Skip to main content
Log in

Role of Orphan Nuclear Receptors in the Regulation of Drug-Metabolising Enzymes

Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

During the past several years, important advances have been made in our understanding of the mechanisms that regulate the expression of genes that determine drug clearance, including phase I and phase II drug-metabolising enzymes and drug transporters. Orphan nuclear receptors have been recognised as key mediators of drug-induced changes in both metabolism and efflux mechanisms.

In this review, we summarise recent findings regarding the function of nuclear receptors in regulating drug-metabolising and transport systems, and the relevance of these receptors to clinical drug-drug interactions and the development of new drugs. Emphasis is given to two newly recognised ‘orphan’ receptors (the pregnane X receptor [PXR] and the constitutive androstane receptor [CAR]) and their regulation of cytochrome P450 enzymes, such as CYP3A4, CYP2Cs and CYP2B6; and transporters, such as P-glycoprotein (MDR1), multidrug resistance-associated proteins (MRPs) and organic anion transporter peptide 2 (OATP2). Although ‘cross-talk’ occurs between these two receptors and their target sequences, significant species differences exist between ligand-binding and activation profiles for both receptors, and PXR appears to be the predominant or ‘master’ regulator of hepatic drug disposition in humans. Several important physiological processes, such as cholesterol synthesis and bile acid metabolism, are also tightly controlled by certain ligand-activated orphan nuclear receptors (farnesoid X receptor [FXR] and liver X receptor [LXR]).

In general, their ability to bind a broad range of ligands and regulate an extensive array of genes that are involved in drug clearance and disposition makes these orphan receptors attractive targets for drug development. Drugs have the capacity to alter nuclear receptor expression (modulators) and/or serve as ligands for the receptors (agonists or antagonists), and thus can have synergistic or antagonistic effects on the expression of drug-metabolising enzymes and transporters. Coadministration of drugs that are nuclear receptor agonists or antagonists can lead to severe toxicity, a loss of therapeutic efficacy or an imbalance in physiological substrates, providing a novel molecular mechanism for drug-drug interactions.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Table I

Similar content being viewed by others

References

  1. Kastner PM, Mark M, Chambon P. Nonsteroid nuclear receptors: what are genetic studies telling us about their role in real life? Cell 1995; 83(6): 859–69

    Article  PubMed  CAS  Google Scholar 

  2. Mangelsdorf DJ, Thummel C, Beato M, et al. The nuclear receptor superfamily: the second decade. Cell 1995; 83: 835–9

    Article  PubMed  CAS  Google Scholar 

  3. Moore LB, Parks DJ, Jones SA, et al. Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. J Biol Chem 2000; 275: 15122–7

    Article  PubMed  CAS  Google Scholar 

  4. Honkakoski P, Negishi M. Regulation of cytochrome P450 (CYP) genes by nuclear receptors. Biochem J 2000; 347: 321–37

    Article  PubMed  CAS  Google Scholar 

  5. Evans RM. The steroid and thyroid hormone receptor superfamily. Science 1988; 240: 889–95

    Article  PubMed  CAS  Google Scholar 

  6. Colston KW, Berger U, Coombes RC. Possible role for vitamin D in controlling breast cancer cell proliferation. Lancet 1989; I: 188–91

    Article  Google Scholar 

  7. Rupprecht R, Frolich L, Mergenthaler T, et al. Glucocorticoid receptors on mononuclear leukocytes in Alzheimers’s disease. Psychiatry Res 1990; 34: 237–41

    Article  PubMed  CAS  Google Scholar 

  8. Rupprecht R, Wodarz N, Kornhuber J, et al. In vivo and in vitro effects of glucocorticoids on lymphocyte proliferation in man: relationship to glucocorticoid receptors. Neuropsychobiology 1990; 24: 61–6

    Article  PubMed  CAS  Google Scholar 

  9. Giguere V, Yang N, Segui P, et al. Identification of a new class of steroid hormone receptors. Nature 1988; 331: 91–4

    Article  PubMed  CAS  Google Scholar 

  10. Blumberg B, Sabbagh Jr W, Juguilon H, et al. SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Genes Dev 1998; 12: 3195–205

    Article  PubMed  CAS  Google Scholar 

  11. Giguere V. Orphan nuclear receptors: from gene to function. Endocr Rev 1999; 20: 689–725

    Article  PubMed  CAS  Google Scholar 

  12. Blumberg B, Evans RM. Orphan nuclear receptors -new ligands and new possibilities. Genes Dev 1998; 12: 3149–55

    Article  PubMed  CAS  Google Scholar 

  13. Heyman RA, Mangelsdorf DJ, Dyck JA, et al. 9-cis-retinoic acid is a high affinity ligand for the retinoid X receptor. Cell 1992; 68: 397–406

    Article  PubMed  CAS  Google Scholar 

  14. Cairns W, Cairns C, Pongratz I, et al. Assembly of a glucocorticoid receptor complex prior to DNA binding enhances its specific interaction with a glucocorticoid response element. J Biol Chem 1991; 266: 11221–6

    PubMed  CAS  Google Scholar 

  15. Lee MS, Kliewer SA, Provencal J, et al. Structure of the retinoid X receptor alpha DNA binding domain: a helix required for homodimeric DNA binding. Science 1993; 260: 1117–21

    Article  PubMed  CAS  Google Scholar 

  16. Lanz RB, McKenna NJ, Onate SA, et al. A steroid receptor coactivator, SRA, functions as an RNA and is present in an SRC-1 complex. Cell 1999; 97: 17–27

    Article  PubMed  CAS  Google Scholar 

  17. Freedman LP. Increasing the complexity of coactivation in nuclear receptor signaling. Cell 1999; 97: 5–8

    Article  PubMed  CAS  Google Scholar 

  18. Parkinson A. Biotransformation of xenobiotics. In: Klaassen CD, editor. Casarett and Doulls’s toxicology: the basic science of poisons. Vol. 5. New York: McGraw Hill, 2001: 113–86

    Google Scholar 

  19. Vaz AD. Multiple oxidants in cytochrome P450 catalyzed reactions: implications for drug metabolism. Curr Drug Metab 2001; 2: 1–16

    Article  PubMed  CAS  Google Scholar 

  20. Ghanayem BI, Wang H, Sumner S. Using cytochrome P-450 gene knock-out mice to study chemical metabolism, toxicity, and carcinogenicity. Toxicol Pathol 2000; 28: 839–50

    Article  PubMed  CAS  Google Scholar 

  21. Kumar GN, Surapaneni S. Role of drug metabolism in drug discovery and development. Med Res Rev 2001; 21: 397–411

    Article  PubMed  CAS  Google Scholar 

  22. Waxman DJ. P450 gene induction by structurally diverse xenochemicals: central role of nuclear receptors CAR, PXR, and PPAR. Arch Biochem Biophys 1999; 369: 11–23

    Article  PubMed  CAS  Google Scholar 

  23. Rendic S, Di Carlo FJ. Human cytochrome P450 enzymes: a status report summarizing their reactions, substrates, inducers, and inhibitors. Drug Metab Rev 1997; 29: 413–580

    Article  PubMed  CAS  Google Scholar 

  24. Nelson DR, Kamataki T, Waxman DJ, et al. The P450 superfamily: update on new sequences, gene mapping, accession numbers, early trivial names of enzymes, and nomenclature. DNA Cell Biol 1993; 12: 1–51

    Article  PubMed  CAS  Google Scholar 

  25. Goldstein JA. Clinical relevance of genetic polymorphisms in the human CYP2C subfamily. Br J Clin Pharmacol 2001; 52: 349–55

    Article  PubMed  CAS  Google Scholar 

  26. Xie W, Evans RM. Orphan nuclear receptors: the exotics of xenobiotics. J Biol Chem 2001; 276: 37739–42

    PubMed  CAS  Google Scholar 

  27. Synold TW, Dussault I, Forman BM. The orphan nuclear receptor SXR coordinately regulates drug metabolism and efflux. Nat Med 2001; 7: 584–90

    Article  PubMed  CAS  Google Scholar 

  28. Le Cluyse EL. Pregnane X receptor: molecular basis for species differences in CYP3A induction by xenobiotics. Chem Biol Interact 2001; 134: 283–9

    Article  Google Scholar 

  29. Kliewer SA, Moore JT, Wade L, et al. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 1998; 92: 73–82

    Article  PubMed  CAS  Google Scholar 

  30. Goodwin B, Hodgson E, Liddle C. The orphan human pregnane X receptor mediates the transcriptional activation of CYP3A4 by rifampicin through a distal enhancer module. Mol Pharmacol 1999; 56: 1329–39

    PubMed  CAS  Google Scholar 

  31. Xie W, Barwick JL, Simon CM, et al. Reciprocal activation of xenobiotic response genes by nuclear receptors SXR/PXR and CAR. Genes Dev 2000; 14: 3014–23

    Article  PubMed  CAS  Google Scholar 

  32. Bertilsson G, Heidrich J, Svensson K, et al. Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc Natl Acad Sci USA 1998; 95: 12208–13

    Article  PubMed  CAS  Google Scholar 

  33. Zhang H, Le Cluyse E, Liu L, et al. Rat pregnane X receptor: molecular cloning, tissue distribution, and xenobiotic regulation. Arch Biochem Biophys 1999; 368: 14–22

    Article  PubMed  CAS  Google Scholar 

  34. Jones SA, Moore LB, Shenk JL, et al. The pregnane X receptor: a promiscuous xenobiotic receptor that has diverged during evolution. Mol Endocrinol 2000; 14: 27–39

    Article  PubMed  CAS  Google Scholar 

  35. Willson TM, Jones SA, Moore JT, et al. Chemical genomics: functional analysis of orphan nuclear receptors in the regulation of bile acid metabolism. Med Res Rev 2001; 21: 513–22

    Article  PubMed  CAS  Google Scholar 

  36. Anon. A unified nomenclature system for the nuclear receptor superfamily [letter]. Cell 1999; 97: 161–3

    Article  Google Scholar 

  37. Moore JT, Kliewer SA. Use of the nuclear receptor PXR to predict drug interactions. Toxicology 2000; 153: 1–10

    Article  PubMed  CAS  Google Scholar 

  38. Lehmann JM, McKee DD, Watson MA, et al. The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Invest 1998; 102: 1016–23

    Article  PubMed  CAS  Google Scholar 

  39. Schuetz JD, Beach DL, Guzelian PS. Selective expression of cytochrome P450 CYP3A mRNAs in embryonic and adult human liver. Pharmacogenetics 1994; 4: 11–20

    Article  PubMed  CAS  Google Scholar 

  40. Watkins RE, Wisely GB, Moore LB, et al. The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity. Science 2001; 292: 2329–33

    Article  PubMed  CAS  Google Scholar 

  41. Xie W, Barwick JL, Downes M, et al. Humanized xenobiotic response in mice expressing nuclear receptor SXR. Nature 2000; 406: 435–9

    Article  PubMed  CAS  Google Scholar 

  42. Staudinger JL, Goodwin B, Jones SA, et al. The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc Natl Acad Sci USA 2001; 98: 3369–74

    Article  PubMed  CAS  Google Scholar 

  43. Goodwin B, Moore LB, Stoltz CM, et al. Regulation of the human CYP2B6 gene by the nuclear pregnane X receptor. Mol Pharmacol 2001; 60: 427–31

    PubMed  CAS  Google Scholar 

  44. Traber PG, Wang W, McDonnell M, et al. P450IIB gene expression in rat small intestine: cloning of intestinal P450IIB1 mRNA using the polymerase chain reaction and transcriptional regulation of induction. Mol Pharmacol 1990; 37: 810–9

    PubMed  CAS  Google Scholar 

  45. Honkakoski P, Moore R, Washburn KA, et al. Activation by diverse xenochemicals of the 51-base pair phenobarbital-responsive enhancer module in the CYP2B10 gene. Mol Pharmacol 1998; 53: 597–601

    PubMed  CAS  Google Scholar 

  46. Honkakoski P, Zelko I, Sueyoshi T, et al. The nuclear orphan receptor CAR-retinoid X receptor heterodimer activates the phenobarbital-responsive enhancer module of the CYP2B gene. Mol Cell Biol 1998; 18: 5652–8

    PubMed  CAS  Google Scholar 

  47. Luo G, Cunningham M, Burn T, et al. CYP3A4 induction by xenobiotics: correlation between PXR activation and CYP3A4 expression. Drug Metab Dispos 2002; 30(7): 795–804

    Article  PubMed  CAS  Google Scholar 

  48. Faucette S, Wang HB, Hamilton g, et al. Regulation of CYP2B6 in primary human hepatocytes by prototypical inducers. Drug Metab Dispos. 2004; In Press.

  49. Wang H, Faucette S, Sueyoshi T, 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; 278(16): 14146–52

    Article  PubMed  CAS  Google Scholar 

  50. Wang H, Faucette SR, Gilbert D, et al. Glucocorticoid receptor enhancement of pregnane X receptor-mediated CYP2B6 regulation in primary human hepatocytes. Drug Metab Dispos 2003; 31: 620–30

    Article  PubMed  CAS  Google Scholar 

  51. Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by the multidrug transporter. Annu Rev Biochem 1993; 62: 385–427

    Article  PubMed  CAS  Google Scholar 

  52. Wacher VJ, Wu CY, Benet LZ. Overlapping substrate specificities and tissue distribution of cytochrome P450 3A and P-glycoprotein: implications for drug delivery and activity in cancer chemotherapy. Mol Carcinog 1995; 13: 129–34

    Article  PubMed  CAS  Google Scholar 

  53. Geick A, Eichelbaum M, Burk O. Nuclear receptor response elements mediate induction of intestinal MDR1 by rifampin. J Biol Chem 2001; 276: 14581–7

    Article  PubMed  CAS  Google Scholar 

  54. Kast HR, Goodwin B, Tarr PT, 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; 277: 2908–15

    Article  PubMed  CAS  Google Scholar 

  55. Dussault I, Lin M, Hollister K, et al. Peptide mimetic HIV protease inhibitors are ligands for the orphan receptor SXR. J Biol Chem 2001; 276: 33309–12

    Article  PubMed  CAS  Google Scholar 

  56. Fromm MF, Kauffmann HM, Fritz P, et al. The effect of rifampin treatment on intestinal expression of human MRP transporters. Am J Pathol 2000; 157: 1575–80

    Article  PubMed  CAS  Google Scholar 

  57. Staudinger J, Liu Y, Madan A, et al. Coordinate regulation of xenobiotic and bile acid homeostasis by pregnane X receptor. Drug Metab Dispos 2001; 29: 1467–72

    PubMed  CAS  Google Scholar 

  58. Luo G, Zeldin DC, Blaisdell JA, et al. Cloning and expression of murine CYP2Cs and their ability to metabolize arachidonic acid. Arch Biochem Biophys 1998; 357: 45–57

    Article  PubMed  CAS  Google Scholar 

  59. Goldstein JA, de Morais SM. Biochemistry and molecular biology of the human CYP2C subfamily. Pharmacogenetics 1994; 4: 285–99

    Article  PubMed  CAS  Google Scholar 

  60. Goldstein JA, Faletto MB, Romkes-Sparks M, et al. Evidence that CYP2C19 is the major (S)-mephenytoin 4′-hydroxylase in humans. Biochemistry 1994; 33: 1743–52

    Article  PubMed  CAS  Google Scholar 

  61. Pascussi JM, Gerbal-Chaloin S, Fabre JM, et al. Dexamethasone enhances constitutive androstane receptor expression in human hepatocytes: consequences on cytochrome P450 gene regulation. Mol Pharmacol 2000; 58: 1441–50

    PubMed  CAS  Google Scholar 

  62. Gerbal-Chaloin S, Daujat M, Pascussi JM, et al. Transcriptional regulation of CYP2C9 gene. Role of glucocorticoid receptor and constitutive androstane receptor. J Biol Chem 2002; 277: 209–17

    Article  PubMed  CAS  Google Scholar 

  63. Ferguson S, Le Cluyse EL, Negishi M, et al. Regulation of human CYP2C9 by the constitutive androstane receptor (CAR): discovery of a new distal binding site. Mol Pharmacol 2002 Sep; 62(3): 737–46

    Article  PubMed  CAS  Google Scholar 

  64. Heuman DM, Gallagher EJ, Barwick JL, et al. Immunochemical evidence for induction of a common form of hepatic cytochrome P-450 in rats treated with pregnenolone-16 alpha-carbonitrile or other steroidal or non-steroidal agents. Mol Pharmacol 1982; 21: 753–60

    PubMed  CAS  Google Scholar 

  65. Schuetz EG, Wrighton SA, Barwick JL, et al. Induction of cytochrome P-450 by glucocorticoids in rat liver: I. evidence that glucocorticoids and pregnenolone 16 alpha-carbonitrile regulate de novo synthesis of a common form of cytochrome P-450 in cultures of adult rat hepatocytes and in the liver in vivo. J Biol Chem 1984; 259: 1999–2006

    PubMed  CAS  Google Scholar 

  66. Schuetz EG, Guzelian PS. Induction of cytochrome P-450 by glucocorticoids in rat liver. II. Evidence that glucocorticoids regulate induction of cytochrome P-450 by a nonclassical receptor mechanism. J Biol Chem 1984; 259: 2007–12

    PubMed  CAS  Google Scholar 

  67. Quattrochi LC, Mills AS, Barwick JL, et al. A novel cis-acting element in a liver cytochrome P450 3A gene confers synergistic induction by glucocorticoids plus antiglucocorticoids. J Biol Chem 1995; 270: 28917–23

    Article  PubMed  CAS  Google Scholar 

  68. Huss JM, Kasper CB. Two-stage glucocorticoid induction of CYP3A23 through both the glucocorticoid and pregnane X receptors. Mol Pharmacol 2000; 58: 48–57

    PubMed  CAS  Google Scholar 

  69. Schuetz EG, Schmid W, Schutz G, et al. The glucocorticoid receptor is essential for induction of cytochrome P-4502B by steroids but not for drug or steroid induction of CYP3A or P-450 reductase in mouse liver. Drug Metab Dispos 2000; 28: 268–78

    PubMed  CAS  Google Scholar 

  70. Pascussi JM, Drocourt L, Gerbal-Chaloin S, et al. Dual effect of dexamethasone on CYP3A4 gene expression in human hepatocytes: sequential role of glucocorticoid receptor and pregnane X receptor. Eur J Biochem 2001; 268: 6346–58

    Article  PubMed  CAS  Google Scholar 

  71. El-Sankary W, Bombail V, Gibson GG, et al. Glucocorticoid-mediated induction of CYP3A4 is decreased by disruption of a protein-DNA interaction distinct from the pregnane X receptor response element. Drug Metab Dispos 2002; 30: 1029–34

    Article  PubMed  CAS  Google Scholar 

  72. Clarke SE, Jones BC. Human cytochromes P450 and their role in metabolism-based drug-drug interactions. In: Rodrigues AD, editor. Drug-drug interactions. Vol. 116. New York: Marcel Dekker Inc, 2002: 55–88

    Google Scholar 

  73. Ozdemir V, Kalowa W, Tang BK, et al. Evaluation of the genetic component of variability in CYP3A4 activity: a repeated drug administration method. Pharmacogenetics 2000; 10: 373–88

    Article  PubMed  CAS  Google Scholar 

  74. Sata F, Sapone A, Elizondo G, et al. CYP3A4 allelic variants with amino acid substitutions in exons 7 and 12: evidence for an allelic variant with altered catalytic activity. Clin Pharmacol Ther 2000; 67: 48–56

    Article  PubMed  CAS  Google Scholar 

  75. Westlind A, Lofberg L, Tindberg N, et al. Interindividual differences in hepatic expression of CYP3A4: relationship to genetic polymorphism in the 5′-upstream regulatory region. Biochem Biophys Res Commun 1999; 259: 201–5

    Article  PubMed  CAS  Google Scholar 

  76. Zhang J, Kuehl P, Green ED, et al. The human pregnane X receptor: genomic structure and identification and functional characterization of natural allelic variants. Pharmacogenetics 2001; 11: 555–72

    Article  PubMed  CAS  Google Scholar 

  77. Hustert E, Zibat A, Presecan-Siedel E, et al. Natural protein variants of pregnane X receptor with altered transactivation activity toward CYP3A4. Drug Metab Dispos 2001; 29: 1454–9

    PubMed  CAS  Google Scholar 

  78. Eichelbaum M, Burk O. CYP3A genetics in drug metabolism. Nat Med 2001; 7: 285–7

    Article  PubMed  CAS  Google Scholar 

  79. Kyriazopoulou V, Vagenakis AG. Abnormal overnight dexamethasone suppression test in subjects receiving rifampicin therapy. J Clin Endocrinol Metab 1992; 75: 315–7

    Article  PubMed  CAS  Google Scholar 

  80. Terzolo M, Borretta G, Ali A, et al. Misdiagnosis of Cushings’s syndrome in a patient receiving rifampicin therapy for tuberculosis. Horm Metab Res 1995; 27: 148–50

    Article  PubMed  CAS  Google Scholar 

  81. Zawawi TH, al-Hadramy MS, Abdelwahab SM. The effects of therapy with rifampicin and isoniazid on basic investigations for Cushings’s syndrome. Ir J Med Sci 1996; 165: 300–2

    Article  PubMed  CAS  Google Scholar 

  82. Baes M, Gulick T, Choi HS, et al. A new orphan member of the nuclear hormone receptor superfamily that interacts with a subset of retinoic acid response elements. Mol Cell Biol 1994; 14: 1544–51

    PubMed  CAS  Google Scholar 

  83. Choi HS, Chung M, Tzameli I, et al. Differential transactivation by two isoforms of the orphan nuclear hormone receptor CAR. J Biol Chem 1997; 272: 23565–71

    Article  PubMed  CAS  Google Scholar 

  84. Forman BM, Tzameli I, Choi HS, et al. Androstane metabolites bind to and deactivate the nuclear receptor CAR-beta. Nature 1998; 395: 612–5

    Article  PubMed  CAS  Google Scholar 

  85. Mizukami Y, Sogawa K, Suwa Y, et al. Gene structure of a phenobarbital-inducible cytochrome P-450 in rat liver. Proc Natl Acad Sci U S A 1983; 80: 3958–62

    Article  PubMed  CAS  Google Scholar 

  86. Trottier E, Belzil A, Stoltz C, et al. Localization of a phenobarbital-responsive element (PBRE) in the 5′-flanking region of the rat CYP2B2 gene. Gene 1995; 158: 263–8

    Article  PubMed  CAS  Google Scholar 

  87. Honkakoski P, Negishi M. Characterization of a phenobarbital-responsive enhancer module in mouse P450 Cyp2b10 gene. J Biol Chem 1997; 272: 14943–9

    Article  PubMed  CAS  Google Scholar 

  88. Sueyoshi T, Kawamoto T, Zelko I, et al. The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene. J Biol Chem 1999; 274: 6043–6

    Article  PubMed  CAS  Google Scholar 

  89. Ramsden R, Beck NB, Sommer KM, et al. Phenobarbital responsiveness conferred by the 5′-flanking region of the rat CYP2B2 gene in transgenic mice. Gene 1999; 228: 169–79

    Article  PubMed  CAS  Google Scholar 

  90. Poland A, Mak I, Glover E, et al. 1,4-Bis[-(3,5-dichloropyridyloxy)]benzene, a potent phenobarbital-like inducer of microsomal monooxygenase activity. Mol Pharmacol 1980; 18: 571–80

    PubMed  CAS  Google Scholar 

  91. Kawamoto T, Sueyoshi T, Zelko I, et al. Phenobarbital-responsive nuclear translocation of the receptor CAR in induction of the CYP2B gene. Mol Cell Biol 1999; 19: 6318–22

    PubMed  CAS  Google Scholar 

  92. Walker D, Htun H, Hager GL. Using inducible vectors to study intracellular trafficking of GFP-tagged steroid/nuclear receptors in living cells. Methods 1999; 19: 386–93

    Article  PubMed  CAS  Google Scholar 

  93. Picard D, Yamamoto KR. Two signals mediate hormone-dependent nuclear localization of the glucocorticoid receptor. EMBO J 1987; 6: 3333–40

    PubMed  CAS  Google Scholar 

  94. Guiochon-Mantel A, Lescop P, Christin-Maitre S, et al. Nucleocytoplasmic shuttling of the progesterone receptor. EMBO J 1991; 10: 3851–9

    PubMed  CAS  Google Scholar 

  95. Tzameli I, Pissios P, Schuetz EG, et al. The xenobiotic compound 1,4-bis[2-(3,5-dichloropyridyloxy)]benzene is an agonist ligand for the nuclear receptor CAR. Mol Cell Biol 2000; 20: 2951–8

    Article  PubMed  CAS  Google Scholar 

  96. Muangmoonchai R, Smirlis D, Wong SC, et al. Xenobiotic induction of cytochrome P450 2B1 (CYP2B1) is mediated by the orphan nuclear receptor constitutive androstane receptor (CAR) and requires steroid co-activator 1 (SRC-1) and the transcription factor Sp1. Biochem J 2001; 355: 71–8

    Article  PubMed  CAS  Google Scholar 

  97. Smirlis D, Muangmoonchai R, Edwards M, et al. Orphan receptor promiscuity in the induction of cytochromes p450 by xenobiotics. J Biol Chem 2001; 276: 12822–6

    Article  PubMed  CAS  Google Scholar 

  98. Honkakoski P, Negishi M. Protein serine/threonine phosphatase inhibitors suppress phenobarbital-induced Cyp2b10 gene transcription in mouse primary hepatocytes. Biochem J 1998; 330: 889–95

    PubMed  CAS  Google Scholar 

  99. Negishi M. Nuclear receptor CAR as a phenobarbital induction signal of CYP2B gene [abstract]. FASEB J 2000; 14: 1306

    Google Scholar 

  100. Kemper B. Regulation of cytochrome P450 gene transcription by phenobarbital. Prog Nucleic Acid Res Mol Biol 1998; 61: 23–64

    PubMed  CAS  Google Scholar 

  101. Wei P, Zhang J, Egan-Hafley M, et al. The nuclear receptor CAR mediates specific xenobiotic induction of drug metabolism. Nature 2000; 407: 920–3

    Article  PubMed  CAS  Google Scholar 

  102. Ueda A, Hamadeh HK, Webb HK, et al. Diverse roles of the nuclear orphan receptor CAR in regulating hepatic genes in response to phenobarbital. Mol Pharmacol 2002; 61: 1–6

    Article  PubMed  CAS  Google Scholar 

  103. Yoshinari K, Sueyoshi T, Moore R, et al. Nuclear receptor CAR as a regulatory factor for the sexually dimorphic induction of CYB2B1 gene by phenobarbital in rat livers. Mol Pharmacol 2001; 59: 278–84

    PubMed  CAS  Google Scholar 

  104. Iyanagi T, Emi Y, Ikushiro S. Biochemical and molecular aspects of genetic disorders of bilirubin metabolism. Biochim Biophys Acta 1998; 1407: 173–84

    Article  PubMed  CAS  Google Scholar 

  105. Fisher MB, Paine MF, Strelevitz TJ, et al. The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev 2001; 33: 273–97

    Article  PubMed  CAS  Google Scholar 

  106. Arias IM, Gartner LM, Cohen M, et al. Chronic nonhemolytic unconjugated hyperbilirubinemia with glucuronyl transferase deficiency: clinical, biochemical, pharmacologic and genetic evidence for heterogeneity. Am J Med 1969; 47: 395–409

    Article  PubMed  CAS  Google Scholar 

  107. Yaffe SJ, Levy G, Matsuzawa T, et al. Enhancement of glucuronide-conjugating capacity in a hyperbilirubinemic infant due to apparent enzyme induction by phenobarbital. N Engl J Med 1966; 275: 1461–6

    Article  PubMed  CAS  Google Scholar 

  108. Ritter JK, Kessler FK, Thompson MT, et al. Expression and inducibility of the human bilirubin UDP-glucuronosyltransferase UGT1A1 in liver and cultured primary hepatocytes: evidence for both genetic and environmental influences. Hepatology 1999; 30: 476–84

    Article  PubMed  CAS  Google Scholar 

  109. Sutherland L, Ebner T, Burchell B. The expression of UDP-glucuronosyltransferases of the UGT1 family in human liver and kidney and in response to drugs. Biochem Pharmacol 1993; 45: 295–301

    Article  PubMed  CAS  Google Scholar 

  110. Sugatani J, Kojima H, Ueda A, et al. The phenobarbital response enhancer module in the human bilirubin UDP-glucuronosyltransferase UGT1A1 gene and regulation by the nuclear receptor CAR. Hepatology 2001; 33: 1232–8

    Article  PubMed  CAS  Google Scholar 

  111. Issemann I, Green S. Activation of a member of the steroid hormone receptor superfamily by peroxisome proliferators. Nature 1990; 347: 645–50

    Article  PubMed  CAS  Google Scholar 

  112. Braissant O, Foufelle F, Scotto C, et al. Differential expression of peroxisome proliferator-activated receptors (PPARs): tissue distribution of PPAR-alpha, -beta, and -gamma in the adult rat. Endocrinology 1996; 137: 354–66

    Article  PubMed  CAS  Google Scholar 

  113. Amri EZ, Bonino F, Ailhaud G, et al. Cloning of a protein that mediates transcriptional effects of fatty acids in preadipocytes: homology to peroxisome proliferator-activated receptors. J Biol Chem 1995; 270: 2367–71

    Article  PubMed  CAS  Google Scholar 

  114. Lemberger T, Braissant O, Juge-Aubry C, et al. PPAR tissue distribution and interactions with other hormone-signaling pathways. Ann N Y Acad Sci 1996; 804: 231–51

    Article  PubMed  CAS  Google Scholar 

  115. Corton JC, Anderson SP, Stauber A. Central role of peroxisome proliferator-activated receptors in the actions of peroxisome proliferators. Annu Rev Pharmacol Toxicol 2000; 40: 491–518

    Article  PubMed  CAS  Google Scholar 

  116. Corton JC, Lapinskas PJ, Gonzalez FJ. Central role of PPARalpha in the mechanism of action of hepatocarcinogenic peroxisome proliferators. Mutat Res 2000; 448: 139–51

    Article  PubMed  CAS  Google Scholar 

  117. Tugwood JD, Aldridge TC, Lambe KG, et al. Peroxisome proliferator-activated receptors: structures and function. Ann N Y Acad Sci 1996; 804: 252–65

    Article  PubMed  CAS  Google Scholar 

  118. Osumi T, Osada S, Tsukamoto T. Analysis of peroxisome proliferator-responsive enhancer of the rat acyl-CoA oxidase gene. Ann N Y Acad Sci 1996; 804: 202–13

    Article  PubMed  CAS  Google Scholar 

  119. Johnson EF, Palmer CN, Griffin KJ, et al. Role of the peroxisome proliferator-activated receptor in cytochrome P450 4A gene regulation. FASEB J 1996; 10: 1241–8

    PubMed  CAS  Google Scholar 

  120. Johnson EF, Palmer CN, Hsu MH. The peroxisome proliferator-activated receptor: transcriptional activation of the CYP4A6 gene. Ann N Y Acad Sci 1996; 804: 373–86

    Article  PubMed  CAS  Google Scholar 

  121. Aldridge TC, Tugwood JD, Green S. Identification and characterization of DNA elements implicated in the regulation of CYP4A1 transcription. Biochem J 1995; 306: 473–9

    PubMed  CAS  Google Scholar 

  122. Palmer CN, Hsu MH, Griffin HJ, et al. Novel sequence determinants in peroxisome proliferator signaling. J Biol Chem 1995; 270: 16114–21

    Article  PubMed  CAS  Google Scholar 

  123. Zhu Y, Qi C, Korenberg JR, et al. Structural organization of mouse peroxisome proliferator-activated receptor gamma (mPPAR gamma) gene: alternative promoter use and different splicing yield two mPPAR gamma isoforms. Proc Natl Acad Sci U S A 1995; 92: 7921–5

    Article  PubMed  CAS  Google Scholar 

  124. MacDougald OA, Lane MD. Transcriptional regulation of gene expression during adipocyte differentiation. Annu Rev Biochem 1995; 64: 345–73

    Article  PubMed  CAS  Google Scholar 

  125. Moras D, Gronemeyer H. The nuclear receptor ligand-binding domain: structure and function. Curr Opin Cell Biol 1998; 10: 384–91

    Article  PubMed  CAS  Google Scholar 

  126. Willy PJ, Umesono K, Ong ES, et al. LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 1995; 9: 1033–45

    Article  PubMed  CAS  Google Scholar 

  127. Song C, Hiipakka RA, Kokontis JM, et al. Ubiquitous receptor: structures, immunocytochemical localization, and modulation of gene activation by receptors for retinoic acids and thyroid hormones. Ann N Y Acad Sci 1995; 761: 38–49

    Article  PubMed  CAS  Google Scholar 

  128. Kainu T, Enmark E, Gustafsson JA, et al. Localization of the Rev-ErbA orphan receptors in the brain. Brain Res 1996; 743: 315–9

    Article  PubMed  CAS  Google Scholar 

  129. Peet DJ, Janowski BA, Mangelsdorf DJ. The LXRs: a new class of oxysterol receptors. Curr Opin Genet Dev 1998; 8: 571–5

    Article  PubMed  CAS  Google Scholar 

  130. Lehmann JM, Kliewer SA, Moore LB, et al. Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. J Biol Chem 1997; 272: 3137–40

    Article  PubMed  CAS  Google Scholar 

  131. Peet DJ, Turley SD, Ma W, et al. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 1998; 93: 693–704

    Article  PubMed  CAS  Google Scholar 

  132. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature 1990; 343: 425–30

    Article  PubMed  CAS  Google Scholar 

  133. Accad M, Farese Jr RV. Cholesterol homeostasis: a role for oxysterols. Curr Biol 1998; 8: R601–4

    Article  PubMed  CAS  Google Scholar 

  134. Jelinek DF, Andersson S, Slaughter CA, et al. Cloning and regulation of cholesterol 7α-hydroxylase, the rate- limiting enzyme in bile acid biosynthesis. J Biol Chem 1990; 265: 8190–7

    PubMed  CAS  Google Scholar 

  135. Jelinek DF, Russell DW. Structure of the rat gene encoding cholesterol 7α-hydroxylase. Biochemistry 1990; 29: 7781–5

    Article  PubMed  CAS  Google Scholar 

  136. Li YC, Wang DP, Chiang JY. Regulation of cholesterol 7α-hydroxylase in the liver. Cloning, sequencing, and regulation of cholesterol 7α-hydroxylase mRNA. J Biol Chem 1990; 265: 12012–9

    PubMed  CAS  Google Scholar 

  137. Laffitte BA, Joseph SB, Walczak R, et al. Autoregulation of the human liver X receptor alpha promoter. Mol Cell Biol 2001; 21: 7558–68

    Article  PubMed  CAS  Google Scholar 

  138. Whitney KD, Watson MA, Goodwin B, et al. Liver X receptor (LXR) regulation of the LXRalpha gene in human macrophages. J Biol Chem 2001; 276: 43509–15

    Article  PubMed  CAS  Google Scholar 

  139. Laffitte BA, Repa JJ, Joseph SB, et al. LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc Natl Acad Sci U S A 2001; 98: 507–12

    Article  PubMed  CAS  Google Scholar 

  140. Miyata KS, McCaw SE, Patel HV, et al. The orphan nuclear hormone receptor LXRα interacts with the peroxisome proliferator-activated receptor and inhibits peroxisome proliferator signaling. J Biol Chem 1996; 271: 9189–92

    Article  PubMed  CAS  Google Scholar 

  141. Forman BM, Goode E, Chen J, et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 1995; 81: 687–93

    Article  PubMed  CAS  Google Scholar 

  142. Grober J, Zaghini I, Fujii H, et al. Identification of a bile acid-responsive element in the human ileal bile acid-binding protein gene: involvement of the farnesoid X receptor/9-cis-retinoic acid receptor heterodimer. J Biol Chem 1999; 274: 29749–54

    Article  PubMed  CAS  Google Scholar 

  143. Wang H, Chen J, Hollister K, et al. Endogenous bile acids are ligands for the nuclear receptor FXR/BAR. Mol Cell 1999; 3: 543–53

    Article  PubMed  CAS  Google Scholar 

  144. Makishima M, Okamoto AY, Repa JJ, et al. Identification of a nuclear receptor for bile acids. Science 1999; 284: 1362–5

    Article  PubMed  CAS  Google Scholar 

  145. Tu H, Okamoto AY, Shan B. FXR, a bile acid receptor and biological sensor. Trends Cardiovasc Med 2000; 10: 30–5

    Article  PubMed  CAS  Google Scholar 

  146. Parks DJ, Blanchard SG, Bledsoe RK, et al. Bile acids: natural ligands for an orphan nuclear receptor. Science 1999; 284: 1365–8

    Article  PubMed  CAS  Google Scholar 

  147. Goodwin B, Jones SA, Price RR, et al. A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 2000; 6: 517–26

    Article  PubMed  CAS  Google Scholar 

  148. Kanda T, Foucand L, Nakamura Y, et al. Regulation of expression of human intestinal bile acid-binding protein in Caco-2 cells. Biochem J 1998; 330: 261–5

    PubMed  CAS  Google Scholar 

  149. Hofmann AF. The continuing importance of bile acids in liver and intestinal disease. Arch Intern Med 1999; 159: 2647–58

    Article  PubMed  CAS  Google Scholar 

  150. Brown MS, Goldstein JL. A receptor-mediated pathway for cholesterol homeostasis. Science 1986; 232: 34–47

    Article  PubMed  CAS  Google Scholar 

  151. Mangelsdorf DJ, Ong ES, Dyck JA, et al. Nuclear receptor that identifies a novel retinoic acid response pathway. Nature 1990; 345: 224–9

    Article  PubMed  CAS  Google Scholar 

  152. Mangelsdorf DJ, Borgmeyer U, Heyman RA, et al. Characterization of three RXR genes that mediate the action of 9-cisretinoic acid. Genes Dev 1992; 6: 329–44

    Article  PubMed  CAS  Google Scholar 

  153. Krezel W, Dupe V, Mark M, et al. RXRγ null mice are apparently normal and compound RXRα +/-RXRβ -/-RXRγ -/-mutant mice are viable. Proc Natl Acad Sci U S A 1996; 93: 9010–4

    Article  PubMed  CAS  Google Scholar 

  154. Kurokawa R, DiRenzo J, Boehm M, et al. Regulation of retinoid signalling by receptor polarity and allosteric control of ligand binding. Nature 1994; 371: 528–31

    Article  PubMed  CAS  Google Scholar 

  155. Kliewer SA, Umesono K, Mangelsdorf DJ, et al. Retinoid X receptor interacts with nuclear receptors in retinoic acid, thyroid hormone and vitamin D3 signalling. Nature 1992; 355: 446–9

    Article  PubMed  CAS  Google Scholar 

  156. Forman BM, Evans RM. Nuclear hormone receptors activate direct, inverted, and everted repeats. Ann N Y Acad Sci 1995; 761: 29–37

    Article  PubMed  CAS  Google Scholar 

  157. Schulman IG, Evans RM. Retinoid receptors in development and disease. Leukemia 1997; 11 Suppl. 3: 376–7

    PubMed  Google Scholar 

  158. Ressel A, Schmitt O, Weiss C, et al. Therapeutic outcome and side-effects after radiotherapy, chemotherapy and/or hyperthermia treatment of head and neck tumour xenografts. Eur J Cancer 2002; 38: 594–601

    Article  PubMed  CAS  Google Scholar 

  159. Shiau YC, Tsai SC, Wang JJ, et al. Predicting chemotherapy response and comparing with P-glycoprotein expression using technetium-99m tetrofosmin scan in untreated malignant lymphomas. Cancer Lett 2001; 170: 139–46

    Article  PubMed  CAS  Google Scholar 

  160. van Heeswijk RP, Veldkamp A, Mulder JW, et al. Combination of protease inhibitors for the treatment of HIV-1-infected patients: a review of pharmacokinetics and clinical experience. Antivir Ther 2001; 6: 201–29

    PubMed  Google Scholar 

  161. Moyle GJ, Back D. Principles and practice of HIV-protease inhibitor pharmacoenhancement. HIV Med 2001; 2: 105–13

    Article  PubMed  CAS  Google Scholar 

  162. Pozniak A. Multidrug-resistant tuberculosis and HIV infection. Ann N Y Acad Sci 2001; 953: 192–8

    Article  PubMed  CAS  Google Scholar 

  163. Guo GL, Johnson DR, Klaassen CD. Postnatal expression and induction by pregnenolone-16alpha-carbonitrile of the organic anion-transporting polypeptide 2 in rat liver. Drug Metab Dispos 2002; 30: 283–8

    Article  PubMed  CAS  Google Scholar 

  164. Kauffmann HM, Pfannschmidt S, Zoller H, et al. Influence of redox-active compounds and PXR-activators on human MRP1 and MRP2 gene expression. Toxicology 2002; 171: 137–46

    Article  PubMed  CAS  Google Scholar 

  165. Kivisto KT, Kroemer HK, Eichelbaum M. The role of human cytochrome P450 enzymes in the metabolism of anticancer agents: implications for drug interactions. Br J Clin Pharmacol 1995; 40: 523–30

    Article  PubMed  CAS  Google Scholar 

  166. Michalets EL. Update: clinically significant cytochrome P-450 drug interactions. Pharmacotherapy 1998; 18: 84–112

    PubMed  CAS  Google Scholar 

  167. Backman JT, Olkkola KT, Neuvonen PJ. Rifampin drastically reduces plasma concentrations and effects of oral midazolam. Clin Pharmacol Ther 1996; 59: 7–13

    Article  PubMed  CAS  Google Scholar 

  168. Izzo AA, Ernst E. Interactions between herbal medicines and prescribed drugs: a systematic review. Drugs 2001; 61: 2163–75

    Article  PubMed  CAS  Google Scholar 

  169. Fugh-Berman A, Ernst E. Herb-drug interactions: review and assessment of report reliability. Br J Clin Pharmacol 2001; 52: 587–95

    Article  PubMed  CAS  Google Scholar 

  170. Ernst E. Second thoughts about safety of St Johns’s wort. Lancet 1999; 354: 2014–6

    Article  PubMed  CAS  Google Scholar 

  171. de Maat MM, Hoetelmans RM, Mathot RA, et al. Drug interaction between St Johns’s wort and nevirapine. AIDS 2001; 15: 420–1

    Article  PubMed  Google Scholar 

  172. Moschella C, Jaber BL. Interaction between cyclosporine and Hypericum perforatum (St Johns’s wort) after organ transplantation. Am J Kidney Dis 2001; 38: 1105–7

    Article  PubMed  CAS  Google Scholar 

  173. Moore L, Goodwin B, Jones SA, et al. St Johns’s wort induces hepatic drug metabolism through activation of the pregnane X receptor. Proc Natl Acad Sci U S A 2000; 97: 7500–2

    Article  PubMed  CAS  Google Scholar 

  174. Huang Z, Roy P, Waxman DJ. Role of human liver microsomal CYP3A4 and CYP2B6 in catalyzing N-dechloroethylation of cyclophosphamide and ifosfamide. Biochem Pharmacol 2000; 59: 961–72

    Article  PubMed  CAS  Google Scholar 

  175. Lindley C, Hamilton G, Mccune JS, et al. The effect of cyclophosphamide with and without dexamethasone on cytochrome P450 3A4 and 2B6 in human hepatocytes. Drug Metab Dispos 2002; 30: 814–22

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the contributions of Stephanie Faucette, Rick Graham, Cornelia Smith, Lynn Johnson and Tom Lutz from the Division of Drug Delivery and Disposition, School of Pharmacy, University of North Carolina at Chapel Hill. This work was partly supported by National Institutes of Health Grant RO1: DK061652.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Edward L. LeCluyse.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, H., LeCluyse, E.L. Role of Orphan Nuclear Receptors in the Regulation of Drug-Metabolising Enzymes. Clin Pharmacokinet 42, 1331–1357 (2003). https://doi.org/10.2165/00003088-200342150-00003

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-200342150-00003

Keywords

Navigation