Elsevier

Advanced Drug Delivery Reviews

Volume 62, Issue 13, 30 October 2010, Pages 1238-1249
Advanced Drug Delivery Reviews

Regulation of drug-metabolizing enzymes by xenobiotic receptors: PXR and CARā˜†

https://doi.org/10.1016/j.addr.2010.08.006Get rights and content

Abstract

Drug-metabolizing enzymes (DMEs) and transporters play pivotal roles in the disposition and detoxification of numerous foreign and endogenous chemicals. To accommodate chemical challenges, the expression of many DMEs and transporters is up-regulated by a group of ligand-activated transcription factors namely nuclear receptors (NRs). The importance of NRs in xenobiotic metabolism and clearance is best exemplified by the most promiscuous xenobiotic receptors: pregnane X receptor (PXR, NR1I2) and constitutive androstane/activated receptor (CAR, NR1I3). Together, these two receptors govern the inductive expression of a largely overlapping array of target genes encoding phase I and II DMEs, and drug transporters. Moreover, PXR and CAR also represent two distinctive mechanisms of NR activation, whereby CAR demonstrates both constitutive and ligand-independent activation. In this review, recent advances in our understanding of PXR and CAR as xenosensors are discussed with emphasis placed on the differences rather than similarities of these two xenobiotic receptors in ligand recognition and target gene regulation.

Introduction

Exposure to xenobiotics such as drugs and environmental chemicals has profound influence on human health. In order to modulate their own metabolism and excretion, xenobiotics alter the transcription of a broad array of genes expressed in multiple tissues and vital organs such as the liver, kidney, intestine, lungs, brain, placenta, and pancreas [1], [2], [3]. Mechanistically speaking however, it is majorly the mammalian nuclear receptor (NR) superfamily of transcription factors that makes xenobiotic regulation of gene expression at the transcriptional level possible. The characteristic structural features of NRs include a highly-conserved DNA-binding domain (DBD), which links the receptors to specific promoter regions of their target genes, and a less conserved ligand-binding domain (LBD) that permits them to directly interact with hormones and/or xenobiotics [4], [5]. Moreover, the extraordinary flexibility in the size and shape of LBDs provides the basis for a number of NRs being able to accommodate a myriad of ligands with diverse chemical structuresĀ [6], [7]. Notably, the first human nuclear receptor, glucocorticoid receptor (GR), was cloned starting from the purification and characterization of the receptor protein from the cortisone-producing adrenal glands, before specific antibodies were used to isolate the corresponding cDNA [8], [9]. Utilizing these classical endocrinology approaches, a number of endocrine receptors such as GR, estrogen receptor (ER), and thyroid hormone receptor (TR), were isolated soon thereafter. Typically all these receptors have relatively compact LDBs and are capable of binding unique high-affinity endogenous ligands at nanomolar concentrations.

Taking advantage of the fact that nearly all NRs share a highly conserved cysteine-rich DBD, researchers strive to search for unrecognized NRs using this common segment as bait to screen recombinant DNA libraries at low stringency [10]. This approach has led to the identification of a group of proteins that resemble NRs on the basis of structure and sequence, but lack identified endogenous ligands. Coined ā€œorphan receptors,ā€ this group of NRs accounts for approximately 60% of the over 70 distinct members of the NR superfamily [11], [12], [13]. The availability of this large number of orphan receptors also triggered a shift of the classic endocrinology approach into the so called ā€œreverse endocrinologyā€ one, whereby instead of using a purified hormone to identify its partner receptor, novel bioactive molecules were recognized as selective ligands of these orphan receptors [14]. As such, a number of the orphan receptors were termed ā€˜adopted.ā€™ Nevertheless, in contrast to the prototypical endocrine receptors, members of the orphan receptors are typically activated by abundant but low-affinity lipophilic molecules at micromolar concentrations [15], [16].

Notably, the majority of ligands for orphan or adopted receptors are xenobiotics including drugs, carcinogens, food additives, pesticides and environmental pollutants [17]. Functioning as sensors of toxic byproducts derived from both endogenous and exogenous chemical breakdowns, a number of these receptors were also termed xenobiotic receptors, which include but are not limited to: farnesoid X receptor (FXR), liver X receptor (LXR), proxisome proliferator activation receptors (PPARs), constitutive androstane/active receptor (CAR), pregnane X receptor (PXR), nuclear factor-erythroid 2-related factor 2 (Nrf2), and aryl hydrocarbon receptor (AhR). Also worth mentioning, though primarily responsive to xenobiotics, AhR belongs to the basic helixā€“loopā€“helix protein of the PAS Per-ARNT-Sim (PAS) family, not to the NR superfamily [18]. Among these xenobiotic receptors, PXR and CAR exhibit promiscuous xenobiotic activation capability; and collaboratively they govern the transcription of a broad spectrum of distinct and overlapping genes encoding phase I, phase II drug-metabolizing enzymes (DMEs), as well as uptake and efflux transporters [17], [19], [20], [21], [22] (Fig.Ā 1).

The purpose of this review is to highlight the recent advances in our understanding of the regulation of DMEs and transporters by xenobiotic receptors: PXR and CAR. Emphasis is given to the distinct, rather than the overlapping roles, of PXR and CAR in gene regulation. To a lesser extent, we also discuss the role of AhR as a xenosensor. This review however is by no means a comprehensive coverage of CAR, PXR, and AhR research findings.

Section snippets

Pregnane X receptor (PXR)

The pregnane X receptor (PXR, NR1I2) is an approximately 434-amino acid, 50-kDa protein, primarily expressed in the liver and intestine [23]. In 1998, three research groups independently isolated cDNAs encoding a novel orphan receptor, PXR, which was subsequently shown to play a central role in the transcriptional regulation of CYP3A genes across multiple species [24], [25], [26]. Prior to being designated NR1I2, this receptor was also named SXR (steroid and xenobiotic receptor) and PAR

Constitutive androstane/activated receptor (CAR)

In the nuclear receptor superfamily tree, CAR (NR1I3) is the closest relative to the abovementioned PXR and is expressed primarily in the liver and intestine. Initially named MB67 in 1994, this receptor was designated as constitutive activated receptor (CAR), because it forms a heterodimer with retinoid X receptor (RXR) that binds to retinoic acid response elements (RAREs) and transactivates target genes in the absence of ligand stimulation [105], [106]. In 1998, the first class of CAR ligands

Aryl hydrocarbon receptor (AhR)

In addition to PXR and CAR, a ligand-activated transcription factor AhR has been frequently referred to as another important xenosensor dictating the inductive expression of many DMEs and transporters. Although AhR was classified into the basic helixā€“loopā€“helix protein of the PER-ARNT-SIM (PAS) family, it shares several important characteristics comparable to CAR and PXR as xenobiotic receptors [18], [141]. In response to chemical activation, AhR enhances the expression of drug-metabolizing

Concluding remarks

Over the past ten years, remarkable advances have been achieved in our understanding of CAR and PXR-governed inductive expression of drug metabolism and disposition genes. It is clear now that, as the major xenobiotic receptors, CAR and PXR control a largely overlapping array of genes coding hepatic DMEs and transporters, thereby affecting the pharmacokinetics and toxicity of many drugs and environmental chemicals. The impact of these xenobiotic receptors on the induction-related DDIs has been

References (188)

  • T. Sueyoshi et al.

    The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene

    J. Biol. Chem.

    (1999)
  • H. Wang et al.

    A novel distal enhancer module regulated by pregnane X receptor/constitutive androstane receptor is essential for the maximal induction of CYP2B6 gene expression

    J. Biol. Chem.

    (2003)
  • R.E. Watkins et al.

    Coactivator binding promotes the specific interaction between ligand and the pregnane X receptor

    J. Mol. Biol.

    (2003)
  • S. Bhalla et al.

    Ligand-activated pregnane X receptor interferes with HNF-4 signaling by targeting a common coactivator PGC-1alpha. Functional implications in hepatic cholesterol and glucose metabolism

    J. Biol. Chem.

    (2004)
  • T. Satoh et al.

    Structure, function and regulation of carboxylesterases

    Chem. Biol. Interact.

    (2006)
  • D. Shi et al.

    Dexamethasone suppresses the expression of multiple rat carboxylesterases through transcriptional repression: evidence for an involvement of the glucocorticoid receptor

    Toxicology

    (2008)
  • T. Iyanagi

    Molecular mechanism of phase I and phase II drug-metabolizing enzymes: implications for detoxification

    Int. Rev. Cytol.

    (2007)
  • J. Sugatani et al.

    The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR

    Hepatology

    (2001)
  • M.F. Yueh et al.

    Involvement of the xenobiotic response element (XRE) in Ah receptor-mediated induction of human UDP-glucuronosyltransferase 1A1

    J. Biol. Chem.

    (2003)
  • C.A. Vyhlidal et al.

    Development and refinement of pregnane X receptor (PXR) DNA binding site model using information theory: insights into PXR-mediated gene regulation

    J. Biol. Chem.

    (2004)
  • C.B. Pickett et al.

    Differential induction of rat hepatic cytochrome P-448 and glutathione S-transferase B messenger RNAs by 3-methylcholanthrene

    Biochem. Biophys. Res. Commun.

    (1982)
  • A. Geick et al.

    Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin

    J. Biol. Chem.

    (2001)
  • H.R. Kast et al.

    Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor

    J. Biol. Chem.

    (2002)
  • S.C. Dogra et al.

    Transcriptional activation of cytochrome P450 genes by different classes of chemical inducers

    Clin. Exp. Pharmacol. Physiol.

    (1998)
  • P. Pavek et al.

    Xenobiotic-induced transcriptional regulation of xenobiotic metabolizing enzymes of the cytochrome P450 superfamily in human extrahepatic tissues

    Curr. Drug Metab.

    (2008)
  • C. Xu et al.

    Induction of phase I, II and III drug metabolism/transport by xenobiotics

    Arch. Pharm. Res.

    (2005)
  • R.M. Evans

    The steroid and thyroid hormone receptor superfamily

    Science

    (1988)
  • W. Bourguet et al.

    Crystal structure of the ligand-binding domain of the human nuclear receptor RXR-alpha

    Nature

    (1995)
  • R.E. Watkins et al.

    The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity

    Science

    (2001)
  • S.M. Hollenberg et al.

    Primary structure and expression of a functional human glucocorticoid receptor cDNA

    Nature

    (1985)
  • R. Miesfeld et al.

    Characterization of a steroid hormone receptor gene and mRNA in wild-type and mutant cells

    Nature

    (1984)
  • V. Giguere et al.

    Identification of a new class of steroid hormone receptors

    Nature

    (1988)
  • E. Enmark et al.

    Orphan nuclear receptorsā€“the first eight years

    Mol. Endocrinol.

    (1996)
  • S.A. Kliewer et al.

    Orphan nuclear receptors: shifting endocrinology into reverse

    Science

    (1999)
  • C. Handschin et al.

    Induction of drug metabolism: the role of nuclear receptors

    Pharmacol. Rev.

    (2003)
  • K.M. Burbach et al.

    Cloning of the Ah-receptor cDNA reveals a distinctive ligand-activated transcription factor

    Proc. Natl Acad. Sci. USA

    (1992)
  • B.L. Urquhart et al.

    Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interindividual variability in response to drugs

    J. Clin. Pharmacol.

    (2007)
  • C.D. Klaassen et al.

    Regulation of hepatic transporters by xenobiotic receptors

    Curr. Drug Metab.

    (2005)
  • P. Ramadoss et al.

    Role of the aryl hydrocarbon receptor in drug metabolism

    Expert Opin. Drug Metab. Toxicol.

    (2005)
  • J.M. Lehmann et al.

    The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions

    J. Clin. Investig.

    (1998)
  • B. Blumberg et al.

    Orphan nuclear receptors ā€“ new ligands and new possibilities

    Genes Dev.

    (1998)
  • G.r. Bertilsson et al.

    Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction

    Proc. Natl Acad. Sci. USA

    (1998)
  • N.R.N. Committee

    A unified nomenclature system for the nuclear receptor superfamily

    Cell

    (1999)
  • C. Zhou et al.

    The steroid and xenobiotic receptor (SXR), beyond xenobiotic metabolism

    Nucl. Recept. Signal.

    (2009)
  • S. Ekins et al.

    Challenges predicting ligand-receptor interactions of promiscuous proteins: the nuclear receptor PXR

    PLoS Comput. Biol.

    (2009)
  • R.E. Watkins et al.

    The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity

    Science

    (2001)
  • R.E. Watkins et al.

    A crystal structure of human PXR in complex with the St. John's Wort compound hyperforinae

    Biochemistry

    (2003)
  • J.E. Chrencik et al.

    Structural disorder in the complex of human pregnane X receptor and the macrolide antibiotic rifampicin

    Mol. Endocrinol.

    (2005)
  • C.-H. Ngan et al.

    The structural basis of pregnane X receptor binding promiscuity

    Biochemistry

    (2009)
  • J. Maglich et al.

    Comparison of complete nuclear receptor sets from the human, Caenorhabditis elegans and Drosophila genomes

    Genome Biol.

    (2001)
  • Cited by (302)

    • Inhibitory effects of traditional Chinese medicine colquhounia root tablet on the pharmacokinetics of tacrolimus in rats

      2022, Journal of Ethnopharmacology
      Citation Excerpt :

      On the contrary, our results indicated that the associated NRs (CAR and PXR) showed an increase in the protein and mRNA expressions. Given that NRs had an obvious activating effect on the expressions of various DMEs and DTs, including rCYP3A1, rCYP3A2, P-gp and MRP2, etc., as confirmed in previous studies (Chen et al., 2012; Tolson and Wang, 2010), the elevated NRs after multiple doses of CRT, presumably, might partially offset the promoting effect of CRT on the bioavailability of TAC. With protracted pretreatment, the accumulated plasma exposure of key components in CRT may elevate the mRNA and protein expressions of rCYP3A1/2, P-gp and MRP2 by activating NRs transcription, and finally, enhance the enzyme activities.

    • Nuclear Receptors

      2022, Comprehensive Pharmacology
    View all citing articles on Scopus
    ā˜†

    This review is part of the Advanced Drug Delivery Reviews theme issue on ā€œDevelopment of Novel Therapeutic Strategy by Regulating the Nuclear Hormone Receptorsā€.

    View full text