Skip to main content

Advertisement

Log in

U0126, a mitogen-activated protein kinase kinase 1 and 2 (MEK1 and 2) inhibitor, selectively up-regulates main isoforms of CYP3A subfamily via a pregnane X receptor (PXR) in HepG2 cells

  • Molecular Toxicology
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

Abstract

Hepatocyte tumor cell lines lack the expression or induction properties of major cytochrome P450 (CYP) enzymes compared to primary human hepatocytes. The Ras/Raf/MEK/ERK signaling cascade contributes to hepatocarcinogenesis, dedifferentiation and loss of hepatocyte drug metabolism in hepatocyte tumors. In the present study, we examined whether MEK1/2 inhibitors can restore the expression of CYP genes in hepatocarcinoma HepG2 cells. We found that U0126, a prototype dual MEK1/2 inhibitor, is a potent inducer of CYP3A4, CYP3A5 and CYP3A7 mRNA expression (>100-fold) in HepG2 cells and CYP3A4 mRNA expression in primary human hepatocytes. This U0126-mediated induction is sensitive to the transcriptional inhibitor actinomycin D and was not detected for CYP2B6 or MDR1 mRNA expression. In gene reporter assays, U0126 activates a CYP3A4 promoter luciferase reporter construct containing PXR response elements (PXREs), but not a construct containing mutated PXREs. Based on a ligand binding assay and the examination of a PXR mutant expressing an obstructed ligand binding pocket, we found that U0126 is a ligand of PXR. We also found that U0126 up-regulates the mRNA expression of the nuclear receptors HNF4α, CAR, VDR and PXR but abolishes small heterodimer partner (SHP) corepressor expression in HepG2 cells. The MEK1/2 inhibitors PD0325901 and PD184352, as well as dominant-negative MEK1 expression, also down-regulate SHP mRNA expression. In contrast, dominant-negative MEK1 expression does not significantly induce CYP3A4 gene in HepG2 cells. In conclusion, we found that U0126 is an atypical PXR ligand that via direct (binding and activation of PXR) and indirect (SHP dowregulation) mechanisms selectively restores CYP3A genes in HepG2 cells.

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

Similar content being viewed by others

Abbreviations

CAR:

Constitutive androstane receptor

CDCA:

Chenodeoxycholic acid

CDK:

Cyclin-dependent kinase

CYP:

Cytochrome P450

DMEs:

Drug-metabolizing enzymes

DN:

Dominant negative

ERK1/2:

Extracellular signal-regulated kinases 1/2

FXR:

Farnesoid X receptor

HNF4α:

Hepatocyte nuclear factor-4 alpha

LBD:

Ligand binding domain

MAPK:

Mitogen-activated protein kinase

MDR1:

Multi-drug resistance 1; ABCB1 gen; P-glycoprotein

MEK1/2:

Mitogen-activated/extracellular signal-regulated kinase kinases 1/2

NR:

Nuclear receptor

PKA:

Cyclic AMP-dependent protein kinase

PKC:

Protein kinase C

PXR:

Pregnane X receptor

PXRE:

Pregnane X receptor response element

SHP:

Short/small heterodimer partner (N0B2)

SRC1:

Steroid receptor coactivator-1

UTR:

Untranslated region

References

  • Andrieux L, Langouet S, Fautrel A et al (2004) Aryl hydrocarbon receptor activation and cytochrome P450 1A induction by the mitogen-activated protein kinase inhibitor U0126 in hepatocytes. Mol Pharmacol 65(4):934–943. doi:10.1124/mol.65.4.934

    Article  CAS  PubMed  Google Scholar 

  • Bachleda P, Dvorak Z (2008) Pharmacological inhibitors of JNK and ERK kinases SP600125 and U0126 are not appropriate tools for studies of drug metabolism because they activate aryl hydrocarbon receptor. Gen Physiol Biophys 27(2):143–145

    CAS  PubMed  Google Scholar 

  • Bertilsson G, Heidrich J, Svensson K et al (1998) Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc Natl Acad Sci USA 95(21):12208–12213

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Blumberg B, Sabbagh W Jr, Juguilon H et al (1998) SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev 12(20):3195–3205

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Braeuning A (2009) Regulation of cytochrome P450 expression by Ras- and β-catenin-dependent signaling. Curr Drug Metab 10(2):138–158

    Article  CAS  PubMed  Google Scholar 

  • Castell JV, Jover R, Martinez-Jimenez CP, Gomez-Lechon MJ (2006) Hepatocyte cell lines: their use, scope and limitations in drug metabolism studies. Expert Opin Drug Metab Toxicol 2(2):183–212

    Article  CAS  PubMed  Google Scholar 

  • De Luca A, Maiello MR, D’Alessio A, Pergameno M, Normanno N (2012) The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin Ther Targets 16(Suppl 2):S17–S27. doi:10.1517/14728222.2011.639361

    Article  PubMed  Google Scholar 

  • Ding X, Staudinger JL (2005a) Induction of drug metabolism by forskolin: the role of the pregnane X receptor and the protein kinase a signal transduction pathway. J Pharmacol Exp Ther 312(2):849–856

    Article  CAS  PubMed  Google Scholar 

  • Ding X, Staudinger JL (2005b) Repression of PXR-mediated induction of hepatic CYP3A gene expression by protein kinase C. Biochem Pharmacol 69(5):867–873

    Article  CAS  PubMed  Google Scholar 

  • Donato MT, Lahoz A, Castell JV, Gomez-Lechon MJ (2008) Cell lines: a tool for in vitro drug metabolism studies. Curr Drug Metab 9(1):1–11

    Article  CAS  PubMed  Google Scholar 

  • Dong H, Lin W, Wu J, Chen T (2010) Flavonoids activate pregnane X receptor-mediated CYP3A4 gene expression by inhibiting cyclin-dependent kinases in HepG2 liver carcinoma cells. BMC Biochem 11:23. doi:10.1186/1471-2091-11-23

    Article  PubMed Central  PubMed  Google Scholar 

  • Doricakova A, Novotna A, Vrzal R, Pavek P, Dvorak Z (2013) The role of residues T248, Y249 and T422 in the function of human pregnane X receptor. Arch Toxicol 87(2):291–301. doi:10.1007/s00204-012-0937-9

    Article  CAS  PubMed  Google Scholar 

  • Fasinu P, Bouic PJ, Rosenkranz B (2012) Liver-based in vitro technologies for drug biotransformation studies—a review. Curr Drug Metab 13(2):215–224

    Article  CAS  PubMed  Google Scholar 

  • Favata MF, Horiuchi KY, Manos EJ et al (1998) Identification of a novel inhibitor of mitogen-activated protein kinase kinase. J Biol Chem 273(29):18623–18632

    Article  CAS  PubMed  Google Scholar 

  • Goodwin B, Hodgson E, Liddle C (1999) The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol Pharmacol 56(6):1329–1339

    CAS  PubMed  Google Scholar 

  • Goodwin B, Jones SA, Price RR et al (2000) A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 6(3):517–526

    Article  CAS  PubMed  Google Scholar 

  • Goodwin B, Moore LB, Stoltz CM, McKee DD, Kliewer SA (2001) Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol 60(3):427–431

    CAS  PubMed  Google Scholar 

  • Haas S, Merkelbach-Bruse S, Justenhoven C, Brauch H, Fischer HP (2009) Expression of xenobiotic and steroid hormone metabolizing enzymes in hepatocellular tumors of the non-cirrhotic liver. Pathol Res Pract 205(10):716–725. doi:10.1016/j.prp.2009.06.003

    Article  PubMed  Google Scholar 

  • Harmsen S, Koster AS, Beijnen JH, Schellens JH, Meijerman I (2008) Comparison of two immortalized human cell lines to study nuclear receptor-mediated CYP3A4 induction. Drug Metab Dispos 36(6):1166–1171. doi:10.1124/dmd.107.017335

    Article  CAS  PubMed  Google Scholar 

  • Healan-Greenberg C, Waring JF, Kempf DJ, Blomme EA, Tirona RG, Kim RB (2008) A human immunodeficiency virus protease inhibitor is a novel functional inhibitor of human pregnane X receptor. Drug Metab Dispos 36(3):500–507. doi:10.1124/dmd.107.019547

    Article  CAS  PubMed  Google Scholar 

  • Lee HC, Tian B, Sedivy JM, Wands JR, Kim M (2006) Loss of Raf kinase inhibitor protein promotes cell proliferation and migration of human hepatoma cells. Gastroenterology 131(4):1208–1217. doi:10.1053/j.gastro.2006.07.012

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lehmann JM, McKee DD, Watson MA, Willson TM, Moore JT, Kliewer SA (1998) The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Invest 102(5):1016–1023

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Li T, Chiang JY (2006) Rifampicin induction of CYP3A4 requires pregnane X receptor cross talk with hepatocyte nuclear factor 4alpha and coactivators, and suppression of small heterodimer partner gene expression. Drug Metab Dispos 34(5):756–764

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lichti-Kaiser K, Brobst D, Xu C, Staudinger JL (2009a) A systematic analysis of predicted phosphorylation sites within the human pregnane X receptor protein. J Pharmacol Exp Ther 331(1):65–76

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lichti-Kaiser K, Xu C, Staudinger JL (2009b) Cyclic AMP-dependent protein kinase signaling modulates pregnane x receptor activity in a species-specific manner. J Biol Chem 284(11):6639–6649

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Lin W, Wu J, Dong H, Bouck D, Zeng FY, Chen T (2008) Cyclin-dependent kinase 2 negatively regulates human pregnane X receptor-mediated CYP3A4 gene expression in HepG2 liver carcinoma cells. J Biol Chem 283(45):30650–30657. doi:10.1074/jbc.M806132200

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Martinez-Jimenez CP, Jover R, Donato MT, Castell JV, Gomez-Lechon MJ (2007) Transcriptional regulation and expression of CYP3A4 in hepatocytes. Curr Drug Metab 8(2):185–194

    Article  CAS  PubMed  Google Scholar 

  • Meloche S, Pouyssegur J (2007) The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1- to S-phase transition. Oncogene 26(22):3227–3239. doi:10.1038/sj.onc.1210414

    Article  CAS  PubMed  Google Scholar 

  • Min L, He B, Hui L (2010) Mitogen-activated protein kinases in hepatocellular carcinoma development. Semin Cancer Biol. doi:10.1016/j.semcancer.2010.10.011

  • Min L, He B, Hui L (2011) Mitogen-activated protein kinases in hepatocellular carcinoma development. Semin Cancer Biol 21(1):10–20. doi:10.1016/j.semcancer.2010.10.011

    Article  CAS  PubMed  Google Scholar 

  • Novotna A, Doricakova A, Vrzal R, Pavek P, Dvorak Z (2011) Construction and characterization of hepatocyte nuclear factor HNF4alpha1 over-expressing cell line derived from human hepatoma HepG2 cells. Eur J Pharmacol 669(1–3):45–50. doi:10.1016/j.ejphar.2011.07.049

    Article  CAS  PubMed  Google Scholar 

  • Osabe M, Sugatani J, Takemura A et al (2009) Up-regulation of CAR expression through Elk-1 in HepG2 and SW480 cells by serum starvation stress. FEBS Lett 583(5):885–889. doi:10.1016/j.febslet.2009.01.051

    Article  CAS  PubMed  Google Scholar 

  • Ourlin JC, Lasserre F, Pineau T et al (2003) The small heterodimer partner interacts with the pregnane X receptor and represses its transcriptional activity. Mol Endocrinol 17(9):1693–1703. doi:10.1210/me.2002-0383

    Article  CAS  PubMed  Google Scholar 

  • Pan YZ, Gao W, Yu AM (2009) MicroRNAs regulate CYP3A4 expression via direct and indirect targeting. Drug Metab Dispos 37(10):2112–2117. doi:10.1124/dmd.109.027680

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Pavek P, Pospechova K, Svecova L et al (2010) Intestinal cell-specific vitamin D receptor (VDR)-mediated transcriptional regulation of CYP3A4 gene. Biochem Pharmacol 79(2):277–287. doi:10.1016/j.bcp.2009.08.017

    Article  CAS  PubMed  Google Scholar 

  • Pavek P, Stejskalova L, Krausova L, Bitman M, Vrzal R, Dvorak Z (2012) Rifampicin does not significantly affect the expression of small heterodimer partner in primary human hepatocytes. Front Pharmacol 3:1. doi:10.3389/fphar.2012.00001

    Article  PubMed Central  PubMed  Google Scholar 

  • Plant N (2007) The human cytochrome P450 sub-family: transcriptional regulation, inter-individual variation and interaction networks. Biochim Biophys Acta 1770(3):478–488. doi:10.1016/j.bbagen.2006.09.024

    Article  CAS  PubMed  Google Scholar 

  • Pondugula SR, Brimer-Cline C, Wu J, Schuetz EG, Tyagi RK, Chen T (2009) A phosphomimetic mutation at threonine-57 abolishes transactivation activity and alters nuclear localization pattern of human pregnane X receptor. Drug Metab Dispos 37(4):719–730

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  • Pospechova K, Rozehnal V, Stejskalova L et al (2009) Expression and activity of vitamin D receptor in the human placenta and in choriocarcinoma BeWo and JEG-3 cell lines. Mol Cell Endocrinol 299(2):178–187. doi:10.1016/j.mce.2008.12.003

    Article  CAS  PubMed  Google Scholar 

  • Rochette-Egly C (2003) Nuclear receptors: integration of multiple signalling pathways through phosphorylation. Cell Signal 15(4):355–366

    Article  CAS  PubMed  Google Scholar 

  • Rodriguez-Antona C, Donato MT, Boobis A et al (2002) Cytochrome P450 expression in human hepatocytes and hepatoma cell lines: molecular mechanisms that determine lower expression in cultured cells. Xenobiotica 32(6):505–520. doi:10.1080/00498250210128675

    Article  CAS  PubMed  Google Scholar 

  • Rulcova A, Prokopova I, Krausova L et al (2010) Stereoselective interactions of warfarin enantiomers with the pregnane X nuclear receptor in gene regulation of major drug-metabolizing cytochrome P450 enzymes. J Thromb Haemost 8(12):2708–2717

    Article  CAS  PubMed  Google Scholar 

  • Shaul YD, Seger R (2007) The MEK/ERK cascade: from signaling specificity to diverse functions. Biochim Biophys Acta 1773(8):1213–1226. doi:10.1016/j.bbamcr.2006.10.005

    Article  CAS  PubMed  Google Scholar 

  • Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich FP (1994) Interindividual variations in human liver cytochrome P-450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30 Japanese and 30 Caucasians. J Pharmacol Exp Ther 270(1):414–423

    CAS  PubMed  Google Scholar 

  • Smutny T, Mani S, Pavek P (2013) Post-translational and post-transcriptional modifications of pregnane X receptor (PXR) in regulation of the cytochrome P450 superfamily. Curr Drug Metab 14(10):1059–1069

  • Svecova L, Vrzal R, Burysek L et al (2008) Azole antimycotics differentially affect rifampicin-induced pregnane X receptor-mediated CYP3A4 gene expression. Drug Metab Dispos 36(2):339–348. doi:10.1124/dmd.107.018341

    Article  CAS  PubMed  Google Scholar 

  • Takeshita A, Taguchi M, Koibuchi N, Ozawa Y (2002) Putative role of the orphan nuclear receptor SXR (steroid and xenobiotic receptor) in the mechanism of CYP3A4 inhibition by xenobiotics. J Biol Chem 277(36):32453–32458

    Article  CAS  PubMed  Google Scholar 

  • Vrzal R, Stejskalova L, Monostory K et al (2009) Dexamethasone controls aryl hydrocarbon receptor (AhR)-mediated CYP1A1 and CYP1A2 expression and activity in primary cultures of human hepatocytes. Chem Biol Interact 179(2–3):288–296

    Article  CAS  PubMed  Google Scholar 

  • Wang H, Faucette S, Sueyoshi T et al (2003) A novel distal enhancer module regulated by pregnane X receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression. J Biol Chem 278(16):14146–14152. doi:10.1074/jbc.M212482200

    Article  CAS  PubMed  Google Scholar 

  • Wang H, Huang H, Li H et al (2007) Activated pregnenolone X-receptor is a target for ketoconazole and its analogs. Clin Cancer Res 13(8):2488–2495

    Article  CAS  PubMed  Google Scholar 

  • Wiesenauer CA, Yip-Schneider MT, Wang Y, Schmidt CM (2004) Multiple anticancer effects of blocking MEK-ERK signaling in hepatocellular carcinoma. J Am Coll Surg 198(3):410–421. doi:10.1016/j.jamcollsurg.2003.10.004

    Article  PubMed  Google Scholar 

  • Wilkinson MG, Millar JB (2000) Control of the eukaryotic cell cycle by MAP kinase signaling pathways. FASEB J 14(14):2147–2157. doi:10.1096/fj.00-0102rev

    Article  CAS  PubMed  Google Scholar 

  • Zhou C, Poulton EJ, Grun F et al (2007) The dietary isothiocyanate sulforaphane is an antagonist of the human steroid and xenobiotic nuclear receptor. Mol Pharmacol 71(1):220–229

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This research has been supported by the Czech Scientific Agency GACR303/12/0472 (to P.P.) and by SVV 170/50/33904-3 Project.

Conflict of interest

None.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Petr Pavek.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Smutny, T., Bitman, M., Urban, M. et al. U0126, a mitogen-activated protein kinase kinase 1 and 2 (MEK1 and 2) inhibitor, selectively up-regulates main isoforms of CYP3A subfamily via a pregnane X receptor (PXR) in HepG2 cells. Arch Toxicol 88, 2243–2259 (2014). https://doi.org/10.1007/s00204-014-1254-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-014-1254-2

Keywords

Navigation