Critical role of free cytosolic calcium, but not uncoupling, in mitochondrial permeability transition and cell death induced by diclofenac oxidative metabolites in immortalized human hepatocytes

https://doi.org/10.1016/j.taap.2006.09.012Get rights and content

Abstract

Diclofenac is a widely used nonsteroidal anti-inflammatory drug that has been associated with rare but serious hepatotoxicity. Experimental evidence indicates that diclofenac targets mitochondria and induces the permeability transition (mPT) which leads to apoptotic cell death in hepatocytes. While the downstream effector mechanisms have been well characterized, the more proximal pathways leading to the mPT are not known. The purpose of this study was to explore the role of free cytosolic calcium (Ca2+c) in diclofenac-induced cell injury in immortalized human hepatocytes. We show that exposure to diclofenac caused time- and concentration-dependent cell injury, which was prevented by the specific mPT inhibitor cyclosporin A (CsA, 5 μM). At 8 h, diclofenac caused increases in [Ca2+]c (Fluo-4 fluorescence), which was unaffected by CsA. Combined exposure to diclofenac/BAPTA (Ca2+ chelator) inhibited cell injury, indicating that Ca2+ plays a critical role in precipitating mPT. Diclofenac decreased the mitochondrial membrane potential, ΔΨm (JC-1 fluorescence), even in the presence of CsA or BAPTA, indicating that mitochondrial depolarization was not a consequence of the mPT or elevated [Ca2+]c. The CYP2C9 inhibitor sulphaphenazole (10 μM) protected from diclofenac-induced cell injury and prevented increases in [Ca2+]c, while it had no effect on the dissipation of the ΔΨm. Finally, diclofenac exposure greatly increased the mitochondria-selective superoxide levels secondary to the increases in [Ca2+]c. In conclusion, these data demonstrate that diclofenac has direct depolarizing effects on mitochondria which does not lead to cell injury, while CYP2C9-mediated bioactivation causes increases in [Ca2+]c, triggering the mPT and precipitating cell death.

Introduction

Diclofenac is a widely used nonsteroidal anti-inflammatory drug. It is generally safe but, in view of the large consumer population worldwide, the drug has been associated with significant adverse effects including gastrointestinal bleeding and ulceration, but also small increases in plasma alanine aminotransferase (ALT) activity, suggesting mild hepatic injury. In rare cases, diclofenac causes more severe idiosyncratic hepatotoxicity (Banks et al., 1995, Boelsterli et al., 1995, Boelsterli, 2003). The mechanisms underlying diclofenac liver liability have not been established as the susceptibility factors determining the human health risk are currently unknown. However, a number of studies have addressed the hazard of diclofenac in cellular models. For example, reactive metabolites have been invoked to account for some of these effects (Kretz-Rommel and Boelsterli, 1993, Tang, 2003); in fact, diclofenac can be glucuronidated to a protein-reactive acyl glucuronide that can reach high concentrations in the biliary tract due to active uphill secretion from the hepatocytes into the bile canaliculi (Kretz-Rommel and Boelsterli, 1994, Boelsterli, 2002). In addition, diclofenac has also been shown to be oxidatively metabolized by CYP2C9/19 to mono- or dihydroxylated derivatives, and these ring-hydroxylated metabolites have been suggested to be involved in producing oxidant stress and glutathione adducts due to their ability to form benzoquinone imines with the amino group in the para position (Shen et al., 1999, Tang et al., 1999a, Poon et al., 2001). However, the role of these metabolites has primarily been studied in rat hepatocytes, and their significance in diclofenac-associated liver injury in humans has remained unknown.

Apart from these possible mechanisms that involve reactive intermediates, diclofenac has also been demonstrated to target mitochondria and induce mitochondrial dysfunction (Masubuchi et al., 1998, Gomez-Lechon et al., 2003b, Inoue et al., 2004). Specifically, in isolated rat or mouse liver mitochondria, diclofenac readily uncouples oxidative phosphorylation (ox-phos), leading to a collapse of the mitochondrial inner transmembrane potential (ΔΨm). Furthermore, diclofenac can induce the mitochondrial permeability transition (mPT) in isolated mitochondria (Masubuchi et al., 2002, Gomez-Lechon et al., 2003a), which may lead to the release of proapoptotic factors into the cytosol and nucleus and activate the cell death pathways. Indeed, in-depth studies have characterized the diclofenac-mediated activation of caspase cascades in hepatocytes and have shown that caspase-8 and -9 are apical caspases that activate executioner caspase-3 and lead to apoptotic cell death (Gomez-Lechon et al., 2003b).

While these distal pathways are well known, relatively little is known about the more proximal mechanisms of diclofenac-induced cell injury. For example, the mPT can be triggered by a number of stress factors. Among these factors are not only reactive oxygen species (ROS) and increases in Ca2+, but also thiol-reactive metabolites that can oxidize critical sulfhydryl groups in the protein components that are part of the mPT pore (Petronilli et al., 1994, He and Lemasters, 2002). In addition, the role of diclofenac metabolic activation and their relationship to cell injury has not been adequately addressed. Since most of the studies on the induction of apoptosis were performed in isolated mitochondria or cultured rat hepatocytes, it is important to consider the species-specific bioactivation pathways and to explore the upstream mechanisms in human hepatocytes.

The purpose of this study was therefore to examine the role of [Ca2+]c and oxidant stress in diclofenac-induced mitochondrial toxicity in an immortalized human hepatocellular cell line (HC-04) (Sattabongkot et al., 2006). We chose this metabolism-competent cell line to obtain a consistent experimental background, as primary human hepatocytes exhibit great variation in CYP expression. The second aim of the study was to explore whether oxidative metabolites of diclofenac were involved. While we found that diclofenac readily causes uncoupling, it is the oxidative metabolites that cause increased [Ca2+]c, which is a critical factor for the initiation of the mPT and cell death in these immortalized human hepatocytes.

Section snippets

Chemicals

All chemicals were purchased from Sigma (St. Louis, MO) unless otherwise stated.

Culture of human immortalized HC-04 cells

HC-04 cells (Siam Life Science Ltd, Bangkok, Thailand) were grown in Hepatocyte Basal Medium (HBM) and supplemented with Hepatocyte Culture Medium Bulletkit (Cambrex, Baltimore, MD), supplemented with 10% fetal bovine serum (Hyclone, Logan, UT). Primaria flasks (Falcon) were coated with 0.01 mg/ml each of fibronectin and BSA as well as 0.03 mg/ml collagen in HBM for 2 h at 37 °C. The cells were maintained at 37 °C

Diclofenac induces mPT-mediated cell injury in cultured human HC-04 cells

Exposure of HC-04 cells to diclofenac resulted in time and concentration-dependent cell injury, as assessed by LDH release into the extracellular medium (Fig. 1). Diclofenac at concentrations ≥ 500 μM resulted in significant cytotoxicity as compared to solvent controls, which became apparent at a late time point (48 h). This delayed toxicity suggests that time-dependent intracellular signaling pathways were activated, or that generation of (a) reactive intermediate(s) was required before the

Discussion

The aim of this study was to explore the upstream mechanisms of diclofenac-induced cell injury in cultured immortalized human hepatocytes. In particular, we investigated the roles of cytosolic free calcium ([Ca2+]c) and mitochondrial depolarization in triggering the more downstream events leading to the mPT and lethal cell injury. An important question was whether these potential changes were dependent on CYP-mediated oxidative biotransformation of diclofenac. We found that diclofenac at high

Acknowledgments

We thank Dr. J.S. Bian and Dr. C. Latchoumycandane for kind help and discussions. This work was supported by grants from the National Medical Research Council (R-184-000-087-213, to U.A.B.), the Biomedical Research Council (R-184-000-096-305, to U.A.B.), and the Toxicology Program of the NUS Office of Life Sciences (R-184-000-079-712, to U.A.B.).

References (41)

  • A. Inoue et al.

    Molecular mechanisms of diclofenac-induced apoptosis of promyelocytic leukemia: dependency on reactive oxygen species, Akt, Bid, cytochrome c, and caspase pathway

    Free Radical Biol. Med.

    (2004)
  • A. Kretz-Rommel et al.

    Diclofenac covalent protein binding is dependent on acyl glucuronide formation and is inversely related to acute cell injury in cultured rat hepatocytes

    Toxicol. Appl. Pharmacol.

    (1993)
  • P.B. Limaye et al.

    Calpain released from dying hepatocytes mediates progression of acute liver injury induced by model hepatotoxicants

    Toxicol. Appl. Pharmacol.

    (2003)
  • Y. Masubuchi et al.

    Role of mitochondrial permeability transition in diclofenac-induced hepatocyte injury in rats

    Hepatology

    (2002)
  • R. Moreno-Sanchez et al.

    Inhibition and uncoupling of oxidative phosphorylation by nonsteroidal anti-inflammatory drugs

    Biochem. Pharmacol.

    (1999)
  • I. Petrescu et al.

    Uncoupling effects of diclofenac and aspirin in the perfused liver and isolated hepatic mitochondria of rat

    Biochim. Biophys. Acta (Bioenergetics)

    (1997)
  • V. Petronilli et al.

    The voltage sensor of the mitochondrial permeability transition pore is tuned by the oxidation-reduction state of vicinal thiols. Increase of the gating potential by oxidants and its reversal by reducing agents

    J. Biol. Chem.

    (1994)
  • S.A. Uyemura et al.

    Diclofenac sodium and mefenamic acid: potent inducers of the membrane permeability transition in renal cortex mitochondria

    Arch. Biochem. Biophys.

    (1997)
  • U.A. Boelsterli

    Xenobiotic acyl glucuronides and acyl CoA thioesters as protein-reactive metabolites with the potential to cause idiosyncratic drug reactions

    Curr. Drug Metab.

    (2002)
  • U.A. Boelsterli et al.

    Idiosyncratic liver toxicity of nonsteroidal antiinflammatory drugs: molecular mechanisms and pathology

    Crit. Rev. Toxicol.

    (1995)
  • Cited by (0)

    View full text