DOI QR코드

DOI QR Code

Regulation of CYP1A1 and Inflammatory Cytokine by NCOA7 Isoform 4 in Response to Dioxin Induced Airway Inflammation

  • Cho, Sung-Hwan (Regional Center for Respiratory Diseases, Kangwon National University Hospital) ;
  • Park, Shin Young (Regional Center for Respiratory Diseases, Kangwon National University Hospital) ;
  • Lee, Eun Jeong (Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine) ;
  • Cho, Yo Han (Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine) ;
  • Park, Hyun Sun (Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine) ;
  • Hong, Seok-Ho (Department of Internal Medicine and Environmental Health Center, Kangwon National University Hospital, Kangwon National University School of Medicine) ;
  • Kim, Woo Jin (Regional Center for Respiratory Diseases, Kangwon National University Hospital)
  • Received : 2014.07.17
  • Accepted : 2014.12.10
  • Published : 2015.02.28

Abstract

Background: Aryl hydrocarbon receptor (AhR), a ligand-dependent transcription factor, binds to a wide variety of synthetic and naturally occurring compounds. AhR is involved in the regulation of inflammatory response during acute and chronic respiratory diseases. We investigated whether nuclear receptor coactivator 7 (NCOA7) could regulate transcriptional levels of AhR target genes and inflammatory cytokines in 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-treated human bronchial epithelial cells. This study was based on our previous study that NCOA7 was differentially expressed between normal and chronic obstructive pulmonary disease lung tissues. Methods: BEAS-2B and A549 cells grown under serum-free conditions were treated with or without TCDD (0.15 nM and 6.5 nM) for 24 hours after transfection of pCMV-NCOA7 isoform 4. Expression levels of cytochrome P4501A1 (CYP1A1), IL-6, and IL-8 were measured by quantitative real-time polymerase chain reaction. Results: The transcriptional activities of CYP1A1 and inflammatory cytokines were strongly induced by TCDD treatment in both BEAS-2B and A549 cell lines. The NCOA7 isoform 4 oppositely regulated the transcriptional activities of CYP1A1 and inflammatory cytokines between BEAS-2B and A549 cell lines. Conclusion: Our results suggest that NCOA7 could act as a regulator in the TCDD-AhR signaling pathway with dual roles in normal and abnormal physiological conditions.

Keywords

References

  1. Das SK. Harmful health effects of cigarette smoking. Mol Cell Biochem 2003;253:159-65. https://doi.org/10.1023/A:1026024829294
  2. Kitamura M, Kasai A. Cigarette smoke as a trigger for the dioxin receptor-mediated signaling pathway. Cancer Lett 2007;252:184-94. https://doi.org/10.1016/j.canlet.2006.11.015
  3. Poland A, Knutson JC. 2,3,7,8-Tetrachlorodibenzo-p-dioxin and related halogenated aromatic hydrocarbons: examination of the mechanism of toxicity. Annu Rev Pharmacol Toxicol 1982;22:517-54. https://doi.org/10.1146/annurev.pa.22.040182.002505
  4. Lai H, Rogers DF. New pharmacotherapy for airway mucus hypersecretion in asthma and COPD: targeting intracellular signaling pathways. J Aerosol Med Pulm Drug Deliv 2010;23:219-31. https://doi.org/10.1089/jamp.2009.0802
  5. Barnes PJ. Immunology of asthma and chronic obstructive pulmonary disease. Nat Rev Immunol 2008;8:183-92. https://doi.org/10.1038/nri2254
  6. Mauad T, Dolhnikoff M. Pathologic similarities and differences between asthma and chronic obstructive pulmonary disease. Curr Opin Pulm Med 2008;14:31-8. https://doi.org/10.1097/MCP.0b013e3282f19846
  7. Hylkema MN, Sterk PJ, de Boer WI, Postma DS. Tobacco use in relation to COPD and asthma. Eur Respir J 2007;29:438-45. https://doi.org/10.1183/09031936.00124506
  8. Lai ZW, Hundeiker C, Gleichmann E, Esser C. Cytokine gene expression during ontogeny in murine thymus on activation of the aryl hydrocarbon receptor by 2,3,7,8-tetrachlorodibenzo-p-dioxin. Mol Pharmacol 1997;52:30-7. https://doi.org/10.1124/mol.52.1.30
  9. Vogel C, Abel J. Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on growth factor expression in the human breast cancer cell line MCF-7. Arch Toxicol 1995;69:259-65. https://doi.org/10.1007/s002040050168
  10. Vogel C, Donat S, Dohr O, Kremer J, Esser C, Roller M, et al. Effect of subchronic 2,3,7,8-tetrachlorodibenzo-p-dioxin exposure on immune system and target gene responses in mice: calculation of benchmark doses for CYP1A1 and CYP1A2 related enzyme activities. Arch Toxicol 1997;71:372-82. https://doi.org/10.1007/s002040050401
  11. Warren TK, Mitchell KA, Lawrence BP. Exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) suppresses the humoral and cell-mediated immune responses to influenza A virus without affecting cytolytic activity in the lung. Toxicol Sci 2000;56:114-23. https://doi.org/10.1093/toxsci/56.1.114
  12. Mimura J, Fujii-Kuriyama Y. Functional role of AhR in the expression of toxic effects by TCDD. Biochim Biophys Acta 2003;1619:263-8. https://doi.org/10.1016/S0304-4165(02)00485-3
  13. Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu Rev Pharmacol Toxicol 2003;43:309-34. https://doi.org/10.1146/annurev.pharmtox.43.100901.135828
  14. Tojo M, Matsuzaki K, Minami T, Honda Y, Yasuda H, Chiba T, et al. The aryl hydrocarbon receptor nuclear transporter is modulated by the SUMO-1 conjugation system. J Biol Chem 2002;277:46576-85. https://doi.org/10.1074/jbc.M205987200
  15. Beischlag TV, Wang S, Rose DW, Torchia J, Reisz-Porszasz S, Muhammad K, et al. Recruitment of the NCoA/SRC-1/p160 family of transcriptional coactivators by the aryl hydrocarbon receptor/aryl hydrocarbon receptor nuclear translocator complex. Mol Cell Biol 2002;22:4319-33. https://doi.org/10.1128/MCB.22.12.4319-4333.2002
  16. Kasai A, Hiramatsu N, Hayakawa K, Yao J, Maeda S, Kitamura M. High levels of dioxin-like potential in cigarette smoke evidenced by in vitro and in vivo biosensing. Cancer Res 2006;66:7143-50. https://doi.org/10.1158/0008-5472.CAN-05-4541
  17. Gebremichael A, Tullis K, Denison MS, Cheek JM, Pinkerton KE. Ah-receptor-dependent modulation of gene expression by aged and diluted sidestream cigarette smoke. Toxicol Appl Pharmacol 1996;141:76-83. https://doi.org/10.1016/S0041-008X(96)80011-1
  18. Shao W, Halachmi S, Brown M. ERAP140, a conserved tissuespecific nuclear receptor coactivator. Mol Cell Biol 2002;22:3358-72. https://doi.org/10.1128/MCB.22.10.3358-3372.2002
  19. Paramanik V, Thakur MK. Interaction of estrogen receptor associated protein (ERAP) 140 with ER beta decreases but its expression increases in aging mouse cerebral cortex. Cell Mol Neurobiol 2010;30:961-6. https://doi.org/10.1007/s10571-010-9526-8
  20. Li H, Gomes PJ, Chen JD. RAC3, a steroid/nuclear receptorassociated coactivator that is related to SRC-1 and TIF2. Proc Natl Acad Sci U S A 1997;94:8479-84. https://doi.org/10.1073/pnas.94.16.8479
  21. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, et al. The nuclear receptor superfamily: the second decade. Cell 1995;83:835-9. https://doi.org/10.1016/0092-8674(95)90199-X
  22. Naar AM, Beaurang PA, Zhou S, Abraham S, Solomon W, Tjian R. Composite co-activator ARC mediates chromatindirected transcriptional activation. Nature 1999;398:828-32. https://doi.org/10.1038/19789
  23. Norman AW, Litwack G. Estrogens and progestins. In: Litwack G, editor. Hormones. London: Academic Press; 1987. p. 550-60.
  24. Vogel CF, Matsumura F. A new cross-talk between the aryl hydrocarbon receptor and RelB, a member of the NF-kappaB family. Biochem Pharmacol 2009;77:734-45. https://doi.org/10.1016/j.bcp.2008.09.036
  25. Badal S, Delgoda R. Role of the modulation of CYP1A1 expression and activity in chemoprevention. J Appl Toxicol 2014;34:743-53. https://doi.org/10.1002/jat.2968
  26. Lee AJ, Cai MX, Thomas PE, Conney AH, Zhu BT. Characterization of the oxidative metabolites of 17beta-estradiol and estrone formed by 15 selectively expressed human cytochrome p450 isoforms. Endocrinology 2003;144:3382-98. https://doi.org/10.1210/en.2003-0192
  27. Ma X, Idle JR, Krausz KW, Gonzalez FJ. Metabolism of melatonin by human cytochromes p450. Drug Metab Dispos 2005;33:489-94. https://doi.org/10.1124/dmd.104.002410
  28. Androutsopoulos VP, Tsatsakis AM, Spandidos DA. Cytochrome P450 CYP1A1: wider roles in cancer progression and prevention. BMC Cancer 2009;9:187. https://doi.org/10.1186/1471-2407-9-187
  29. Buterin T, Hess MT, Luneva N, Geacintov NE, Amin S, Kroth H, et al. Unrepaired fjord region polycyclic aromatic hydrocarbon- DNA adducts in ras codon 61 mutational hot spots. Cancer Res 2000;60:1849-56.
  30. Rodriguez M, Potter DA. CYP1A1 regulates breast cancer proliferation and survival. Mol Cancer Res 2013;11:780-92. https://doi.org/10.1158/1541-7786.MCR-12-0675

Cited by

  1. The Evolutionarily Conserved Tre2/Bub2/Cdc16 (TBC), Lysin Motif (LysM), Domain Catalytic (TLDc) Domain Is Neuroprotective against Oxidative Stress vol.291, pp.6, 2015, https://doi.org/10.1074/jbc.m115.685222
  2. TLDc proteins: new players in the oxidative stress response and neurological disease vol.28, pp.9, 2017, https://doi.org/10.1007/s00335-017-9706-7
  3. Identification of a unique gene expression signature in mercury and 2,3,7,8-tetrachlorodibenzo-p-dioxin co-exposed cells vol.6, pp.3, 2015, https://doi.org/10.1039/c6tx00432f
  4. Synergistic cellular effects including mitochondrial destabilization, autophagy and apoptosis following low-level exposure to a mixture of lipophilic persistent organic pollutants vol.7, pp.None, 2015, https://doi.org/10.1038/s41598-017-04654-0
  5. Mmp12 Is Upregulated by in utero Second-Hand Smoke Exposures and Is a Key Factor Contributing to Aggravated Lung Responses in Adult Emphysema, Asthma, and Lung Cancer Mouse Models vol.12, pp.None, 2021, https://doi.org/10.3389/fphys.2021.704401