885
Views
34
CrossRef citations to date
0
Altmetric
Review Article

Nuclear receptors in the cross-talk of drug metabolism and inflammation

, , &
Pages 122-144 | Received 25 Sep 2012, Accepted 29 Nov 2012, Published online: 21 Jan 2013

References

  • Adcock, I. M., Caramori, G. (2001). Cross-talk between pro-inflammatory transcription factors and glucocorticoids. Immunol Cell Biol 79:376–384.
  • Ahn, S. H., Shah, Y. M., Inoue, J., Morimura, K., Kim, I., Yim, S. et al. (2008). Hepatocyte nuclear factor 4alpha in the intestinal epithelial cells protects against inflammatory bowel disease. Inflamm Bowel Dis 14:908–920.
  • Aitken, A. E., Richardson, T. A., Morgan, E. T. (2006). Regulation of drug-metabolizing enzymes and transporters in inflammation. Annu Rev Pharmacol Toxicol 46:123–149.
  • Anwar-Mohamed, A., Degenhardt, O. S., El Gendy, M. A., Seubert, J. M., Kleeberger, S. R., El-Kadi, A. O. (2011). The effect of Nrf2 knockout on the constitutive expression of drug metabolizing enzymes and transporters in C57Bl/6 mice livers. Toxicol In Vitro 25:785–795.
  • Anzenbacher, P. Anzenbacherová, E. (2001). Cytochromes P450 and metabolism of xenobiotics. Cell Mol Life Sci 58:737–747.
  • Ashino, T., Oguro, T., Shioda, S., Horai, R., Asano, M., Sekikawa, K. et al. (2004). Involvement of interleukin-6 and tumor necrosis factor alpha in CYP3A11 and 2C29 down-regulation by Bacillus Calmette-Guerin and lipopolysaccharide in mouse liver. Drug Metab Dispos 32:707–714.
  • Assenat, E., Gerbal-Chaloin, S., Larrey, D., Saric, J., Fabre, J. M., Maurel, P. et al. (2004). Interleukin 1beta inhibits CAR-induced expression of hepatic genes involved in drug and bilirubin clearance. Hepatology 40:951–960.
  • Bachs, L., Parés, A., Elena, M., Piera, C., Rodés, J. (1992). Effects of long-term rifampicin administration in primary biliary cirrhosis. Gastroenterology 102:2077–2080.
  • Barnes, P. J. (1997). Nuclear factor-kappa B. Int J Biochem Cell Biol 29:867–870.
  • Barnes, P. J. (1998). Anti-inflammatory actions of glucocorticoids: molecular mechanisms. Clin Sci (Lond) 94:557–572.
  • Barnes, P. J., Karin, M. (1997). Nuclear factor-kappaB: a pivotal transcription factor in chronic inflammatory diseases. N Engl J Med 336:1066–1071.
  • Beigneux, A. P., Moser, A. H., Shigenaga, J. K., Grunfeld, C., Feingold, K. R. (2000). The acute phase response is associated with retinoid X receptor repression in rodent liver. J Biol Chem 275:16390–16399.
  • Beigneux, A. P., Moser, A. H., Shigenaga, J. K., Grunfeld, C., Feingold, K. R. (2002). Reduction in cytochrome P-450 enzyme expression is associated with repression of CAR (constitutive androstane receptor) and PXR (pregnane X receptor) in mouse liver during the acute phase response. Biochem Biophys Res Commun 293:145–149.
  • Beischlag, T. V., Luis Morales, J., Hollingshead, B. D., Perdew, G. H. (2008). The aryl hydrocarbon receptor complex and the control of gene expression. Crit Rev Eukaryot Gene Expr 18:207–250.
  • Bleau, A. M., Fradette, C., El-Kadi, A. O., Côté, M. C., du Souich, P. (2001). Cytochrome P450 down-regulation by serum from humans with a viral infection and from rabbits with an inflammatory reaction. Drug Metab Dispos 29:1007–1012.
  • Bleau, A. M., Levitchi, M. C., Maurice, H., du Souich, P. (2000). Cytochrome P450 inactivation by serum from humans with a viral infection and serum from rabbits with a turpentine-induced inflammation: the role of cytokines. Br J Pharmacol 130:1777–1784.
  • Bleau, A. M., Maurel, P., Pichette, V., Leblond, F., du Souich, P. (2003). Interleukin-1beta, interleukin-6, tumour necrosis factor-alpha and interferon-gamma released by a viral infection and an aseptic inflammation reduce CYP1A1, 1A2 and 3A6 expression in rabbit hepatocytes. Eur J Pharmacol 473:197–206.
  • Blokzijl, H., Vander Borght, S., Bok, L. I., Libbrecht, L., Geuken, M., van den Heuvel, F. A. et al. (2007). Decreased P-glycoprotein (P-gp/MDR1) expression in inflamed human intestinal epithelium is independent of PXR protein levels. Inflamm Bowel Dis 13:710–720.
  • Bolotin, E., Liao, H., Ta, T. C., Yang, C., Hwang-Verslues, W., Evans, J. R. et al. (2010). Integrated approach for the identification of human hepatocyte nuclear factor 4alpha target genes using protein binding microarrays. Hepatology 51:642–653.
  • Bonizzi, G., Bebien, M., Otero, D. C., Johnson-Vroom, K. E., Cao, Y., Vu, D. et al. (2004). Activation of IKKalpha target genes depends on recognition of specific kappaB binding sites by RelB:p52 dimers. EMBO J 23:4202–4210.
  • Bonizzi, G., Karin, M. (2004). The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol 25:280–288.
  • Cain, B. S., Meldrum, D. R., Harken, A. H. (1999). Protein kinase C in normal and pathologic myocardial states. J Surg Res 81:249–259.
  • Celius, T., Pansoy, A., Matthews, J., Okey, A. B., Henderson, M. C., Krueger, S. K. et al. (2010). Flavin-containing monooxygenase-3: induction by 3-methylcholanthrene and complex regulation by xenobiotic chemicals in hepatoma cells and mouse liver. Toxicol Appl Pharmacol 247:60–69.
  • Celius, T., Roblin, S., Harper, P. A., Matthews, J., Boutros, P. C., Pohjanvirta, R. et al. (2008). Aryl hydrocarbon receptor-dependent induction of flavin-containing monooxygenase mRNAs in mouse liver. Drug Metab Dispos 36:2499–2505.
  • Chang, L., Karin, M. (2001). Mammalian MAP kinase signalling cascades. Nature 410:37–40.
  • Chen, K. J., Lin, S. Z., Zhou, L., Xie, H. Y., Zhou, W. H., Taki-Eldin, A. et al. (2011). Selective recruitment of regulatory T cell through CCR6-CCL20 in hepatocellular carcinoma fosters tumor progression and predicts poor prognosis. Plos One 6:e24671.
  • Chen, S., Operaña, T., Bonzo, J., Nguyen, N., Tukey, R. H. (2005a). ERK kinase inhibition stabilizes the aryl hydrocarbon receptor: implications for transcriptional activation and protein degradation. J Biol Chem 280:4350–4359.
  • Chen, Y., Ferguson, S. S., Negishi, M., Goldstein, J. A. (2003). Identification of constitutive androstane receptor and glucocorticoid receptor binding sites in the CYP2C19 promoter. Mol Pharmacol 64:316–324.
  • Chen, Y. H., Wang, J. P., Wang, H., Sun, M. F., Wei, L. Z., Wei, W. et al. (2005b). Lipopolysaccharide treatment downregulates the expression of the pregnane X receptor, cyp3a11 and mdr1a genes in mouse placenta. Toxicology 211:242–252.
  • Chou, W. C., Prokova, V., Shiraishi, K., Valcourt, U., Moustakas, A., Hadzopoulou-Cladaras, M. et al. (2003). Mechanism of a transcriptional cross talk between transforming growth factor-beta-regulated Smad3 and Smad4 proteins and orphan nuclear receptor hepatocyte nuclear factor-4. Mol Biol Cell 14:1279–1294.
  • Copple, I. M., Goldring, C. E., Kitteringham, N. R., Park, B. K. (2008). The Nrf2-Keap1 defence pathway: role in protection against drug-induced toxicity. Toxicology 246:24–33.
  • Coutinho, A. E., Chapman, K. E. (2011). The anti-inflammatory and immunosuppressive effects of glucocorticoids, recent developments and mechanistic insights. Mol Cell Endocrinol 335:2–13.
  • Dalton, T. P., Puga, A., Shertzer, H. G. (2002). Induction of cellular oxidative stress by aryl hydrocarbon receptor activation. Chem Biol Interact 141:77–95.
  • De Bosscher, K., Haegeman, G. (2009). Minireview: latest perspectives on antiinflammatory actions of glucocorticoids. Mol Endocrinol 23:281–291.
  • De Bosscher, K., Vanden Berghe, W., Haegeman, G. (2003). The interplay between the glucocorticoid receptor and nuclear factor-kappaB or activator protein-1: molecular mechanisms for gene repression. Endocr Rev 24:488–522.
  • De Fabiani, E., Mitro, N., Anzulovich, A. C., Pinelli, A., Galli, G., Crestani, M. (2001). The negative effects of bile acids and tumor necrosis factor-alpha on the transcription of cholesterol 7alpha-hydroxylase gene (CYP7A1) converge to hepatic nuclear factor-4: a novel mechanism of feedback regulation of bile acid synthesis mediated by nuclear receptors. J Biol Chem 276:30708–30716.
  • De Souza, V. R., Cabrera, W. K., Galvan, A., Ribeiro, O. G., De Franco, M., Vorraro, F. et al. (2009). Aryl hydrocarbon receptor polymorphism modulates DMBA-induced inflammation and carcinogenesis in phenotypically selected mice. Int J Cancer 124:1478–1482.
  • Deng, Y., Theken, K. N., Lee, C. R. (2010). Cytochrome P450 epoxygenases, soluble epoxide hydrolase, and the regulation of cardiovascular inflammation. J Mol Cell Cardiol 48:331–341.
  • Denison, M. S., Nagy, S. R. (2003). Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol 43:309–334.
  • Denson, L. A., Auld, K. L., Schiek, D. S., McClure, M. H., Mangelsdorf, D. J., Karpen, S. J. (2000). Interleukin-1beta suppresses retinoid transactivation of two hepatic transporter genes involved in bile formation. J Biol Chem 275:8835–8843.
  • Dickmann, L. J., Patel, S. K., Wienkers, L. C., Slatter, J. G. (2012). Effects of interleukin 1ß (IL-1ß) and IL-1ß/interleukin 6 (IL-6) combinations on drug metabolizing enzymes in human hepatocyte culture. Curr Drug Metab 13:930–937.
  • DiNatale, B. C., Schroeder, J. C., Perdew, G. H. (2011). Ah receptor antagonism inhibits constitutive and cytokine inducible IL6 production in head and neck tumor cell lines. Mol Carcinog 50:173–183.
  • Ding, X., Staudinger, J. L. (2005). Repression of PXR-mediated induction of hepatic CYP3A gene expression by protein kinase C. Biochem Pharmacol 69:867–873.
  • Diry, M., Tomkiewicz, C., Koehle, C., Coumoul, X., Bock, K. W., Barouki, R. et al. (2006). Activation of the dioxin/aryl hydrocarbon receptor (AhR) modulates cell plasticity through a JNK-dependent mechanism. Oncogene 25:5570–5574.
  • Dou, W., Mukherjee, S., Li, H., Venkatesh, M., Wang, H., Kortagere, S. et al. (2012). Alleviation of gut inflammation by Cdx2/Pxr pathway in a mouse model of chemical colitis. Plos One 7:e36075.
  • Dring, M. M., Goulding, C. A., Trimble, V. I., Keegan, D., Ryan, A. W., Brophy, K. M. et al. (2006). The pregnane X receptor locus is associated with susceptibility to inflammatory bowel disease. Gastroenterology 130:341–8; quiz 592.
  • Dubrac, S., Elentner, A., Ebner, S., Horejs-Hoeck, J., Schmuth, M. (2010). Modulation of T lymphocyte function by the pregnane X receptor. J Immunol 184:2949–2957.
  • Duret, C., Daujat-Chavanieu, M., Pascussi, J. M., Pichard-Garcia, L., Balaguer, P., Fabre, J. M. et al. (2006). Ketoconazole and miconazole are antagonists of the human glucocorticoid receptor: consequences on the expression and function of the constitutive androstane receptor and the pregnane X receptor. Mol Pharmacol 70:329–339.
  • Duret, C., Gerbal-Chaloin, S., Ramos, J., Fabre, J. M., Jacquet, E., Navarro, F. et al. (2007). Isolation, characterization, and differentiation to hepatocyte-like cells of nonparenchymal epithelial cells from adult human liver. Stem Cells 25:1779–1790.
  • El-Kadi, A. O., Bleau, A. M., Dumont, I., Maurice, H., du Souich, P. (2000). Role of reactive oxygen intermediates in the decrease of hepatic cytochrome P450 activity by serum of humans and rabbits with an acute inflammatory reaction. Drug Metab Dispos 28:1112–1120.
  • Ernst, P. (1999). Review article: the role of inflammation in the pathogenesis of gastric cancer. Aliment Pharmacol Ther 13 Suppl 1:13–18.
  • Fan, J., Maeng, H. J., Du, Y., Kwan, D., Pang, K. S. (2011). Transport of 5,5-diphenylbarbituric acid and its precursors and their effect on P-gp, MRP2 and CYP3A4 in Caco-2 and LS180 cells. Eur J Pharm Sci 42:19–29.
  • Fang, C., Yoon, S., Tindberg, N., Järveläinen, H. A., Lindros, K. O., Ingelman-Sundberg, M. (2004). Hepatic expression of multiple acute phase proteins and down-regulation of nuclear receptors after acute endotoxin exposure. Biochem Pharmacol 67:1389–1397.
  • Favreau, L. V., Pickett, C. B. (1991). Transcriptional regulation of the rat NAD(P)H:quinone reductase gene. Identification of regulatory elements controlling basal level expression and inducible expression by planar aromatic compounds and phenolic antioxidants. J Biol Chem 266:4556–4561.
  • Feghali, C. A., Wright, T. M. (1997). Cytokines in acute and chronic inflammation. Front Biosci 2:d12–d26.
  • Ferguson, S. S., Chen, Y., LeCluyse, E. L., Negishi, M., Goldstein, J. A. (2005). Human CYP2C8 is transcriptionally regulated by the nuclear receptors constitutive androstane receptor, pregnane X receptor, glucocorticoid receptor, and hepatic nuclear factor 4alpha. Mol Pharmacol 68:747–757.
  • Fisher, C. D., Lickteig, A. J., Augustine, L. M., Ranger-Moore, J., Jackson, J. P., Ferguson, S. S. et al. (2009). Hepatic cytochrome P450 enzyme alterations in humans with progressive stages of nonalcoholic fatty liver disease. Drug Metab Dispos 37:2087–2094.
  • Fujii-Kuriyama, Y., Kawajiri, K. (2010). Molecular mechanisms of the physiological functions of the aryl hydrocarbon (dioxin) receptor, a multifunctional regulator that senses and responds to environmental stimuli. Proc Jpn Acad Ser B Phys Biol Sci 86:40–53.
  • Gabay, C., Kushner, I. (1999). Acute-phase proteins and other systemic responses to inflammation. N Engl J Med 340:448–454.
  • Gajjar, K., Martin-Hirsch, P. L., Martin, F. L. (2012). CYP1B1 and hormone-induced cancer. Cancer Lett 324:13–30.
  • Gao, B., Wang, H., Lafdil, F., Feng, D. (2012). STAT proteins - key regulators of anti-viral responses, inflammation, and tumorigenesis in the liver. J Hepatol 57:430–441.
  • Geier, A., Dietrich, C. G., Voigt, S., Kim, S. K., Gerloff, T., Kullak-Ublick, G. A. et al. (2003). Effects of proinflammatory cytokines on rat organic anion transporters during toxic liver injury and cholestasis. Hepatology 38:345–354.
  • Geiss-Friedlander, R., Melchior, F. (2007). Concepts in sumoylation: a decade on. Nat Rev Mol Cell Biol 8:947–956.
  • Gerbal-Chaloin, S., Daujat, M., Pascussi, J. M., Pichard-Garcia, L., Vilarem, M. J., Maurel, P. (2002). Transcriptional regulation of CYP2C9 gene. Role of glucocorticoid receptor and constitutive androstane receptor. J Biol Chem 277:209–217.
  • Germain, P., Staels, B., Dacquet, C., Spedding, M., Laudet, V. (2006). Overview of nomenclature of nuclear receptors. Pharmacol Rev 58:685–704.
  • Gharavi, N., El-Kadi, A. O. (2005). Down-regulation of aryl hydrocarbon receptor-regulated genes by tumor necrosis factor-alpha and lipopolysaccharide in murine hepatoma Hepa 1c1c7 cells. J Pharm Sci 94:493–506.
  • Gharavi, N., El-Kadi, A. O. (2007). Role of nitric oxide in downregulation of cytochrome P450 1a1 and NADPH: Quinone oxidoreductase 1 by tumor necrosis factor-alpha and lipopolysaccharide. J Pharm Sci 96:2795–2807.
  • Ghose, R., Mulder, J., von Furstenberg, R. J., Thevananther, S., Kuipers, F., Karpen, S. J. (2007). Rosiglitazone attenuates suppression of RXRalpha-dependent gene expression in inflamed liver. J Hepatol 46:115–123.
  • Ghose, R., Zimmerman, T. L., Thevananther, S., Karpen, S. J. (2004). Endotoxin leads to rapid subcellular re-localization of hepatic RXRalpha: A novel mechanism for reduced hepatic gene expression in inflammation. Nucl Recept 2:4.
  • Giannini, E., Fasoli, A., Botta, F., Romagnoli, P., Malfatti, F., Chiarbonello, B. et al. (2003). Helicobacter pylori infection is associated with greater impairment of cytochrome P-450 liver metabolic activity in anti-HCV positive cirrhotic patients. Dig Dis Sci 48:802–808.
  • Glas, J., Seiderer, J., Fischer, D., Tengler, B., Pfennig, S., Wetzke, M. et al. (2011). Pregnane X receptor (PXR/NR1I2) gene haplotypes modulate susceptibility to inflammatory bowel disease. Inflamm Bowel Dis 17:1917–1924.
  • Gu, X., Ke, S., Liu, D., Sheng, T., Thomas, P. E., Rabson, A. B. et al. (2006). Role of NF-kappaB in regulation of PXR-mediated gene expression: a mechanism for the suppression of cytochrome P-450 3A4 by proinflammatory agents. J Biol Chem 281:17882–17889.
  • Haack, M. J., Bak, M. L., Beurskens, R., Maes, M., Stolk, L. M., Delespaul, P. A. (2003). Toxic rise of clozapine plasma concentrations in relation to inflammation. Eur Neuropsychopharmacol 13:381–385.
  • Hankinson, O. (1995). The aryl hydrocarbon receptor complex. Annu Rev Pharmacol Toxicol 35:307–340.
  • Henkler, F., Stolpmann, K., Luch, A. (2012). Exposure to polycyclic aromatic hydrocarbons: bulky DNA adducts and cellular responses. Exs 101:107–131.
  • Henklova, P., Vrzal, R., Papouskova, B., Bednar, P., Jancova, P., Anzenbacherova, E. et al. (2008a). SB203580, a pharmacological inhibitor of p38 MAP kinase transduction pathway activates ERK and JNK MAP kinases in primary cultures of human hepatocytes. Eur J Pharmacol 593:16–23.
  • Henklová, P., Vrzal, R., Ulrichová, J., Dvorák, Z. (2008b). Role of mitogen-activated protein kinases in aryl hydrocarbon receptor signaling. Chem Biol Interact 172:93–104.
  • Ho, G. T., Soranzo, N., Tate, S. K., Drummond, H., Nimmo, E. R., Tenesa, A. et al. (2006). Lack of association of the pregnane X receptor (PXR/NR1I2) gene with inflammatory bowel disease: parallel allelic association study and gene wide haplotype analysis. Gut 55:1676–1677.
  • Hoffer, A., Chang, C. Y., Puga, A. (1996). Dioxin induces transcription of fos and jun genes by Ah receptor-dependent and -independent pathways. Toxicol Appl Pharmacol 141:238–247.
  • Hollingshead, B. D., Beischlag, T. V., Dinatale, B. C., Ramadoss, P., Perdew, G. H. (2008). Inflammatory signaling and aryl hydrocarbon receptor mediate synergistic induction of interleukin 6 in MCF-7 cells. Cancer Res 68:3609–3617.
  • Holt, M. P., Ju, C. (2006). Mechanisms of drug-induced liver injury. AAPS J 8:E48–E54.
  • Hsu, H. Y., Wen, M. H. (2002). Lipopolysaccharide-mediated reactive oxygen species and signal transduction in the regulation of interleukin-1 gene expression. J Biol Chem 277:22131–22139.
  • Hu, G., Xu, C., Staudinger, J. L. (2010). Pregnane X receptor is SUMOylated to repress the inflammatory response. J Pharmacol Exp Ther 335:342–350.
  • Huang, H., Wang, H., Sinz, M., Zoeckler, M., Staudinger, J., Redinbo, M. R. et al. (2007). Inhibition of drug metabolism by blocking the activation of nuclear receptors by ketoconazole. Oncogene 26:258–268.
  • Hybertson, B. M., Gao, B., Bose, S. K., McCord, J. M. (2011). Oxidative stress in health and disease: the therapeutic potential of Nrf2 activation. Mol Aspects Med 32:234–246.
  • Ikuta, T., Kobayashi, Y., Kawajiri, K. (2004). Cell density regulates intracellular localization of aryl hydrocarbon receptor. J Biol Chem 279:19209–19216.
  • Johnson, C., Han, Y., Hughart, N., McCarra, J., Alpini, G., Meng, F. (2012). Interleukin-6 and its receptor, key players in hepatobiliary inflammation and cancer. Transl Gastrointest Cancer 1:58–70.
  • Kacevska, M., Robertson, G. R., Clarke, S. J., Liddle, C. (2008). Inflammation and CYP3A4-mediated drug metabolism in advanced cancer: impact and implications for chemotherapeutic drug dosing. Expert Opin Drug Metab Toxicol 4:137–149.
  • Kamiya, A., Inoue, Y., Gonzalez, F. J. (2003). Role of the hepatocyte nuclear factor 4alpha in control of the pregnane X receptor during fetal liver development. Hepatology 37:1375–1384.
  • Kassel, O., Herrlich, P. (2007). Crosstalk between the glucocorticoid receptor and other transcription factors: molecular aspects. Mol Cell Endocrinol 275:13–29.
  • Kawamoto, T., Sueyoshi, T., Zelko, I., Moore, R., Washburn, K., Negishi, M. (1999). Phenobarbital-responsive nuclear translocation of the receptor CAR in induction of the CYP2B gene. Mol Cell Biol 19:6318–6322.
  • Ke, S., Rabson, A. B., Germino, J. F., Gallo, M. A., Tian, Y. (2001). Mechanism of suppression of cytochrome P-450 1A1 expression by tumor necrosis factor-alpha and lipopolysaccharide. J Biol Chem 276:39638–39644.
  • Kensler, T. W., Wakabayashi, N., Biswal, S. (2007). Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol 47:89–116.
  • Kerkvliet, N. I., Shepherd, D. M., Baecher-Steppan, L. (2002). T lymphocytes are direct, aryl hydrocarbon receptor (AhR)-dependent targets of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD): AhR expression in both CD4+ and CD8+ T cells is necessary for full suppression of a cytotoxic T lymphocyte response by TCDD. Toxicol Appl Pharmacol 185:146–152.
  • Kim, D. W., Gazourian, L., Quadri, S. A., Romieu-Mourez, R., Sherr, D. H., Sonenshein, G. E. (2000). The RelA NF-kappaB subunit and the aryl hydrocarbon receptor (AhR) cooperate to transactivate the c-myc promoter in mammary cells. Oncogene 19:5498–5506.
  • Kim, J. Y., Lee, S. M. (2006). Vitamins C and E protect hepatic cytochrome P450 dysfunction induced by polymicrobial sepsis. Eur J Pharmacol 534:202–209.
  • Kimura, A., Naka, T., Nakahama, T., Chinen, I., Masuda, K., Nohara, K. et al. (2009). Aryl hydrocarbon receptor in combination with Stat1 regulates LPS-induced inflammatory responses. J Exp Med 206:2027–2035.
  • Kimura, A., Naka, T., Nohara, K., Fujii-Kuriyama, Y., Kishimoto, T. (2008). Aryl hydrocarbon receptor regulates Stat1 activation and participates in the development of Th17 cells. Proc Natl Acad Sci U S A 105:9721–9726.
  • Kobayashi, A., Numayama-Tsuruta, K., Sogawa, K., Fujii-Kuriyama, Y. (1997). CBP/p300 functions as a possible transcriptional coactivator of Ah receptor nuclear translocator (Arnt). J Biochem 122:703–710.
  • Kobayashi, S., Okamoto, H., Iwamoto, T., Toyama, Y., Tomatsu, T., Yamanaka, H. et al. (2008). A role for the aryl hydrocarbon receptor and the dioxin TCDD in rheumatoid arthritis. Rheumatology (Oxford) 47:1317–1322.
  • Köhle, C. Bock, K. W. (2007). Coordinate regulation of Phase I and II xenobiotic metabolisms by the Ah receptor and Nrf2. Biochem Pharmacol 73:1853–1862.
  • Köhle, C., Bock, K. W. (2009). Coordinate regulation of human drug-metabolizing enzymes, and conjugate transporters by the Ah receptor, pregnane X receptor and constitutive androstane receptor. Biochem Pharmacol 77:689–699.
  • Koike, C., Moore, R., Negishi, M. (2007). Extracellular signal-regulated kinase is an endogenous signal retaining the nuclear constitutive active/androstane receptor (CAR) in the cytoplasm of mouse primary hepatocytes. Mol Pharmacol 71:1217–1221.
  • Korashy, H. M., Anwar-Mohamed, A., Soshilov, A. A., Denison, M. S., El-Kadi, A. O. (2011). The p38 MAPK inhibitor SB203580 induces cytochrome P450 1A1 gene expression in murine and human hepatoma cell lines through ligand-dependent aryl hydrocarbon receptor activation. Chem Res Toxicol 24:1540–1548.
  • Korashy, H. M., El-Kadi, A. O. (2008). The role of redox-sensitive transcription factors NF-kappaB and AP-1 in the modulation of the Cyp1a1 gene by mercury, lead, and copper. Free Radic Biol Med 44:795–806.
  • Kosters, A., White, D. D., Sun, H., Thevananther, S., Karpen, S. J. (2009). Redundant roles for cJun-N-terminal kinase 1 and 2 in interleukin-1beta-mediated reduction and modification of murine hepatic nuclear retinoid X receptor alpha. J Hepatol 51:898–908.
  • Kumar, M. B., Perdew, G. H. (1999). Nuclear receptor coactivator SRC-1 interacts with the Q-rich subdomain of the AhR and modulates its transactivation potential. Gene Expr 8:273–286.
  • Kyriakis, J. M., Avruch, J. (2012). Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Physiol Rev 92:689–737.
  • Lambert, C. B., Spire, C., Claude, N., Guillouzo, A. (2009). Dose- and time-dependent effects of phenobarbital on gene expression profiling in human hepatoma HepaRG cells. Toxicol Appl Pharmacol 234:345–360.
  • Lecureur, V., Ferrec, E. L., N’diaye, M., Vee, M. L., Gardyn, C., Gilot, D. et al. (2005). ERK-dependent induction of TNFalpha expression by the environmental contaminant benzo(a)pyrene in primary human macrophages. Febs Letters 579:1904–1910.
  • Lee, J. H., Park, S. M., Kim, O. S., Lee, C. S., Woo, J. H., Park, S. J. et al. (2009). Differential SUMOylation of LXRalpha and LXRbeta mediates transrepression of STAT1 inflammatory signaling in IFN-gamma-stimulated brain astrocytes. Mol Cell 35:806–817.
  • Lee, J. S., Kim, K. I., Baek, S. H. (2008). Nuclear receptors and coregulators in inflammation and cancer. Cancer Lett 267:189–196.
  • Lehmann, J. M., McKee, D. D., Watson, M. A., Willson, T. M., Moore, J. T., Kliewer, S. A. (1998). The human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Invest 102:1016–1023.
  • Li, N., Klaassen, C. D. (2004). Role of liver-enriched transcription factors in the down-regulation of organic anion transporting polypeptide 4 (oatp4; oatplb2; slc21a10) by lipopolysaccharide. Mol Pharmacol 66:694–701.
  • Li, T., Jahan, A., Chiang, J. Y. (2006). Bile acids and cytokines inhibit the human cholesterol 7 alpha-hydroxylase gene via the JNK/c-jun pathway in human liver cells. Hepatology 43:1202–1210.
  • Li, T., Yu, R. T., Atkins, A. R., Downes, M., Tukey, R. H., Evans, R. M. (2012). Targeting the pregnane X receptor in liver injury. Expert Opin Ther Targets 16:1075–1083.
  • Li, W., Vogel, C. F., Wu, D., Matsumura, F. (2010). Non-genomic action of TCDD to induce inflammatory responses in HepG2 human hepatoma cells and in liver of C57BL/6J mice. Biol Chem 391:1205–1219.
  • Long, W. P., Pray-Grant, M., Tsai, J. C., Perdew, G. H. (1998). Protein kinase C activity is required for aryl hydrocarbon receptor pathway-mediated signal transduction. Mol Pharmacol 53:691–700.
  • Ma, Q. (2008). Xenobiotic-activated receptors: from transcription to drug metabolism to disease. Chem Res Toxicol 21:1651–1671.
  • Ma, Q. (2011). Influence of light on aryl hydrocarbon receptor signaling and consequences in drug metabolism, physiology and disease. Expert Opin Drug Metab Toxicol 7:1267–1293.
  • Ma, Q., Kinneer, K., Bi, Y., Chan, J. Y., Kan, Y. W. (2004). Induction of murine NAD(P)H:quinone oxidoreductase by 2,3,7,8-tetrachlorodibenzo-p-dioxin requires the CNC (cap ‘n’ collar) basic leucine zipper transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2): cross-interaction between AhR (aryl hydrocarbon receptor) and Nrf2 signal transduction. Biochem J 377:205–213.
  • Majkova, Z., Smart, E., Toborek, M., Hennig, B. (2009). Up-regulation of endothelial monocyte chemoattractant protein-1 by coplanar PCB77 is caveolin-1-dependent. Toxicol Appl Pharmacol 237:1–7.
  • Malaplate-Armand, C., Ferrari, L., Masson, C., Siest, G., Batt, A. M. (2003). Astroglial CYP1B1 up-regulation in inflammatory/oxidative toxic conditions: IL-1beta effect and protection by N-acetylcysteine. Toxicol Lett 138:243–251.
  • Mannering, G. J. (1986). Measurement of effect of interferon on drug metabolism. Methods Enzymol 119:718–725.
  • Mannering, G. J., Renton, K. W., el Azhary, R., Deloria, L. B. (1980). Effects of interferon-inducing agents on hepatic cytochrome P-450 drug metabolizing systems. Ann N Y Acad Sci 350:314–331.
  • Marchand, A., Barouki, R., Garlatti, M. (2004). Regulation of NAD(P)H:quinone oxidoreductase 1 gene expression by CYP1A1 activity. Mol Pharmacol 65:1029–1037.
  • Masubuchi, Y., Horie, T. (2004). Endotoxin-mediated disturbance of hepatic cytochrome P450 function and development of endotoxin tolerance in the rat model of dextran sulfate sodium-induced experimental colitis. Drug Metab Dispos 32:437–441.
  • Masubuchi, Y., Sugiyama, S., Horie, T. (2009). Th1/Th2 cytokine balance as a determinant of acetaminophen-induced liver injury. Chem Biol Interact 179:273–279.
  • Matsumura, F. (2009). The significance of the nongenomic pathway in mediating inflammatory signaling of the dioxin-activated Ah receptor to cause toxic effects. Biochem Pharmacol 77:608–626.
  • McFadyen, M. C., Murray, G. I. (2005). Cytochrome P450 1B1: a novel anticancer therapeutic target. Future Oncology (London, England) 1:259–263.
  • McKay, L. I., Cidlowski, J. A. (1999). Molecular control of immune/inflammatory responses: interactions between nuclear factor-kappa B and steroid receptor-signaling pathways. Endocr Rev 20:435–459.
  • McKay, L. I., Cidlowski, J. A. (2000). CBP (CREB binding protein) integrates NF-kappaB (nuclear factor-kappaB) and glucocorticoid receptor physical interactions and antagonism. Mol Endocrinol 14:1222–1234.
  • Mencarelli, A., Renga, B., Palladino, G., Claudio, D., Ricci, P., Distrutti, E. et al. (2011). Inhibition of NF-?B by a PXR-dependent pathway mediates counter-regulatory activities of rifaximin on innate immunity in intestinal epithelial cells. Eur J Pharmacol 668:317–324.
  • Miao, W., Hu, L., Scrivens, P. J., Batist, G. (2005). Transcriptional regulation of NF-E2 p45-related factor (NRF2) expression by the aryl hydrocarbon receptor-xenobiotic response element signaling pathway: direct cross-talk between phase I and II drug-metabolizing enzymes. J Biol Chem 280:20340–20348.
  • Monteiro, P., Gilot, D., Langouet, S., Fardel, O. (2008). Activation of the aryl hydrocarbon receptor by the calcium/calmodulin-dependent protein kinase kinase inhibitor 7-oxo-7H-benzimidazo[2,1-a]benz[de]isoquinoline-3-carboxylic acid (STO-609). Drug Metab Dispos 36:2556–2563.
  • Monteleone, I., MacDonald, T. T., Pallone, F., Monteleone, G. (2012). The aryl hydrocarbon receptor in inflammatory bowel disease: linking the environment to disease pathogenesis. Curr Opin Gastroenterol 28:310–313.
  • Moore, L. B., Parks, D. J., Jones, S. A., Bledsoe, R. K., Consler, T. G., Stimmel, J. B. et al. (2000). Orphan nuclear receptors constitutive androstane receptor and pregnane X receptor share xenobiotic and steroid ligands. J Biol Chem 275:15122–15127.
  • Moreau, A., Vilarem, M. J., Maurel, P., Pascussi, J. M. (2008). Xenoreceptors CAR and PXR activation and consequences on lipid metabolism, glucose homeostasis, and inflammatory response. Mol Pharm 5:35–41.
  • Morgan, E. T. (1997). Regulation of cytochromes P450 during inflammation and infection. Drug Metab Rev 29:1129–1188.
  • Morgan, E. T. (2001). Regulation of cytochrome p450 by inflammatory mediators: why and how? Drug Metab Dispos 29:207–212.
  • Morgan, E. T. (2009). Impact of infectious and inflammatory disease on cytochrome P450-mediated drug metabolism and pharmacokinetics. Clin Pharmacol Ther 85:434–438.
  • Morgan, E. T., Goralski, K. B., Piquette-Miller, M., Renton, K. W., Robertson, G. R., Chaluvadi, M. R. et al. (2008). Regulation of drug-metabolizing enzymes and transporters in infection, inflammation, and cancer. Drug Metab Dispos 36:205–216.
  • Moriya, N., Kataoka, H., Fujino, H., Nishikawa, J., Kugawa, F. (2012). Effect of lipopolysaccharide on the xenobiotic-induced expression and activity of hepatic cytochrome P450 in mice. Biol Pharm Bull 35:473–480.
  • Moshage, H. (1997). Cytokines and the hepatic acute phase response. J Pathol 181:257–266.
  • Mottet, C., Golshayan, D. (2007). CD4+CD25+Foxp3+ regulatory T cells: from basic research to potential therapeutic use. Swiss Med Wkly 137:625–634.
  • Muntané-Relat, J., Ourlin, J. C., Domergue, J., Maurel, P. (1995). Differential effects of cytokines on the inducible expression of CYP1A1, CYP1A2, and CYP3A4 in human hepatocytes in primary culture. Hepatology 22(4 Pt 1):1143–1153.
  • Murray, G. I. (2000). The role of cytochrome P450 in tumour development and progression and its potential in therapy. J Pathol 192:419–426.
  • Murray, G. I., Melvin, W. T., Greenlee, W. F., Burke, M. D. (2001). Regulation, function, and tissue-specific expression of cytochrome P450 CYP1B1. Annu Rev Pharmacol Toxicol 41:297–316.
  • Mutoh, S., Osabe, M., Inoue, K., Moore, R., Pedersen, L., Perera, L. et al. (2009). Dephosphorylation of threonine 38 is required for nuclear translocation and activation of human xenobiotic receptor CAR (NR1I3). J Biol Chem 284:34785–34792.
  • N’Diaye, M., Le Ferrec, E., Lagadic-Gossmann, D., Corre, S., Gilot, D., Lecureur, V. et al. (2006). Aryl hydrocarbon receptor- and calcium-dependent induction of the chemokine CCL1 by the environmental contaminant benzo[a]pyrene. J Biol Chem 281:19906–19915.
  • Nikolaidou-Neokosmidou, V., Zannis, V. I., Kardassis, D. (2006). Inhibition of hepatocyte nuclear factor 4 transcriptional activity by the nuclear factor kappaB pathway. Biochem J 398:439–450.
  • Nolin, T. D. (2010). On warfarin use in kidney disease: a therapeutic window of opportunity? Am J Kidney Dis 56:805–808.
  • Nones, K., Dommels, Y. E., Martell, S., Butts, C., McNabb, W. C., Park, Z. A. et al. (2009). The effects of dietary curcumin and rutin on colonic inflammation and gene expression in multidrug resistance gene-deficient (mdr1a-/-) mice, a model of Br J Nutr 101:169–181.
  • Opitz, C. A., Litzenburger, U. M., Sahm, F., Ott, M., Tritschler, I., Trump, S. et al. (2011). An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478:197–203.
  • Osabe, M., Negishi, M. (2011). Active ERK1/2 protein interacts with the phosphorylated nuclear constitutive active/androstane receptor (CAR; NR1I3), repressing dephosphorylation and sequestering CAR in the cytoplasm. J Biol Chem 286:35763–35769.
  • Papa, S., Zazzeroni, F., Bubici, C., Jayawardena, S., Alvarez, K., Matsuda, S. et al. (2004). Gadd45 beta mediates the NF-kappa B suppression of JNK signalling by targeting MKK7/JNKK2. Nat Cell Biol 6:146–153.
  • Park, S. J., Yoon, W. K., Kim, H. J., Son, H. Y., Cho, S. W., Jeong, K. S. et al. (2005). 2,3,7,8-Tetrachlorodibenzo-p-dioxin activates ERK and p38 mitogen-activated protein kinases in RAW 264.7 cells. Anticancer Res 25:2831–2836.
  • Pascual, G., Fong, A. L., Ogawa, S., Gamliel, A., Li, A. C., Perissi, V. et al. (2005). A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature 437:759–763.
  • Pascussi, J. M., Gerbal-Chaloin, S., Drocourt, L., Assénat, E., Larrey, D., Pichard-Garcia, L. et al. (2004). Cross-talk between xenobiotic detoxication and other signalling pathways: clinical and toxicological consequences. Xenobiotica 34:633–664.
  • Pascussi, J. M., Gerbal-Chaloin, S., Duret, C., Daujat-Chavanieu, M., Vilarem, M. J., Maurel, P. (2008). The tangle of nuclear receptors that controls xenobiotic metabolism and transport: crosstalk and consequences. Annu Rev Pharmacol Toxicol 48:1–32.
  • Pascussi, J. M., Gerbal-Chaloin, S., Fabre, J. M., Maurel, P., Vilarem, M. J. (2000a). Dexamethasone enhances constitutive androstane receptor expression in human hepatocytes: consequences on cytochrome P450 gene regulation. Mol Pharmacol 58:1441–1450.
  • Pascussi, J. M., Gerbal-Chaloin, S., Pichard-Garcia, L., Daujat, M., Fabre, J. M., Maurel, P. et al. (2000b). Interleukin-6 negatively regulates the expression of pregnane X receptor and constitutively activated receptor in primary human hepatocytes. Biochem Biophys Res Commun 274:707–713.
  • Pascussi, J. M., Robert, A., Moreau, A., Ramos, J., Bioulac-Sage, P., Navarro, F. et al. (2007). Differential regulation of constitutive androstane receptor expression by hepatocyte nuclear factor4alpha isoforms. Hepatology 45:1146–1153.
  • Patel, R. D., Murray, I. A., Flaveny, C. A., Kusnadi, A., Perdew, G. H. (2009). Ah receptor represses acute-phase response gene expression without binding to its cognate response element. Lab Invest 89:695–707.
  • Paunescu, E. (1970). In vivo and in vitro suppression of humoral and cellular immunological response by rifampicin. Nature 228:1188–1190.
  • Pedruzzi, L. M., Stockler-Pinto, M. B., Leite, M. Jr., Mafra, D. (2012). Nrf2-keap1 system versus NF-?B: the good and the evil in chronic kidney disease? Biochimie 94:2461–2466.
  • Pereira, T. M., Carlstedt-Duke, J., Lechner, M. C., Gustafsson, J. A. (1998). Identification of a functional glucocorticoid response element in the CYP3A1/IGC2 gene. DNA Cell Biol 17:39–49.
  • Piscaglia, F., Knittel, T., Kobold, D., Barnikol-Watanabe, S., Di Rocco, P., Ramadori, G. (1999). Cellular localization of hepatic cytochrome 1B1 expression and its regulation by aromatic hydrocarbons and inflammatory cytokines. Biochem Pharmacol 58:157–165.
  • Pondugula, S. R., Dong, H., Chen, T. (2009). Phosphorylation and protein-protein interactions in PXR-mediated CYP3A repression. Expert Opin Drug Metab Toxicol 5:861–873.
  • Puga, A., Ma, C., Marlowe, J. L. (2009). The aryl hydrocarbon receptor cross-talks with multiple signal transduction pathways. Biochem Pharmacol 77:713–722.
  • Puga, A., Nebert, D. W., Carrier, F. (1992). Dioxin induces expression of c-fos and c-jun proto-oncogenes and a large increase in transcription factor AP-1. DNA Cell Biol 11:269–281.
  • Puga, A., Xia, Y., Elferink, C. (2002). Role of the aryl hydrocarbon receptor in cell cycle regulation. Chem Biol Interact 141:117–130.
  • Radjendirane, V., Jaiswal, A. K. (1999). Antioxidant response element-mediated 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) induction of human NAD(P)H:quinone oxidoreductase 1 gene expression. Biochem Pharmacol 58:1649–1655.
  • Rawlings, J. S., Rosler, K. M., Harrison, D. A. (2004). The JAK/STAT signaling pathway. J Cell Sci 117(Pt 8):1281–1283.
  • Reiners, J. J. Jr, Schöller, A., Bischer, P., Cantu, A. R., Pavone, A. (1993). Suppression of cytochrome P450 Cyp1a-1 induction in murine hepatoma 1c1c7 cells by 12-O-tetradecanoylphorbol-13-acetate and inhibitors of protein kinase C. Arch Biochem Biophys 301:449–454.
  • Renton, K. W. (2001). Alteration of drug biotransformation and elimination during infection and inflammation. Pharmacol Ther 92:147–163.
  • Renton, K. W. (2005). Regulation of drug metabolism and disposition during inflammation and infection. Expert Opin Drug Metab Toxicol 1:629–640.
  • Renton, K. W., Keyler, D. E., Mannering, G. J. (1979). Suppression of the inductive effects of phenobarbital and 3-methylcholanthrene on ascorbic acid synthesis and hepatic cytochrome P-450-linked monooxygenase systems by the interferon inducers, poly rI.rC and tilorone. Biochem Biophys Res Commun 88:1017–1023.
  • Richardson, T. A., Morgan, E. T. (2005). Hepatic cytochrome P450 gene regulation during endotoxin-induced inflammation in nuclear receptor knockout mice. J Pharmacol Exp Ther 314:703–709.
  • Ricote, M., Li, A. C., Willson, T. M., Kelly, C. J., Glass, C. K. (1998). The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 391:79–82.
  • Rizzardini, M., Zappone, M., Villa, P., Gnocchi, P., Sironi, M., Diomede, L. et al. (1998). Kupffer cell depletion partially prevents hepatic heme oxygenase 1 messenger RNA accumulation in systemic inflammation in mice: role of interleukin 1beta. Hepatology 27:703–710.
  • Roberts, R. A., Ganey, P. E., Ju, C., Kamendulis, L. M., Rusyn, I., Klaunig, J. E. (2007). Role of the Kupffer cell in mediating hepatic toxicity and carcinogenesis. Toxicol Sci 96:2–15.
  • Rochette-Egly, C. (2003). Nuclear receptors: integration of multiple signalling pathways through phosphorylation. Cellular Signalling 15:355–366.
  • Roos, P. H., Bolt, H. M. (2005). Cytochrome P450 interactions in human cancers: new aspects considering CYP1B1. Expert Opin Drug Metab Toxicol 1:187–202.
  • Ross, D. (2004). Quinone reductases multitasking in the metabolic world. Drug Metab Rev 36:639–654.
  • Roy-Chowdhury, J., Locker, J., Roy-Chowdhury, N. (2003). Nuclear receptors orchestrate detoxification pathways. Developmental Cell 4:607–608.
  • Ryu, S. D., Yi, H. G., Cha, Y. N., Kang, J. H., Kang, J. S., Jeon, Y. C. et al. (2004). Flavin-containing monooxygenase activity can be inhibited by nitric oxide-mediated S-nitrosylation. Life Sciences 75:2559–2572.
  • Sachdeva, K., Yan, B., Chichester, C. O. (2003). Lipopolysaccharide and cecal ligation/puncture differentially affect the subcellular distribution of the pregnane X receptor but consistently cause suppression of its target genes CYP3A. Shock (Augusta, Ga.) 19:469–474.
  • Samaras, S. C., Dietz, N. (1953). Physiopathology of detoxication of pentobarbital sodium. Fed Am Soc Exp Biol 12:400.
  • Schmith, V. D., Foss, J. F. (2008). Effects of inflammation on pharmacokinetics/pharmacodynamics: increasing recognition of its contribution to variability in response. Clin Pharmacol Ther 83:809–811.
  • Schote, A. B., Turner, J. D., Schiltz, J., Muller, C. P. (2007). Nuclear receptors in human immune cells: expression and correlations. Mol Immunol 44:1436–1445.
  • Schuetz, J. D., Schuetz, E. G., Thottassery, J. V., Guzelian, P. S., Strom, S., Sun, D. (1996). Identification of a novel dexamethasone responsive enhancer in the human CYP3A5 gene and its activation in human and rat liver cells. Mol Pharmacol 49:63–72.
  • Sciullo, E. M., Vogel, C. F., Li, W., Matsumura, F. (2008). Initial and extended inflammatory messages of the nongenomic signaling pathway of the TCDD-activated Ah receptor in U937 macrophages. Arch Biochem Biophys 480:143–155.
  • Sekine, H., Mimura, J., Oshima, M., Okawa, H., Kanno, J., Igarashi, K. et al. (2009). Hypersensitivity of aryl hydrocarbon receptor-deficient mice to lipopolysaccharide-induced septic shock. Mol Cell Biol 29:6391–6400.
  • Shah, Y. M., Ma, X., Morimura, K., Kim, I., Gonzalez, F. J. (2007). Pregnane X receptor activation ameliorates DSS-induced inflammatory bowel disease via inhibition of NF-kappaB target gene expression. Am J Physiol Gastrointest Liver Physiol 292:G1114–G1122.
  • Sheppard, K. A., Phelps, K. M., Williams, A. J., Thanos, D., Glass, C. K., Rosenfeld, M. G. et al. (1998). Nuclear integration of glucocorticoid receptor and nuclear factor-kappaB signaling by CREB-binding protein and steroid receptor coactivator-1. J Biol Chem 273:29291–29294.
  • Sheppard, K. A., Rose, D. W., Haque, Z. K., Kurokawa, R., McInerney, E., Westin, S. et al. (1999). Transcriptional activation by NF-kappaB requires multiple coactivators. Mol Cell Biol 19:6367–6378.
  • Shi, L. Z., Faith, N. G., Nakayama, Y., Suresh, M., Steinberg, H., Czuprynski, C. J. (2007). The aryl hydrocarbon receptor is required for optimal resistance to Listeria monocytogenes infection in mice. J Immunol 179:6952–6962.
  • Shibazaki, M., Takeuchi, T., Ahmed, S., Kikuchia, H. (2004). Blockade by SB203580 of Cyp1a1 induction by 2,3,7,8-tetrachlorodibenzo-p-dioxin, and the possible mechanism: possible involvement of the p38 mitogen-activated protein kinase pathway in shuttling of Ah receptor overexpressed in COS-7 cells. Ann N Y Acad Sci 1030:275–281.
  • Shin, S., Wakabayashi, N., Misra, V., Biswal, S., Lee, G. H., Agoston, E. S. et al. (2007). NRF2 modulates aryl hydrocarbon receptor signaling: influence on adipogenesis. Mol Cell Biol 27:7188–7197.
  • Siewert, E., Bort, R., Kluge, R., Heinrich, P. C., Castell, J., Jover, R. (2000). Hepatic cytochrome P450 down-regulation during aseptic inflammation in the mouse is interleukin 6 dependent. Hepatology 32:49–55.
  • Sissung, T. M., Price, D. K., Sparreboom, A., Figg, W. D. (2006). Pharmacogenetics and regulation of human cytochrome P450 1B1: implications in hormone-mediated tumor metabolism and a novel target for therapeutic intervention. Molecular Cancer Research: Mcr 4:135–150.
  • Slaviero, K. A., Clarke, S. J., Rivory, L. P. (2003). Inflammatory response: an unrecognised source of variability in the pharmacokinetics and pharmacodynamics of cancer chemotherapy. Lancet Oncol 4:224–232.
  • Sorrell, T. C., Forbes, I. J. (1975). Depression of immune competence by phenytoin and carbamazepine. Studies in vivo and in vitro. Clinical and Experimental Immunology 20:273–285.
  • Staudinger, J. L., Xu, C., Biswas, A., Mani, S. (2011). Post-translational modification of pregnane x receptor. Pharmacol Res 64:4–10.
  • Stevens, E. A., Mezrich, J. D., Bradfield, C. A. (2009). The aryl hydrocarbon receptor: a perspective on potential roles in the immune system. Immunology 127:299–311.
  • Stoltz, C., Anderson, A. (1999). Positive regulation of the rat CYP2B2 phenobarbital response unit by the nuclear receptor hexamer half-site.nuclear factor 1 complex. Biochem Pharmacol 57:1073–1076.
  • Sueyoshi, T., Kawamoto, T., Zelko, I., Honkakoski, P., Negishi, M. (1999). The repressed nuclear receptor CAR responds to phenobarbital in activating the human CYP2B6 gene. J Biol Chem 274:6043–6046.
  • Sugita-Konishi, Y., Kobayashi, K., Naito, H., Miura, K., Suzuki, Y. (2003). Effect of lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on the susceptibility to Listeria infection. Biosci Biotechnol Biochem 67:89–93.
  • Sun, S. C. (2012). The noncanonical NF-?B pathway. Immunol Rev 246:125–140.
  • Tan, Z., Chang, X., Puga, A., Xia, Y. (2002). Activation of mitogen-activated protein kinases (MAPKs) by aromatic hydrocarbons: role in the regulation of aryl hydrocarbon receptor (AHR) function. Biochem Pharmacol 64:771–780.
  • Tan, Z., Huang, M., Puga, A., Xia, Y. (2004). A critical role for MAP kinases in the control of Ah receptor complex activity. Toxicol Sci 82:80–87.
  • Teng, S., Piquette-Miller, M. (2005). The involvement of the pregnane X receptor in hepatic gene regulation during inflammation in mice. J Pharmacol Exp Ther 312:841–848.
  • Theken, K. N., Deng, Y., Kannon, M. A., Miller, T. M., Poloyac, S. M., Lee, C. R. (2011). Activation of the acute inflammatory response alters cytochrome P450 expression and eicosanoid metabolism. Drug Metab Dispos 39:22–29.
  • Tian, Y. (2009). Ah receptor and NF-kappaB interplay on the stage of epigenome. Biochem Pharmacol 77:670–680.
  • Tian, Y., Ke, S., Chen, M., Sheng, T. (2003). Interactions between the aryl hydrocarbon receptor and P-TEFb. Sequential recruitment of transcription factors and differential phosphorylation of C-terminal domain of RNA polymerase II at cyp1a1 promoter. J Biol Chem 278:44041–44048.
  • Tian, Y., Ke, S., Denison, M. S., Rabson, A. B., Gallo, M. A. (1999). Ah receptor and NF-kappaB interactions, a potential mechanism for dioxin toxicity. J Biol Chem 274:510–515.
  • Tian, Y., Rabson, A. B., Gallo, M. A. (2002). Ah receptor and NF-kappaB interactions: mechanisms and physiological implications. Chem Biol Interact 141:97–115.
  • Tirona, R. G., Lee, W., Leake, B. F., Lan, L. B., Cline, C. B., Lamba, V. et al. (2003). The orphan nuclear receptor HNF4alpha determines PXR- and CAR-mediated xenobiotic induction of CYP3A4. Nat Med 9:220–224.
  • Umannová, L., Machala, M., Topinka, J., Nováková, Z., Milcová, A., Kozubík, A. et al. (2008). Tumor necrosis factor-alpha potentiates genotoxic effects of benzo[a]pyrene in rat liver epithelial cells through upregulation of cytochrome P450 1B1 expression. Mutat Res 640:162–169.
  • Umannová, L., Machala, M., Topinka, J., Schmuczerová, J., Krcmár, P., Neca, J. et al. (2011). Benzo[a]pyrene and tumor necrosis factor-a coordinately increase genotoxic damage and the production of proinflammatory mediators in alveolar epithelial type II cells. Toxicol Lett 206:121–129.
  • Umannová, L., Zatloukalová, J., Machala, M., Krcmár, P., Májková, Z., Hennig, B. et al. (2007). Tumor necrosis factor-alpha modulates effects of aryl hydrocarbon receptor ligands on cell proliferation and expression of cytochrome P450 enzymes in rat liver “stem-like” cells. Toxicol Sci 99:79–89.
  • Urquhart, B. L., Tirona, R. G., Kim, R. B. (2007). Nuclear receptors and the regulation of drug-metabolizing enzymes and drug transporters: implications for interindividual variability in response to drugs. J Clin Pharmacol 47:566–578.
  • Van Bogaert, T., De Bosscher, K., Libert, C. (2010). Crosstalk between TNF and glucocorticoid receptor signaling pathways. Cytokine Growth Factor Rev 21:275–286.
  • Van Voorhis, B. J., Anderson, D. J., Hill, J. A. (1989). The effects of RU 486 on immune function and steroid-induced immunosuppression in vitro. J Clin Endocrinol Metab 69:1195–1199.
  • Veldhoen, M., Hirota, K., Westendorf, A. M., Buer, J., Dumoutier, L., Renauld, J. C. et al. (2008). The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453:106–109.
  • Venteclef, N., Jakobsson, T., Steffensen, K. R., Treuter, E. (2011). Metabolic nuclear receptor signaling and the inflammatory acute phase response. Trends Endocrinol Metab 22:333–343.
  • Vogel, C. F., Matsumura, F. (2009). A new cross-talk between the aryl hydrocarbon receptor and RelB, a member of the NF-kappaB family. Biochem Pharmacol 77:734–745.
  • Vogel, C. F., Nishimura, N., Sciullo, E., Wong, P., Li, W., Matsumura, F. (2007a). Modulation of the chemokines KC and MCP-1 by 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice. Arch Biochem Biophys 461:169–175.
  • Vogel, C. F., Sciullo, E., Li, W., Wong, P., Lazennec, G., Matsumura, F. (2007b). RelB, a new partner of aryl hydrocarbon receptor-mediated transcription. Mol Endocrinol 21:2941–2955.
  • Vogel, C. F., Sciullo, E., Matsumura, F. (2007c). Involvement of RelB in aryl hydrocarbon receptor-mediated induction of chemokines. Biochem Biophys Res Commun 363:722–726.
  • Vondrácek, J., Umannová, L., Machala, M. (2011). Interactions of the aryl hydrocarbon receptor with inflammatory mediators: beyond CYP1A regulation. Curr Drug Metab 12:89–103.
  • Wagner, M., Zollner, G., Trauner, M. (2011). Nuclear receptors in liver disease. Hepatology 53:1023–1034.
  • Wallace, K., Cowie, D. E., Konstantinou, D. K., Hill, S. J., Tjelle, T. E., Axon, A. et al. (2010). The PXR is a drug target for chronic inflammatory liver disease. J Steroid Biochem Mol Biol 120:137–148.
  • Wan, Y. J., An, D., Cai, Y., Repa, J. J., Hung-Po Chen, T., Flores, M. et al. (2000). Hepatocyte-specific mutation establishes retinoid X receptor alpha as a heterodimeric integrator of multiple physiological processes in the liver. Mol Cell Biol 20:4436–4444.
  • Warren, G. W., Poloyac, S. M., Gary, D. S., Mattson, M. P., Blouin, R. A. (1999). Hepatic cytochrome P-450 expression in tumor necrosis factor-alpha receptor (p55/p75) knockout mice after endotoxin administration. J Pharmacol Exp Ther 288:945–950.
  • Watkins, L. R., Maier, S. F., Goehler, L. E. (1995). Immune activation: the role of pro-inflammatory cytokines in inflammation, illness responses and pathological pain states. Pain 63:289–302.
  • Watt, A. J., Garrison, W. D., Duncan, S. A. (2003). HNF4: a central regulator of hepatocyte differentiation and function. Hepatology 37:1249–1253.
  • Weiss, C., Faust, D., Dürk, H., Kolluri, S. K., Pelzer, A., Schneider, S. et al. (2005). TCDD induces c-jun expression via a novel Ah (dioxin) receptor-mediated p38-MAPK-dependent pathway. Oncogene 24:4975–4983.
  • Wu, D., Li, W., Lok, P., Matsumura, F., Vogel, C. F. (2011). AhR deficiency impairs expression of LPS-induced inflammatory genes in mice. Biochem Biophys Res Commun 410:358–363.
  • Wu, R., Cui, X., Dong, W., Zhou, M., Simms, H. H., Wang, P. (2006). Suppression of hepatocyte CYP1A2 expression by Kupffer cells via AhR pathway: the central role of proinflammatory cytokines. Int J Mol Med 18:339–346.
  • Xu, D. X., Wei, W., Sun, M. F., Wei, L. Z., Wang, J. P. (2005). Melatonin attenuates lipopolysaccharide-induced down-regulation of pregnane X receptor and its target gene CYP3A in mouse liver. J Pineal Res 38:27–34.
  • Xu, D. X., Wei, W., Sun, M. F., Wu, C. Y., Wang, J. P., Wei, L. Z. et al. (2004). Kupffer cells and reactive oxygen species partially mediate lipopolysaccharide-induced downregulation of nuclear receptor pregnane x receptor and its target gene CYP3a in mouse liver. Free Radic Biol Med 37:10–22.
  • Yamamoto, Y., Moore, R., Flavell, R. A., Lu, B., Negishi, M. (2010). Nuclear receptor CAR represses TNFalpha-induced cell death by interacting with the anti-apoptotic GADD45B. Plos One 5:e10121.
  • Yamamoto, Y., Negishi, M. (2008). The antiapoptotic factor growth arrest and DNA-damage-inducible 45 beta regulates the nuclear receptor constitutive active/androstane receptor-mediated transcription. Drug Metab Dispos 36:1189–1193.
  • Yang-Yen, H. F., Chambard, J. C., Sun, Y. L., Smeal, T., Schmidt, T. J., Drouin, J. et al. (1990). Transcriptional interference between c-Jun and the glucocorticoid receptor: mutual inhibition of DNA binding due to direct protein-protein interaction. Cell 62:1205–1215.
  • Yang, J., Hao, C., Yang, D., Shi, D., Song, X., Luan, X. et al. (2010). Pregnane X receptor is required for interleukin-6-mediated down-regulation of cytochrome P450 3A4 in human hepatocytes. Toxicol Lett 197:219–226.
  • Yang, Q., Shi, Y., He, J., Chen, Z. (2012). The evolving story of macrophages in acute liver failure. Immunol Lett 147:1–9.
  • Yeager, R. L., Reisman, S. A., Aleksunes, L. M., Klaassen, C. D. (2009). Introducing the “TCDD-inducible AhR-Nrf2 gene battery”. Toxicol Sci 111:238–246.
  • Zassadowski, F., Rochette-Egly, C., Chomienne, C., Cassinat, B. (2012). Regulation of the transcriptional activity of nuclear receptors by the MEK/ERK1/2 pathway. Cell Signal 24:2369–2377.
  • Zhang, J., Chaluvadi, M. R., Reddy, R., Motika, M. S., Richardson, T. A., Cashman, J. R. et al. (2009). Hepatic flavin-containing monooxygenase gene regulation in different mouse inflammation models. Drug Metab Dispos 37:462–468.
  • Zhou, C., Tabb, M. M., Nelson, E. L., Grün, F., Verma, S., Sadatrafiei, A. et al. (2006). Mutual repression between steroid and xenobiotic receptor and NF-kappaB signaling pathways links xenobiotic metabolism and inflammation. J Clin Invest 116:2280–2289.
  • Zhou, M., Maitra, S. R., Wang, P. (2008). The potential role of transcription factor aryl hydrocarbon receptor in downregulation of hepatic cytochrome P-450 during sepsis. Int J Mol Med 21:423–428.
  • Zimmerman, T. L., Thevananther, S., Ghose, R., Burns, A. R., Karpen, S. J. (2006). Nuclear export of retinoid X receptor alpha in response to interleukin-1beta-mediated cell signaling: roles for JNK and SER260. J Biol Chem 281:15434–15440.
  • Zollner, G., Wagner, M., Trauner, M. (2010). Nuclear receptors as drug targets in cholestasis and drug-induced hepatotoxicity. Pharmacol Ther 126:228–243.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.