Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter February 25, 2013

Mitragyna speciosa Korth leaves extracts induced the CYP450 catalyzed aminopyrine-N-demethylase (APND) and UDP-glucuronosyl transferase (UGT) activities in male Sprague-Dawley rat livers

  • Juzaili Azizi , Sabariah Ismail EMAIL logo and Sharif Mahsufi Mansor

Abstract

Background:Mitragyna speciosa leaves have been abused by drug addicts as some of the alkaloids (mainly mitragynine) from the plant possess opiate and cocaine-like effects. These bring to its prohibition in Malaysia in 2004 as consumption of M. speciosa leaves has been perceived to lead to the abuse of other drugs such as cannabis and heroin.

Methods: In the current study, the in vitro and in vivo effects of M. speciosa methanolic, aqueous and total alkaloid leaves extracts on drug metabolizing enzymes, namely, cytochrome P450s (CYP450s) and UDP-glucuronosyl transferase (UGT) had been evaluated in rat liver cytosolic fraction and microsomes. Aminopyrine and p-nitrophenol (pNP) were employed as probe substrates in aminopyrine N-demethylase (APND) and UGT enzyme assays, respectively. Furthermore, mitragynine was also tested in vitro for its likelihood to inhibit APND and UGT activity. The assessment of the enzyme activity was conducted using spectrophotometric methods.

Results:In vitro, the IC50 value could only be obtained for the methanolic extract in APND study (595.30±30.78 µg/mL) and not in other studies due to the enzyme percentage inhibitions being <70%. In contrast to the in vitro study, the oral treatment of male Sprague-Dawley rats for 14 days with 50, 100 and 200 mg/kg of methanolic and aqueous extracts and with 5, 10 and 20 mg/kg of total alkaloid extract showed a profound increment on the APND and UGT activities.

Conclusions: The current findings showed that possibilities exist for herb-drug interaction with increased clearance of drugs, which are primarily metabolized by CYP450s and UGT1A6 among M. speciosa leaves extract users.


Corresponding author: Sabariah Ismail, Centre for Drug Research, Universiti Sains Malaysia, 11800 USM Penang, Malaysia, Phone: +60-46533269, Fax: +60-46568669

Conflict of interest statement

Authors’ conflict of interest disclosure: The authors stated that there are no conflicts of interest regarding the publication of this article. Research funding played no role in thestudy design; in the collection, analysis, and interpretationof data; in the writing of the report; or in the decision tosubmit the report for publication.

Research funding: This project was funded by the Ministry of Science, Technology and Innovation (MOSTI) Grant and the Universiti Sains Malaysia (USM) Research University Grant.

Employment or leadership: None declared.

Honorarium: None declared.

Fellowship: Juzaili Azizi is supported by the USM Fellowship Scheme from the Institute of Postgraduate Studies (IPS) of the Universiti Sains Malaysia.

References

1. Reanmongkol W, Keawpradub N, Sawangjaroen K. Effects of the extracts from Mitragyna speciosa Korth. leaves on analgesic and behavioral activities in experimental animals. Songklanakarin J Sci Technol 2007;29:39–48.Search in Google Scholar

2. Tsuchiya S, Miyashita S, Yamamoto M, Horie S, Sakai S-I, Aimi N, etal. Effect of mitragynine, derived from Thai folk medicine, on gastric acid secretion through opioid receptor in anesthetized rats. Eur J Pharmacol 2002;443:185–8.10.1016/S0014-2999(02)01588-1Search in Google Scholar

3. Babu KM, Mccurdy CR, Boyer EW. Opioid receptors and legal highs: Salvia divinorum and kratom opioid receptors and legal highs. Clin Toxicol 2008;46:146–52.10.1080/15563650701241795Search in Google Scholar

4. Chittrakarn S, Sawangjaroen K, Prasettho S, Janchawee B, Keawpradu N. Inhibitory effects of kratom leaf extract (Mitragyna speciosa Korth.) on the rat gastrointestinal tract. J Ethnopharmacol 2008;116:173–8.10.1016/j.jep.2007.11.032Search in Google Scholar

5. Takayama H, Ishikawa H, Kurihara M, Kitajima M, Aimi N, Ponglux D, etal. Studies on the synthesis and opioid aganostic activities of mitragynine-related indole alkaloid: discovery of opioid agonistic structurally different from other opioid ligands. J Med Chem 2002;45:1949–56.10.1021/jm010576eSearch in Google Scholar

6. Yamamoto LT, Horie S, Takayama H, Aimi N, Sakai S, Ponglux D, etal. Opioid receptor agonistic characteristics of mitragynine pseudoindoxyl in comparison with mitragynine derived from Thai medicinal plant Mitragyna speciosa. Gen Pharmacol 1999;33:73–81.10.1016/S0306-3623(98)00265-1Search in Google Scholar

7. Aziz Z, Tey NP. Herbal medicines: prevalence and predictors of use among Malaysian adults. Complement Ther Med 2009;17:44–50.10.1016/j.ctim.2008.04.008Search in Google Scholar PubMed

8. Lynch N, Berry D. Differences in perceived risks and benefits of herbal, over-the-counter conventional, and prescribed conventional, medicines, and the implications of this for the safe and effective use of herbal products. Complement Ther Med 2007;15:84–91.10.1016/j.ctim.2006.06.007Search in Google Scholar PubMed

9. Venkataramanan R, Komoroski B, Strom S. In vitro and in vivo assessment of herb drug interactions. Life Sci 2006;78: 2105–15.10.1016/j.lfs.2005.12.021Search in Google Scholar PubMed

10. Woolf TF. In: Hooper WD, editor. Handbook of drug metabolism. New York: Marcel Dekker, Inc., 1999:229–38.Search in Google Scholar

11. Schensula JJ, Conveya M, Burkholder G. Challenges in measuring concurrency, agency and intentionality in polydrug research. Addict Behav 2005;30:571–4.10.1016/j.addbeh.2004.05.022Search in Google Scholar PubMed

12. Hill SL, Thomas SH. What’s new in. Toxicity of drugs of abuse. Medicine 2009;37:621–6.10.1016/j.mpmed.2009.08.009Search in Google Scholar

13. Axelrod J, Cochin J. The inhibitory action of nalorphine on the enzymatic N-demethylation of narcotic drugs. J Pharmacol Exp Ther 1957;121:107–12.Search in Google Scholar

14. Milthers K. The in vivo transformation of morphine and nalorphine into normorphine in the brain of rats. Acta Pharmacol Toxicol 1962;19:235–40.10.1111/j.1600-0773.1962.tb01699.xSearch in Google Scholar

15. Caraco Y, Tateishi T, Guengerich FP, Wood AJ. Microsomal codeine N-demethylation: cosegregation with cytochrome P4503A4 activity. Drug Metab Dispos 1996;24:761–4.Search in Google Scholar

16. Iribarne C, Berthou F, Baird S, Dreano Y, Picart D, Bail JP, etal. Involvement of cytochrome P450 3A4 enzyme in the N-demethylation of methadone in human liver microsomes. Chem Res Toxicol 1996;9:365–73.10.1021/tx950116mSearch in Google Scholar

17. Alkharfy KM, Frye RF. Effect of valerian, valerian/hops extracts, and valerenic acid on glucuronidation in vitro. Xenobiotica 2007;37:113–23.10.1080/00498250601050420Search in Google Scholar

18. Meech R, Mackenzie PI. Structure and function of uridine diphosphate glucuronosyltransferases. Clin Exp Pharmacol Physiol 1997;24:907–15.10.1111/j.1440-1681.1997.tb02718.xSearch in Google Scholar

19. Bock KW, Kohle C. Topological aspects of oligomeric UDP-glucuronosyltransferases in endoplasmic reticulum membranes: advances and open questions. Biochem Pharmacol 2009;77:1458–65.10.1016/j.bcp.2008.12.004Search in Google Scholar

20. Nishiyama T, Kobori T, Arai K, Ogura K, Ohnuma T, Ishii K, etal. Identification of human UDP-glucuronosyltransferase isoform(s) responsible for the C-glucuronidation of phenylbutazone. Arch Biochem Biophys 2006;454:72–9.10.1016/j.abb.2006.07.010Search in Google Scholar

21. Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, etal. Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos 2004;32:1201–8.10.1124/dmd.104.000794Search in Google Scholar

22. Correia MA. Drug biotransformation. In: Katzung BG, editor. Basic and clinical pharmacology, 9th ed. New York: McGraw-Hill Companies, 2004.Search in Google Scholar

23. Plant N. Strategies for using in vitro screens in drug metabolism. Drug Discov Today 2004;9:328–35.10.1016/S1359-6446(03)03019-8Search in Google Scholar

24. Pomory CM. Color development time of the Lowry protein assay. Anal Biochem 2008;378:216–7.10.1016/j.ab.2008.04.015Search in Google Scholar

25. Omura T, Sato R. The carbon monoxide-binding pigment of liver microsomes: evidence for its hemoprotein nature. J Biol Chem 1964;239:2370–8.10.1016/S0021-9258(20)82244-3Search in Google Scholar

26. Matsubara T, Touchi A, Tochino Y. Hepatic aminopyrine N-demethylase system: further studies of assay procedure. Jpn J Pharmacol 1977;27:127–36.10.1254/jjp.27.127Search in Google Scholar

27. Bock KW, Burchell B, Dutton GD, Hanninen O, Mulder GJ, Owens IS, etal. UDP-glucuronosyl transferase activities: guideline for consistent interim terminology and assay conditions. Biochem Pharmacol 1983;32:953–5.10.1016/0006-2952(83)90610-XSearch in Google Scholar

28. Gibson GG, Skett P. Introduction to drug metabolism. London: Chapman and Hall Ltd, 1986.10.1007/978-1-4899-3188-7Search in Google Scholar

29. Parthasarathy S, Bin Azizi J, Ramanathan S, Ismail S, Sasidharan S, Said MI, etal. Evaluation of antioxidant and antibacterial activities of aqueous, methanolic and alkaloid extracts from Mitragyna speciosa (Rubiaceae family) leaves. Molecules 2009;14:3964–74.10.3390/molecules14103964Search in Google Scholar

30. Moon YJ, Wang X, Morris ME. Dietary flavonoids: effects on xenobiotic and carcinogen metabolism. Toxicol Vitro 2006;20:187–210.10.1016/j.tiv.2005.06.048Search in Google Scholar

31. Wood AW, Smith DS, Chang RL, Huang MT, Conney AH. Effects of flavonoids on the metabolism of xenobiotics. Prog Clin Biol Res 1986;213:195–210.Search in Google Scholar

32. Eastman DF, Segall HJ. Effects of the pyrrolizidine alkaloids senecionine, retrorsine and seneciphylline on aminopyrine N-demethylase activity of the rat liver S-10 fraction. Toxicol Lett 1981;8:217–22.10.1016/0378-4274(81)90104-1Search in Google Scholar

33. Burlingham BT, Widlanski TS. An intuitive look at the relationship of ki and IC50: a more general use for the Dixon plot. J Chem Educ 2003;80:214–8.10.1021/ed080p214Search in Google Scholar

34. Coon MJ. Cytochrome P450: nature’s most versatile biological catalyst. Annu Rev Pharmacol Toxicol 2005;45:1–25.10.1146/annurev.pharmtox.45.120403.100030Search in Google Scholar

35. Liu GT. Effects of some compounds isolated from Chinese medicinal herbs on hepatic microsomal cytochrome P-450 and their potential biological consequences. Drug Metab Rev 1991;23:439–65.10.3109/03602539109029767Search in Google Scholar

36. Yang XF, Wang NP, Lu WH, Zeng FD. Effects of Ginkgo biloba extract and tanshinone on cytochrome P-450 isozymes and glutathione transferase in rats. Acta Pharmacol Sin 2003;24:1033–8.Search in Google Scholar

37. Anwar R, Hussin AH, Ismail S, Mansor SM. In vitro effect of mitragynine (a major alkaloid of Mitragyna speciosa korth) on aminopyrine metabolism in rat hepatocytes. Int J Pharm Sci Res 2012;3:2238–42.Search in Google Scholar

38. Anwar R, Hussin AH, Ismail S, Mansor SM. In vitro effect of mitragynine on activity of drug metabolizing enzymes, N-demethylase and glutathione S-transferase in streptozotocin-induced diabetic rats. Pharmacologyonline 2012;1:68–75.Search in Google Scholar

39. Hodek P, Trefil P, Stiborova M. Flavonoids-potent and versatile biologically active compounds interacting with cytochromes P450. Chem Biol Interact 2002;139:1–21.10.1016/S0009-2797(01)00285-XSearch in Google Scholar

40. Lin JH, Lu AY. Inhibition and induction of cytochrome P450 and the clinical implications. Clin Pharmacokinet 1998;35: 361–90.10.2165/00003088-199835050-00003Search in Google Scholar

41. Oguri K, Yamada H, Yoshimura H. Regiochemistry of cytochrome P450 isozymes. Annu Rev Pharmacol Toxicol 1994;34:251–79.10.1146/annurev.pa.34.040194.001343Search in Google Scholar

42. Imaoka S, Inoue K, Funae Y. Aminopyrine metabolisms by multiple forms of cytochrome P450 from rat liver microsomes: simultaneous quantitation on four aminopyrine metabolites by high-performance liquid chromatography. Arch Biochem Biophys 1988;265:159–70.10.1016/0003-9861(88)90381-5Search in Google Scholar

43. Tsyrlov IB, Gerasimov KE. Aminopyrine-N-demethylase. I. Directed modification of substrates’ structure as a way of production of inducer of the monooxygenase isoform P-450b. Eur J Drug Metabol Pharmacokinet 1991;16:207–12.10.1007/BF03189961Search in Google Scholar

44. Yang CS, Brady JE, Hong JY. Dietary effects on cytochromes P450, xenobiotic metabolism, and toxicity. FASEB J 1992;6:737–44.10.1096/fasebj.6.2.1537464Search in Google Scholar

45. Rodeiro I, Donato MT, Lahoz A, Gonzalez-Lavaut JA, Laguna A, Castell JV, etal. Modulation of P450 enzymes by Cuban natural products rich in polyphenolic compounds in rat hepatocytes. Chem Biol Interact 2008;172:1–10.10.1016/j.cbi.2007.10.004Search in Google Scholar

46. Rodeiro I, Donato MT, Jimenez N, Garrido G, Delgado R, Gomez-Lechon MJ. Effects of Mangifera indica L. aqueous extract (Vimang) on primary culture of rat hepatocytes. Food Chem Toxicol 2007;45:2526–32.10.1016/j.fct.2007.05.027Search in Google Scholar

47. Ikushiro S, Emi Y, Iyanagi T. Identification and analysis of drug responsive expression of UDP-glucuronosyltransferase family 1(UGT1) isozyme in rat hepatic microsomes using anti-peptides antibodies. Arch Biochem Biophys 1995;324:267–72.10.1006/abbi.1995.0039Search in Google Scholar

48. Bock KW, Gschaidmeier H, Heel H, Lehmkoster T, Munzel PA, Bock-Hennig BS. Functions and transcriptional regulation of PAH-inducible human UDP-glucuronosyltransferases. Drug Metab Rev 1999;31:411–22.10.1081/DMR-100101927Search in Google Scholar

49. Hanioka N, Jinno H, Tanaka-Kagawa T, Nishimura T, Ando M. Determination of UDP-glucuronosyltransferase UGT1A6 activity in human and rat liver microsomes by HPLC with UV detection. J Pharma Biomed Anal 2001;25:65–75.10.1016/S0731-7085(00)00491-XSearch in Google Scholar

50. Ueng Y-F, Ko H-C, Chen C-F, Wang J-J, Chen K-T. Modulation of drug-metabolizing enzymes by extracts of a herbal medicine Evodia rutaecarpa in C57BL/6J mice. Life Sci 2002;71:1267–77.10.1016/S0024-3205(02)01854-4Search in Google Scholar

51. Debersac P, Heydel JM, Amiot MJ, Goudonnet H, Artur Y, Suschetet M, etal. Induction of cytochrome P450 and/or detoxication enzymes by various extracts of rosemary: description of specific patterns. Food Chem Toxicol 2001;39:907–18.10.1016/S0278-6915(01)00034-5Search in Google Scholar

52. Teyssier C, Amiot M-J, Mondy N, Auger J, Kahane R, Siess M-H. Effect of onion consumption by rats on hepatic drug-metabolizing enzymes. Food Chem Toxicol 2001;39:981–7.10.1016/S0278-6915(01)00056-4Search in Google Scholar

53. Tepsuwan A, Kupradinun P, Kusamran WR. Effect of Siamese cassia leaves on the activities of chemical carcinogen metabolizing enzymes and on mammary gland carcinogenesis in the rat. Mutat Res 1999;428:363–73.10.1016/S1383-5742(99)00062-9Search in Google Scholar

54. Bock KW, Forstera A, Gschaidmeiera H, Brücka M, Münzela P, Scharecka W, etal. Paracetamol glucuronidation by recombinant rat and human phenol UDP-glucuronosyltransferases. Biochem Pharmacol 1993;45:1809–14.10.1016/0006-2952(93)90437-2Search in Google Scholar

55. Ciotti M, Marrone A, Potter C, Owens IS. Genetic polymorphism in the human UGT1A6 (planar phenol) UDP-glucuronosyltransferase: pharmacological implications. Pharmacogenetics 1997;7:485–95.10.1097/00008571-199712000-00007Search in Google Scholar

56. Ethell BT, Anderson GD, Burchell B. The effect of valproic acid on drug and steroid glucuronidation by expressed human UDP-glucuronosyltransferases. Biochem Pharmacol 2003;65:1441–9.10.1016/S0006-2952(03)00076-5Search in Google Scholar

57. Orzechowski A, Schrenk D, Bock-Hennig BS, Bock KW. Glucuronidation of carcinogenic arylamines and their N-hydroxy derivatives by rat and human phenol UDP-glucuronosyltransferase of the UGT1 gene complex. Carcinogenesis 1994;15:1549–53.10.1093/carcin/15.8.1549Search in Google Scholar PubMed

Received: 2012-11-9
Accepted: 2013-1-23
Published Online: 2013-02-25
Published in Print: 2013-06-01

©2013 by Walter de Gruyter Berlin Boston

Downloaded on 20.4.2024 from https://www.degruyter.com/document/doi/10.1515/dmdi-2012-0039/html
Scroll to top button