Skip to main content
Log in

Species and Gender Differences Affect the Metabolism of Emodin via Glucuronidation

  • Research Article
  • Published:
The AAPS Journal Aims and scope Submit manuscript

Abstract

The aim of the present study was to define the mechanisms responsible for poor bioavailability of emodin by determining its metabolism using in vitro and in situ disposition models of the intestine and liver. Liver microsomes of mice, rats, guinea pigs, dogs, and humans were used along with the rat intestinal perfusion model and the rat intestinal microsomes. In the rat intestine, excretion rates of emodin-3-O-glucuronide were significantly different (p < 0.05) in four regions of the intestine and were higher in males than in females (p < 0.01). Emodin glucuronidation in liver microsomes was species-dependent, and K m values varied 5.7-fold (3.2–18.2 μM) in males and 2.8-fold (4.6–13.0 μM) in females. The male intrinsic clearance (CLint) values differed by 5-fold (27.6–138.3 mL h−1 mg−1 protein), and female CLint values differed by 4.3-fold (24.3–103.5 mL h−1 mg−1 protein). Since CLint values of emodin glucuronidation were 10-fold higher than that of isoflavones, emodin was considered rapidly glucuronidated. In contrast to the large species-dependent effects on K m and CLint values, gender had a smaller effect on these kinetic parameters (2-fold, p < 0.05). Lastly, glucuronidation rates obtained using liver microsomes from various experimental animals of the same gender correlated well with those in human liver microsomes. In conclusion, Rapid metabolism by UDP-glucuronosyltransferase is the major reason why emodin has poor bioavailability. Species and gender affected emodin metabolism to a different degree, and experimental animals are expected to be useful in predicting emodin glucuronidation in humans.

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
Fig. 8
Fig. 9

Similar content being viewed by others

References

  1. Garg AK, Buchholz TA, Aggarwal BB. Chemosensitization and radiosensitization of tumors by plant polyphenols. Antioxid Redox Signal. 2005;7:1630–47.

    Article  PubMed  CAS  Google Scholar 

  2. Huang Q, Lu G, Shen HM, Chung MC, Ong CN. Anti-cancer properties of anthraquinones from rhubarb. Med Res Rev. 2007;27:609–30.

    Article  PubMed  CAS  Google Scholar 

  3. Srinivas G, Babykutty S, Sathiadevan PP, Srinivas P. Molecular mechanism of emodin action: transition from laxative ingredient to an antitumor agent. Med Res Rev. 2007;27:591–608.

    Article  PubMed  CAS  Google Scholar 

  4. Sakakibara H, Honda Y, Nakagawa S, Ashida H, Kanazawa K. Simultaneous determination of all polyphenols in vegetables, fruits, and teas. J Agric Food Chem. 2003;51:571–81.

    Article  PubMed  CAS  Google Scholar 

  5. Riahi S, Reza Ganjali M, Dinarvand R, Karamdoust S, Bagherzadeh K, Norouzi P. A theoretical study on interactions between mitoxantrone as an anticancer drug and DNA: application in drug design. Chem Biol Drug Des. 2008;71:474–82.

    Article  PubMed  CAS  Google Scholar 

  6. Matsuda Y, Yokohira M, Suzuki S, Hosokawa K, Yamakawa K, Zeng Y, et al. One-year chronic toxicity study of Aloe arborescens Miller var. natalensis Berger in Wistar Hannover rats. A pilot study. Food Chem Toxicol. 2008;46:733–9.

    Article  PubMed  CAS  Google Scholar 

  7. Srinivas G, Anto RJ, Srinivas P, Vidhyalakshmi S, Senan VP, Karunagaran D. Emodin induces apoptosis of human cervical cancer cells through poly(ADP-ribose) polymerase cleavage and activation of caspase-9. Eur J Pharmacol. 2003;473:117–25.

    Article  PubMed  CAS  Google Scholar 

  8. Yi J, Yang J, He R, Gao F, Sang H, Tang X, et al. Emodin enhances arsenic trioxide-induced apoptosis via generation of reactive oxygen species and inhibition of survival signaling. Cancer Res. 2004;64:108–16.

    Article  PubMed  CAS  Google Scholar 

  9. Huang Q, Shen HM, Ong CN. Emodin inhibits tumor cell migration through suppression of the phosphatidylinositol 3-kinase-Cdc42/Rac1 pathway. Cell Mol Life Sci. 2005;62:1167–75.

    Article  PubMed  CAS  Google Scholar 

  10. Huang Q, Shen HM, Ong CN. Inhibitory effect of emodin on tumor invasion through suppression of activator protein-1 and nuclear factor-kappaB. Biochem Pharmacol. 2004;68:361–71.

    Article  PubMed  CAS  Google Scholar 

  11. Wang SC, Zhang L, Hortobagyi GN, Hung MC. Targeting HER2: recent developments and future directions for breast cancer patients. Semin Oncol. 2001;28:21–9.

    Article  PubMed  Google Scholar 

  12. Cha TL, Qiu L, Chen CT, Wen Y, Hung MC. Emodin down-regulates androgen receptor and inhibits prostate cancer cell growth. Cancer Res. 2005;65:2287–95.

    Article  PubMed  CAS  Google Scholar 

  13. Su YT, Chang HL, Shyue SK, Hsu SL. Emodin induces apoptosis in human lung adenocarcinoma cells through a reactive oxygen species-dependent mitochondrial signaling pathway. Biochem Pharmacol. 2005;70:229–41.

    Article  PubMed  CAS  Google Scholar 

  14. Muller SO, Eckert I, Lutz WK, Stopper H. Genotoxicity of the laxative drug components emodin, aloe-emodin and danthron in mammalian cells: topoisomerase II mediated? Mutat Res. 1996;371:165–73.

    Article  PubMed  CAS  Google Scholar 

  15. Huang XZ, Wang J, Huang C, Chen YY, Shi GY, Hu QS, et al. Emodin enhances cytotoxicity of chemotherapeutic drugs in prostate cancer cells: the mechanisms involve ROS-mediated suppression of multidrug resistance and hypoxia inducible factor-1. Cancer Biol Ther. 2008;7:468–75.

    Article  PubMed  CAS  Google Scholar 

  16. Masuda T, Ueno Y. Microsomal transformation of emodin into a direct mutagen. Mutat Res. 1984;125:135–44.

    PubMed  CAS  Google Scholar 

  17. Masuda T, Haraikawa K, Morooka N, Nakano S, Ueno Y. 2-Hydroxyemodin, an active metabolite of emodin in the hepatic microsomes of rats. Mutat Res. 1985;149:327–32.

    PubMed  CAS  Google Scholar 

  18. Morita H, Umeda M, Masuda T, Ueno Y. Cytotoxic and mutagenic effects of emodin on cultured mouse carcinoma FM3A cells. Mutat Res. 1988;204:329–32.

    Article  PubMed  CAS  Google Scholar 

  19. Krivobok S, Seigle-Murandi F, Steiman R, Marzin DR, Betina V. Mutagenicity of substituted anthraquinones in the Ames/Salmonella microsome system. Mutat Res. 1992;279:1–8.

    Article  PubMed  CAS  Google Scholar 

  20. Huang HC, Chang JH, Tung SF, Wu RT, Foegh ML, Chu SH. Immunosuppressive effect of emodin, a free radical generator. Eur J Pharmacol. 1992;211:359–64.

    Article  PubMed  CAS  Google Scholar 

  21. Shia CS, Hou YC, Tsai SY, Huieh PH, Leu YL, Chao PD. Differences in pharmacokinetics and ex vivo antioxidant activity following intravenous and oral administrations of emodin to rats. J Pharm Sci. 2010;99:2185–95.

    PubMed  CAS  Google Scholar 

  22. Teng ZH, Zhou SY, Ran YH, Liu XY, Yang RT, Yang X, et al. Cellular absorption of anthraquinones emodin and chrysophanol in human intestinal Caco-2 cells. Biosci Biotechnol Biochem. 2007;71:1636–43.

    Article  PubMed  CAS  Google Scholar 

  23. Song R, Xu F, Zhang Z, Liu Y, Dong H, Tian Y. Structural elucidation of in vitro metabolites of emodin by liquid chromatography–tandem mass spectrometry. Biomed Chromatogr. 2008;22:1230–6.

    Article  PubMed  CAS  Google Scholar 

  24. Le Guellec C, Lacarelle B, Villard PH, Point H, Catalin J, Durand A. Glucuronidation of propofol in microsomal fractions from various tissues and species including humans: effect of different drugs. Anesth Analg. 1995;81:855–61.

    Article  PubMed  CAS  Google Scholar 

  25. Hu M, Roland K, Ge L, Chen J, Li Y, Tyle P, et al. Determination of absorption characteristics of AG337, a novel thymidylate synthase inhibitor, using a perfused rat intestinal model. J Pharm Sci. 1998;87:886–90.

    Article  PubMed  CAS  Google Scholar 

  26. Hu M, Sinko PJ, deMeere AL, Johnson DA, Amidon GL. Membrane permeability parameters for some amino acids and beta-lactam antibiotics: application of the boundary layer approach. J Theor Biol. 1988;131:107–14.

    Article  PubMed  CAS  Google Scholar 

  27. Tang L, Singh R, Liu Z, Hu M. Structure and concentration changes affect characterization of UGT isoform-specific metabolism of isoflavones. Mol Pharm. 2009;6:1466–82.

    Article  PubMed  CAS  Google Scholar 

  28. Wang SW, Chen J, Jia X, Tam VH, Hu M. Disposition of flavonoids via enteric recycling: structural effects and lack of correlations between in vitro and in situ metabolic properties. Drug Metab Dispos. 2006;34:1837–48.

    Article  PubMed  CAS  Google Scholar 

  29. Hu M, Krausz K, Chen J, Ge X, Li J, Gelboin HL, et al. Identification of CYP1A2 as the main isoform for the phase I hydroxylated metabolism of genistein and a prodrug converting enzyme of methylated isoflavones. Drug Metab Dispos. 2003;31:924–31.

    Article  PubMed  CAS  Google Scholar 

  30. Houston JB, Kenworthy KE. In vitro–in vivo scaling of CYP kinetic data not consistent with the classical Michaelis–Menten model. Drug Metab Dispos. 2000;28:246–54.

    PubMed  CAS  Google Scholar 

  31. Hutzler JM, Tracy TS. Atypical kinetic profiles in drug metabolism reactions. Drug Metab Dispos. 2002;30:355–62.

    Article  PubMed  CAS  Google Scholar 

  32. Bachmann M, Schlatter C. Metabolism of [14C]emodin in the rat. Xenobiotica. 1981;11:217–25.

    Article  PubMed  CAS  Google Scholar 

  33. Chen J, Wang S, Jia X, Bajimaya S, Lin H, Tam VH, et al. Disposition of flavonoids via recycling: comparison of intestinal versus hepatic disposition. Drug Metab Dispos. 2005;33:1777–84.

    PubMed  CAS  Google Scholar 

  34. Coldham NG, Sauer MJ. Pharmacokinetics of [(14)C]Genistein in the rat: gender-related differences, potential mechanisms of biological action, and implications for human health. Toxicol Appl Pharmacol. 2000;164:206–15.

    Article  PubMed  CAS  Google Scholar 

  35. Amidon GL, Sinko PJ, Fleisher D. Estimating human oral fraction dose absorbed: a correlation using rat intestinal membrane permeability for passive and carrier-mediated compounds. Pharm Res. 1988;5:651–54.

    Article  PubMed  CAS  Google Scholar 

  36. Wang SW, Kulkarni KH, Tang L, Wang JR, Yin T, Daidoji T, et al. Disposition of flavonoids via enteric recycling: UDP-glucuronosyltransferase (UGT) 1As deficiency in Gunn rats is compensated by increases in UGT2Bs activities. J Pharmacol Exp Ther. 2009;329:1023–31.

    Article  PubMed  CAS  Google Scholar 

  37. Buckley DB, Klaassen CD. Mechanism of gender-divergent UDP-glucuronosyltransferase mRNA expression in mouse liver and kidney. Drug Metab Dispos. 2009;37:834–40.

    Article  PubMed  CAS  Google Scholar 

  38. Takeuchi T, Tsutsumi O, Nakamura N, Ikezuki Y, Takai Y, Yano T, et al. Gender difference in serum bisphenol A levels may be caused by liver UDP-glucuronosyltransferase activity in rats. Biochem Biophys Res Commun. 2004;325:549–54.

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

This work was mainly supported by Key Project of Chinese National Programs for Fundamental Research and Development (973 program) 2009CB522800, and the Grant of Science and Technology of Guangzhou 2006Z1-E6021, both to ZL. MH was also supported by NIH GM070737.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Ming Hu or Zhongqiu Liu.

Additional information

WL and LT contributed equally to this paper.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Liu, W., Tang, L., Ye, L. et al. Species and Gender Differences Affect the Metabolism of Emodin via Glucuronidation. AAPS J 12, 424–436 (2010). https://doi.org/10.1208/s12248-010-9200-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-010-9200-6

Key words

Navigation