Comparison of Immortalized Fa2N-4 Cells and Human Hepatocytes as in Vitro Models for Cytochrome P450 Induction

Fa2N-4 cells have been proposed as a tool to identify CYP3A4 inducers. To evaluate whether Fa2N-4 cells are a reliable surrogate for cryopreserved human hepatocytes, we assessed the basal mRNA expression of 64 drug disposition genes in Fa2N-4 cells. Significant differences were found in the expression of major drug-metabolizing enzymes, nuclear receptors, and transporters between both cell types. Importantly, the expression of constitutive androstane receptor (CAR) and several hepatic uptake transporters was significantly lower (>50-fold) in Fa2N-4 cells, whereas the expression of pregnane X-receptor (PXR) and aryl hydrocarbon receptor (AhR) was similar between Fa2N-4 cells and human hepatocytes. By using an optimized induction assay for Fa2N-4 cells, CYP3A4 induction by rifampicin, the prototypical PXR activator, increased from 1.5- to 7-fold at the level of functional activity. With nine selected compounds, which are known inducers of CYP3A4 either via activation of PXR, CAR, or both, we evaluated CYP3A4 and CYP2B6 mRNA induction using Fa2N-4 cells and human hepatocytes. No response was observed in Fa2N-4 cells treated with the selective CAR activators 6-(4-chlorophenyl)imidazo[2,1-b][1,3]-thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime and artemisinin. CYP3A4 and CYP2B6 induction in Fa2N-4 cells were also low for phenytoin, phenobarbital, and efavirenz, which are dual activators of PXR/CAR. This finding was in agreement with the lack of expression of CAR. The EC50 value for rifampicin-mediated CYP3A4 induction was 10-fold higher than that in human hepatocytes. This result could be attributed to the low expression of hepatic organic anion-transporting polypeptides OATP1B1 and OATP1B3 in Fa2N-4 cells. In summary, our findings identify limitations of Fa2N-4 cells as a predictive induction model.

[1]  R. Evans,et al.  Nuclear receptor that identifies a novel retinoic acid response pathway , 1990, Nature.

[2]  R. Fisher,et al.  Regulation of human liver cytochromes P‐450 in family 3A in primary and continuous culture of human hepatocytes , 1993, Hepatology.

[3]  K. Umesono,et al.  The nuclear receptor superfamily: The second decade , 1995, Cell.

[4]  W. Sabbagh,et al.  SXR, a novel steroid and xenobiotic-sensing nuclear receptor. , 1998, Genes & development.

[5]  J. Lehmann,et al.  Orphan nuclear receptors: shifting endocrinology into reverse. , 1999, Science.

[6]  E L LeCluyse,et al.  Pregnane X receptor: molecular basis for species differences in CYP3A induction by xenobiotics. , 2001, Chemico-biological interactions.

[7]  M. Pfaffl,et al.  A new mathematical model for relative quantification in real-time RT-PCR. , 2001, Nucleic acids research.

[8]  R. Kim,et al.  Pharmacogenomics of organic anion-transporting polypeptides (OATP). , 2002, Advanced drug delivery reviews.

[9]  Sean Kim,et al.  CYP3A4 induction by drugs: correlation between a pregnane X receptor reporter gene assay and CYP3A4 expression in human hepatocytes. , 2002, Drug metabolism and disposition: the biological fate of chemicals.

[10]  Bosentan, a dual endothelin receptor antagonist, activates the pregnane X nuclear receptor. , 2002, European journal of pharmacology.

[11]  L. Moore,et al.  Identification of a Novel Human Constitutive Androstane Receptor (CAR) Agonist and Its Use in the Identification of CAR Target Genes* , 2003, The Journal of Biological Chemistry.

[12]  D. Meijer,et al.  Drug uptake systems in liver and kidney. , 2003, Current drug metabolism.

[13]  Darrell R. Abernethy,et al.  International Union of Pharmacology: Approaches to the Nomenclature of Voltage-Gated Ion Channels , 2003, Pharmacological Reviews.

[14]  J. Sahi,et al.  Induction of Drug Metabolism Enzymes and MDR1 Using a Novel Human Hepatocyte Cell Line , 2004, Journal of Pharmacology and Experimental Therapeutics.

[15]  E. Schuetz Lessons from the CYP3A4 promoter. , 2004, Molecular pharmacology.

[16]  E. Carlini,et al.  Comparative Effects of Fibrates on Drug Metabolizing Enzymes in Human Hepatocytes , 2004, Pharmaceutical Research.

[17]  N. Hariparsad,et al.  Induction of CYP3A4 by Efavirenz in Primary Human Hepatocytes: Comparison With Rifampin and Phenobarbital , 2004, Journal of clinical pharmacology.

[18]  Hongbing Wang,et al.  Human Constitutive Androstane Receptor Mediates Induction of CYP2B6 Gene Expression by Phenytoin* , 2004, Journal of Biological Chemistry.

[19]  J. Raucy,et al.  HIGH VOLUME BIOASSAYS TO ASSESS CYP3A4-MEDIATED DRUG INTERACTIONS: INDUCTION AND INHIBITION IN A SINGLE CELL LINE , 2005, Drug Metabolism and Disposition.

[20]  R. Kim,et al.  Nuclear receptors and drug disposition gene regulation. , 2005, Journal of pharmaceutical sciences.

[21]  J. Goldstein,et al.  The Nuclear Receptors Constitutive Androstane Receptor and Pregnane X Receptor Cross-Talk with Hepatic Nuclear Factor 4α to Synergistically Activate the Human CYP2C9 Promoter , 2005, Journal of Pharmacology and Experimental Therapeutics.

[22]  T. Maurer,et al.  Use of Immortalized Human Hepatocytes to Predict the Magnitude of Clinical Drug-Drug Interactions Caused by CYP3A4 Induction , 2006, Drug Metabolism and Disposition.

[23]  L. Benet,et al.  IN VITRO AND IN VIVO CORRELATION OF HEPATIC TRANSPORTER EFFECTS ON ERYTHROMYCIN METABOLISM: CHARACTERIZING THE IMPORTANCE OF TRANSPORTER-ENZYME INTERPLAY , 2006, Drug Metabolism and Disposition.

[24]  M. Lang,et al.  In vivo and mechanistic evidence of nuclear receptor CAR induction by artemisinin , 2006, European journal of clinical investigation.

[25]  C. Smith,et al.  Differential Regulation of Hepatic CYP2B6 and CYP3A4 Genes by Constitutive Androstane Receptor but Not Pregnane X Receptor , 2006, Journal of Pharmacology and Experimental Therapeutics.

[26]  Jiunn H. Lin,et al.  CYP Induction-Mediated Drug Interactions: in Vitro Assessment and Clinical Implications , 2006, Pharmaceutical Research.

[27]  A. Bell,et al.  Phenobarbital regulates nuclear expression of HNF‐4α in mouse and rat hepatocytes independent of CAR and PXR , 2006, Hepatology.

[28]  Wen Xie,et al.  International Union of Pharmacology. LXII. The NR1H and NR1I receptors: constitutive androstane receptor, pregnene X receptor, farnesoid X receptor alpha, farnesoid X receptor beta, liver X receptor alpha, liver X receptor beta, and vitamin D receptor. , 2006, Pharmacological reviews.

[29]  Yuichi Sugiyama,et al.  Transporters as a determinant of drug clearance and tissue distribution. , 2006, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[30]  B. Stieger,et al.  Bosentan Is a Substrate of Human OATP1B1 and OATP1B3: Inhibition of Hepatic Uptake as the Common Mechanism of Its Interactions with Cyclosporin A, Rifampicin, and Sildenafil , 2007, Drug Metabolism and Disposition.

[31]  Hongbing Wang,et al.  Relative Activation of Human Pregnane X Receptor versus Constitutive Androstane Receptor Defines Distinct Classes of CYP2B6 and CYP3A4 Inducers , 2007, Journal of Pharmacology and Experimental Therapeutics.

[32]  Hayley S. Brown,et al.  Primary Hepatocytes: Current Understanding of the Regulation of Metabolic Enzymes and Transporter Proteins, and Pharmaceutical Practice for the Use of Hepatocytes in Metabolism, Enzyme Induction, Transporter, Clearance, and Hepatotoxicity Studies , 2007, Drug metabolism reviews.

[33]  J. Pascussi,et al.  The tangle of nuclear receptors that controls xenobiotic metabolism and transport: crosstalk and consequences. , 2008, Annual review of pharmacology and toxicology.

[34]  O. Trubetskoy,et al.  A simultaneous assessment of CYP3A4 metabolism and induction in the DPX-2 cell line , 2005, The AAPS Journal.

[35]  M. Ogg,et al.  Development of an in vitro reporter gene assay to assess xenobiotic induction of the human CYP3A4 gene , 1997, European Journal of Drug Metabolism and Pharmacokinetics.