions and pharmacokinetic interaction between herb compounds based on LC-MS, the third is phytochemical metabolomic studies of ginseng based on LC-MS, and the fourth deals with metabonomic studies of ginseng intervention effects based on LC-MS. RESULTS LC-MS based metabonomic research on ginseng include analysis of single ginsenoside and total ginsenosides. The theory of multi-components and multi-targeted mechanisms helps to explain ginseng effects. CONCLUSION LC-MS based metabonomics is a promising way to comprehensively assess ginseng. It is valuable for quality control and mechanism studies of ginseng.
Collapse
Affiliation(s)
- Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Chuanxi Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Yue Ma
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Lili Jiao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| | - Shuying Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, P. R. China
| |
Collapse
|
32
|
So SH, Lee JW, Kim YS, Hyun SH, Han CK. Red ginseng monograph. J Ginseng Res 2018; 42:549-561. [PMID: 30337816 PMCID: PMC6190493 DOI: 10.1016/j.jgr.2018.05.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 04/12/2018] [Accepted: 05/08/2018] [Indexed: 12/22/2022] Open
Abstract
Ginseng has been traditionally used for several millennia in Asian countries, including Korea, China, and Japan, not only as a nourishing and tonifying agent but also as a therapeutic agent for a variety of diseases. In recent years, the various effects of red ginseng including immunity improvement, fatigue relief, memory improvement, blood circulation improvement, antioxidation, mitigation of menopausal women's symptoms, and anticancer an effect have been reported in clinical as well as basic research. Around the world, there is a trend of the rising consumption of health functional foods on the level of disease prevention along with increased interest in maintaining health because of population aging and the awareness of lifestyle diseases and chronic diseases. Red ginseng occupies an important position as a health functional food. But till now, international ginseng monographs including those of the World Health Organization have been based on data on white ginseng and have mentioned red ginseng only partly. Therefore, the red ginseng monograph is needed for component of red ginseng, functionality certified as a health functional food in the Korea Food and Drug Administration, major efficacy, action mechanism, and safety. The present red ginseng monograph will contribute to providing accurate information on red ginseng to agencies, businesses, and consumers both in South Korea and abroad.
Collapse
Affiliation(s)
- Seung-Ho So
- Laboratory of Fundamental Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Jong Won Lee
- Laboratory of Fundamental Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Young-Sook Kim
- Laboratory of Fundamental Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Sun Hee Hyun
- Laboratory of Fundamental Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| | - Chang-Kyun Han
- Laboratory of Fundamental Research, Korea Ginseng Corporation, Daejeon, Republic of Korea
| |
Collapse
|
33
|
Zang X, Wang G, Cai Q, Zheng X, Zhang J, Chen Q, Wu B, Zhu X, Hao H, Zhou F. A Promising Microtubule Inhibitor Deoxypodophyllotoxin Exhibits Better Efficacy to Multidrug-Resistant Breast Cancer than Paclitaxel via Avoiding Efflux Transport. Drug Metab Dispos 2018. [DOI: 10.1124/dmd.117.079442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
34
|
Chen L, Yang H, Yu C, Yuan M, Li H. High hepatic exposure of furanocoumarins in Radix Angelica dahuricae is associated with transporter mediated active uptake. JOURNAL OF ETHNOPHARMACOLOGY 2018; 212:74-85. [PMID: 29055720 DOI: 10.1016/j.jep.2017.10.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 05/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radix Angelica dahuricae (RAD), the roots of Angelica dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav, is a well-known traditional Chinese medicine (TCM) and has been used for centuries to treat headaches, toothaches, nose congestion, abscesses, furunculoses, and acne. This herb is also one of frequently reported TCMs showing the herb-drug interaction potential. Furanocoumarins are main bioactive components of RAD. AIM OF THE STUDY This study is designed to characterize the tissue distribution profiles of furanocoumarins after oral administration of RAD extract in rats and to explore the mechanism underlying the high hepatic exposure of the major furanocoumarins. MATERIALS AND METHODS The tissue distribution of nine furanocoumarins was determined in rats after an oral dose of 0.46g/kg RAD extract using high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Unbound fractions (ƒu) of major furanocoumarins, including imperatorin (IM), isoimperatorin (IIM), bergapten (BER) and oxypeucedanin hydrate (OXYH), were measured in rat plasma and selected tissue homogenates (liver, kidney, lung and brain) with Rapid Equilibrium Dialysis (RED) method. The temperature dependent hepatic uptake of IM, IIM, BER and OXYH were evaluated in suspended rat primary hepatocytes at 4°C or 37°C by the oil-spin method. The uptake kinetics was conducted in the cells over a wide concentration range. The furanocoumarins were co-incubated with a panel of transporter inhibitors to investigate the involvement of uptake transporters in the hepatic uptake. The transcellular transport characteristics of IM, IIM, BER and OXYH were further assessed using Caco-2 cell monolayer model. RESULTS IM, IIM, BER and OXYH were found to be the major bioactive furanocoumarins in rat plasma and tissues, representing more than 90% exposure for all the detected furanocoumarins. The most concentrative organ of major furanocoumarins was the liver, with liver-to-plasma exposure ratio (Kp,AUC) of 5.1, 6.5 and 4.7 for IM, IIM and BER, and 2.3 for OXYH, respectively. IM, IIM and BER also showed higher concentrations in the kidney with Kp above 2.2. The higher protein binding of the furanocoumarins partially contributed to their higher tissue exposure. In suspended rat primary hepatocyte, the hepatic uptake of IM, IIM, BER and OXYH was temperature-dependent, with considerably higher uptake at 37°C than at 4°C. Uptake kinetics indicated that the hepatic uptake of IM, IIM, BER and OXYH involved both active transport and passive diffusion processes. For IM, IIM and BER, the contribution of the active transport was greater than the passive process, with the CLactive/CLuptake > 72%. Ritonavir (RTN) and cyclosporine A (CsA), the known inhibitors of organic anion transporting polypeptide (Oatp) significantly inhibited the hepatic uptake of IM and BER, while the inhibitor of the organic anion transporters (Oat) probenecid (PBC) remarkably reduced IIM uptake. In the Caco-2 cell model, the furanocoumarins were highly permeable in the apical to basolateral direction without notable active efflux. CONCLUSION The furanocoumarins rapidly and widely distributed into various tissues after oral dose of the RAD extract. IM, IIM, BER and OXYH were the major components detected in both plasma and tissues. Liver was the most distributed tissue of the total and free furanocoumarins. Non-specific protein binding contributed partially to the higher tissue exposures of these bioactive components. The Oatp and Oat mediated active uptake played the primal role in the high hepatic exposure of the furanocoumarins.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Haiying Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Chenchen Yu
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Mei Yuan
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| | - Hua Li
- State Key Laboratory of Toxicology and Medical Countermeasures, 100850 Beijing, China; Beijing Institute of Pharmacology and Toxicology, 100850 Beijing, China.
| |
Collapse
|
35
|
Yuan ZW, Li YZ, Liu ZQ, Feng SL, Zhou H, Liu CX, Liu L, Xie Y. Role of tangeretin as a potential bioavailability enhancer for silybin: Pharmacokinetic and pharmacological studies. Pharmacol Res 2018; 128:153-166. [DOI: 10.1016/j.phrs.2017.09.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 09/05/2017] [Accepted: 09/24/2017] [Indexed: 01/23/2023]
|
36
|
Sun B, Zhong Z, Wang F, Xu J, Xu F, Kong W, Ling Z, Shu N, Li Y, Wu T, Zhang M, Zhu L, Liu X, Liu L. Atorvastatin impaired glucose metabolism in C2C12 cells partly via inhibiting cholesterol-dependent glucose transporter 4 translocation. Biochem Pharmacol 2018; 150:108-119. [PMID: 29338971 DOI: 10.1016/j.bcp.2018.01.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 01/08/2018] [Indexed: 10/18/2022]
Abstract
Skeletal muscle accounts for approximately 75% of glucose disposal in body and statins impair glucose metabolism. We aimed to investigate the effect of atorvastatin on glucose metabolism in C2C12 cells. Glucose metabolism and expression of glucose transporter 4 (GLUT4) and hexokinase II (HXKII) were measured following incubation with atorvastatin or pravastatin. Roles of cholesterol in atorvastatin-induced glucose metabolism impairment were investigated via adding cholesterol or mevalonic acid and confirmed by cholesterol depletion with methyl-β-cyclodextrin. Hypercholesterolemia mice induced by high fat diet (HFD) feeding, orally received atorvastatin (6 and 12 mg/kg) or pravastatin (12 mg/kg) for 22 days. Results showed that atorvastatin not pravastatin concentration-dependently impaired glucose consumption, glucose uptake and GLUT4 membrane translocation in C2C12 cells without affecting expression of HXKII or total GLUT4 protein. The atorvastatin-induced alterations were reversed by cholesterol or mevalonic acid. Cholesterol depletion exerted similar impact to atorvastatin, which could be alleviated by cholesterol supplement. Glucose consumption or GLUT4 translocation was positively associated with cellular cholesterol levels. In HFD mice, atorvastatin not pravastatin significantly increased blood glucose levels following glucose or insulin dose and decreased expression of membrane not total GLUT4 protein in muscle. Glucose exposure following glucose or insulin dose was negatively correlated to muscular free cholesterol concentration. Expression of membrane GLUT4 protein was positively related to free cholesterol in muscle. In conclusion, atorvastatin impaired glucose utilization in muscle cells partly via inhibiting GLUT4 membrane translocation due to inhibition of cholesterol synthesis by atorvastatin, at least, partly contributing to glucose intolerance in HFD mice.
Collapse
Affiliation(s)
- Binbin Sun
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zeyu Zhong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Fan Wang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Jiong Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Feng Xu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Weimin Kong
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhaoli Ling
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Shu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Ying Li
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tong Wu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Mian Zhang
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Liang Zhu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, College of Pharmacy, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Zha W. Transporter-mediated natural product-drug interactions for the treatment of cardiovascular diseases. J Food Drug Anal 2017; 26:S32-S44. [PMID: 29703385 PMCID: PMC9326887 DOI: 10.1016/j.jfda.2017.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/18/2017] [Accepted: 11/21/2017] [Indexed: 12/17/2022] Open
Abstract
The growing use of natural products in cardiovascular (CV) patients has been greatly raising the concerns about potential natural product–CV drug interactions. Some of these may lead to unexpected cardiovascular adverse effects and it is, therefore, essential to identify or predict potential natural product–CV drug interactions, and to understand the underlying mechanisms. Drug transporters are important determinants for the pharmacokinetics of drugs and alterations of drug transport has been recognized as one of the major causes of natural product–drug interactions. In last two decades, many CV drugs (e.g., angiotensin II receptor blockers, beta-blockers and statins) have been identified to be substrates and inhibitors of the solute carrier (SLC) transporters and the ATP-binding cassette (ABC) transporters, which are two major transporter superfamilies. Meanwhile, in vitro and in vivo studies indicate that a growing number of natural products showed cardioprotective effects (e.g., gingko biloba, danshen and their active ingredients) are also substrates and inhibitors of drug transporters. Thus, to understand transporter-mediated natural product–CV drug interactions is important and some transporter-mediated interactions have already shown to have clinical relevance. In this review, we review the current knowledge on the role of ABC and SLC transporters in CV therapy, as well as transporter modulation by natural products used in CV diseases and their induced natural product–CV drug interactions through alterations of drug transport. We hope our review will aid in a comprehensive summary of transporter-mediated natural product–CV drug interactions and help public and physicians understand these type of interactions.
Collapse
Affiliation(s)
- Weibin Zha
- MyoKardia, South San Francisco, CA, USA.
| |
Collapse
|
38
|
Yu J, Zhou Z, Tay-Sontheimer J, Levy RH, Ragueneau-Majlessi I. Intestinal Drug Interactions Mediated by OATPs: A Systematic Review of Preclinical and Clinical Findings. J Pharm Sci 2017; 106:2312-2325. [DOI: 10.1016/j.xphs.2017.04.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/05/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023]
|
39
|
Simultaneous Determination of Seven Components in Human Plasma by LC–ESI–MS/MS After Oral Administration of Danqi Tablets with Application to a Pharmacokinetic Study. Chromatographia 2017. [DOI: 10.1007/s10337-017-3365-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
40
|
Aalinkeel R, Kutscher HL, Singh A, Cwiklinski K, Khechen N, Schwartz SA, Prasad PN, Mahajan SD. Neuroprotective effects of a biodegradable poly(lactic-co-glycolic acid)-ginsenoside Rg3 nanoformulation: a potential nanotherapy for Alzheimer’s disease? J Drug Target 2017; 26:182-193. [DOI: 10.1080/1061186x.2017.1354002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ravikumar Aalinkeel
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, NY, USA
| | - Hilliard L. Kutscher
- Department of Chemistry, Institute for Lasers, Photonics and Biophotonics, University at Buffalo – The State University of New York, Buffalo, NY, USA
| | - Ajay Singh
- Department of Chemistry, Institute for Lasers, Photonics and Biophotonics, University at Buffalo – The State University of New York, Buffalo, NY, USA
| | - Katherine Cwiklinski
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, NY, USA
| | - Noor Khechen
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, NY, USA
| | - Stanley A. Schwartz
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, NY, USA
| | - Paras N. Prasad
- Department of Chemistry, Institute for Lasers, Photonics and Biophotonics, University at Buffalo – The State University of New York, Buffalo, NY, USA
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology, and Rheumatology, State University of New York at Buffalo, Clinical Translational Research Center, Buffalo, NY, USA
| |
Collapse
|
41
|
Liu WY, Zhang JW, Yao XQ, Jiang C, He JC, Ni P, Liu JL, Chen QY, Li QR, Zang XJ, Yao L, Liu YZ, Wang ML, Shen PQ, Wang GJ, Zhou F. Shenmai injection enhances the cytotoxicity of chemotherapeutic drugs against colorectal cancers via improving their subcellular distribution. Acta Pharmacol Sin 2017; 38:264-276. [PMID: 27867186 DOI: 10.1038/aps.2016.99] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 08/26/2016] [Indexed: 12/16/2022] Open
Abstract
Shenmai injection (SMI) is a Chinese patent-protected injection, which was mainly made of Red Ginseng and Radix Ophiopogonis and widely used for treating coronary heart disease and tumors by boosting Qi and nourishing Yin. In this study we examined whether SMI could produce direct synergetic effects on the cytoxicity of adriamycin (ADR) and paclitaxel (PTX) in colorectal cancers in vivo and in vitro, and explored the underlying pharmacokinetic mechanisms. BALB/c nude mice with LoVo colon cancer xenografts were intraperitoneally injected with ADR (2 mg·kg-1·3d-1) or PTX (7.5 mg·kg-1·3d-1) with or without SMI (0.01 mL·g-1·d-1) for 13 d. Co-administration of SMI significantly enhanced the chemotherapeutic efficacy of ADR and PTX, whereas administration of SMI alone at the given dosage did not produce visible anti-cancer effects, The chemosensitizing action of SMI was associated with increased concentrations of ADR and PTX in the plasma and tumors. In Caco-2 and LoVo cells in vitro, co-treatment with SMI (2 μL/mL) significantly enhanced the cytotoxicity of ADR and PTX, and resulted in some favorable pharmacokinetic changes in the subcellular distribution of ADR and PTX. In addition, SMI-induced intracellular accumulation of ADR was closely correlated with the increased expression levels of P-glycoprotein in 4 colon cancer cell lines (r2=+0.8558). SMI enhances the anti-cancer effects of ADR and PTX in colon cancers in vivo and in vitro by improving the subcellular distributions of ADR and PTX.
Collapse
|
42
|
Zhao J, Zeng Z, Sun J, Zhang Y, Li D, Zhang X, Liu M, Wang X. A Novel Model of P-Glycoprotein Inhibitor Screening Using Human Small Intestinal Organoids. Basic Clin Pharmacol Toxicol 2016; 120:250-255. [PMID: 27657920 DOI: 10.1111/bcpt.12680] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/19/2016] [Indexed: 01/31/2023]
Abstract
P-glycoprotein (P-gp), an important efflux transporter in intestine, regulates the bioavailability of orally taken drugs. To develop an in vitro model that preferably mimics the physiological microenvironment of human intestine, we employed the three-dimensionally (3D) cultured organoids from human normal small intestinal epithelium. It was observed that the intestinal crypts could efficiently form cystic organoid structure with the extension of culture time. Furthermore, the physiological expression of ABCB1 was detected at both mRNA and protein levels in cultured organoids. Rhodamine 123 (Rh123), a typical substrate of P-gp, was actively transported across 3D organoids and accumulated in the luminal space. This transport process was also inhibited by verapamil and mitotane. In summary, the above-mentioned model based on human small intestinal 3D organoids is suitable to imitate the small intestinal epithelium and could be used as a novel in vitro model especially for P-gp inhibitor screening.
Collapse
Affiliation(s)
- Junfang Zhao
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhiyang Zeng
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jialiang Sun
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Yuanjin Zhang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Dali Li
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Xueli Zhang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Shanghai Fengxian District Central Hospital, Shanghai, China
| | - Mingyao Liu
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.,Department of Molecular and Cellular Medicine, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Xin Wang
- East China Normal University and Shanghai Fengxian District Central Hospital Joint Research Center for Translational Medicine, Shanghai Key laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
43
|
Kim MG, Kim Y, Jeon JY, Kim DS. Effect of fermented red ginseng on cytochrome P450 and P-glycoprotein activity in healthy subjects, as evaluated using the cocktail approach. Br J Clin Pharmacol 2016; 82:1580-1590. [PMID: 27495955 PMCID: PMC5099554 DOI: 10.1111/bcp.13080] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/22/2016] [Accepted: 08/01/2016] [Indexed: 11/28/2022] Open
Abstract
Aims We assessed the drug interaction profile of fermented red ginseng with respect to the activity of major cytochrome (CYP) P450 enzymes and of a drug transporter protein, P‐glycoprotein (P‐gp), in healthy volunteers. Methods This study was an open‐label crossover study. The CYP probe cocktail drugs caffeine, losartan, dextromethorphan, omeprazole, midazolam and fexofenadine were administered before and after 2 weeks of fermented red ginseng administration. Plasma samples were collected, and tolerability was assessed. Pharmacokinetic parameters were calculated, and the 90% confidence intervals (CIs) of the geometric mean ratios of the parameters were determined from logarithmically transformed data. Values were compared between before and after fermented red ginseng administration using analysis of variance (anova). Results Fifteen healthy male subjects were evaluated, none of whom were genetically defined as a poor CYP2C9, CYP2C19 or CYP2D6 metabolizer based on genotyping. Before and after fermented red ginseng administration, the geometric least‐square mean metabolic ratio (90% CI) was 0.901 (0.830–0.979) for caffeine (CYP1A2) to paraxanthine, 0.774 (0.720–0.831) for losartan (CYP2C9) to EXP3174, 1.052 (0.925–1.197) for omeprazole (CYP2C19) to 5‐hydroxyomeprazole, 1.150 (0.860–1.538) for dextromethorphan (CYP2D6) to dextrorphan, and 0.816 (0.673–0.990) for midazolam (CYP3A4) to 1‐hydroxymidazolam. The geometric mean ratio of the area under the curve of the last sampling time (AUClast) for fexofenadine (P‐gp) was 1.322 (1.112–1.571). Conclusion No significantly different drug interactions were observed between fermented red ginseng and the CYP probe substrates following the two‐week administration of concentrated fermented red ginseng. However, the inhibition of P‐gp was significantly different between fermented red ginseng and the CYP probe substrates. The use of fermented red ginseng requires close attention due to the potential for increased systemic exposure when it is used in combination with P‐gp substrate drugs.
Collapse
Affiliation(s)
- Min-Gul Kim
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Yunjeong Kim
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Ji-Young Jeon
- Research Institute of Clinical Medicine of Chonbuk National University, Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Republic of Korea
| | - Dal-Sik Kim
- Department of Laboratory Medicine, Chonbuk National University Medical School, Jeonju, Republic of Korea
| |
Collapse
|
44
|
Xu XH, Li T, Fong CMV, Chen X, Chen XJ, Wang YT, Huang MQ, Lu JJ. Saponins from Chinese Medicines as Anticancer Agents. Molecules 2016; 21:molecules21101326. [PMID: 27782048 PMCID: PMC6272920 DOI: 10.3390/molecules21101326] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022] Open
Abstract
Saponins are glycosides with triterpenoid or spirostane aglycones that demonstrate various pharmacological effects against mammalian diseases. To promote the research and development of anticancer agents from saponins, this review focuses on the anticancer properties of several typical naturally derived triterpenoid saponins (ginsenosides and saikosaponins) and steroid saponins (dioscin, polyphyllin, and timosaponin) isolated from Chinese medicines. These saponins exhibit in vitro and in vivo anticancer effects, such as anti-proliferation, anti-metastasis, anti-angiogenesis, anti-multidrug resistance, and autophagy regulation actions. In addition, related signaling pathways and target proteins involved in the anticancer effects of saponins are also summarized in this work.
Collapse
Affiliation(s)
- Xiao-Huang Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Chi Man Vivienne Fong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Xiao-Jia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Yi-Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| | - Ming-Qing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
45
|
Wu X, Ma J, Ye Y, Lin G. Transporter modulation by Chinese herbal medicines and its mediated pharmacokinetic herb–drug interactions. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1026:236-253. [DOI: 10.1016/j.jchromb.2015.11.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 10/22/2022]
|
46
|
Liu F, Wang X, Li Z, Li J, Zhuang X, Zhang Z. P-Glycoprotein (ABCB1) limits the brain distribution of YQA-14, a novel dopamine D3 receptor antagonist. Chem Pharm Bull (Tokyo) 2016; 63:512-8. [PMID: 26133067 DOI: 10.1248/cpb.c15-00089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
YQA-14 is a promising agent for treating addiction to cocaine and opioids. However, previous studies have showed there is marked contrast between the relatively small differences in pharmacological action in vivo and the large differences in their respective receptor binding properties in vitro. We hypothesized that the conflict between the in vivo and in vitro outcomes was attributable to poor brain exposure to YQA-14 caused by drug efflux transporters. To address this issue, we investigated the directional flux of YQA-14 across Caco-2 cells at 37°C or 4°C and the bidirectional transport in the presence and absence of transporter chemical inhibitors. These phenomena were further investigated by an in vivo determination of the brain and blood pharmacokinetics (PK) profile of YQA-14 following intraperitoneal administration with and without inhibitor. The efflux ratio of YQA-14 on Caco-2 cell monolayers was 2.39 and the efflux was temperature-dependent. When co-incubated with GF120918 or LY335979, the efflux of YQA-14 was markedly decreased. However, there was no significant difference in the permeability of YQA-14 when the cells were treated with Ko143. In vivo experiments showed that the brain-to-plasma ratio increased by more than 75-fold and 20-fold with co-administration of GF120918 and LY335979, respectively. Use of Ko143 did not change the brain-to-blood ratio of YQA-14. The results indicate that the brain distribution of YQA-14 was restricted because of active efflux transport at the blood brain barrier. In addition, P-glycoprotein (P-gp) played a dominant role in limiting the distribution of YQA-14 to the brain.
Collapse
Affiliation(s)
- Fei Liu
- Department of Pharmacy, The First Affiliated Hospital of PLA
| | | | | | | | | | | |
Collapse
|
47
|
Wang Q, Jiang C, Zheng X, Zhu X, Yan S, Wang H, Fu R, Fan H, Chen Y. Insight into the pharmacokinetic behavior of tanshinone IIA in the treatment of Crohn's disease: comparative data for tanshinone IIA and its two glucuronidated metabolites in normal and recurrent colitis models after oral administration. Xenobiotica 2016; 47:66-76. [PMID: 27045386 DOI: 10.3109/00498254.2016.1160158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
1. Previous reports implied that tanshinone IIA (TSA) may offer potential benefits for Crohn's disease (CD). However, the detailed pharmacokinetic behavior of TSA in the treatment of colitis remain unclear. Herein, a recurrent trinitrobenzene sulfonic acid (TNBS)-colitis mouse model was used to investigate whether TSA possesses favorable pharmacokinetic and colonic distribution profiles to serve as a candidate drug. 2. Although the systemic TSA exposures were low (AUC0-t approximately 330 ng*h/ml) in both the normal and colitis models after oral administration TSA 20 mg/kg, high levels of TSA were found in the gastrointestinal tract (GI). Such a GI exposure of TSA in colitis mice is adequate to exert anti-inflammatory effects as observed in various in vitro studies. 3. Interestingly, colonic TSA exposure in the colitis mouse model was much lower than that in the normal mice, which may be explained by a significant upregulation of colonic UDP-glucuronosyltransferase (Ugt)1a9 expression and a higher plasma concentration of TSA glucuronides in the model mice at 0.5, 1 and 2 h after TSA administration. 4. Together, these results reveal high accumulation at the site of inflammation and minimal systemic concentration of TSA, which are favorable pharmacokinetic behaviors to meet the requirements for CD treatment.
Collapse
Affiliation(s)
| | - Chao Jiang
- b Department of Digestive Tumor Surgery , and
| | - Xiao Zheng
- c Department of Pharmacy , Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , China
| | | | | | | | - Rui Fu
- a Department of Pharmacology
| | - Hongwei Fan
- d Department of Clinical Pharmacology Laboratory , Nanjing First Hospital of Nanjing Medical University , Nanjing , China , and
| | - Yugen Chen
- e Department of Colorectal Surgery , Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , China
| |
Collapse
|
48
|
Kim DS, Kim Y, Jeon JY, Kim MG. Effect of Red Ginseng on cytochrome P450 and P-glycoprotein activities in healthy volunteers. J Ginseng Res 2015; 40:375-381. [PMID: 27746690 PMCID: PMC5052446 DOI: 10.1016/j.jgr.2015.11.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 10/15/2015] [Accepted: 11/28/2015] [Indexed: 12/05/2022] Open
Abstract
Background We evaluated the drug interaction profile of Red Ginseng (RG) with respect to the activities of major cytochrome P450 (CYP) enzymes and the drug transporter P-glycoprotein (P-gp) in healthy Korean volunteers. Methods This article describes an open-label, crossover study. CYP probe cocktail drugs, caffeine, losartan, dextromethorphan, omeprazole, midazolam, and fexofenadine were administered before and after RG supplementation for 2 wk. Plasma samples were collected, and tolerability was assessed. Pharmacokinetic parameters were calculated, and 90% confidence intervals (CIs) of the geometric mean ratios of the parameters were determined from logarithmically transformed data using analysis of variance after RG administration versus before RG administration. Results Fourteen healthy male participants were evaluated, none of whom were genetically defined as poor CYP2C9, 2C19, and CYP2D6 metabolizers based on genotyping. Before and after RG administration, the geometric least-square mean metabolic ratio (90% CI) was 0.870 (0.805–0.940) for caffeine to paraxanthine (CYP1A2), 0.871 (0.800–0.947) for losartan (CYP2C9) to EXP3174, 1.027 (0.938–1.123) for omeprazole (CYP2C19) to 5-hydroxyomeprazole, 1.373 (0.864–2.180) for dextromethorphan to dextrorphan (CYP2D6), and 0.824 (0.658–1.032) for midazolam (CYP3A4) to 1-hydroxymidazolam. The geometric mean ratio of the area under the curve of the last sampling time (AUClast) for fexofenadine (P-gp) was 0.963 (0.845–1.098). Administration of concentrated RG for 2 wk weakly inhibited CYP2C9 and CYP3A4 and weakly induced CYP2D6. However, no clinically significant drug interactions were observed between RG and CYP and P-gp probe substrates. Conclusion RG has no relevant potential to cause CYP enzyme- or P-gp-related interactions. Clinical trial registration number (ClinicalTrials.gov): NCT02056743.
Collapse
Affiliation(s)
- Dal-Sik Kim
- Department of Laboratory Medicine, Chonbuk National University Medical School, Jeonju, Korea
| | - Yunjeong Kim
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Ji-Young Jeon
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Min-Gul Kim
- Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
49
|
Chen L, Jian Y, Wei N, Yuan M, Zhuang X, Li H. Separation and simultaneous quantification of nine furanocoumarins from Radix Angelicae dahuricae using liquid chromatography with tandem mass spectrometry for bioavailability determination in rats. J Sep Sci 2015; 38:4216-24. [PMID: 26496866 DOI: 10.1002/jssc.201500840] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/15/2015] [Accepted: 10/12/2015] [Indexed: 12/20/2022]
Abstract
Radix Angelicae dahuricae is a well-known medicinal herb in a number of herb preparations for medical uses. In this study, a rapid and selective method using liquid chromatography with tandem mass spectrometry was developed for the separation and simultaneous quantitation of nine furanocoumarins from Radix A. dahuricae, namely imperatorin, isoimperatorin, oxypeucedanin hydrate, bergapten, oxypeucedanin, xanthotoxol, xanthotoxin, isopimpinellin, and psoralen. Chromatographic separation was achieved on a CAPCELL PAK MG II C18 analytical column. Detection was performed using positive electrospray ion source in the multiple reaction monitoring mode. The method was fully validated for analyzing these principles in rat plasma with a lower limit of quantification from 0.5 to 5 ng/mL. The intra- and interbatch precisions were less than 10%, and the accuracies ranged from -7.5 to 8.0%. The extraction recovery of the analytes was above 70% without a significant matrix effect. The method was used to determine the oral and intravenous pharmacokinetic profiles of these furanocoumarins after dosing with Radix A. dahurica extract. The bioavailability of these furanocoumarins ranged from 10.1 to 82.8%. These data provide critical information for a better understanding of the pharmacological mechanisms and herb-drug interaction potential of Radix A. dahurica.
Collapse
Affiliation(s)
- Lin Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Jian
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ning Wei
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Mei Yuan
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Hua Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing, China.,Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
50
|
Wong AST, Che CM, Leung KW. Recent advances in ginseng as cancer therapeutics: a functional and mechanistic overview. Nat Prod Rep 2015; 32:256-72. [PMID: 25347695 DOI: 10.1039/c4np00080c] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Ginseng, a key ingredient in traditional Chinese medicine, shows great promise as a new treatment option. As listed by the U.S. National Institutes of Health as a complementary and alternative medicine, its anti-cancer functions are being increasingly recognized. This review covers the mechanisms of action of ginsenosides and their metabolites, which can modulate signaling pathways associated with inflammation, oxidative stress, angiogenesis, metastasis, and stem/progenitor-like properties of cancer cells. The emerging use of structurally modified ginsenosides and recent clinical studies on the use of ginseng either alone or in combination with other herbs or Western medicines which are exploited as novel therapeutic strategies will also be explored.
Collapse
Affiliation(s)
- Alice S T Wong
- State Key Laboratory of Oncogenes and Related Genes, and School of Biological Sciences, The University of Hong Kong, Hong Kong.
| | | | | |
Collapse
|