Up-regulation of Heme Oxygenase-1 Expression by cAMP-elevating Agents in RAW 264.7 cells

  • Ko, Young-Shin (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences) ;
  • Park, Min-Kyu (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences) ;
  • Kang, Young-Jin (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences) ;
  • Lee, Young-Soo (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences) ;
  • Seo, Han-Geuk (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences) ;
  • Lee, Duck-Hyung (Department of Chemistry, Sogang University) ;
  • Yunchoi, Hye-Sook (Natural Products Research Institute, Seoul National University) ;
  • Chong, Won-Seog (Department of pharmacology, College of Medicine, Sonam University) ;
  • Chang, Ki-Churl (Department of pharmacology, College of Medicine, Gyeongsang National University, Institute of Health Sciences)
  • Published : 2002.06.01

Abstract

Heme oxygenase-1 (HO-1) is the inducible from of the rate-limiting enzyme of heme degradation; it regulates the cellular contents of heme. HO-1 is up-regulated by various stimuli including oxidative stress so that it is thought to participate in general cellular defense mechanisms against oxidative stress in mammalian cells. To investigate the role of the cAMP-dependent protein kinase A (PKA) signaling pathway on nitrogen oxidative stress-induced HO-1 gene expression, RAW 264.7 cell cultures were treated with sodium nitroprusside (SNP). SNP increased the expression of HO-1 mRNA and protein, time- and concentration-dependently. Treatment with H89, PKA inhibitor, but not LY83583, guanylate cyclase inhibitor, significantly diminished the HO-1 expression by SNP, indicating that cAMP plays a crucial role in the induction of HO-1. Incubation with cAMP-elevating agents, such as forskolin or isoproterenol resulted in up-regulation of the expression of HO-1. Forskolin-induced expression of HO-1 was inhibited by H89. Furthermore, propranolol, $\beta$-adrenoceptor blocker, inhibited the isoproterenol-induced HO-1 expression, supporting the importance of cAMP in the induction of HO-1 expression. Higenamine-S, but not higenamineR, enhanced the HO-1 expression induced by SNP. Furthermore, cellular toxicity induced by hydrogen peroxide was attenuated by the presence of SNP, which was further increased by the presence of ZnPPIX, HO-1 inhibitor. Collectively, these results strongly suggest that up-regulation of HO-1 expression in RAW 264.7 cells involves PKA signal pathway.

Keywords

References

  1. Abraham, N. G., Lavrovsky, Y., Schwartzman, M. L., Stoltx, R. A., Levere R. D., Gerritsen M. E., Shibahara S. and Kappas A. (1995). Transfection of the human heme oxygenase gene into rabbit coronary microvessel endo-thelial cells: protective effect against home and hemoglobin toxicity. Proc Natl Acad Sci USA., 92, 6798-6802 https://doi.org/10.1073/pnas.92.15.6798
  2. Ahmed, A., Rahman, M., Zhang, X., Acevedo, C. H., Nijjar S., Rushton I., Bussolati B. and St John J. (2000). Induction of placental heme oxygenase-1 is protective against TNF-alpha-induced cytotoxicity and promotes vessel relaxation. MoIMed,. 6,391-409
  3. Applegate, L. A, Luscher, P, and Tyrrell, R. M. (1991). Induction of heme oxygenase: a general response to oxidant stress in cultured mammalian cells. Cancer Res., 51, 974-978
  4. Camhi, S. L., Alam, J., Wiegand, G. W., Chin, B. Y. and Choi, A. M. (1998). Transcriptional activation of the HO-l gene by lipopolysaccharide is mediated by 5' distal enhancers: role of reactive oxygen intermediates and AP-l. Am J Respir Cell Mol. Biol., 18, 226-234 https://doi.org/10.1165/ajrcmb.18.2.2910
  5. Chang, K. C., Chong, W. S. and Lee, I. J. (1994). Different pharmacological characteristics of structurally similar benzylisoqumoline analogs, papaverine, higenamine, and GS 389, on isolated rat aorta and heart. Can. J. PhysioI. Pharmacol., 72, 327-3345 https://doi.org/10.1139/y94-049
  6. Chang, K. C., Lim, J. K. and Park, C. W. (1986). Synthesis of higenamine and its cardiovascular effects in rabbit:evidence for $\beta$-adrenoceptor agonist. Kor. J. PharmacoI., 22,96-104
  7. Chen, K. and Maines, M. D. (2000). Nitric oxide induces heme oxygenase-1 via mitogen-activated protein kinases ERK and P38. Cell. Mol.. Biol., 46, 609-617
  8. Christou, H., Morita, T., Hsich, C. M., Koike, H., Arkonac, B., Perrella, M. A. and Kourembanas, S. (2000). Prevention of hypoxia-induced pulmonary hypertension by enhancement of endogenous heme oxygenase-1 in the rat. Circ Res,. 86, 1224-1229 https://doi.org/10.1161/01.RES.86.12.1224
  9. Gross, S. S. and Levi, R. (1992). Tetrahydrobiopterin synthesis. An absolute requirement for cytokine-induced nitric oxide generation by vascular smooth muscle. J BioI Chem., 25, 267(36):25722-9
  10. Immenschuh, S., Kietzmann, T., Hinke, V., Wiederhold, M., Katz, N. and Mullcr-Eberhard, U. (1998). The rat heme oxygenase-1 gene is transcriptionally induced via the protein kinase A signaling pathway in rat hepatocyte cultures. Mol Pharmacol., 53, 483-91 https://doi.org/10.1124/mol.53.3.483
  11. Johnson, R. A, Lavesa, M., Askari, B., Abraham, N. G. and Nasjletti, A. (1995). A heme oxygenase product, Presu-mably carbon monoxide, mediates a vasodepressor role in rats. Hypertension., 25, 166-169 https://doi.org/10.1161/01.HYP.25.2.166
  12. Johnson, R. A., Lavesa, M., DeSeyn, K., Scholer, M. J. and Nasjletti A. (1996). Heme oxygenase substrates acutely lower blood pressure in hypertensive rats. Am J PhysioI., 271,H1131-H1138 https://doi.org/10.1152/ajpcell.1996.271.4.C1131
  13. Kang, Y. J., Lee, Y. S., Lee, G. W., Lee, D. H., Ryu, J. C., Yun-Choi, H. S. and Cbang, K. C. (1999). Inhibition of activation of nuclear factor kB is responsible for inhibition of inducible nitric oxide synthase expression by higen-amine, and active component of aconite root. J. Pharmacol. Exp. Ther., 291, 314-320
  14. Kyokane, T., Norimizu, S., Taniai, H., Yamaguchi, T., Takeoka, S., Tsuchida, E., Naito, M., Nimura, Y., Ishimura, Y., Suematsu, M. (2001). Carbon monoxide from heme catabolism protects against hepatobiliary dysfunction in endotoxin-treated rat liver. Gastroenterology, 120, 1227-1240 https://doi.org/10.1053/gast.2001.23249
  15. Lautier, D., Luscher, P. and Tyrrcll, R. M. (1992). Endogenous glutathione levels modulate both constitutive and UVA radiation/oxidant-inducible expression of the human home oxygenase gene- Carcinogenesis., 13, 227-232 https://doi.org/10.1093/carcin/13.2.227
  16. Lee, P. J, Alam, J., Wiegand, G. W. and Choi, A. M. (1996). Overexpression of heme oxygenase-1 in pulmonary epithelial cells results in cell growth arrest and increased resistance to hyperoxia. Proc NatI Acad Sci USA., 93, 10393-10398 https://doi.org/10.1073/pnas.93.19.10393
  17. Lee, Y. S., Kang, Y. J., Lee, B. K., Ko, Y. S., Park, M. K., Seo, H.G., Yim-Choi, H. S. and Chang, K. C. (2001). Down-regulation of TNF-a and IL-6 by higenamine is responsible for reduction ofinfarct size and myocardial ischemic injury in the rat. Kor. J. Appl. Pharmacol., 9, 167-175
  18. McCoubrey, W. K., Huang, T. J. and Maines, M. D. (1997). Isolation and characterization of a cDNA from the rat brain that encodes hemoprotein heme oxygenase-3. Eur J Biochem., 247, 725-732 https://doi.org/10.1111/j.1432-1033.1997.00725.x
  19. Miano, L. L. J. M., Mercer, B, and Olson, E, N. (1996). Expression of the SM22a promoter in transgenic inice provides evidence for distinct transcriptional regulatory programs in vascular and visceral smooth muscle cells. JCell Biol., 132, 849-859 https://doi.org/10.1083/jcb.132.5.849
  20. Shibahara, S., Muller, R., Taguchi, H. and Yoshida, T. (1985). Cloning and expression of cDNA for rat heme oxygenase. Proc Natl Acad Sci USA., 82, 7865-7869 https://doi.org/10.1073/pnas.82.23.7865
  21. Suematsu, M. and Ishimura, Y. (2000). The heme oxygenase-carbon monoxide system: a regulator of hepatobiliary functions. Hepatology., 31, 3-6 https://doi.org/10.1002/hep.510310102
  22. Trakshel, G. M., Kutty, R. K. and Maines, M. D. (1986). Purification and characterization of the major constitutive form of testicular heme oxygenase. J BioI Chem., 261, 11131-11137
  23. Yun-Choi, H. S., Lee, D. H. and Chang, K. C. (2001). Synthesis of optical isomers of tetrahydroisoquinoline compounds and its pharmaceutical composition. Korean Patent Application, 2001-21500