Skip to main content
Log in

Prediction of total propofol clearance based on enzyme activities in microsomes from human kidney and liver

  • Pharmacokinetics and Disposition
  • Published:
European Journal of Clinical Pharmacology Aims and scope Submit manuscript

Abstract

Objective

Propofol is commonly used for anesthesia and sedation in intensive care units. Approximately 53% of injected propofol is excreted in the urine as the glucuronide and 38% as hydroxylated metabolites. Liver, kidneys and intestine are suspected as clearance tissues. We investigated the contribution of the liver and kidneys to propofol metabolism in humans using an in vitro–in vivo scale up approach.

Methods

Renal tissue was obtained from five patients who received nephrectomies. Each renal hydroxylation and glucuronidation enzymatic activities in microsomal fractions from patients were performed discretely and their estimation based on the decrease of propofol concentration. Hepatic hydroxylation and glucuronidation activities were also performed separately using human liver microsomes. This estimation is based on the decrease of propofol concentration, assuming that the contribution of hydroxylation activity without NADPH-generating system and glucuronidation activity without UDPGA in each microsomal fraction are negligible. Both renal and hepatic clearances were estimated assuming a well-stirred model.

Results

Enzymatic activity of propofol oxidation in renal microsomes was negligible. Although glucuronidation activity in microsomes from kidneys was comparable to that from liver, the hepatic intrinsic clearance predicted from in vitro study was higher than that in kidneys due to the larger tissue volume and higher protein concentration. However, glucuronidation clearance in kidney is relatively similar to that in liver because of blood flow limitation of clearance in both tissues.

Conclusion

The high degree of hydroxylation activity in liver microsomes is consistent with the blood flow-limited hepatic clearance of propofol. Although the activity of propofol glucuronidation in liver is higher, glucuronidation in kidney may be a substantial contributor.

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.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Hiraoka H, Yamamoto K, Okano N, Morita T, Goto F, Horiuchi R (2004) Changes in drug plasma concentrations of extensively bound and highly extracted drug, propofol, in response to altered plasma binding. Clin Pharmacol Ther 75:324–330

    Article  PubMed  CAS  Google Scholar 

  2. Takizawa D, Hiraoka H, Nakamura K, Yamamoto K, Horiuchi R (2004) Propofol concentrations during the anhepatic phase of living-related donor liver transplantation. Clin Pharmacol Ther 76:648–649

    Article  PubMed  CAS  Google Scholar 

  3. Takizawa D, Hiraoka H, Goto F, Yamamoto K, Horiuchi R (2005) Human kidneys play an important role in the elimination of propofol. Anesthesiology 102:327–330

    Article  PubMed  CAS  Google Scholar 

  4. Hiraoka H, Yamamoto K, Miyoshi S, Morita T, Nakamura K, Kadoi Y, Kunimoto F, Horiuchi R (2005) Kidneys contribute to the extrahepatic clearance of propofol in humans but not lungs or brain. Br J Clin Pharmacol 60:176–182

    Article  PubMed  CAS  Google Scholar 

  5. Favetta P, Degoute CS, Perdrix JP, Dufresne C, Boulieu R, Guitton J (2002) Propofol metabolites in man following propofol induction and maintenance. Br J Anaesth 88:653–658

    Article  PubMed  CAS  Google Scholar 

  6. Court MH, Duan SX, Hesse LM, Venkatakrishnan K, Greenblatt DJ (2001) Cytochrome P-450 2B6 is responsible for interindividual variability of propofol hydroxylation by human liver microsomes. Anesthesiology 94:110–119

    Article  PubMed  CAS  Google Scholar 

  7. Oda Y, Hamaoka N, Hiroi T, Imaoka S, Hase I, Tanaka K, Funae Y, Ishizaki T, Asada A (2001) Involvement of human liver cytochrome P4502B6 in the metabolism of propofol. Br J Clin Pharmacol 51:281–285

    Article  PubMed  CAS  Google Scholar 

  8. Ethell BT, Beaumont K, Rance DJ, Burchell B (2001) Use of cloned and expressed human UDP-glucuronosyltransferases for the assessment of human drug conjugation and identification of potential drug interactions. Drug Metab Dispos 29:48–53

    PubMed  CAS  Google Scholar 

  9. Cheng Z, Radominska-Pandya A, Tephly TR (1999) Studies on the substrate specificity of human intestinal UDP-glucuronosyltransferases 1A8 and 1A10. Drug Metab Dispos 27:1165–1170

    PubMed  CAS  Google Scholar 

  10. Raoof AA, van Obbergh LJ, de Ville de Goyet J, Verbeeck RK (1996) Extrahepatic glucuronidation of propofol in man: possible contribution of gut wall and kidney. Eur J Clin Pharmacol 50:91–96

    Article  PubMed  CAS  Google Scholar 

  11. McGurk KA, Brierley CH, Burchell B (1998) Drug glucuronidation by human renal UDP-glucuronosyltransferases. Biochem Pharmacol 55:1005–1012

    Article  PubMed  CAS  Google Scholar 

  12. Kitagawa H, Kamataki T (1971) Studies on drug metabolism. XII. Activity of liver microsomal drug-metabolizing enzymes in human liver. Chem Pharm Bull (Tokyo) 19:827–830

    CAS  Google Scholar 

  13. Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein measurement with the folin phenol reagent. J Biol Chem 193:265–275

    PubMed  CAS  Google Scholar 

  14. Fisher MB, Campanale K, Ackermann BL, VandenBranden M, Wrighton SA (2000) In vitro glucuronidation using Human Liver microsomes and the pore-forming peptide alamethicin. Drug Metab Dispos 28:560–566

    PubMed  CAS  Google Scholar 

  15. Vree TB, Baars AM, de Grood PM (1987) High performance liquid chromatographic determination and preliminary pharmacokinetics of propofol and its metabolites in human plasma and urine. J Chromatog 417:458–464

    Article  CAS  Google Scholar 

  16. Carlile DJ, Stevens AJ, Ashforth EI, Waghela D, Houston JB (1998) In vivo clearance of ethoxycoumarin and its prediction from In vitro systems. Use Of drug depletion and metabolite formation methods in hepatic microsomes and isolated hepatocytes. Drug Metab Dispos 26:216–221

    PubMed  CAS  Google Scholar 

  17. Omura T, Sato R (1964) The carbon monoxide-binding pigment of liver microsomes. I. Evidence for its hemoprotein nature. J Biol Chem 239:2370–2378

    PubMed  CAS  Google Scholar 

  18. Strobel H, Dignam D (1978) Purification and properties of NADPH-cytochrome P450 reductase. In: Fleischer S, Packer L (eds) Methods in enzymology, vol. 52. Academic Press, New York, pp 89–96

  19. Wilson ZE, Rostami-Hodjegan A, Burn JL, Tooley A, Boyle J, Ellis SW, Tucker GT (2003) Inter-individual variability in levels of human microsomal protein and hepatocellularity per gram of liver. Br J Clin Pharmacol 56:433–40. Erratum in: Br J Clin Pharmacol 2004 Dec;58:680

    Article  PubMed  CAS  Google Scholar 

  20. Gillette J (1963) Drug metabolism by enzyme mechanism. Prog Drug Res 6:55–57

    Google Scholar 

  21. Bickel MH, Steele JW (1974) Binding of basic and acidic drugs to rat tissue subcellular fractions. Chem-Biol Interact 8:151–162

    Article  PubMed  CAS  Google Scholar 

  22. Di Francesco C, Bickel MH (1977) Membrane lipids as intracellular binders of chlorpromazine and related drugs. Chem-Biol Interact 16:335–346

    Article  PubMed  CAS  Google Scholar 

  23. Obach RS (1997) Non specific binding to microsomes: Impact on scale-up of in vitro intrinsic clearance to hepatic clearance as assessed through examination of warfarin, imipramine and propranolol. Drug Metab Dispos 25:1359–1369

    PubMed  CAS  Google Scholar 

  24. Soars MG, Burchell B, Riley RJ (2002) In vitro analysis of human drug glucuronidation and prediction of in vivo metabolic clearance. J Pharmacol ExpTher 30:382–390

    Google Scholar 

  25. Iwatsubo T, Hirota N, Ooie T, Suzuki H, Shimada N, Chiba K, Ishizaki T, Green CE, Tyson CA, Sugiyama Y (1997) Prediction of in vivo drug metabolism in the human liver from in vitro metabolism data. Pharmacol Ther 73:147–171

    Article  PubMed  CAS  Google Scholar 

  26. Houston JB (1994) Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol 47:1469–1479

    Article  PubMed  CAS  Google Scholar 

  27. Veroli P, O’Kelly B, Bertrand F, Trouvin JH, Farinotti R, Ecoffey C (1992) Extrahepatic metabolism of propofol in man during the anhepatic phase of orthotopic liver transplantation. Br J Anaesth 68:183–186

    Article  PubMed  CAS  Google Scholar 

  28. Aleksa K, Matsell D, Krausz K, Gelboin H, Ito S, Koren G (2005) Cytochrome P450 3A and 2B6 in the developing kidney: implications for ifosfamide nephrotoxicity. Pediatr Nephrol 20:872–885

    Article  PubMed  Google Scholar 

  29. Berg CL, Radominska A, Lester R, Gollan JL (1995) Membrane translocation and regulation of uridine diphosphate-glucuronic acid uptake in rat liver microsomal vesicles. Gastroenterology 108:183–192

    Article  PubMed  CAS  Google Scholar 

  30. Yokota H, Ando F, Iwano H, Yuasa A (1998) Inhibitory effect of uridine diphosphate on UDP- glucuronosyltransferase. Life Sci 63:1693–1699

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank Daisuke Yoshikawa, MD, PhD, and Daisuke Takizawa, MD, for their technical assistance. The experiments comply with the current law of the country in which they were performed inclusive of ethics approval. Financial support was provided solely from institutional and/or departmental sources.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ryuya Horiuchi.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Al-Jahdari, W.S., Yamamoto, K., Hiraoka, H. et al. Prediction of total propofol clearance based on enzyme activities in microsomes from human kidney and liver. Eur J Clin Pharmacol 62, 527–533 (2006). https://doi.org/10.1007/s00228-006-0130-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00228-006-0130-2

Keywords

Navigation