Induction of cellular oxidative stress by aryl hydrocarbon receptor activation

https://doi.org/10.1016/S0009-2797(02)00067-4Get rights and content

Abstract

The aryl hydrocarbon receptor (AHR) has long been associated with the induction of a battery of genes involved in the metabolism of foreign and endogenous compounds. Depending on experimental conditions, AHR can mediate either activation or amelioration of chemical toxicity. For the past decade, evidence has mounted that AHR is associated with a cellular oxidative stress response that must be considered when evaluating the mechanism of action of xenobiotics capable of activating AHR, or capable of metabolic activation by enzymes encoded by genes under control of AHR. In this review, we have evaluated the diverse mechanisms by which AHR generates an oxidative stress response, including inflammation, antioxidant and prooxidant enzymes and cytochrome P450. A review of the regulation of Ahr transcription and functional polymorphisms especially related to oxidative stress is also included. We have carefully avoided placing a value judgment on the degree of toxicity produced by such a response, in view of the realization that an oxidative response is involved in many normal physiological processes. Since the interface between physiological, adaptive and toxicological responses elicited by the AHR-mediated oxidative stress response is not clearly defined, it behooves the researcher to evaluate both toxicological and physiological features of the response.

Introduction

The aryl hydrocarbon receptor (AHR) is a ligand-activated member of the per-arnt-sim (PAS) family of basic-helix-loop-helix transcription factors [1], [2]. Prior to activation, AHR is found in the cytosol in association with heat shock protein (HSP)90 and HSP90 accessory proteins. Ligand binding releases it from this complex and promotes nuclear translocation. In the nucleus, the ligand-activated AHR forms a heterodimeric complex with the aryl hydrocarbon nuclear translocator (ARNT) [1]. The complex binds to aryl hydrocarbon response elements (AhRE)—also known as xenobiotic response elements (XRE), and dioxin response elements (DRE)—which function as cis-acting enhancers in the regulatory domains of a growing number of genes collectively known as the AHR gene battery[3]. Ligands for AHR include planar polycyclic and halogenated aromatic hydrocarbons and diverse classes of plant-derived chemicals. It has been hypothesized that the AHR–ARNT transcriptional complex and the genes that it regulates evolved for defense against an increasingly diverse array of plant toxins and as a result it is unlikely to serve endogenous physiological functions [4]. Recent studies, however, suggest that many important physiological and developmental processes are regulated by AHR. The activation of AHR results in rapid transcriptional activation of a large number of genes whose products control a broad spectrum of cellular functions [3], [5], [6]. This is likely, in part, due to interactions between AHR and transcription factors other than ARNT, some of which are involved in the control of complex cellular programs, such as cell division and cell fate [6], [7], [8], [9], [10] (see also Puga, Xia and Elferink herein). Furthermore, AHR-null mutant mice, although viable, suffer numerous age-related pathologies involving multiple organ systems [11]. In light of such studies, it is likely that AHR has also an important role in cellular homeostasis and that its activation by environmental chemicals may have the dual effect of triggering its homeostatic functions in some cases and disrupting them in others. Whether or not AHR activation leads to toxic effects will probably be determined by the diversity of AHR interactions, the complexity of the cellular transcriptome, the persistence of AHR activation, and the nature of the AHR ligand.

In addition to the control of physiological processes, AHR is mechanistically involved in toxicological processes involving oxidative stress. In this review, we will use the term oxidative stress for any condition that increases the cellular oxidation state to produce an oxidative stress response. This generally results from increasing the production of reactive oxygen species (ROS) (superoxide, hydrogen peroxide, hydroxyl radical, peroxynitrite, singlet oxygen) relative to cellular antioxidant defenses (antioxidants, antioxidant enzymes). Although an oxidative stress response does not necessarily result in toxicity, it is a component in the mechanism of many toxic events. In this review, we will only deal in passing with the classical concept that AHR regulates the expression of phase I and phase II detoxification enzymes that, by the nature of their enzymatic activities, generate electrophilic reactions, which cause oxidative stress, and conjugation reactions, which combat it. We chose to focus on systems in which AHR activation results in a shift in the cellular redox balance, resulting in an oxidative stress response. The literature reporting this phenomenon is vast and diverse; we apologize to those colleagues whose work is not cited.

Section snippets

The role of AHR in the induction of genes associated with inflammation

The inflammatory response involves an extremely complex system of cross-talk interactions between cells of the immune system and non-immune cells. This response is generally beneficial to the organism; however, under certain conditions, an aggressive inflammatory response can be pathological, and much of this pathology has been attributed to the production of inflammatory cytokines. For example, high levels of TNFα, such as those seen as a result of bacterial sepsis, have been demonstrated to

Superoxide dismutase

The incomplete reduction of O2 by several enzyme systems results in the generation of superoxide. The primary cellular defense against superoxide is reduction by superoxide dismutase (SOD). Eukaryotic cells have two genes that encode two different SOD enzymes. Following translation, the Mn-dependent SOD, MnSOD, is imported into the mitochondria, while the Cu/Zn-dependent enzyme, Cu/ZnSOD, remains in the cytoplasm. Although endogenous expression analysis of the gene was not reported, the rat

AHR activation alters estrogen metabolism to produce an oxidative stress response

In long-term bioassays, TCDD significantly increased the incidence of liver tumors in female, but not in male, rats [79], [80]. It is a general observation that female rats are more susceptible to TCDD-induced oxidative stress [81], DNA oxidative damage [82] and hepatocarcinogenesis [83]. These responses are mediated, at least in part, by estrogen [84], [85]. The presumption that oxidative stress and carcinogenesis is mediated by estrogen was confirmed in Syrian hamsters, which show 100% kidney

Oxidant stress downregulates AHR-dependent transcription

The numerous studies cited above support the notion that AHR activation shifts the cellular redox state towards oxidizing conditions, perhaps as a consequence of the increased expression of CYP1 family genes. In this regard, it is interesting to note that oxidant stress in turn downregulates CYP1A1 expression [119], [120]. Under physiological oxidant stress conditions, the AHR–ARNT transcriptional complex is not redox-sensitive; however, AHR–ARNT acts in synergy with a redox-sensitive

Functional polymorphisms in AHR

Toxic responses, including oxidative stress, are modified by naturally occurring polymorphisms of AHR. Like many other transcription factors, AHR has been amenable to dissection into functional domains. In broad terms, the N-terminal half of AHR consists of the basic-region-helix-loop-helix domain and of two PAS domains which overlap in function and are responsible for DNA binding, ligand binding and dimerization [1]. The C-terminal half of AHR is responsible for transactivation. In the context

Concluding remarks

Studies aimed at understanding AHR-mediated toxicity have led to the discovery of AHR variants that appear to maintain physiological function and yet confer greatly to diminish toxicity. This is probably due to the remarkable structural plasticity of AHR, as the studies in inbred mouse strains and rats have shown. The human AHRs thus far studied demonstrate ligand-binding affinity characteristics similar to those of the low-affinity mouse strains, which might be an important factor explaining

Acknowledgements

Preparation of this review and the work done in our labs cited herein were supported in part by NIH Grants RO1 ES10133, ES06273, ES10807, and P30 ES06096.

References (145)

  • M.J. Taylor et al.

    Inhibition of acute TCDD toxicity by treatment with anti-tumor necrosis factor antibody or dexamethasone

    Toxicol. Appl. Pharmacol.

    (1992)
  • A.B. Moos et al.

    Inhibition of tumor necrosis factor activity fails to restore 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced suppression of the antibody response to sheep red blood cells

    Toxicol. Lett.

    (1995)
  • H.G. Jeong

    Cytokine-mediated suppression of cytochrome P450 1A1 in Hepa-1c1c7 cells by pokeweed mitogen

    Toxicol. Lett.

    (2001)
  • S. Ke et al.

    Mechanism of suppression of cytochrome P450 1A1 expression by TNF-alpha and lipopolysaccharide

    J. Biol. Chem.

    (2001)
  • A. Puga et al.

    Activation of transcription factors activator protein-1 and nuclear factor-kappa B by 2,3,7,8-tetrachlorodibenzo-p-dioxin

    Biochem. Pharmacol.

    (2000)
  • J.A. Maier et al.

    Cyclooxygenase is an immediate-early gene induced by interleukin-1 in human endothelial cells

    J. Biol. Chem.

    (1990)
  • A. Puga et al.

    Sustained increase in intracellular free calcium and activation of cyclooxygenase-2 expression in mouse hepatoma cells treated with dioxin

    Biochem. Pharmacol.

    (1997)
  • S.A. Kraemer et al.

    Regulation of prostaglandin endoperoxide H synthase-2 expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin

    Arch. Biochem. Biophys.

    (1996)
  • B.P. Lawrence et al.

    Role of altered arachidonic acid metabolism in 2,3,7,8-tetrachlorodibenzo-p-dioxin-induced immune suppression in C57Bl/6 mice

    Toxicol. Sci.

    (1998)
  • H. Giles et al.

    The biology and pharmacology of PGD2

    Prostaglandins

    (1988)
  • M. Fukushima

    Biological activities and mechanisms of action of PGJ2 and related compounds: an update

    Prostaglandins Leukot. Essent. Fatty Acids

    (1992)
  • M. Kondo et al.

    Cyclopentenone prostaglandins as potential inducers of intracellular oxidative stress

    J. Biol. Chem.

    (2001)
  • L. Li et al.

    15-Deoxy-delta 12,14-prostaglandin J2 induces apoptosis of human hepatic myofibroblasts. A pathway involving oxidative stress independently of peroxisome-proliferator-activated receptors

    J. Biol. Chem.

    (2001)
  • G. Lenaz

    Role of mitochondria in oxidative stress and aging

    Biochim. Biophys. Acta

    (1998)
  • H.Y. Yoo et al.

    Xenobiotic-responsive element for the transcriptional activation of the rat Cu/Zn superoxide dismutase gene

    Biochem. Biophys. Res. Commun.

    (1999)
  • Y. Groner et al.

    Cell damage by excess Cu/ZnSOD and Down's syndrome

    Biomed. Pharmacother.

    (1994)
  • B.J. Zimmerman et al.

    Mechanisms of reperfusion injury

    Am. J. Med. Sci.

    (1994)
  • C.A. Pritsos

    Cellular distribution, metabolism and regulation of the xanthine oxidoreductase enzyme system

    Chem. Biol. Interact.

    (2000)
  • K. Sugihara et al.

    Aryl hydrocarbon receptor (AhR)-mediated induction of xanthine oxidase/xanthine dehydrogenase activity by 2,3,7,8-tetrachlorodibenzo-p-dioxin

    Biochem. Biophys. Res. Commun.

    (2001)
  • S.C. Bondy et al.

    Contribution of hepatic cytochrome P450 systems to the generation of reactive oxygen species

    Biochem. Pharmacol.

    (1994)
  • J. Capdevila et al.

    Inhibitors of cytochrome P450-dependent arachidonic acid metabolism

    Arch. Biochem. Biophys.

    (1988)
  • A.B. Rifkind et al.

    Arachidonic metabolism by dioxin-induced cytochrome P450: a new hypothesis on the role of P450 in dioxin toxicity

    Biochem. Biophys. Res. Commun.

    (1990)
  • K. Nakai et al.

    Naphthoflavone induction of a cytochrome P450 arachidonic acid epoxygenase in chick embryo liver distinct from the aryl hydrocarbon hydroxylase and from phenobarbital-induced arachidonate epoxygenase

    J. Biol. Chem.

    (1992)
  • J.J. Schlezinger et al.

    Induction and suppression of cytochrome P450 1A by 3,3′,4,4′,5-pentachlorobiphenyl and its relationship to oxidative stress in the marine fish scup (Stenotomus chrysops)

    Aquat. Toxicol.

    (2001)
  • H.G. Shertzer et al.

    TCDD causes a sustained oxidative stress response in C57BL/6J mice

    Biochem. Biophys. Res. Commun.

    (1998)
  • M.E. Andersen et al.

    Regional hepatic CYP1A1 and CYP1A2 induction with 2,3,7,8-tetrachlorodibenzo-p-dioxin evaluated with a multicompartment geometric model of hepatic zonation

    Toxicol. Appl. Pharmacol.

    (1997)
  • J.J. Diliberto et al.

    Role of CYP1A2 in hepatic sequestration of dioxin: studies using CYP1A2 knockout mice

    Biochem. Biophys. Res. Commun.

    (1997)
  • M.J. Santostefano et al.

    A pharmacodynamic analysis of TCDD-induced cytochrome P450 gene expression in multiple tissues: dose- and time-dependent effects

    Toxicol. Appl. Pharmacol.

    (1998)
  • X. Wang et al.

    Determination of parameters responsible for pharmacokinetic behavior of TCDD in female Sprague–Dawley rats

    Toxicol. Appl. Pharmacol.

    (1997)
  • S.V. Bhagwat et al.

    Dual targeting property of the N-terminal signal sequence of P450 1A1. Targeting of heterologous proteins to endoplasmic reticulum and mitochondria

    J. Biol. Chem.

    (1999)
  • E. Boopathi et al.

    Accumulation of mitochondrial P450 MT2, NH2-terminal truncated cytochrome P450 1A1 in rat brain during chronic treatment with beta-naphthoflavone. A role in the metabolism of neuroactive drugs

    J. Biol. Chem.

    (2000)
  • H.K. Anandatheerthavarada et al.

    Localization of multiple forms of inducible cytochromes P450 in rat liver mitochondria: immunological characteristics and patterns of xenobiotic substrate metabolism

    Arch. Biochem. Biophys.

    (1997)
  • M.A. Robin et al.

    Mitochondrial targeted cytochrome P450 2E1 (P450 MT5) contains an intact N terminus and requires mitochondrial specific electron transfer proteins for activity

    J. Biol. Chem.

    (2001)
  • S.V. Bhagwat et al.

    Constitutive and inducible cytochromes P450 in rat lung mitochondria: xenobiotic induction, relative abundance, and catalytic properties

    Toxicol. Appl. Pharmacol.

    (1999)
  • C. Giulivi et al.

    Hydroxyl radical generation during mitochondrial electron transfer and the formation of 8-hydroxydesoxyguanosine in mitochondrial DNA

    Arch. Biochem. Biophys.

    (1995)
  • Y. Li et al.

    Biochemical characterization of lucigenin (bis-N-methylacridinium) as a chemiluminescent probe for detecting intramitochondrial superoxide anion radical production

    Biochem. Biophys. Res. Commun.

    (1999)
  • Y. Li et al.

    Detection of mitochondria-derived reactive oxygen species production by the chemilumigenic probes lucigenin and luminol

    Biochim. Biophys. Acta

    (1999)
  • E. Cadenas et al.

    Mitochondrial free radical generation, oxidative stress, and aging

    Free Radic. Biol. Med.

    (2000)
  • E.A. Hassoun et al.

    The relative abilities of TCDD and its congeners to induce oxidative stress in the hepatic and brain tissues of rats after subchronic exposure

    Toxicology

    (2000)
  • A. Senft et al.

    Dioxin increases reactive oxygen production in mouse liver mitochondria

    Toxicol. Appl. Pharmacol.

    (2002)
  • Cited by (156)

    View all citing articles on Scopus
    View full text