DOI QR코드

DOI QR Code

Human Liver Microsomes과 HepG2 세포를 이용한 약물유래 간독성 평가 방법의 최적화

The Optimization of Method for Prediction of Drug-Induced Liver Injury Using HepG2 Cells Cultured with Human Liver Microsomes

  • 투고 : 2015.04.24
  • 심사 : 2015.07.17
  • 발행 : 2015.10.31

초록

The aim of the present study was to optimize in vitro method for the prediction of drug-induced liver injury using human liver microsomes (HLM). Cytotoxicity test of cyclophosphamide and acetaminophen in HepG2 cells cultured with HLM showed that the newly established condition using 0.375 mg/ml HLM for 24 hr incubation was comparable or more sensitive than the previously established condition using 0.75 mg/ml HLM for 12 hr incubation. Although the cytotoxic effect of troglitazone was completely attenuated by 0.75 mg/ml HLM, it was augmented by 0.375 mg/ml HLM in the presence of the NADPH-generating system. The cytotoxic effect of chlormezanone, a withdrawn drug due to hepatotoxicity in human, was increased by HLM in the presence of the NADPH-generating system. In contrast, the cytotoxic effect of methapyrilene, a withdrawn drug due to hepatotoxicity in rats, was decreased by HLM in the presence of the NADPH-generating system. The present study suggests that the optimized in vitro method using HLM can be useful for the prediction of drug-induced hepatotoxicity.

키워드

참고문헌

  1. Kaplowitz, N. : Idiosyncratic drug hepatotoxicity. Nat. Rev. Drug Discov. 4, 489 (2005). https://doi.org/10.1038/nrd1750
  2. Guengerich, F. P. and MacDonald, J. S. : Applying mechanisms of chemical toxicity to predict drug safety. Chem. Res. Toxicol. 20, 344 (2007). https://doi.org/10.1021/tx600260a
  3. Park, B. K., Kitteringham, N. R., Maggs, J. L., Pirmohamed, M. and Williams, D. P. : The role of metabolic activation in druginduced hepatotoxicity. Annu. Rev. Pharmacol. Toxicol. 45, 177 (2005). https://doi.org/10.1146/annurev.pharmtox.45.120403.100058
  4. Donato, M. T., Lahoz, A., Castell, J. V. and Gomez-Lechon, M. J. : Cell lines: a tool for in vitro drug metabolism studies. Curr. Drug Metab. 9, 1 (2008). https://doi.org/10.2174/138920008783331086
  5. Guillouzo, A. : Liver cell models in in vitro toxicology. Environ. Health Perspect. 106 Suppl. 2, 511 (1998). https://doi.org/10.1289/ehp.98106511
  6. Aninat, C., Piton, A., Glaise, D., Le Charpentier, T., Langouet, S., Morel, F., Guguen-Guillouzo, C. and Guillouzo, A. : Expression of cytochromes P450, conjugating enzymes and nuclear receptors in human hepatoma HepaRG cells. Drug Metab. Dispos. 34, 75 (2006).
  7. Doehmer, J., Dogra, S., Friedberg, T., Monier, S., Adesnik, M., Glatt, H. and Oesch, F. : Stable expression of rat cytochrome P-450IIB1 cDNA in Chinese hamster cells (V79) and metabolic activation of aflatoxin B1. Proc. Natl. Acad. Sci. U.S.A. 85, 5769 (1988). https://doi.org/10.1073/pnas.85.16.5769
  8. Gomez-Lechon, M. J., Donato, T., Jover, R., Rodriguez, C., Ponsoda, X., Glaise, D., Castell, J. V. and Guguen-Guillouzo, C. : Expression and induction of a large set of drug-metabolizing enzymes by the highly differentiated human hepatoma cell line BC2. Eur. J. Biochem. 268, 1448 (2001). https://doi.org/10.1046/j.1432-1327.2001.02011.x
  9. Hosomi, H., Fukami, T., Iwamura, A., Nakajima, M. and Yokoi, T. : Development of a highly sensitive cytotoxicity assay system for CYP3A4-mediated metabolic activation. Drug Metab. Dispos. 39, 1388 (2011). https://doi.org/10.1124/dmd.110.037077
  10. Choi, J. M., Oh, S. J., Lee, J. Y., Jeon, J. S., Ryu, C. S., Kim, Y. M., Lee, K. H. and Kim, S. K. : Prediction of drug-induced liver injury in HepG2 cells cultured with human liver microsomes. Chem. Res. Toxicol. 28, 872 (2015). https://doi.org/10.1021/tx500504n
  11. Mosmann, T. : Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55 (1983). https://doi.org/10.1016/0022-1759(83)90303-4
  12. Lee, K. S. and Kim, S. K. : Direct and metabolism-dependent cytochrome P450 inhibition assays for evaluating drug-drug interactions. J. Appl. Toxicol. 33, 100 (2013). https://doi.org/10.1002/jat.1720
  13. Smith, M. T. : Mechanisms of troglitazone hepatotoxicity. Chem. Res. Toxicol. 16, 679 (2003). https://doi.org/10.1021/tx034033e
  14. Ikeda, T. : Drug-induced idiosyncratic hepatotoxicity: prevention strategy developed after the troglitazone case. Drug Metab. Pharmacokinet. 26, 60 (2011). https://doi.org/10.2133/dmpk.DMPK-10-RV-090
  15. Graham, E. E., Walsh, R. J., Hirst, C. M., Maggs, J. L., Martin, S., Wild, M. J., Wilson, I. D., Harding, J. R., Kenna, J. G., Peter, R. M., Williams, D. P. and Park, B. K. : Identification of the thiophene ring of methapyrilene as a novel bioactivation-dependent hepatic toxicophore. J. Pharmacol. Exp. Ther. 326, 657 (2008). https://doi.org/10.1124/jpet.107.135483
  16. Howell, B. A., Yang, Y., Kumar, R., Woodhead, J. L., Harrill, A. H., Clewell, H. J., 3rd, Andersen, M. E., Siler, S. Q. and Watkins, P. B. : In vitro to in vivo extrapolation and species response comparisons for drug-induced liver injury (DILI) using DILIsym: a mechanistic, mathematical model of DILI. J. Pharmacokinet. Pharmacodyn. 39, 527 (2012). https://doi.org/10.1007/s10928-012-9266-0