Functional expression of sinusoidal and canalicular hepatic drug transporters in the differentiated human hepatoma HepaRG cell line

https://doi.org/10.1016/j.ejps.2006.01.004Get rights and content

Abstract

Functional expression of both sinusoidal and canalicular hepatic drug transporters was investigated in the highly differentiated human hepatoma HepaRG cell line and also, for comparison, in primary human hepatocytes and in the hepatoma HepG2 cell line. Using RT-qPCR assays, differentiated HepaRG cells were found to display a pattern of transporter expression close to that found in primary human hepatocytes, i.e. they exhibit substantial mRNA levels of the influx transporters OCT1, OATP-B, OATP-C and NTCP, and of the secretion transporters MRP2, MRP3, BSEP and P-glycoprotein. By contrast, expression of influx transporters was not present or very weak in HepG2 cells. Drug transport assays allowed to detect functional activities of OCT1, OATPs/OAT2, NTCP, MRPs and P-glycoprotein in differentiated HepaRG cells as in primary human hepatocytes whereas HepG2 cells only showed notable MRP and P-glycoprotein activities. In addition, expression of canalicular transporters in HepaRG cells was found to be up-regulated by known inducers of transporters such as rifampicin, phenobarbital and chenodeoxycholate acting on P-glycoprotein, MRP2 and BSEP, respectively. HepaRG cells thus exhibit functional expression of both sinusoidal and canalicular drug transporters and have retained regulatory pathways controlling transporter levels. These data, associated with the known high expression of drug metabolizing enzymes in HepaRG cells, highlight the interest of such hepatoma cells for analysing hepatic drug detoxification pathways.

Introduction

Membrane transport proteins, belonging to the solute carrier (SLC) and ATP-binding cassette (ABC) superfamilies of transporters, play a major role in biliary elimination of drugs (Kim, 2002). Indeed, sinusoidal transporters mediate the initial step of hepatic elimination, i.e. drug uptake from blood in hepatocytes (van Montfoort et al., 2003), whereas canalicular transporters are involved in the final stage of hepatic elimination, i.e. secretion of drugs or their metabolites from hepatocytes into bile canaliculi (Schinkel and Jonker, 2003). Major hepatic influx transporters correspond to organic cation transporter 1 (OCT1/SLC22A1), handling organic cations such as tetra-ethylammonium (TEA) and the hypoglycemic agent metformin (Jonker and Schinkel, 2004), organic anion transporting polypeptides (OATP-A/SLCO1A2, OATP-B/SLCO2B1, OATP-C/SLCO1B1 and OATP8/SLCO1B3) and organic anion transporter 2 (OAT2/SLC22A7), mediating the transport of organic anions such as estrone-3-sulfate (ES) (Rippin et al., 2001, Kobayashi et al., 2005) and also of amphipathic organic cations like digoxin (Kim, 2003, Hagenbuch and Meier, 2004), and Na+-taurocholate co-transporting polypeptide (NTCP/SLC10A1) involved in Na+-dependent uptake of biliary acids (Trauner and Boyer, 2003). Major hepatic canalicular transporters are ABC proteins. They include P-glycoprotein (ABCB1), encoded by MDR1 gene and responsible for efflux of hydrophobic organic cations including anticancer drugs such as doxorubicin, HIV antiprotease inhibitors such as saquinavir and dyes like rhodamine 123 (Schinkel and Jonker, 2003). Multidrug resistance protein 2 (MRP2/ABCC2) constitutes another major canalicular transporter, mediating the secretion of organic anions and drug conjugates, including pravastatin and the fluorescent dye carboxy-2′,7′-dichlorofluorescein (CF) (Fardel et al., 2005). The bile salt export pump (BSEP/ABCB11) is involved in canalicular efflux of biliary acids (Arrese and Ananthanarayanan, 2004). The ABC pump MRP3, located at the sinusoidal pole of hepatocytes, is thought to mediate secretion of drug metabolites into the bloodstream for subsequent urinary elimination (Zelcer et al., 2005). Interestingly, expression of several liver drug transporters are thought to be regulated by xenobiotic treatments through nuclear receptor activation as already described for drug metabolizing enzymes. Thus, rifampin, a pregnane X receptor ligand, and phenobarbital, an activator of the constitutive androstane receptor, have been shown to enhance expression of P-glycoprotein and MRP2 (Geick et al., 2001, Courtois et al., 2002), respectively, whereas BSEP levels can be up-regulated by bile salts via the farnesoid X receptor (Plass et al., 2002). Xenobiotics may also inhibit activity of transporters, through competitive or non-competitive mechanisms (Leonard et al., 2002), which may lead to drug–drug interactions and/or drug adverse effects such as cholestasis or hyperbilirubinemia (Campbell et al., 2004, Shitara et al., 2005).

Primary human hepatocytes have been recently demonstrated to constitute an adequate and useful tool for analysing activity and regulation of drug transporters. Indeed, they exhibit substantial activity of both uptake and efflux transporters (Jigorel et al., 2005, Payen et al., 2000) and retain regulatory ways of transporter expression (Payen et al., 2002). Unfortunately, their unpredictable and scarce availability limits their use, especially for analyses during pre-clinical development of pharmaceutical compounds. Human immortalized hepatic cell lines have been proposed as an alternative to primary hepatocytes in toxicological studies (Knasmuller et al., 2004, Xu et al., 2004). In this context, the newly characterized hepatoma HepaRG cell line may be important to consider since it has been shown to express specific liver functions, including drug detoxifying enzymes, at relatively high levels, in contrast to other hepatoma cell lines such as HepG2 (Gripon et al., 2002, Aninat et al., 2006). Due to the increasing importance of transporters in pharmacokinetic, drug–drug interactions and drug adverse effects, the present study was therefore designed to investigate functional expression of both sinusoidal and canalicular drug transporters in HepaRG cells. Our results indicated that activity and regulation of major hepatic drug transporters can be detected in this cell line, underlining its potential interest as an alternative to primary human hepatocytes for toxicological studies.

Section snippets

Chemicals

[3H(G)]taurocholic acid (TC) (sp. act. 1.19 Ci/mmol), [6,7-3H(N)]ES (sp. act. 57.30 Ci/mmol), and [1-14C]TEA (sp. act. 2.4 mCi/mmol) were purchased from Perkin-Elmer Life Sciences (Boston, MA). Probenecid, verapamil, phenobarbital, rifampicin, chenodeoxycholic acid and rhodamine 123 were obtained from Sigma–Aldrich (St. Louis, MO) whereas CF diacetetate was provided by Molecular Probes (Interchim, Montluçon, France).

Cell culture

Human hepatoma HepaRG cells, initially isolated from a liver tumor of a female

Transporter mRNA levels in HepaRG cells

Sinusoidal and canalicular transporter mRNA levels were analysed in proliferating (3 days old after seeding), confluent (2 weeks old after seeding) and DMSO-treated (4 weeks old after seeding with addition of DMSO during the two last weeks) HepaRG cells, since confluence and addition of DMSO have been shown to constitute critical parameters for the expression of liver-specific functions in these hepatoma cells (Gripon et al., 2002). For comparison purposes, transporter expression was determined

Discussion

In addition to drug metabolizing enzymes, hepatic transporters are now recognized as major players contributing to pharmacokinetic, drug–drug interactions and drug adverse effects, underlining the need of convenient cellular models to analyse drug-transporter interactions, especially in pharmaceutical industries during preclinical evaluation of drug candidates (Mizuno and Sugiyama, 2002). In this context, we have analysed in the present study functional expression and regulation of transporters

References (43)

  • R.B. Kim

    Transporters and xenobiotic disposition

    Toxicology

    (2002)
  • S. Knasmuller et al.

    Use of human-derived liver cell lines for the detection of environmental and dietary genotoxicants; current state of knowledge

    Toxicology

    (2004)
  • G.A. Kullak-Ublick et al.

    Organic anion-transporting polypeptide B (OATP-B) and its functional comparison with three other OATPs of human liver

    Gastroenterology

    (2001)
  • N. Mizuno et al.

    Drug transporters: their role and importance in the selection and development of new drugs

    Drug Metab. Pharmacokinet.

    (2002)
  • L. Payen et al.

    Characterization and inhibition by a wide range of xenobiotics of organic anion excretion by primary human hepatocytes

    Biochem. Pharmacol.

    (2000)
  • J.R. Plass et al.

    Farnesoid X receptor and bile salts are involved in transcriptional regulation of the gene encoding the human bile salt export pump

    Hepatology

    (2002)
  • S.J. Rippin et al.

    Cholestatic expression pattern of sinusoidal and canalicular organic anion transport systems in primary cultured rat hepatocytes

    Hepatology

    (2001)
  • M. Rousset

    The human colon carcinoma cell lines HT-29 and Caco-2: two in vitro models for the study of intestinal differentiation

    Biochimie

    (1986)
  • M. Sasaki et al.

    Transcellular transport of organic anions across a double-transfected Madin-Darby canine kidney II cell monolayer expressing both human organic anion-transporting polypeptide (OATP2/SLC21A6) and multidrug resistance-associated protein 2 (MRP2/ABCC2)

    J. Biol. Chem.

    (2002)
  • A.H. Schinkel et al.

    Mammalian drug efflux transporters of the ATP binding cassette (ABC) family: an overview

    Adv. Drug Deliv. Rev.

    (2003)
  • T. Su et al.

    Impact of dimethyl sulfoxide on expression of nuclear receptors and drug-inducible cytochromes P450 in primary rat hepatocytes

    Arch. Biochem. Biophys.

    (2004)
  • Cited by (158)

    • Antidotal effect of cyclosporine A against α-amanitin toxicity in CD-1 mice, at clinical relevant doses

      2022, Food and Chemical Toxicology
      Citation Excerpt :

      Conversely, when given concomitantly, cyclosporine A gave some protection in the hepatic model, suggesting a protective/inhibition mechanism possibly involving uptake mechanisms. Regarding the expression of α-amanitin uptake transporters, HepaRG cells have lower expression of organic anion transporting polypeptide (OATP) 1B3 transporters (2–15%) but express higher levels of Na + -taurocholate cotransporter polypeptide (NTCP) transporter (191–247%) when compared to human hepatocytes (Kotani et al., 2012; Le Vee et al., 2006). As reported, α-amanitin enters hepatocytes via the OATP1B3 transporter protein located on the sinusoidal membrane (Letschert et al., 2006), but NTCP transporters can also facilitate hepatocellular α-amanitin uptake (Gundala et al., 2004; Kroncke et al., 1986).

    View all citing articles on Scopus
    View full text