Skip to main content
Log in

Effects and Mechanism of Action of Panax notoginseng Saponins on the Pharmacokinetics of Warfarin

  • Original Research Article
  • Published:
European Journal of Drug Metabolism and Pharmacokinetics Aims and scope Submit manuscript

A Correction to this article was published on 22 February 2022

This article has been updated

Abstract

Backgroud

The interactions between Chinese herbs and drugs pose a great challenge to the combined clinical application of Chinese herbs and drugs. Chinese medicinal products contain complex pharmacologically active components that may influence the in vivo processes of drugs in a variety of ways. In China, drugs based on Panax ginseng total saponins (PNS) are often combined with warfarin for the treatment of cardiovascular diseases.

Objectives

To assess the effects of Panax notoginseng saponins (PNS) on the pharmacokinetics of warfarin and its mechanism.

Method

Blood was collected for the determination of the prothrombin time (PT) and international normalized ratio (INR) from rats treated with warfarin alone or with warfarin + PNS. The plasma concentration of warfarin was determined by high-performance liquid chromatography. Western blot was used to detect the expression of cytochrome P450 (CYP) enzymes.

Results

When warfarin and PNS were co-administered, the PT and INR increased compared to when warfarin was given alone. 72 hours after administration, compared to the warfarin alone group, the warfarin + low-dose PNS, warfarin + medium-dose PNS, and warfarin + high-dose PNS groups showed 110%, 122%, and 126% increases in PT, respectively (all P < 0.05), as well as 111%, 124%, and 128% increases in INR (all P < 0.05). Compared with the warfarin alone group, the clearance rate (CL/F) of warfarin in the warfarin + low-dose PNS, warfarin + medium-dose PNS, and warfarin + high-dose PNS groups was 10% (P  > 0.05), 23% (P  < 0.05), and 33% (P  < 0.05) lower, respectively, while the systemic exposure (area under the concentration-time curve, AUC0–t) increased by 106% (P  > 0.05), 119% (P  < 0.05), and 134% (P  < 0.05), respectively, and the blood concentration of warfarin incresed by 112%, 113%, and 114%, respectively (all P > 0.05). After combined treatment of HepG2 cells with warfarin + PNS, CYP1A2 expression was upregulated (P  < 0.05) and CYP3A4 was downregulated (P  < 0.05) but there was no effect on CYP2C9. In animal experiments, PNS had different effect on the expression of CYP1A2 in different doses. While a low dose of PNS resulted in downregulated CYP1A2 expression (P  < 0.05), a medium dose resulted in upregulation (P  < 0.05), and CYP1A2 expression was not significantly affected by a high dose of PNS (P  > 0.05). Meanwhile, PNS at all doses downregulated the expression of CYP3A4 (P  < 0.05) but had no effect on the expression of CYP2C9 (P >  0.05).

Conclusion

PNS can increase the blood concentration of warfarin, as well as the exposure time, and it can enhance the anticoagulant effect of warfarin by inhibiting the expression of the liver enzyme CYP3A4.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Change history

References

  1. Wen MS, Lee MT. Warfarin pharmacogenetics: new life for an old drug. Acta Cardiol Sin. 2013;29:235–42. https://doi.org/10.5935/abc.20130106.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Kearon C, Akl EA, Ornelas J, Blaivas A, et al. Antithrombotic therapy for VTE disease: chest guideline and expert panel report. Chest. 2016;149:315–52. https://doi.org/10.1016/j.chest.2015.11.026.

    Article  PubMed  Google Scholar 

  3. Zhang J-H, Liu M-B, Guan C-F, et al. Establishment and practice of online anticoagulation clinic. Chin J Pharm. 2014;16:1476–8. https://doi.org/10.11669/cpj.2014.16.024.

    Article  Google Scholar 

  4. Wang Y, Tan X-P, Yan P-K. A case of abnormal INR caused by warfarin in patients with atrial fibrillation complicated with coronary heart disease. China Pharmacist. 2015;18(1):105–8.

  5. Zhang J, Chen Z, Chen C. Impact of CYP2C9, VKORC1 and CYP4F2 genetic polymorphisms on maintenance warfarin dosage in Han-Chinese patients: a systematic review and meta-analysis. Meta Gene. 2016;9:197–209. https://doi.org/10.1016/j.mgene.2016.07.002.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Hirsh JV, Fuster V, Halperin JL. American Heart Association/American College of Cardiology Foundation guide to warfarin therapy. J Am Coll Cardiol. 2003;4l(9):1633–52. https://doi.org/10.1161/01.CIR.0000063575.17904.4E.

    Article  Google Scholar 

  7. Breckenridge A, Orme M. Kinetics of warfarin absorption in man. Clin Pharmacol Ther. 1973;14(6):955–61. https://doi.org/10.1002/cpt1973146955.

    Article  CAS  PubMed  Google Scholar 

  8. Miners JO, Birketc JO. Cytochrome P4502C9: an enzyme of major importance in human drug metabolism. Br J Clin Pharmaeol. 1998;45(6):525–38. https://doi.org/10.1046/j.1365-2125.1998.00721.x.

  9. Ngui JS, Chen Q, Shou M, et al. In vitro stimulation of warfarin metabolism by quinidine: increases in the formation of 4′- and 10-hydroxywarfarin. Drug Metab Dispos. 2001;29(6):877–86.

  10. Zhang Z, Fasco MJ, Huang Z, Guengerich FP, Kaminsky LS. Human cytochromes P4501A1 and P450 1A2: R-warfarin metabolism as a probe. Drug Metab Dispos. 1995;23(12):1339–46.

  11. Wienkers LC, Wurden CJ, Storch E, et al. Formation of (R)-8-hydroxywarfarin in human liver microsomes. A new metabolic marker for the (S)-mephenytoin hydroxylase, P4502C19. Drug Metab Dispos. 1996;24(5):610–4.

    CAS  PubMed  Google Scholar 

  12. Ye H, Sui D, Liu W, Yuan Y, Ouyang Z, Wei Y. Effects of CYP2C11 gene knockout on the pharmacokinetics and pharmacodynamics of warfarin in rats. Xenobiotica. 2019;49(12):1478–84. https://doi.org/10.1080/00498254.2019.1579006.

    Article  CAS  PubMed  Google Scholar 

  13. Jian-mei Wu, Zhang M-S, Zhang J-H. Pharmacokinetics of Panax notoginseng on warfarin in rats. Chin J Clin Pharmacol. 2018;34(04):461–3.

  14. Jiang S, Peng X-S, Huang Z-H, et al. Compatibility of astragalus polysaccharides and notoginseng total saponin in diabetic rat kidney IV collagen type and the influence of layer adhesion protein expression. J Hunan Univ Chin Med. 2015;35(12):18–22.

    CAS  Google Scholar 

  15. Han SY, Li HX, Ma X, et al. Evaluation of the antimyocardial ischemia effect of individual and combined extracts of Panax notoginseng and Carthamus tinctorius in rats. J Ethnopharmacol. 2013;145(3):722–7.

    Article  CAS  Google Scholar 

  16. Liu R, Qin M, Hang P, et al. Effects of Panax notoginseng saponins on the activities of CYP1A2, CYP2C9, CYP2D6 and CYP3A4 in rats in vivo. Phytother Res. 2012;26(8):1113–8.

  17. Xiao-yu L. Study on the expression of CYP450 enzyme in rat liver mediated by total saponins of Panax notoginseng and its mechanism. Shanghai: Shanghai Jiao Tong University; 2009.

    Google Scholar 

  18. Mo Q-Y, Zu-e Lu. Clinical application effect and pharmacological analysis of Panax notoginseng. J Gen Stomatol. 2018;5(21):110–1.

    Google Scholar 

  19. Jin Z, Gao L, Zhang L, et al. Antimicrobial activity of saponins produced by two novel endophytic fungi from Panax notoginseng. Nat Prod Res. 2017;31(22):2700–3. https://doi.org/10.1080/14786419.2017.1292265.

    Article  CAS  PubMed  Google Scholar 

  20. Kitamura K, Takamura Y, Iwamoto T, et al. Dammarane-type triterpene extracts of Panax notoginseng root ameliorates hyperglycemia and insulin sensitivity by enhancing glucose uptake in skeletal muscle. Biosci Biotechnol Biochem. 2017;81(2):335–42. https://doi.org/10.1080/09168451.2016.1246173.

    Article  CAS  PubMed  Google Scholar 

  21. Liao J, Wei B, Chen H, et al. Bioinformatics investigation of therapeutic mechanisms of Xuesaitong capsule treating ischemic cerebrovascular rat model with comparative transcriptome analysis. Am J Transl Res. 2016;8(5):2438–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ratno Budiarto B, Chan WH. Oxidative stresses-mediated apoptotic effects of ginsenoside rb1 on pre- and post-implantation mouse embryos in vitro and in vivo. Environ Toxicol. 2017;32(8):1990–2003. https://doi.org/10.1002/tox.22366.

    Article  CAS  PubMed  Google Scholar 

  23. Tian XP, Zhao HM, Xue MJ. Effect of xuesaitong on elderly patients with severe acute pancreatitis by selective arterial intervention therapy. Biomed Res-India. 2017;28(12):5233–6.

    CAS  Google Scholar 

  24. Xu D, Huang P, Yu Z, et al. Efficacy and safety of Panax notoginseng saponin therapy for acute intracerebral hemorrhage, meta-analysis, and mini review of potential mechanisms of action. Front Neurol. 2014;5:274. https://doi.org/10.3389/fneur.2014.00274.

    Article  PubMed  Google Scholar 

  25. Yang M, Liang EY, Mao LJ, et al. Notoginsenoside R1 has thrombopoietic effect in irradiated mice. Blood. 2016;128(22):4932.

    Article  Google Scholar 

  26. Yang Z-M, Yang X-F. Inhibitory effect of Panax notoginseng saponins on drug metabolizing enzyme CYP3A in rat liver and its kinetic analysis. Chin J Tradit Chin Med. 2012;37(22):3486–9.

    Google Scholar 

  27. Lin Y, Wei Y, Hu X, et al. Evaluation of lentinan effects on cytochrome P450 activity in rats by a cocktail method. Iran J Basic Med Sci. 2019;22(3):296–301. https://doi.org/10.22038/ijbms.2019.31611.7611.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Shimada T, Yamazaki H, Mimura M, et al. Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther. 1994;270:414–23.

    CAS  PubMed  Google Scholar 

  29. Niwa T, Morimoto M, Hirai T, et al. Effect of penicillin-based antibiotics, amoxicillin, ampicillin, and piperacillin, on drug-metabolizing activities of human hepatic cytochromes P450. J Toxicol Sci. 2016;41(1):143–6. https://doi.org/10.2131/jts.41.143.

    Article  CAS  PubMed  Google Scholar 

  30. Han W, Shao Fu, Fang X-L. Study on oral absorption and pharmacokinetics of Ginsenoside Rg1 and Rb1 in Panax notoginseng saponins. J Pharm Sci. 2007;42(8):849–53.

    CAS  Google Scholar 

Download references

Acknowledgements

The authors wish to thank Mrs. Tingting Wu regarding the collection of the data.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jinhua Zhang.

Ethics declarations

Funding

This study was funded by Fujian Provincial Health Technology Project, China (2019-CX-19).

Conflicts of interest

The authors declare that there are no conflicts of interest.

Data availability statement

The data that support the findings of this study are available on request from the corresponding author. The data are not publicly available due to privacy or ethical restrictions.

Code availability

Not applicable.

Author contributions

WJC, JMW, and JFQ, who participated in the design and conduct of the study, contributed equally to the article. MNL, SJJ, ZWZ, ZWF, and MRC analyzed the data and conducted the section on statistical analysis, and JHZ revised the manuscript. All the authors contributed to the final paper.

Ethical approval

The protocol of the study was reviewed and approved by the Ethics Committee of Fujian Medical University. All institutional and national guidelines for the care of the laboratory animals were followed.

Consent for publication

Not applicable.

Consent for publication

Not applicable.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Qian, J., Chen, W., Wu, J. et al. Effects and Mechanism of Action of Panax notoginseng Saponins on the Pharmacokinetics of Warfarin. Eur J Drug Metab Pharmacokinet 47, 331–342 (2022). https://doi.org/10.1007/s13318-022-00753-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13318-022-00753-0

Navigation