Evidence for the Association of Ce11u1ar Iron Loss in Nitric Oxide-induced Apoptosis of HL-60 Cells: Involvement of p38 Kinase, c-Jun N-terminal Kinase, Cytochrome C Release, and Caspases Pathways

  • Choi, Suck-Chei (Department of Digestive Research Unit, Institute of Wonkwang Medical Research Center) ;
  • Kim, Beom-Su (Department of Microbiology and Immunology, School of Medicine, Wonkwang University) ;
  • Yoon, Kwon-Ha (Department of Digestive Research Unit, Institute of Wonkwang Medical Research Center) ;
  • Song, Moon-Young (Department of Microbiology and Immunology, School of Medicine, Wonkwang University) ;
  • Oh, Hyun-Mee (Department of Microbiology and Immunology, School of Medicine, Wonkwang University) ;
  • Han, Weon-Cheol (Department of Digestive Research Unit, Institute of Wonkwang Medical Research Center) ;
  • Kim, Tae-Hyeon (Department of Digestive Research Unit, Institute of Wonkwang Medical Research Center) ;
  • Kim, Eun-Cheol (Department of Pathology, School of Dentistry, Wonkwang University) ;
  • Jun, Chang Duk (Department of Digestive Research Unit, Institute of Wonkwang Medical Research Center, Department of Microbiology and Immunology, School of Medicine, Wonkwang University)
  • Published : 2002.06.01

Abstract

Nitric oxide has high affinity for iron, and thus it can cause intracellular iron loss. We tested the idea that intracellular iron can be the primary target of NO toxicity by comparing the signaling mechanisms involved in cell death caused by iron depletion and that caused by NO. Treatment of HL-60 cells with a NO donor, S-nitroso-N-acetyl-DL-penicillamine (SNAP), decreased the intracellular iron level rapidly as that observed with the iron chelator deferoxamine (DFO). Iron chelators such as DFO and mimosine could induce death of human leukemic HL-60 cells by a mechanism requiring activation of p38 kinase, c-Jun N-terminal kinase, caspase-3 and caspase-8. DFO and SNAP also caused release of cytochrome c from mitochondria. Inhibition of p38 kinase by a selective inhibitor, SB203580, abolished the NO and DFO-induced cell death, release of cytochrome c, and activation of caspase-3 and caspase-8, thus indicating that p38 kinase lies upstream in the cell death processes. In a parallel situation, the cells that are sensitive to NO showed similar sensitivity to DFO. Moreover, simultaneous addition of ferric citrate, an iron-containing compound, inhibited the SNAP and DFO-induced activation of caspases and also blocked the NO-mediated cell cycle arrest at $G_1$ phase. Collectively, our data implicate that the NO-induced cell death of tumor cells including HL-60 cells is mediated by depletion of iron and further suggest that activation of p38 kinase lies upstream of cytochrome c release and caspase activation involved in this apoptotic process.

Keywords

References

  1. Amoroso S, Tortiglione A, Secondo A, Catalano A, Montagnani S, Di Renzo G, and Annunziato L (2000) Sodium nitroprusside prevents chemical hypoxia-induced cell death through iron ions stimulating the activity of the $Na^{+}-Ca^{2+}$ exchanger in C6 glioma cells. J Neurochem 74: 1505-1513 https://doi.org/10.1046/j.1471-4159.2000.0741505.x
  2. Bastian NR, Yim CY, Hibbs JB, Jr., and Samlowski WE (1994) Induction of iron-derived EPR signals in murine cancers by nitric oxide. Evidence for multiple intracellular targets. J Biol Chem 269: 5127-5131
  3. Beckman JS, Beckman TW, Chen J, Marshall PA, and Freeman BA (1990) Apparent hydroxyl radical production by peroxynitrite: implications for endothelial injury from nitric oxide and superoxide. Proc Natl Acad Sci USA 87: 1620-1624 https://doi.org/10.1073/pnas.87.4.1620
  4. Beckman JS and Crow JP (1993) Pathological implications of nitric oxide, superoxide and peroxynitrite formation. Biochem Soc Trans 21: 330-334 https://doi.org/10.1042/bst0210330
  5. Bitangcol JC, Chau AS, Stadnick E, Lohka MJ, Dicken B, and Shibuya EK (1998) Activation of the p42 mitogen-activated protein kinase pathway inhibits Cdc2 activation and entry into M-phase in cycling Xenopus egg extracts. Mol Biol Cell 9: 451-467 https://doi.org/10.1091/mbc.9.2.451
  6. Cheng A, Chan SL, Milhavet O, Wang S, and Mattson MP (2001) p38 MAP kinase mediates nitric oxide-induced apoptosis of neural progenitor cells. J Biol Chem 276: 43320-43327 https://doi.org/10.1074/jbc.M107698200
  7. Chlichlia K, Peter ME, Rocha M, Scaffidi C, Bucur M, Krammer PH, Schirrmacher V, and Umansky V (1998) Caspase activation is required for nitric oxide-mediated, CD95(APO- 1/Fas)-dependent and independent apoptosis in human neoplastic lymphoid cells. Blood 91: 4311-4320
  8. Dawson VL, Dawson TM, London ED, Bredt DS, and Snyder SH (1991) Nitric oxide mediates glutamate neurotoxicity in primary cortical cultures. Proc Natl Acad Sci USA 88: 6368-6371 https://doi.org/10.1073/pnas.88.14.6368
  9. Drapier JC, Pellat C, and Henry Y (1991) Generation of EPR-detectable nitrosyl-iron complexes in tumor target cells cocultured with activated macrophages. J Biol Chem 266: 10162-10167
  10. Feger F, Ferry-Dumazet H, Mamani Matsuda M, Bordenave J, Dupouy M, Nussler AK, Arock M, Devevey L, Nafziger J, Guillosson JJ, Reiffers J, and Mossalayi MD (2001) Role of iron in tumor cell protection from the pro-apoptotic effect of nitric oxide. Cancer Res 61: 5289-5294
  11. Fukuchi K, Tomoyasu S, Tsuruoka N, and Gomi K (1994) Iron deprivation-induced apoptosis in HL-60 cell. FEBS Lett 350: 139-142 https://doi.org/10.1016/0014-5793(94)00755-1
  12. Han J, Lee JD, Bibbs L, and Ulevitch RJ (1994) A MAP kinase targeted by endotoxin and hyperosmolarity in mammalian cells. Science 265: 808-811 https://doi.org/10.1126/science.7914033
  13. Jun CD, Oh CD, Kwak HJ, Pae HO, Yoo JC, Choi BM, Chun JS, Park RK, and Chung HT (1999) Overexpression of protein kinase C isoforms protects RAW 264.7 macrophages from nitric oxide-induced apoptosis: involvement of c-Jun N-terminal kinase/stress-activated protein kinase, p38 kinase, and CPP-32 protease pathways. J Immunol 162: 3395-3401
  14. Jun CD, Pae HO, Kwak HJ, Yoo JC, Choi BM, Oh CD, Chun JS, Paik SG, Park YH, and Chung HT (1999) Modulation of nitric oxide-induced apoptotic death of HL-60 cells by protein kinase C and protein kinase A through mitogen-activated protein kinase and CPP32-like protease pathways. Cell Immunol 194: 36-46 https://doi.org/10.1006/cimm.1999.1480
  15. Jun CD, Pae HO, Yoo JC, Kwak HJ, Park RK, and Chung HT (1998) Cyclic adenosine monophosphate inhibits nitric oxide-induced apoptosis in human leukemic HL-60 cells. Cell Immunol 183: 13-21 https://doi.org/10.1006/cimm.1997.1232
  16. Jun CD, Park SJ, Choi BM, Kwak HJ, Park YC, Kim MS, Park RK, and Chung HT (1997) Potentiation of the activity of nitric oxide by the protein kinase C activator phorbol ester in human myeloid leukemic HL-60 cells: association with enhanced fragmentation of mature genomic DNA. Cell Immunol 176: 41-49 https://doi.org/10.1006/cimm.1996.1064
  17. Kim, YM, Chung HT, Simmons RL, and Billiar TR (2000) Cellular non-heme iron content is a determinant of nitric oxide-mediated apoptosis, necrosis, and caspase inhibition. J Biol Chem 275: 10954-10961 https://doi.org/10.1074/jbc.275.15.10954
  18. Krishan A (1975) Rapid flow cytofluorometric analysis of mammalian cell cycle by propidium iodide staining. J Cell Biol 66: 188-193 https://doi.org/10.1083/jcb.66.1.188
  19. Kulp KS, Green SL, and Vulliet PR (1996) Iron deprivation inhibits cyclin-dependent kinase activity and decreases cyclin D/CDK4 protein levels in asynchronous MDA-MB-453 human breast cancer cells. Exp Cell Res 229: 60-68 https://doi.org/10.1006/excr.1996.0343
  20. Leardi A, Caraglia M, Selleri C, Pepe S, Pizzi C, Notaro R, Fabbrocini A, De Lorenzo S, Musico M, Abbruzzese A, Bianco AR, and Tagliaferri P (1998) Desferioxamine increases iron depletion and apoptosis induced by ara-C of human myeloid leukaemic cells. Br J Haematol 102: 746-752 https://doi.org/10.1046/j.1365-2141.1998.00834.x
  21. Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ, Heys JR, Landvatter SW, and et al. (1994) A protein kinase involved in the regulation of inflammatory cytokine biosynthesis. Nature 372: 739-746 https://doi.org/10.1038/372739a0
  22. Li J, Billiar TR, Talanian RV, and Kim YM (1997) Nitric oxide reversibly inhibits seven members of the caspase family via S-nitrosylation. Biochem Biophys Res Commun 240: 419-424 https://doi.org/10.1006/bbrc.1997.7672
  23. Maciejewski JP, Selleri C, Sato T, Cho HJ, Keefer LK, Nathan CF, and Young NS (1995) Nitric oxide suppression of human hematopoiesis in vitro. Contribution to inhibitory action of interferon-gamma and tumor necrosis factor- alpha. J Clin Invest 96: 1085-1092 https://doi.org/10.1172/JCI118094
  24. Mohr S, Stamler JS, and Brune B (1996) Posttranslational modification of glyceraldehyde-3-phosphate dehydrogenase by S-nitrosylation and subsequent NADH attachment. J Biol Chem 271: 4209-4214 https://doi.org/10.1074/jbc.271.8.4209
  25. Nara K, Konno D, Uchida J, Kiuchi Y, and Oguchi K (1999) Protective effect of nitric oxide against iron-induced neuronal damage. J Neural Transm 106: 835-848 https://doi.org/10.1007/s007020050204
  26. Nathan C (1992) Nitric oxide as a secretory product of mammalian cells. FASEB J 6: 3051-3064 https://doi.org/10.1096/fasebj.6.12.1381691
  27. Rauhala P, Lin AM, and Chiueh CC (1998) Neuroprotetion by S-nitrosoglutathione of brain dopamine neurons from oxidative stress. FASEB J 12: 165-173 https://doi.org/10.1096/fasebj.12.2.165
  28. Rauhala P, Mohanakumar KP, Sziraki I, Lin AM, and Chiueh CC (1996) S-nitrosothiols and nitric oxide, but not sodium nitroprusside, protect nigrostriatal dopamine neurons against iron-induced oxidative stress in vivo. Synapse 23: 58-60 https://doi.org/10.1002/(SICI)1098-2396(199605)23:1<58::AID-SYN7>3.0.CO;2-G
  29. Rouse J, Cohen P, Trigon S, Morange M, Alonso-Llamazares A, Zamanillo D, Hunt T, and Nebreda AR (1994) A novel kinase cascade triggered by stress and heat shock that stimulates MAPKAP kinase-2 and phosphorylation of the small heat shock proteins. Cell 78: 1027-1037 https://doi.org/10.1016/0092-8674(94)90277-1
  30. Schwenger P, Bellosta P, Vietor I, Basilico C, Skolnik EY, and Vilcek J (1997) Sodium salicylate induces apoptosis via p38 mitogen-activated protein kinase but inhibits tumor necrosis factor-induced c-Jun N-terminal kinase/stress-activated protein kinase activation. Proc Natl Acad Sci USA 94: 2869-2873 https://doi.org/10.1073/pnas.94.7.2869
  31. Sergent O, Griffon B, Morel I, Chevanne M, Dubos MP, Cillard P, and Cillard J (1997) Effect of nitric oxide on iron-mediated oxidative stress in primary rat hepatocyte culture. Hepatology 25: 122-127 https://doi.org/10.1002/hep.510250123
  32. Terwel D, Nieland LJ, Schutte B, Reutelingsperger CP, Ramaekers FC, and Steinbusch HW (2000) S-nitroso-N-acetylpenicillamine and nitroprusside induce apopotosis in a neuronal cell line by the production of different reactive molecules. Eur J Pharmacol 400: 19-33 https://doi.org/10.1016/S0014-2999(00)00379-4
  33. Toyokuni S (1996) Iron-induced carcinogenesis: the role of redox regulation. Free Radic Biol Med 20: 553-566 https://doi.org/10.1016/0891-5849(95)02111-6
  34. Weinberg ED (1989) Iron, asbestos, and carcinogenicity. Lancet 1: 1399-1400 https://doi.org/10.1016/S0140-6736(89)92857-2
  35. Weinberg ED (1999) Development of clinical methods of iron deprivation for suppression of neoplastic and intectious diseases. Cancer Invest 17: 507-513 https://doi.org/10.3109/07357909909032860
  36. Wink DA, Kasprzak KS, Maragos CM, Elespuru RK, Misra M, Dunams TM, Cebula TA, Koch WH, Andrews AW, Allen JS, and et al. (1991) DNA deaminating ability and genotoxicity of nitric oxide and its progenitors. Science 254: 1001-1003 https://doi.org/10.1126/science.1948068
  37. Xia Y, Dawson VL, Dawson TM, Snyder SH, and Zweier JL (1996) Nitric oxide synthase generates superoxide and nitric oxide in arginine- depleted cells leading to peroxynitrite-mediated cellular injury. Proc Natl Acad Sci USA 93: 6770-6774 https://doi.org/10.1073/pnas.93.13.6770
  38. Yabuki M, Kariya S, Inai Y, Hamazaki K, Yoshioka T, Yasuda T, Horton AA, and Utsumi K (1997) Molecular mechanisms of apoptosis in HL-60 cells induced by a nitric oxide-releasing compound. Free Radic Res 27: 325-335 https://doi.org/10.3109/10715769709065770
  39. Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP, and Wang X (1997) Prevention of apoptosis by Bcl-2: release of cytochrome c from mitochondria blocked. Science 275: 1129-1132 https://doi.org/10.1126/science.275.5303.1129
  40. Zaman K, Ryu H, Hall D, O'Donovan K, Lin KI, Miller MP, Marquis JC, Baraban JM, Semenza GL, and Ratan RR (1999) Protection from oxidative stress-induced apoptosis in cortical neuronal cultures by iron chelators is associated with enhanced DNA binding of hypoxia-inducible factor-1 and ATF-1/CREB and increased expression of glycolytic enzymes, p21 (waf1/cip1), and erythropoietin. J Neurosci 19: 9821-9830