DOI QR코드

DOI QR Code

Effects of Baicalin on Oral Pharmacokinetics of Caffeine in Rats

  • Received : 2014.12.02
  • Accepted : 2014.12.23
  • Published : 2015.03.01

Abstract

Scutellaria baicalensis is one of the most widely used herbal medicines in East Asia. Because baicalein and baicalin are major components of this herb, it is important to understand the effects of these compounds on drug metabolizing enzymes, such as cytochrome P450 (CYP), for evaluating herb-drug interaction. The effects of baicalin and baicalein on activities of ethoxyresorufin O-deethylase (EROD), methoxyresorufin O-demethylase (MROD), benzyloxyresorufin O-debenzylase (BROD), p-nitrophenol hydroxylase and erythromycin N-demethylase were assessed in rat liver microsomes in the present study. In addition, the pharmacokinetics of caffeine and its three metabolites (i.e., paraxanthine, theobromine and theophylline) in baicalin-treated rats were compared with untreated control. As results, EROD, MROD and BROD activities were inhibited by both baicalin and baicalein. However, there were no significant differences in the pharmacokinetic parameters of oral caffeine and its three metabolites between control and baicalin-treated rats. When the plasma concentration of baicalin was determined, the maximum concentration of baicalin was below the estimated $IC_{50}$ values observed in vitro. In conclusion, baicalin had no effects on the pharmacokinetics of caffeine and its metabolites in vivo, following single oral administration in rats.

Keywords

References

  1. Alsanad, S. M., Williamson, E. M. and Howard, R. L. (2014) Cancer patients at risk of herb/food supplement-drug interations: a systemic review. Phytother. Res. 28, 1749-1755. https://doi.org/10.1002/ptr.5213
  2. Blank, J. A., Tucker, A. N., Sweatlock, J., Gasiewicz, T. A. and Luster, M. I. (1987) $\alpha$-Naphthoflavone antagonism of 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced murine lymphocyte ethoxyresorufin-Odeethylase activity and immunosuppression. Mol. Pharmacol. 32, 169-172.
  3. Chiou, W. L. (1978) Critical evaluation of the potential error in pharmacokinetic studies of using the linear trapezoidal rule method for the calculation of the area under the plasma level--time curve. J. Pharmacokinet. Biopharm. 6, 539-546. https://doi.org/10.1007/BF01062108
  4. Cho, Y. A., Choi, J. S. and Burm, J. P. (2011) Effects of the antioxidant baicalein on the pharmacokinetics of nimodipine in rats: a possible role of P-glycoprotein and CYP3A4 inhibition by baicalein. Pharmacol. Rep. 63, 1066-1073. https://doi.org/10.1016/S1734-1140(11)70624-7
  5. Choi, E. J., Bae, S. H., Park, J. B., Kwon, M. J., Jang, S. M., Zheng, Y. F., Lee, Y. S., Lee, S. J. and Bae, S. K. (2013) Simultaneous quantification of caffeine and its three primary metabolites in rat plasma by liquid chromatography-tandem mass spectrometry. Food Chem. 141, 2735-2742. https://doi.org/10.1016/j.foodchem.2013.05.069
  6. Fan, L., Wang, J. C., Jiang, F., Tan, Z. R., Chen, Y., Li, Q., Zhang, W., Wang, G., Lei, H. P., Hu, D. L., Wang, D. and Zhou, H. H. (2009) Induction of cytochrome P450 2B6 activity by the herbal medicine baicalin as measured by bupropion hydroxylation. Eur. J. Clin. Pharmacol. 65, 403-409. https://doi.org/10.1007/s00228-008-0594-3
  7. FDA (2005) Guidance for industry: estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult healthy volunteers. FDA [online], http://www.fda.gov/downloads/Drugs/Guidances/UCM078932.pdf
  8. Gao, N., Qi, B., Liu, F. J., Fang, Y., Zhou, J., Jia, L. J. and Qiao, H. L. (2014) Inhibition of baicalin on metabolism of phenacetin, a probe of CYP1A2, in human liver microsomes and in rats. PLoS One 9, e89752. https://doi.org/10.1371/journal.pone.0089752
  9. Gao, N., Zou, D. and Qiao, H. L. (2013) Concentration-dependent inhibitory effect of baicalin on the plasma protein binding and metabolism of chlorzoxazone, a CYP2E1 probe substrate, in rats in vitro and in vivo. PLoS One 8, e53038. https://doi.org/10.1371/journal.pone.0053038
  10. Ishimaru, K., Nishikawa, K., Omoto, T., Asai, I., Yoshihira, K. and Shimomura, K. (1995) Two flavone 2'-glucosides from Scutellaria baicalensis. Phytochemistry 40, 279-281. https://doi.org/10.1016/0031-9422(95)00200-Q
  11. Kang, M. J., Ko, G. S., Oh, D. G., Kim, J. S., Noh, K., Kang, W., Yoon, W. K., Kim, H. C., Jeong, H. G., Jeong, T. C. (2014) Role of metabolism by intestinal microbiota in pharmacokinetics of oral baicalin. Arch. Pharm. Res. 37, 371-378. https://doi.org/10.1007/s12272-013-0179-2
  12. Kim, N. H., Lee, S., Kang, M. J., Jeong, H. G., Kang, W. and Jeong, T. C. (2014) Protective effects of diallyl sulfide against thioacetamideinduced toxicity: A possible role of cytochrome P450 2E1. Biomol. Ther. 22, 149-154. https://doi.org/10.4062/biomolther.2014.016
  13. Kim, Y. H., Jeong, D. W., Kim, Y. C., Sohn, D. H., Park, E. S. and Lee, H. S. (2007) Pharmacokinetics of baicalein, baicalin and wogonin after oral administration of a standardized extract of Scutellaria baicalensis, PF-2405 in rats. Arch. Pharm. Res. 30, 260-265. https://doi.org/10.1007/BF02977703
  14. Kim, Y. S., Kim, J. J., Cho, K. H., Jung, W. S., Moon, S. K., Park, E. K., Kim, D. H. (2008) Biotransformation of ginsenoside Rb1, crocin, amygdalin, geniposide, puerarin, ginsenoside Re, hesperidin, poncirin, glycyrrhizin, and baicalin by human fecal microflora and its relation to cytotoxicity against tumor cells. J. Microbiol. Biotechnol. 18, 1109-1114.
  15. Koop, D. R. (1986) Hydroxylation of p-nitrophenol by rabbit ethanolinducible cytochrome P-450 isozyme 3a. Mol. Pharmacol. 29, 399-404.
  16. Lai, M. Y., Hsiu, S. L., Chen, C. C., Hou, Y. C. and Chao, P. D. (2003) Urinary pharmacokinetics of baicalein, wogonin and their glycosides after oral administration of Scutellariae Radix in humans. Biol. Pharm. Bull. 26, 79-83. https://doi.org/10.1248/bpb.26.79
  17. Li, C., Kim, M., Choi, H. and Choi, J. (2011) Effects of baicalein on the pharmacokinetics of tamoxifen and its main metabolite, 4-hydroxytamoxifen, in rats: possible role of cytochrome P450 3A4 and P-glycoprotein inhibition by baicalein. Arch. Pharm. Res. 34, 1965-1972. https://doi.org/10.1007/s12272-011-1117-9
  18. Lowry, O. H., Rosebrough, N. J., Farr, A. L. and Randall, R. J. (1951) Protein measurement with the Folin phenol reagent. J. Biol. Chem. 193, 265-275.
  19. Lu, T., Song, J., Huang, F., Deng, Y., Xie, L., Wang, G. and Liu, X. (2007) Comparative pharmacokinetics of baicalin after oral administration of pure baicalin, Radix Scutellariae extract and Huang-Lian-Jie-Du-Tang to rats. J. Ethnopharmacol. 110, 412-418. https://doi.org/10.1016/j.jep.2006.09.036
  20. Lubet, R. A., Mayer, R. T., Cameron, J. W., Nims, R. W., Burke, M. D., Wolff, T., Guengerich, F. P. (1985) Dealkylation of pentoxyresorufin: a rapid and sensitive assay for measuring induction of cytochrome(s) P-450 by phenobarbital and other xenobiotics in the rat. Arch. Biochem. Biophys. 238, 43-48. https://doi.org/10.1016/0003-9861(85)90138-9
  21. Nash, T. (1953) The colorimetric estimation of formaldehyde by means of the Hantzsch reaction. Biochem. J. 55, 416-421. https://doi.org/10.1042/bj0550416
  22. Noh, K., Seo, Y. M., Lee, S. K., Bista, S. R., Kang, M. J., Jahng, Y., Kim, E., Kang, W. and Jeong, T. C. (2011) Effects of rutaecarpine on the metabolism and urinary excretion of caffeine in rats. Arch. Pharm. Res. 34, 119-125. https://doi.org/10.1007/s12272-011-0114-3
  23. Murray, M. T. (2012) Botanical medicine-a modern perspective. In Textbook of natural medicine (J. E. Pizzorno and M. T. Murray, Ed.), pp. 255-260. Elsevier Health Sciences, St. Louis.
  24. Schneider, H., Ma, L. and Glatt, H. (2003) Extractionless method for the determination of urinary caffeine metabolites using high-performance liquid chromatography coupled with tandem mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 789, 227-237. https://doi.org/10.1016/S1570-0232(03)00065-5
  25. Wang, Y., Yang, J., Li, X. and Wang, J. (2012) The metabolism of baicalin in rat and the biological activities of the metabolites. Evid. Based Complement. Alternat. Med. 2012, 404529.

Cited by

  1. Pharmacokinetic Interaction of Chrysin with Caffeine in Rats vol.24, pp.4, 2016, https://doi.org/10.4062/biomolther.2015.197
  2. Measurement of caffeine and its three primary metabolites in human plasma by HPLC-ESI-MS/MS and clinical application vol.31, pp.6, 2017, https://doi.org/10.1002/bmc.3900
  3. Role of Intestinal Microbiota in Baicalin-Induced Drug Interaction and Its Pharmacokinetics vol.21, pp.3, 2016, https://doi.org/10.3390/molecules21030337
  4. Caffeine and REM sleep deprivation: Effect on basal levels of signaling molecules in area CA1 vol.71, 2016, https://doi.org/10.1016/j.mcn.2015.12.015
  5. Effect of terpinyl acetate on the percutaneous absorption of caffeine in rats vol.26, pp.5, 2017, https://doi.org/10.1111/exd.13219
  6. -Succinyl Macrolactin A Based on Allometric Scaling of Data from Mice, Rats, and Dogs vol.25, pp.6, 2017, https://doi.org/10.4062/biomolther.2016.192
  7. Pharmacokinetics and Bioavailability Enhancement of Baicalin: A Review pp.2107-0180, 2018, https://doi.org/10.1007/s13318-018-0509-3
  8. Role of Intestinal Microbiota in Metabolism of Gastrodin In Vitro and In Vivo vol.9, pp.4, 2015, https://doi.org/10.3390/metabo9040069
  9. Pharmacokinetics of B-Ring Unsubstituted Flavones vol.11, pp.8, 2019, https://doi.org/10.3390/pharmaceutics11080370
  10. Pharmacological Modulation of Sleep Homeostasis in Rat: Novel Effects of an mGluR2/3 Antagonist vol.42, pp.9, 2015, https://doi.org/10.1093/sleep/zsz123
  11. Baicalein inhibits the pharmacokinetics of simvastatin in rats via regulating the activity of CYP3A4 vol.59, pp.1, 2015, https://doi.org/10.1080/13880209.2021.1942927
  12. Human Lanosterol 14-Alpha Demethylase (CYP51A1) Is a Putative Target for Natural Flavonoid Luteolin 7,3′-Disulfate vol.26, pp.8, 2015, https://doi.org/10.3390/molecules26082237
  13. Drug-herb interactions between Scutellaria baicalensis and pharmaceutical drugs: Insights from experimental studies, mechanistic actions to clinical applications vol.138, pp.None, 2015, https://doi.org/10.1016/j.biopha.2021.111445