Home Assessment of cytochrome P450 inhibition and induction potential of lupeol and betulin in rat liver microsomes
Article
Licensed
Unlicensed Requires Authentication

Assessment of cytochrome P450 inhibition and induction potential of lupeol and betulin in rat liver microsomes

  • Madhav Seervi , Shweta Lotankar , Shrikant Barbar and Sadhana Sathaye EMAIL logo
Published/Copyright: March 9, 2016

Abstract

Background: Lupeol and betulin are triterpenoids that are majorly found in dietary substances. The aim of present study was to investigate the inhibition and induction potential of lupeol and betulin on cytochrome P450 (CYP)1A2, CYP2C11, CYP2D6 and CYP3A2 activities in rat liver microsomes.

Methods: The inhibition and induction studies were conducted using ethoxy resorufin-O-deethylase (CYP1A2), tolbutamide hydroxylase (CYP2C9), and midazolam hydroxylase (CYP3A4) activity assays. In vitro inhibition study was evaluated by incubating lupeol and betulin (1, 3, 10, 30 and 100 μM) with rat liver microsomes, and the metabolite formation was analyzed by high-performance liquid chromatography. The induction study was conducted by administering lupeol (20 mg/kg) and betulin (50 mg/kg) intraperitoneally for 14 days to rats followed by liver isolation and microsome preparation.

Results: The IC50 values in inhibition studies were found to be 59.42 μM (CYP1A2), >100 μM (CYP2C11, CYP2D6, CYP3A2) for lupeol, 52.24 μM (CYP1A2), and >100 μM (CYP2C9, CYP2D6, CYP3A2) for betulin. There was no significant modification observed in the CYP450 isoforms, indicating neither inhibition nor induction potential of lupeol and betulin.

Conclusions: Lupeol and betulin have very low propensity to interact with CYP enzyme, suggesting no CYP inhibitory and inducing potential in rat liver microsomes.


Corresponding author: Sadhana Sathaye, Institute of Chemical Technology, Department of Pharmaceutical Science and Technology, Mumbai, Maharashtra 400019, India, E-mail:

Acknowledgments

The authors acknowledge the University Grants Commission (grant/award no. F-63/2007, BSR) for their financial support and Dr. Krishna Iyer, Bombay College of Pharmacy, for his valuable suggestions in study designing.

Author contributions: MS conducted the present study, which included aspects such as laboratory work, interpretation of data, and manuscript writing. SL assisted in the experimental part of study. SB provided lupeol and betulin for the study. SS was involved in planning the study design and also corrected manuscript write-up. All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

Research funding: None declared.

Employment or leadership: None declared.

Honorarium: None declared.

Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: a systemic review. Drugs 2001;61:2163–75.10.2165/00003495-200161150-00002Search in Google Scholar

2. Gohil KJ, Patel JA. Herb-drug interactions: a review and study based on assessment of clinical case reports in literature. Indian J Pharmacol 2007;39:129–39.10.4103/0253-7613.33432Search in Google Scholar

3. Pal D, Mitra AK. MDR- and CYP3A4-mediated drug-herbal interactions. Life Sci 2006;78:2131–45.10.1016/j.lfs.2005.12.010Search in Google Scholar

4. Haq I. Safety of medicinal plants. Pakistan J Med Res 2004;43:203–10.Search in Google Scholar

5. Elvin-Lewis M. Should we be concerned about herbal remedies. J Ethnopharmacol 2001;75:141–64.10.1016/S0378-8741(00)00394-9Search in Google Scholar

6. Choi YH, Chin YW, Kim YG. Herb-drug interactions: focus on metabolic enzymes and transporters. Arch Pharm Res 2011;34:1843–63.10.1007/s12272-011-1106-zSearch in Google Scholar PubMed

7. Na DH, Ji HY, Park EJ, Kim MS, Liu KH, Lee HS. Evaluation of metabolism-mediated herb-drug interactions. Arch Pharm Res 2011;34:1829–42.10.1007/s12272-011-1105-0Search in Google Scholar PubMed

8. Huang SM, Strong JM, Zhang L, Reynolds KS, Nallani S, Temple R, et al. New era in drug interaction evaluation: US Food and Drug Administration update on CYP enzymes, transporters, and the guidance process. J Clin Pharmacol 2008;48:662–70.10.1177/0091270007312153Search in Google Scholar PubMed

9. Guengerich FP. Cytochrome P450s and other enzymes in drug metabolism and toxicity. AAPS J 2006;8:E101–11.10.1208/aapsj080112Search in Google Scholar PubMed PubMed Central

10. Zanger UM, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther 2013;138:103–41.10.1016/j.pharmthera.2012.12.007Search in Google Scholar PubMed

11. Huang SM, Lesko LJ. Drug-drug, drug-dietary supplement, and drug-citrus fruit and other food interactions: what have we learned? J Clin Pharmacol 2004;44:559–69.10.1177/0091270004265367Search in Google Scholar PubMed

12. Zhang L, Zhang YD, Zhao P, Huang SM. Predicting drug–drug interactions: an FDA perspective. AAPS J 2009;11:300–6.10.1208/s12248-009-9106-3Search in Google Scholar

13. Guideline on the investigation of drug interactions. European Medicines Agency, 2010.Search in Google Scholar

14. Guidance for industry: drug interaction studies: study design, data analysis, and implications for dosing and labeling. Rockville, MD: US Food and Drug Administration, 2006 September.Search in Google Scholar

15. Saleem M. Lupeol, a novel anti-inflammatory and anti-cancer dietary triterpene. Cancer Lett 2009;285:109–15.10.1016/j.canlet.2009.04.033Search in Google Scholar

16. Yokoe I, Azuma K, Hata K, Mukaiyama T, Goto T, Tsuka T, et al. Clinical systemic lupeol administration for canine oral malignant melanoma. Mol Clin Oncol 2015;3:89–92.10.3892/mco.2014.450Search in Google Scholar

17. Moghaddam MG, Ahmad FB, Samzadeh-Kermani A. Biological activity of betulinic acid: a review. Pharmacol Pharm 2012;3:119–23.10.4236/pp.2012.32018Search in Google Scholar

18. Kim DS, Pezzuto JM, Pisha E. Synthesis of betulinic acid derivatives with activity against human melanoma. Bioorg Med Chem Lett 1998;8:1707–12.10.1016/S0960-894X(98)00295-9Search in Google Scholar

19. Liu F, He Y, Liang Y, Wen L, Zhu Y, Wu Y, et al. PI3-kinase inhibition synergistically promoted the anti-tumor effect of lupeol in hepatocellular carcinoma. Cancer Cell Int 2013;13:108–14.10.1186/1475-2867-13-108Search in Google Scholar

20. Walawalkar PS, Serai PS, Iyer KR. Isolation and catalytic competence of different animal liver microsomal fractions prepared by calcium-aggregation method. Indian J Pharm Sci 2006;68:262–5.10.4103/0250-474X.25733Search in Google Scholar

21. Omura T, Sato R. The carbon-monoxide binding pigment of liver microsomes. J Biol Chem 1965;239:1867–73.10.1016/S0021-9258(20)82244-3Search in Google Scholar

22. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248–54.10.1016/0003-2697(76)90527-3Search in Google Scholar

23. Pelkonen O, Turpeinen M, Hakkola J, Honkakosk P, Hukkanen J, Raunio H. Inhibition and induction of human cytochrome P450 enzymes: current status. Arch Toxicol 2008;82:667–15.10.1007/s00204-008-0332-8Search in Google Scholar

24. Subash S, Seervi M, Iyer K, Damre A. Assessment of rat hepatic cytochrome P450 induction and inhibition potential of chlorogenic acid. Indian J Pharm Educ 2011;45:32–9.Search in Google Scholar

25. Hanioka N, Tatarazako N, Jinno H, Arizono K, Ando M. Determination of cytochrome P450 1A activities in mammalian liver microsomes by high-performance liquid chromatography with fluorescence detection. J Chromatogr B Biomed Sci Appl 2000;744:399–406.10.1016/S0378-4347(00)00278-4Search in Google Scholar

26. Jurica J, Konecny J, Zahradnikova LZ, Tomandl J. Simultaneous HPLC determination of tolbutamide, phenacetin and their metabolites as markers of cytochromes 1A2 and 2C6/11 in rat liver perfusate. J Pharm Biomed Anal 2010;52:557–64.10.1016/j.jpba.2010.01.028Search in Google Scholar

27. Yu A, Haining RL. Comparative contribution to dextromethorphan metabolism by cytochrome P450 isoforms in vitro: can dextromethorphan be used as a dual probe for both CYP2D6 and CYP3A activities. Drug Metab Dispos 2001;29:1514–20.Search in Google Scholar

28. Fayer JL, Petullo DM, Ring BJ, Wrighton SA, Ruterbories KJ. A novel testosterone 6β-hydroxylase activity assay for the study of CYP3A-mediated metabolism, inhibition, and induction in vitro. J Pharmacol Toxicol Methods 2001;46:117–23.10.1016/S1056-8719(02)00168-5Search in Google Scholar

29. Franco SG, Dominguez G, Pico JC. Alternatives in the induction and preparation of phenobarbital/naphthoflavone-induced S9 and their activation profiles. Mutagenesis 1999;14:323–6.10.1093/mutage/14.3.323Search in Google Scholar PubMed

30. Ramasamy S, Kiew LV, Chung LY. Inhibition of human cytochrome P450 enzymes by Bacopa monnieri standardized extract and constituents. Molecules 2014;19:2588–601.10.3390/molecules19022588Search in Google Scholar PubMed PubMed Central

31. Martignoni M, Groothuis GM, De KR. Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol 2006;2:875–94.10.1517/17425255.2.6.875Search in Google Scholar PubMed

32. Sun M, Tang Y, Ding T, Liu M, Wang X. Investigation of cytochrome P450 inhibitory properties of maslinic acid, a bioactive compound from Olea europaea L., and its structure-activity relationship. Phytomedicine 2015;22:56–65.10.1016/j.phymed.2014.10.003Search in Google Scholar PubMed

33. Forrester LM, Henderson CJ, Glancey MJ, Back DJ, Park BK, Ball SE, et al. Relative expression of cytochrome P450 isoenzymes in human liver and association with the metabolism of drugs and xenobiotics. Biochem J 1992;281:359–68.10.1042/bj2810359Search in Google Scholar PubMed PubMed Central

34. Boobis AR, Sesardic D, Murray BP, Edwards RJ, Singleton AM, Rich KJ, et al. Species variation in the response of the cytochrome P-450-dependent monooxygenase system to inducers and inhibitors. Xenobiotica 1990;20:1139–61.10.3109/00498259009046835Search in Google Scholar PubMed

35. Bailey DG, Malcolm J, Arnold O, David Spence J. Grapefruit juice-drug interactions. Br J Clin Pharmacol 1998;46:101–10.10.1046/j.1365-2125.1998.00764.xSearch in Google Scholar PubMed PubMed Central

36. Ho BE, Shen DD, McCune JS, Bui T, Risler L, Yang Z, et al. Effects of garlic on cytochromes P450 2C9 and 3A4 mediated drug metabolism in human hepatocytes. Sci Pharm 2010;78:473–81.10.3797/scipharm.1002-11Search in Google Scholar PubMed PubMed Central

37. Zhou S, Gao Y, Jiang W, Huang M, Xu A, Paxton JW. Interactions of herbs with cytochrome P450. Drug Metab Rev 2003;35:35–98.10.1081/DMR-120018248Search in Google Scholar

38. Sun M, Tang Y, Ding T, Liu M, Wang X. Inhibitory effects of celastrol on rat liver cytochrome P450 1A2, 2C11, 2D6, 2E1 and 3A2 activity. Fitoterapia 2014;92:1–8.10.1016/j.fitote.2013.10.004Search in Google Scholar PubMed

39. Wang X, Lee WYW, Or PMY, Yeung JHK. Pharmacokinetic interaction studies of tanshinones with tolbutamide, a model CYP2C11 probe substrate, using liver microsomes, primary hepatocytes and in vivo in the rat. Phytomedicine 2010;17:203–11.10.1016/j.phymed.2009.07.013Search in Google Scholar PubMed

40. Wang X, Lee WY, Zhou X, Or PM, Yeung JH. A pharmacodynamic-pharmacokinetic (PD–PK) study on the effects of Danshen (Salvia miltiorrhiza) on midazolam, a model CYP3A probe substrate, in the rat. Phytomedicine 2010;17:876–83.10.1016/j.phymed.2010.05.007Search in Google Scholar PubMed

Received: 2015-12-2
Accepted: 2016-1-19
Published Online: 2016-3-9
Published in Print: 2016-6-1

©2016 by De Gruyter

Downloaded on 15.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/dmpt-2015-0043/html
Scroll to top button