Elsevier

Chemico-Biological Interactions

Volume 255, 5 August 2016, Pages 63-73
Chemico-Biological Interactions

Development of HepG2-derived cells expressing cytochrome P450s for assessing metabolism-associated drug-induced liver toxicity

https://doi.org/10.1016/j.cbi.2015.10.009Get rights and content

Highlights

  • We developed a battery of HepG2-derived cell lines that express 14 individual CYPs.

  • We confirmed the functionality of the CYPs at the mRNA, protein and enzymatic activity levels.

  • We examined the metabolism-related toxicity of three drugs in our cell system.

Abstract

The generation of reactive metabolites from therapeutic agents is one of the major mechanisms of drug-induced liver injury (DILI). In order to evaluate metabolism-related toxicity and improve drug efficacy and safety, we generated a battery of HepG2-derived cell lines that express 14 cytochrome P450s (CYPs) (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5 and 3A7) individually using a lentiviral expression system. The expression/production of a specific CYP in each cell line was confirmed by an increased abundance of the CYP at both mRNA and protein levels. Moreover, the enzymatic activities of representative CYPs in the corresponding cell lines were also measured. Using our CYP-expressed HepG2 cells, the toxicity of three drugs that could induce DILI (amiodarone, chlorpromazine and primaquine) was assessed, and all of them showed altered (increased or decreased) toxicity compared to the toxicity in drug-treated wild-type HepG2 cells. CYP-mediated drug toxicity examined in our cell system is consistent with previous reports, demonstrating the potential of these cells for assessing metabolism-related drug toxicity. This cell system provides a practical in vitro approach for drug metabolism screening and for early detection of drug toxicity. It is also a surrogate enzyme source for the enzymatic characterization of a particular CYP that contributes to drug-induced liver toxicity.

Introduction

Drug-induced liver injury (DILI) is a leading cause of drug failures in clinical trials and the major reason of drug withdrawals from the market [1]. Identification of drugs that cause liver injury at the early stage of drug development poses a challenge to both the pharmaceutical industry and the U.S. Food and Drug Administration (FDA). Preclinical testing is one of the important approaches to early detect drug toxicity, and there is a constant need for the development of improved tools to facilitate toxicity assessment and risk identification.

The causes of DILI are multifactorial, including toxic effects caused by reactive metabolites, reactive oxygen species, inflammatory reactions, mitochondrial dysfunction, and imbalances between cellular damage and protective responses [1], [2], [3], [4], [5]. Metabolism-related toxicity is generally mediated by the generation of reactive metabolites from non-toxic parenteral compounds, particularly via cytochrome P450 (CYP) enzyme pathways [6]. The inter-individual variability in the expression of drug metabolizing genes predisposes certain individuals to increased susceptibility to DILI [7], [8]. Therefore, it is important to examine the roles of drug metabolizing enzymes and identify specific metabolizing enzymes that contribute to drug-induced liver toxicity.

Numerous in vitro models, such as recombinant enzymes, liver microsomes, liver cytosolic fractions, hepatic cells, liver slices and isolated perfused livers, have been used to examine drug-related hepatotoxicity [9]. Traditionally, cell-based assays have been performed using human primary hepatocytes, either freshly isolated or cryopreserved, to evaluate drug metabolism and drug–drug interactions [10], [11]. Indeed, the application of primary human hepatocytes in drug metabolism and toxicity studies is considered as a “gold standard”, because, under appropriate conditions, these cells retain functional activity of the major drug-metabolizing enzymes [12]. However, phenotypic instability, short life span, batch-to-batch variation and limited availability of primary human hepatocytes constrain their broad use.

Human hepatoma cell lines, such as HepG2, Hep3B, and Huh7, have been widely used in toxicity screening and mechanistic studies, owing to their high stability, unlimited life-span and ready availability. However, lower or no expression of the majority of drug-metabolizing genes is the most critical drawback associated with using these cell lines for drug metabolism and toxicity studies [13], [14]. As a strategy to overcome this limitation, genetically modified hepatic cell lines expressing human drug metabolizing genes have been developed and used for assessing drug metabolism and toxicity. For example, using adenoviral or lentiviral infection systems, cells that transiently or stably express individual CYPs, such as CYP1A1, CYP2C8, CYP2C9 or CYP3A4, have been generated [15], [16], [17], [18]. These cells responded appropriately to known toxic chemicals, demonstrating their values for toxicity testing and mechanistic studies. However, not all of them are publicly available.

In this study, we aimed to develop a comprehensive set of cell lines that express the major human CYPs individually, to provide surrogate hepatic cell lines for the study of metabolism-mediated drug hepatotoxicity and the identification of specific CYP isoforms responsible for the metabolism of a drug. Toward this goal, using the lentiviral expression system, HepG2-derived cell lines expressing 14 individual CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5 and 3A7) were generated, and the functionality of these CYPs was confirmed at the mRNA, protein, and enzymatic activity levels. In addition, three drugs that could cause metabolism-mediated DILI were examined to evaluate the utility of these cells in drug metabolism and toxicity screening.

Section snippets

Chemicals and reagents

Dulbecco's modified Eagle's medium (DMEM), amiodarone hydrochloride, chlorpromazine hydrochloride, primaquine bisphosphate, proadifen (SKF-525A, SKF), alpha-naphthoflavone (ANF), ketoconazole (KET) and dimethyl sulfoxide (DMSO) were purchased from Sigma–Aldrich (St. Louis, MO). Fetal bovine serum (FBS) was from Atlanta Biologicals (Lawrenceville, GA). Blasticidin S hydrochloride and antibiotic-antimycotic were from Life Technologies (Grand Island, NY).

Cell culture

The human hepatocellular carcinoma cell

Results and discussion

The central role of the liver in the metabolism and clearance of drugs makes it a major target of drug toxicity. A large number of drugs have been reported to cause hepatotoxicity, posing a profound challenge to the pharmaceutical industry and regulatory agencies [21], [22]. Although the mechanisms underlying drug-induced hepatotoxicity remain poorly understood, it has been widely accepted that in many cases the initial step triggering the progression of hepatotoxicity involves metabolism of a

Summary

In summary, we generated a metabolically competent cell system by stable transduction of CYP cDNAs individually. We also showed the potential of the system for in vitro screening of metabolism-related drug toxicity. Such an in vitro assay system represents a useful paradigm for the evaluation and early detection of drug toxicity. Considering the large variations in metabolic phenotypes observed within and among human populations that impact the use of normal human hepatocytes in pharmacological

Acknowledgments

JX and SC were supported by appointments to the Postgraduate Research Program at the National Center for Toxicological Research administered by the Oak Ridge Institute for Science Education through an interagency agreement between the U.S. Department of Energy and the U.S. FDA.

This article is not an official guidance or policy statement of the U.S. FDA. No official support or endorsement by the U.S. FDA is intended or should be inferred.

References (94)

  • J.K. Lamba et al.

    Genetic contribution to variable human CYP3A-mediated metabolism

    Adv. Drug Deliv. Rev.

    (2002)
  • C. Rodriguez-Antona et al.

    Cytochrome P-450 mRNA expression in human liver and its relationship with enzyme activity

    Arch. Biochem. Biophys.

    (2001)
  • M. Chen et al.

    FDA-approved drug labeling for the study of drug-induced liver injury

    Drug Discov. Today

    (2011)
  • H.R. Ha et al.

    Identification and quantitation of novel metabolites of amiodarone in plasma of treated patients

    Eur. J. Pharm. Sci. Off. J. Eur. Fed. Pharm. Sci.

    (2005)
  • P. Turano et al.

    Thin-layer chromatography of chlorpromazine metabolites. Attempt to identify each of the metabolites appearing in blood, urine and feces of chronically medicated schizophrenics

    J. Chromatogr.

    (1973)
  • K. Yoshii et al.

    Identification of human cytochrome P450 isoforms involved in the 7-hydroxylation of chlorpromazine by human liver microsomes

    Life Sci.

    (2000)
  • J. Wojcikowski et al.

    Main contribution of the cytochrome P450 isoenzyme 1A2 (CYP1A2) to N-demethylation and 5-sulfoxidation of the phenothiazine neuroleptic chlorpromazine in human liver–A comparison with other phenothiazines

    Biochem. Pharmacol.

    (2010)
  • R.G. Watson et al.

    A proposed mechanism for chlorpromazine jaundice–defective hepatic sulphoxidation combined with rapid hydroxylation

    J. Hepatol.

    (1988)
  • J. Vasquez-Vivar et al.

    Hydroxylated metabolites of the antimalarial drug primaquine. Oxidation and redox cycling

    J. Biol. Chem.

    (1992)
  • C.M. Link et al.

    Structure-activity relationships of putative primaquine metabolites causing methemoglobin formation in canine hemolysates

    Toxicol. Appl. Pharmacol.

    (1985)
  • K.A. Fletcher et al.

    Studies on the mechanisms of oxidation in the erythrocyte by metabolites of primaquine

    Biochem. Pharmacol.

    (1988)
  • S. Ganesan et al.

    Cytochrome P(450)-dependent toxic effects of primaquine on human erythrocytes

    Toxicol. Appl. Pharmacol.

    (2009)
  • F. Fontaine et al.

    Cytochrome 1A1 induction by primaquine in human hepatocytes and HepG2 cells: absence of binding to the aryl hydrocarbon receptor

    Biochem. Pharmacol.

    (1999)
  • N. Kaplowitz

    Idiosyncratic drug hepatotoxicity, nature reviews

    Drug Discov.

    (2005)
  • H. Jaeschke et al.

    Oxidant stress, mitochondria, and cell death mechanisms in drug-induced liver injury: lessons learned from acetaminophen hepatotoxicity

    Drug Metab. Rev.

    (2012)
  • C. Ju et al.

    Role of immune reactions in drug-induced liver injury (DILI)

    Drug Metab. Rev.

    (2012)
  • R.J. Andrade et al.

    Pharmacogenomics in drug induced liver injury

    Curr. Drug Metab.

    (2009)
  • S. Russmann et al.

    Pharmacogenetics of drug-induced liver injury

    Hepatology

    (2010)
  • S.M. Huang et al.

    New era in drug interaction evaluation: US food and drug administration update on CYP enzymes, transporters, and the guidance process

    J. Clin. Pharmacol.

    (2008)
  • A.P. Li

    Evaluation of drug metabolism, drug-drug interactions, and in vitro hepatotoxicity with cryopreserved human hepatocytes

    Methods Mol. Biol.

    (2010)
  • P. Godoy et al.

    Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME

    Arch. Toxicol.

    (2013)
  • M.T. Donato et al.

    Cell lines: a tool for in vitro drug metabolism studies

    Curr. Drug Metab.

    (2008)
  • L. Guo et al.

    Similarities and differences in the expression of drug-metabolizing enzymes between human hepatic cell lines and primary human hepatocytes

    Drug Metab. Dispos. Biol. Fate Chem.

    (2011)
  • A. Iwamura et al.

    CYP2C9-mediated metabolic activation of losartan detected by a highly sensitive cell-based screening assay

    Drug Metab. Dispos. Biol. Fate Chem.

    (2011)
  • H. Hosomi et al.

    Development of a highly sensitive cytotoxicity assay system for CYP3A4-mediated metabolic activation

    Drug Metab. Dispos. Biol. Fate Chem.

    (2011)
  • W.M. Lee

    Drug-induced hepatotoxicity

    N. Engl. J. Med.

    (2003)
  • B. Gunawan et al.

    Clinical perspectives on xenobiotic-induced hepatotoxicity

    Drug Metab. Rev.

    (2004)
  • D.J. Antoine et al.

    Understanding the role of reactive metabolites in drug-induced hepatotoxicity: state of the science

    Expert Opin. Drug Metab. Toxicol.

    (2008)
  • S.M. Attia

    Deleterious effects of reactive metabolites

    Oxidative Med. Cell. Longev.

    (2010)
  • A. Srivastava et al.

    Role of Reactive Metabolites in Drug-induced Hepatotoxicity, Handbook of Experimental Pharmacology

    (2010)
  • J. Bai et al.

    Adenovirus mediated overexpression of CYP2E1 increases sensitivity of HepG2 cells to acetaminophen induced cytotoxicity

    Mol. Cell Biochem.

    (2004)
  • Q.G. Yan et al.

    Overexpression of CYP2E1 enhances sensitivity of hepG2 cells to fas-mediated cytotoxicity

    Cancer Biol. Ther.

    (2008)
  • B.B. Knowles et al.

    Human hepatocellular carcinoma cell lines secrete the major plasma proteins and hepatitis B surface antigen

    Science

    (1980)
  • F.J. Gonzalez et al.

    Cytochromes P450 expression systems

    Annu. Rev. Pharmacol. Toxicol.

    (1995)
  • A.S. Cockrell et al.

    Gene delivery by lentivirus vectors

    Mol. Biotechnol.

    (2007)
  • M. Nasri et al.

    Production, purification and titration of a lentivirus-based vector for gene delivery purposes

    Cytotechnology

    (2014)
  • D. Skalamera et al.

    Generation of a genome scale lentiviral vector library for EF1alpha promoter-driven expression of human ORFs and identification of human genes affecting viral titer

    PLoS One

    (2012)
  • Cited by (60)

    • Alisertib shows negligible potential for perpetrating pharmacokinetic drug-drug interactions on ABCB1, ABCG2 and cytochromes P450, but acts as dual-activity resistance modulator through the inhibition of ABCC1 transporter

      2022, Toxicology and Applied Pharmacology
      Citation Excerpt :

      Parent A431 (human squamous carcinoma) cell line with its ABCB1-, ABCG2- and ABCC1-overexpressing relatives were donated by Dr. Balasz Sarkadi (Hungarian Academy of Sciences, Budapest, Hungary) (Elkind et al., 2005). HepG2 (human hepatocarcinoma) cells lentivirally-transduced for the overexpression of human CYP3A4 (HepG2-CYP3A4) were kindly provided by Dr. Lei Guo (National Center for Toxicological Research, U.S. FDA, AR, USA) (Xuan et al., 2016). Caco-2 (human colorectal adenocarcinoma) together with A549 and NCI-H1299 (human non-small cell lung cancer, NSCLC) cells were obtained from the American Type Culture Collection (Manassas, VA, USA).

    View all citing articles on Scopus
    View full text