Skip to main content

Advertisement

Log in

Regioselective Glucuronidation of Andrographolide and Its Major Derivatives: Metabolite Identification, Isozyme Contribution, and Species Differences

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

An Erratum to this article was published on 18 October 2014

Abstract

Andrographolide (AND) and two of its derivatives, deoxyandrographolide (DEO) and dehydroandrographolide (DEH), are widely used in clinical practice as anti-inflammatory agents. However, UDP-glucuronosyltransferase (UGT)-mediated phase II metabolism of these compounds is not fully understood. In this study, glucuronidation of AND, DEO, and DEH was characterized using liver microsomes and recombinant UGT enzymes. We isolated six glucuronides and identified them using 1D and 2D nuclear magnetic resonance (NMR) spectroscopy. We also systematically analyzed various kinetic parameters (K m, V max, and CLint) for glucuronidation of AND, DEO, and DEH. Among 12 commercially available UGT enzymes, UGT1A3, 1A4, 2B4, and 2B7 exhibited metabolic activities toward AND, DEO, and DEH. Further, UGT2B7 made the greatest contribution to glucuronidation of all three anti-inflammatory agents. Regioselective glucuronidation showed considerable species differences. 19-O-Glucuronides were present in liver microsomes from all species except rats. 3-O-Glucuronides were produced by pig and cynomolgus monkey liver microsomes for all compounds, and 3-O-glucuronide of DEH was detected in mouse and rat liver microsomes (RLM). Variations in K m values were 48.6-fold (1.93–93.6 μM) and 49.5-fold (2.01–99.1 μM) for 19-O-glucuronide and 3-O-glucuronide formation, respectively. Total intrinsic clearances (CLint) for 3-O- and 19-O-glucuronidation varied 4.8-fold (22.7–110 μL min−1 mg−1), 10.6-fold (94.2–991 μL min−1 mg−1), and 8.3-fold (122–1,010 μL min−1 mg−1), for AND, DEH, and DEO, respectively. Our results indicate that UGT2B7 is the major UGT enzyme involved in the metabolism of AND, DEO, and DEH. Metabolic pathways in the glucuronidation of AND, DEO, and DEH showed considerable species differences.

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

Similar content being viewed by others

Abbreviations

AND:

Andrographolide

DEH:

Dehydroandrographolide

DEO:

Deoxyandrographolide

HLM:

Human liver microsomes

PLM:

Pig liver microsomes

RLM:

SD rat liver microsomes

MLM:

Mouse liver microsomes

DLM:

Dog liver microsomes

CyLM:

Monkey liver microsomes

UDPGA:

Uridine diphosphate glucuronic acid

UGTs:

UDP-glucuronosyltransferases

AND-1:

Andrographolide-19-O-β-d-glucuronide

AND-2:

Andrographolide-3-O-β-d-glucuronide

DEH-1:

Dehydroandrographolide-19-O-β-d-glucuronide

DEH-2:

Dehydroandrographolide-3-O-β-d-glucuronide

DEO-1:

Deoxyandrographolide-19-O-β-d-glucuronide

DEO-2:

Deoxyandrographolide-3-O-β-d-glucuronide

REFERENCES

  1. Wang J, Yang W, Wang G, Tang P, Sai Y. Determination of six components of Andrographis paniculata extract and one major metabolite of andrographolide in rat plasma by liquid chromatography-tandem mass spectrometry. J Chromatogr B. 2014;951:78–88.

    Article  Google Scholar 

  2. Dai GF, Zhao J, Jiang ZW, Zhu LP, Xu HW, Ma WY, et al. Anti-inflammatory effect of novel andrographolide derivatives through inhibition of NO and PGE2 production. Int Immunopharmacol. 2011;11(12):2144–9.

    Article  CAS  PubMed  Google Scholar 

  3. Chao WW, Kuo YH, Lin BF. Anti-inflammatory activity of new compounds from Andrographis paniculata by NF-κB transactivation inhibition. J Agric Food Chem. 2010;58(4):2505–12.

    Article  CAS  PubMed  Google Scholar 

  4. Thisoda P, Rangkadilok N, Pholphana N, Worasuttayangkurn L, Ruchirawat S, Satayavivad J. Inhibitory effect of Andrographis paniculata extract and its active diterpenoids on platelet aggregation. Eur J Pharmacol. 2006;553:39–45.

    Article  CAS  PubMed  Google Scholar 

  5. Yu BC, Hung CR, Chen WC, Cheng JT. Antihyperglycemic effect of andrographolide in streptozotocin-induced diabetic rats. Planta Med. 2003;69:1075–9.

    Article  CAS  PubMed  Google Scholar 

  6. Shukla B, Visen PKS, Patnaik GK, Dhawan BN. Choleretic effect of andrographolide in rats and guinea pigs. Planta Med. 1992;58:146–9.

    Article  CAS  PubMed  Google Scholar 

  7. Trivedi NP, Rawal UM, Patel BP. Potency of andrographolide as an antitumor compound in BHC-induced liver damage. Integr Cancer Ther. 2009;8:177–89.

    Article  CAS  PubMed  Google Scholar 

  8. Uttekar MM, Das T, Pawar RS, Bhandari B, Menon V, Nutan Gupta SK, et al. Anti-HIV activity of semisynthetic derivatives of andrographolide and computational study of HIV-1 gp120 protein binding. Eur J Med Chem. 2012;56:368–74.

    Article  CAS  PubMed  Google Scholar 

  9. Panossian A, Hovhannisyan A, Mamikonyan G, Abrahamian H, Hambardzumyan E, Gabrielian E, et al. Pharmacokinetic and oral bioavailability of andrographolide from Andrographis paniculata fixed combination Kan Jang in rats and human. Phytomedicine. 2000;7:351–64.

    Article  CAS  PubMed  Google Scholar 

  10. Zhao HY, Hu H, Wang YT. Comparative metabolism and stability of andrographolide in liver microsomes from humans, dogs and rats using ultra-performance liquid chromatography coupled with triple-quadrupole and Fourier transform ion cyclotron resonance mass spectrometry. Rapid Commun Mass Spectrom. 2013;27:1385–92.

    Article  CAS  PubMed  Google Scholar 

  11. Cui L, Qiu F, Yao X. Isolation and identification of seven glucuronide conjugates of andrographolide in human urine. Drug Metab Dispos. 2005;33:555–62.

    Article  CAS  PubMed  Google Scholar 

  12. Cui L, Qiu F, Wang N, Yao X. Four new andrographolide metabolites in human urine. Chem Pharm Bull (Tokyo). 2004;52:772–5.

    Article  CAS  Google Scholar 

  13. Cui L, Chan W, Qiu F, Cai Z, Yao X. Identification of four urea adducts of andrographolide in humans. Drug Metab Lett. 2008;2(4):261–8.

    Article  CAS  PubMed  Google Scholar 

  14. Qiu F, Cui L, Chen L, Sun J, Yao X. Two novel creatinine adducts of andrographolide in human urine. Xenobiotica. 2012;42(9):911–6.

    Article  CAS  PubMed  Google Scholar 

  15. Ritter JK. Roles of glucuronidation and UDP-glucuronosyltransferases in xenobiotic bioactivation reactions. Chem Biol Interact. 2000;129:171–93.

    Article  CAS  PubMed  Google Scholar 

  16. Knights KM, Miners JO. Renal UDP-glucuronosyltransferases and the glucuronidation of xenobiotics and endogenous mediators. Drug Metab Rev. 2010;42(1):63–73.

    Article  CAS  PubMed  Google Scholar 

  17. Wu BJ, Kulkarni K, Basu S, Zhang S, Hu M. First-pass metabolism via UDP-glucuronosyltransferase: barrier to oral bioavailability of phenolics. J Pharm Sci. 2011;100(9):3655–81.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  18. Tukey RH, Strassburg CP. Human UDP-glucuronosyltransferases: metabolism, expression, and disease. Annu Rev Pharmacol Toxicol. 2000;40:581–616.

    Article  CAS  PubMed  Google Scholar 

  19. Stingl JC, Bartels H, Viviani R, Lehmann ML, Brockmöller J. Relevance of UDP-glucuronosyltransferase polymorphisms for drug dosing: a quantitative systematic review. Pharmacol Ther. 2014;141(1):92–116.

    Article  CAS  PubMed  Google Scholar 

  20. Meech R, Miners JO, Lewis BC, Mackenzie PI. The glycosidation of xenobiotics and endogenous compounds: versatility and redundancy in the UDP glycosyltransferase superfamily. Pharmacol Ther. 2012;134(2):200–18.

    Article  CAS  PubMed  Google Scholar 

  21. Lévesque E, Delage R, Benoit-Biancamano MO, Caron P, Bernard O, Couture F, et al. The impact of UGT1A8, UGT1A9, and UGT2B7 genetic polymorphisms on the pharmacokinetic profile of mycophenolic acid after a single oral dose in healthy volunteers. Clin Pharmacol Ther. 2007;81(3):392–400.

    Article  PubMed  Google Scholar 

  22. Yang M, Wang J, Kong L. Quantitative analysis of four major diterpenoids in Andrographis paniculata by 1H NMR and its application. J Pharm Biomed Anal. 2012;70:87–93.

    Article  CAS  PubMed  Google Scholar 

  23. Zhou Q, Zheng ZJ, Xia BJ, Tang L, Lv C, Liu W, et al. Use of isoform-specific UGT metabolism to determine and describe rates and profiles of glucuronidation of wogonin and oroxylin A by human liver and intestinal microsomes. Pharm Res. 2010;27:1568–83.

    Article  PubMed Central  PubMed  Google Scholar 

  24. He YQ, Liu Y, Zhang BF, Liu HX, Lu YL, Yang L, et al. Identification of the UDP-glucuronosyltransferase isozyme involved in senecionine glucuronidation in human liver microsomes. Drug Metab Dispos. 2010;38(4):626–34.

    Article  CAS  PubMed  Google Scholar 

  25. Court MH, Krishnaswamy S, Hao Q, Duan SX, Patten CJ, Von Moltke LL, et al. Evaluation of 3′-azido-3′-deoxythymidine, morphine, and codeine as probe substrates for UDP-glucuronosyltransferase 2B7 (UGT2B7) in human liver microsomes: specificity and influence of the UGT2B7*2 polymorphism. Drug Metab Dispos. 2003;31(9):1125–33.

    Article  CAS  PubMed  Google Scholar 

  26. Chen M, LeDuc B, Kerr S, Howe D, Williams DA. Identification of human UGT2B7 as the major isoform involved in the O-glucuronidation of chloramphenicol. Drug Metab Dispos. 2010;38(3):368–75.

    Article  CAS  PubMed  Google Scholar 

  27. Takeshi I, Miki N, Ryoichi F, Hiroyuki Y, Tatsuki F, Masataka T, et al. Quantitative analysis of UDP-glucuronosyltransferase (UGT) 1A and UGT2B expression levels in human livers. Drug Metab Dispos. 2009;37(8):1759–68.

    Article  Google Scholar 

  28. Uchaipichat V, Mackenzie PI, Elliot DJ, Miners JO. Selectivity of substrate (trifluoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid, hecogenin, phenylbutazone, quinidine, quinine, and sulfinpyrazone) “probes” for human UDP-glucuronosyltransferases. Drug Metab Dispos. 2006;34(3):449–56.

    CAS  PubMed  Google Scholar 

  29. He YJ, Fang ZZ, Ge GB, Jiang P, Jin HZ, Zhang WD, et al. The inhibitory effect of 20(S)-protopanaxatriol (ppt) towards UGT1A1 and UGT2B7. Phytother Res. 2013;27:628–32.

    Article  CAS  PubMed  Google Scholar 

  30. Zhu L, Ge G, Zhang H, Liu H, He G, Liang S, et al. Characterization of hepatic and intestinal glucuronidation of magnolol: application of the relative activity factor approach to decipher the contributions of multiple UDP-glucuronosyltransferase isoforms. Drug Metab Dispos. 2012;40(3):529–38.

    Article  CAS  PubMed  Google Scholar 

  31. Iwuchukwu OF, Nagar S. Resveratrol (trans-resveratrol, 3,5,4’-trihydroxy-trans-stilbene) glucuronidation exhibits atypical enzyme kinetics in various protein sources. Drug Metab Dispos. 2008;36(2):322–30.

    Article  CAS  PubMed  Google Scholar 

  32. Lin Q, Kuang Y, Huang L, Yao X, Wang Z, Wang D, et al. Determination of lactones in Andrographis Herba and its preparation by quantitative analysis of multi-components by single-marker. Chin Tradit Herb Drugs. 2012;43:2406–11.

    CAS  Google Scholar 

  33. Li J, Huang W, Zhang H, Wang X, Zhou H. Synthesis of andrographolide derivatives and their TNF-α and IL-6 expression inhibitory activities. Bioorg Med Chem Lett. 2007;17(24):6891–4.

    Article  CAS  PubMed  Google Scholar 

  34. Jada SR, Subur GS, Matthews C, Hamzah AS, Lajis NH, Saad MS, et al. Semisynthesis and in vivo anticancer activities of andrographolide analogues. Phytochemistry. 2007;68(6):904–12.

    Article  CAS  PubMed  Google Scholar 

  35. Xin XL, Ma XC, Zhang BJ, Su DH, Wu ZM, Wang XJ, et al. Microbial transformation of dehydroandrographolide by Cunninghamella elegans. J Asian Nat Prod Res. 2009;11(2):187–91.

    Article  CAS  PubMed  Google Scholar 

  36. Deng S, Zhang BJ, Wang CY, Tian Y, Yao JH, An L, et al. Microbial transformation of deoxyandrographolide and their inhibitory activity on LPS- induced NO production in RAW 264.7 macrophages. Bioorg Med Chem Lett. 2012;22:1615–8.

    Article  CAS  PubMed  Google Scholar 

  37. Liu W, Tang L, Ye L, Cai Z, Xia B, Zhang J, et al. Species and gender differences affect the metabolism of emodin via glucuronidation. AAPS J. 2010;12(3):424–36.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Coffman BL, Rios GR, King CD, Tephly TR. Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos. 1997;25(1):1–4.

    CAS  PubMed  Google Scholar 

  39. Engtrakul JJ, Foti RS, Strelevitz TJ, Fisher MB. Altered AZT (3-azido-3-deoxythymi-dine) glucuronidation kinetics in liver microsomes as an explanation for underprediction of in vivo clearance: comparison to hepatocytes and effect of incubation environment. Drug Metab Dispos. 2005;33(11):1621–7.

    Article  CAS  PubMed  Google Scholar 

  40. Ge GB, Ning J, Hu LH, Dai ZR, Hou J, Cao YF, et al. A highly selective probe for human cytochrome P450 3A4: isoform selectivity, kinetic characterization and its applications. Chem Commun. 2013;49(84):9779–81.

    Article  CAS  Google Scholar 

Download references

ACKNOWLEDGMENTS

We thank National Natural Science Foundation of China (No. 81073013,81473334, 81274047 and 81202589), Dalian Outstanding Youth Science and Technology Talent, Programs for Liaoning Excellent Talents and New Century Excellent Talents in University (NCET) for financial support.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Xiaochi Ma.

Additional information

Xiangge Tian, Sicheng Liang, and Chao Wang contributed equally to this work.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Fig. S1

Representative HPLC chromatograms (left A, C, E) at 225 nm, 254 nm, and 210 nm and UV spectra (right B, D, F) spectra of Andrographolide, Dehydroandrographolide, and Deoxyandrographolide and their glucuronides among different species, respectively. The UV spectra of their glucuronides were similar with that of substrates. (DOC 217 kb)

Fig. S2

Chemical inhibition of Andrographolide (A), Dehydroandrographolide (B), Deoxyandrographolide (C) O-glucuronidation at different concentration of substrates by four potent inhibitors including diclofenac (400 μM), 20(S)-protopanaxatriol (100 μM), fluconazole (5 mM) and phenylbutazone (500 μM), in HLMs incubations. The incubations without inhibitors but the same volume of solvent were set as the solvent control in which the activity of three substrates O-glucuronidation was normalized to 100%. (DOC 137 kb)

Fig. S3

Correlation analysis between the formation rate of Andrographolide (A, 3 μM; B, 300 μM), Dehydroandrographolide (C, 20 μM; D, 200 μM) and Deoxyandrographolide (E, 2 μM; F, 100 μM) 19-O-glucuronides with UGT2B7-catalyzed AZT glucuronidation in HLMs from 11 individual. (DOC 158 kb)

Fig. S4

Eadie–Hofstee plots of glucuronidation profiles shown in Fig. 5 to determine the best-fit equation. (DOC 254 kb)

Fig. S5

Evaluation of DEO and DEH inhibition toward HLM catalyzed 19-O-glucuronide formation. A and C: Dixon plot of DEO and DEH’s inhibition toward AND 19-O-glucuronidation in HLMs; B and D: Lineweaver–Burk plots of DEO and DEH’s inhibition toward AND 19-O-glucuronidation in HLMs. The data point represents the mean of duplicate experiments. (DOC 165 kb)

Fig. S6

Kinetic profiles (A, B, C, D, E) of 19-O-β-glucuronidation derived from incubation of Andrographolide, Dehydroandrographolide, and Deoxyandrographolide with different species liver microsomes from Pig, Dog, Monkey, SDrat and Mouse, respectively. (DOC 298 kb)

Fig. S7

Kinetic profiles (A, B, C, D) of 3-O-β-glucuronidation derived from incubation of Andrographolide, Dehydroandrographolide, and Deoxyandrographolide with different species liver microsomes from Pig, Monkey, SDrat and Mouse, respectively. (DOC 228 kb)

Fig. S8

Kinetic profiles of 19-O-β-glucuronidation derived from incubation of 3-keton-dehydroandrographolide with UGT2B7. V max = 1.54 nmol/min/mg; K m = 22.1 μM. (DOC 322 kb)

Fig. S9

NMR spectra of in vitro metabolites of AND, DEH and DEO. (DOC 2,911 kb)

Table S1

1H NMR (600 MHz, MeOD) spectral data for metabolites of AND, DEH, and DEO. (DOC 63 kb)

Table S2

Kinetic parameters of 3-O-β-glucuronidation derived from the incubation of Andrographolide, Dehydroandrographolide, and Deoxyandrographolide with different species’ liver microsomes. Vm nmol/min/mg, K m μM, Ki μM. (n = 3) (DOC 37 kb)

Table S3

Inhibitory effects of diclofenac on AND, DEH and DEO glucuronidation in HLM and UGT2B7. The substrate concentrations were about K m values of AND, DEO and DEH. (n = 3) (DOC 30 kb)

Table S4

Inhibitory effects of AND, DEH, DEO and their metabolites against LPS- induced NO production in RAW264.7 macrophages. (n = 3) (DOC 32 kb)

Table S5

Clean of 3-O-β-glucuronidation in different species. CL μl/min · mg. (DOC 32 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tian, X., Liang, S., Wang, C. et al. Regioselective Glucuronidation of Andrographolide and Its Major Derivatives: Metabolite Identification, Isozyme Contribution, and Species Differences. AAPS J 17, 156–166 (2015). https://doi.org/10.1208/s12248-014-9658-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1208/s12248-014-9658-8

KEY WORDS

Navigation