Elsevier

Toxicology in Vitro

Volume 24, Issue 1, February 2010, Pages 297-309
Toxicology in Vitro

Use of cassette dosing in sandwich-cultured rat and human hepatocytes to identify drugs that inhibit bile acid transport

https://doi.org/10.1016/j.tiv.2009.08.009Get rights and content

Abstract

Hepatocellular accumulation of bile acids due to inhibition of the canalicular bile salt export pump (BSEP/ABCB11) is one proposed mechanism of drug-induced liver injury (DILI). Some hepatotoxic compounds also are potent inhibitors of bile acid uptake by Na+-dependent taurocholate cotransporting polypeptide (NTCP/SLC10A1). This study used a cassette dosing approach in rat and human sandwich-cultured hepatocytes (SCH) to determine whether known or suspected hepatotoxic drugs inhibit bile acid transport individually or in combination. [3H]-Taurocholate served as the NTCP/BSEP probe substrate. Individually, cyclosporin A and rifampin decreased taurocholate in vitro biliary clearance (Clbiliary) and biliary excretion index (BEI) by more than 20% in rat SCH, suggesting that these drugs primarily inhibited canalicular efflux. In contrast, ampicillin, carbenicillin, cloxacillin, nafcillin, oxacillin, carbamazepine, pioglitazone, and troglitazone decreased the in vitro Clbiliary by more than 20% with no notable change in BEI, suggesting that these drugs primarily inhibited taurocholate uptake. Cassette dosing (n = 2–4 compounds per cassette) in rat SCH yielded similar findings, and results in human SCH were consistent with rat SCH. In summary, cassette dosing in SCH is a useful in vitro approach to identify compounds that inhibit the hepatic uptake and/or excretion of bile acids, which may cause DILI.

Introduction

Drug-induced liver injury (DILI), a clinical problem worldwide (reviewed in Lucena et al., 2008, Norris et al., 2008), is a leading reason for withdrawal of approved drugs from clinical use or mandated usage restrictions on drug labels, and frequently contributes to attrition of drug candidates during development (Bakke et al., 1995, Kaplowitz, 2005, Kola and Landis, 2004). Numerous pathophysiological mechanisms underlie hepatotoxicity but remain poorly understood, and many cases of DILI are categorized as idiosyncratic (i.e., rare hepatotoxic reactions of unknown etiology). In addition, DILI is difficult to predict with existing animal models and only approximately half of the drugs that cause human hepatotoxicity can be identified during preclinical testing in rodents, dogs, and monkeys (Olson et al., 2000).

Cholestasis is marked by the accumulation of bile acids and bile constituents within hepatocytes; toxicity ensues due, in part, to the detergent-like effects of bile acids, which cause cellular apoptosis or necrosis and mitochondrial dysfunction (reviewed in Sokol et al., 2006). Many drugs and/or metabolites inhibit transport proteins responsible for the hepatobiliary transport of bile acids (reviewed in Kosters and Karpen, 2008, Pauli-Magnus and Meier, 2006). In addition, genetic and environmental factors that modulate the expression and/or activity of these transport proteins have been linked directly to cholestasis or to an increased susceptibility to its development (reviewed in Kosters and Karpen, 2008, Pauli-Magnus and Meier, 2006, Trauner and Boyer, 2003).

Transport proteins are increasingly recognized as pivotal in determining the pharmacokinetics and the pharmacological or toxicological effects of a number of drugs (Hirano et al., 2006, Szakács et al., 2008). Numerous transport proteins are responsible for the movement of bile acids into and out of hepatocytes (reviewed in Kosters and Karpen, 2008, Trauner and Boyer, 2003, Zollner et al., 2006). The sodium-taurocholate cotransporting polypeptide (NTCP/Ntcp; SLC10A1) and the bile salt export pump (BSEP/Bsep; ABCB11) are the predominant proteins involved in bile acid transport and homeostasis in humans and rats. On the basolateral/sinusoidal membrane of hepatocytes, NTCP/Ntcp mediates the uptake of taurine- and glycine-conjugated (i.e., taurocholate) and unconjugated bile acids from the blood in a sodium-dependent manner. Organic anion-transporting polypeptides (OATPs/Oatps; SLCOs) transport predominantly unconjugated bile acids in a sodium-independent manner. Multidrug resistance-associated proteins (MRP/Mrp) 3 (ABCC3) and 4 (ABCC4), and organic solute transporter (OST) α/β efflux bile acids from hepatocytes back into the blood. On the canalicular/apical membrane, BSEP/Bsep effluxes taurine- and glycine-conjugated bile acids into bile canaliculi, serving as a driving force for bile salt-dependent bile flow and for the concentration of bile acids in bile; MRP2/Mrp2 effluxes glucuronidated and sulfated bile acids.

Several in vitro models exist for assessing compound toxicity due to inhibition of transport proteins. Human and rat sandwich-cultured hepatocytes (SCH) are physiologically relevant in vitro models that maintain many in vivo structural and functional characteristics, including canalicular and basolateral membrane domains, expression and localization of liver-specific proteins, and functional bile excretion into sealed canalicular networks (LeCluyse et al., 1994, LeCluyse et al., 2000, Liu et al., 1998, Liu et al., 1999a, Liu et al., 1999b). These models have been used previously to study the inhibitory effects of drugs (e.g., troglitazone, nefazadone, antiretroviral compounds) on bile acid uptake and biliary excretion (Kemp et al., 2005, Kostrubsky et al., 2006, Marion et al., 2007, McRae et al., 2006).

Because of increased attention to DILI (EMEA, 2008, FDA, 2007), a robust assay utilizing SCH for inhibition of bile acid transport would be beneficial during drug development, allowing early identification of drug candidates that may cause cholestasis in humans. Traditionally, compounds are administered individually in preclinical drug development studies, a time-consuming, labor-intensive, and cost-ineffective practice. Cassette dosing, or the simultaneous administration of multiple compounds, was developed as an alternative screening method. This strategy has been used widely in vitro and in vivo to study the pharmacokinetics and metabolism of drug candidates (reviewed in White and Manitpisitkul, 2001, Zhou et al., 2004). To our knowledge, there are no reports in the literature regarding the use of cassette dosing to identify inhibitors of hepatic bile acid transport. The purpose of the present study was two-fold. First, inhibition of bile acid (taurocholate) transport by hepatotoxic drugs was assessed in rat and human SCH to determine whether it was a common mechanism of hepatotoxicity. Second, the feasibility of using a cassette dosing approach with rat and human SCH to screen for inhibition of taurocholate transport was investigated.

Section snippets

Materials

Fetal bovine serum (FBS), dexamethasone (Dex), Hanks’ balanced salts solution (HBSS) supplemented with (H-1387) or without (H-4891) calcium chloride, phenytoin, minocycline, azlocillin, nafcillin, ticarcillin, cloxacillin, carbenicillin, oxacillin, ampicillin, amoxicillin, haloperidol, chlorpromazine, valproic acid, pentamidine, prednisone, ibuprofen, etoposide, ceftriaxone, cefuroxime, carbamazepine, indomethacin, clotrimazole, bezafibrate, bifonazole, colchicine, hydralazine, menadione,

Hepatobiliary disposition of taurocholate in rat and human SCH

The accumulation in cells + bile, accumulation in cells, BEI, and in vitro Clbiliary of taurocholate in untreated rat and human SCH is illustrated in Fig. 2. Human SCH accumulated taurocholate in cells to a greater extent than rat SCH; however, human SCH appeared to have a lower BEI for taurocholate than rat SCH. The in vitro Clbiliary of taurocholate was similar between rat and human SCH.

Modulation of taurocholate hepatobiliary disposition in rat SCH

Table 1 summarizes the effects of known and suspected hepatotoxic drugs from various treatment classes on

Discussion

Modulation of hepatobiliary transport proteins may be an important mechanism underlying DILI. In humans and rodents, BSEP/Bsep and NTCP/Ntcp are largely responsible for hepatic bile acid transport, and are subject to xenobiotic interactions and genetic mutations (reviewed in Kosters and Karpen, 2008, Pauli-Magnus and Meier, 2006, Trauner and Boyer, 2003). Inhibition of BSEP/Bsep can decrease biliary excretion of bile acids, and the detergent-like effects of accumulated bile acids can cause

Conflict of interest

K.L.R.B. is a co-founder and Chair of the Scientific Advisory Board of Qualyst, Inc. Qualyst, Inc. has exclusively licensed the sandwich-cultured hepatocyte technology for quantification of biliary excretion (B-CLEAR®).

Acknowledgements

The authors would like to thank Yi-Wei Rong for her technical expertise in the isolation of rat and human hepatocytes, and Dr. Elaine Leslie for review of the manuscript. The authors would also like to thank Tracy Marion for her assistance with the design of Fig. 1, as well as for review of the manuscript. This work was supported by the National Institutes of Health Grants GM41935 (K.L.R.B.) and CA106101 (K.L.R.B.), and by GlaxoSmithKline, Inc. K.K.W. was funded by a postdoctoral fellowship

References (43)

  • G. Szakács et al.

    The role of ABC transporters in drug absorption, distribution, metabolism, excretion and toxicity (ADME-Tox)

    Drug Discovery Today

    (2008)
  • O.M. Bakke et al.

    Drug safety discontinuations in the United Kingdom, the United States, and Spain from 1974 through 1993: a regulatory perspective

    Clinical Pharmacology and Therapeutics

    (1995)
  • B. Davies et al.

    Physiological parameters in laboratory animals and humans

    Pharmaceutical Research

    (1993)
  • EMEA, 2008. Non-clinical guideline on drug-induced hepatotoxicity. Available at:...
  • FDA, 2007. Drug-induced liver injury: premarketing clinical evaluation. Available at:...
  • G. Ghibellini et al.

    In vitro–in vivo correlation of hepatobiliary drug clearance in humans

    Clinical Pharmacology and Therapeutics

    (2007)
  • H. Hirano et al.

    High-speed screening and QSAR analysis of human ATP-binding cassette transporter ABCB11 (bile salt export pump) to predict drug-induced intrahepatic cholestasis

    Molecular Pharmaceutics

    (2006)
  • N. Kaplowitz

    Idiosyncratic drug hepatotoxicity

    Nature Reviews Drug Discovery

    (2005)
  • D.C. Kemp et al.

    Xenobiotics inhibit hepatic uptake and biliary excretion of taurocholate in rat hepatocytes

    Toxicological Sciences

    (2005)
  • I. Kola et al.

    Can the pharmaceutical industry reduce attrition rates?

    Nature Reviews Drug Discovery

    (2004)
  • A. Kosters et al.

    Bile acid transporters in health and disease

    Xenobiotica

    (2008)
  • Cited by (40)

    • Total hepatocellular disposition profiling of rosuvastatin and pitavastatin in sandwich-cultured human hepatocytes

      2018, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      This study showed that D-PREX analyses can be used for the simultaneous evaluation of hepatic transport processes (uptake, BilEx, and BasEf) of the medicinal drugs in SCHH. D-PREX would also be applicable to evaluation of the cholestatic/hepatotoxic potential of drugs, as demonstrated previously by analysis of the inhibitory effects of target compounds on the efflux rate of exogenous bile acids using SCRH and SCHH [24,25] and of cell viability in the presence of bile acids and target compounds [22,23]. Because these reports suggest that intrahepatic retention of bile acids is caused not only by inhibition of the BSEP but also by the inhibition of BasEf transporters like MRP3/Mrp3 and MRP4/Mrp4, simultaneous evaluation of BilEx and BasEf by D-PREX enables complete clarification of the complex mechanisms of DILI caused by drugs and their metabolites.

    • Establishment of a Drug-Induced, Bile Acid-Dependent Hepatotoxicity Model Using HepaRG Cells

      2016, Journal of Pharmaceutical Sciences
      Citation Excerpt :

      The mRNA expression levels of some transporters (i.e., BSEP, NTCP, and OATP1B1) were lower in HepaRG cells than in SCHHs (Figs. 3a, 3d, and 3f). Other research groups previously reported good [3H]TC uptake in SCHHs (150-200 pmol/mg protein/10 min), with a calculated BEI value of approximately 75%.30,41,42 In contrast, we observed relatively low [3H]TC uptake and BEI values in HepaRG cells (Fig. 2a), consistent with the mRNA expression analysis.

    • Quantitative NTCP pharmacophore and lack of association between DILI and NTCP Inhibition

      2015, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      One mechanism for cholestasis and reduced bile acid excretion is inhibition of the Bile Salt Export Pump (BSEP) (Keppler, 2011). However, one study using the human sandwich-cultured hepatocyte model showed that several known (or suspected) hepatotoxic drugs inhibited taurocholate uptake, without inhibiting taurocholate efflux from hepatocytes (Wolf et al., 2010). Hence, hepatotoxicity of these drugs may be attributed to NTCP inhibition via a mechanism distinct from BSEP inhibition.

    • Age-related differences in reporting of drug-associated liver injury: Data-mining of WHO safety report database

      2014, Regulatory Toxicology and Pharmacology
      Citation Excerpt :

      Relative to adults and the elderly, 29 drugs were associated with a significantly higher reporting frequency of liver injury events in children including: anti-retrovirals (abacavir, indinavir, ritonavir, and zidovudine), CNS agents (pemoline and valproic acid), antimetabolites (methotrexate and mercaptopurine), and antibiotics (oxacillin and minocycline) (Table 2). Drug characteristics were examined for the identified 39 drugs to assess potential associations with differing reporting frequency by age group (Supplementary Table 2) (Chen et al., 2013; Dawson et al., 2012; Thompson et al., 2012; Morgan et al., 2013; DailyMed, 2013; Sakatis et al., 2012; Pessayre et al., 2012; Daly and Day, 2012; Russmann et al., 2010; Yang et al., 2013; Pauli-Magnus and Meier, 2006; Chem Exper Chemical Directory at, 2013; Dykens et al., 2007; Zamek-Gliszczynski et al., 2005; Ramappa and Aithal, 2012; Apostolova et al., 2011; Minuesa et al., 2011; Chowdhury et al., 2006; Ouslander, 1981; Food and Drug Administration Import Refusal Report: Josamycin, 2013; Nadanaciva et al., 2010; Tachibana et al., 1975; Freymond et al., 2003; Tapner et al., 2004; National Institutes of Health, 2013; Wolf et al., 2010; Rosh et al., 1998; Marotta and Roberts, 1998; Karras et al., 2012; Jemnitz et al., 2010; MRHA Drug Safety Update, 2013; Papapetrou et al., 1972; Evered, 1976; Horio, 1975; Kuhnz et al., 1997; Turnheim, 2005; Alfirevic and Pirmohamed, 2010; Drug Information Online, 2013; Bode et al., 2002; Norrby, 1987; Riess and Bruckner-Tuderman, 1990; Boelsterli, 2002; Bernareggi, 1998; Saab et al., 2013; Van Steenbergen et al., 1998; Arrow Roxithromycin, 2013; Puri and Lassman, 1987; Kalliokoski and Niemi, 2009; Chew et al., 2006). A high daily dose (⩾100 mg) was present in the large majority of the identified drugs (87%), which was significantly higher than reported prevalence (<32%) of this daily dose among the 230 drugs most commonly used in the US (p < 0.0001) (Lammert et al., 2008). The prevalence of the high daily dose was similar between the elderly and children (90% vs 86%, respectively).

    • Progress in computational toxicology

      2014, Journal of Pharmacological and Toxicological Methods
      Citation Excerpt :

      This model did not appear to have undergone external testing (Hirano et al., 2006; Saito et al., 2009). Hepatotoxic compounds may also be inhibitors of the bile acid uptake by Na(+)-dependent taurocholate cotransporting polypeptide (NTCP/SLC10A1) which is a key bile acid transporter expressed at the basolateral membrane of hepatocytes (Wolf et al., 2010). NTCP transports bile salts (including 80% of conjugated bile salts) taken up into the liver from the portal blood into the liver (Kullak-Ublick, Stieger, & Meier, 2004).

    View all citing articles on Scopus

    This work was presented at the Society of Toxicology 47th Annual Meeting and ToxExpo™ in Seattle, WA, March 2008.

    View full text