DOI QR코드

DOI QR Code

Nitric Oxide-Induced Apoptosis of Human Dental Pulp Cells Is Mediated by the Mitochondria-Dependent Pathway

  • Park, Min Young (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University) ;
  • Jeong, Yeon Jin (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University) ;
  • Kang, Gi Chang (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University) ;
  • Kim, Mi-Hwa (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University) ;
  • Kim, Sun Hun (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Anatomy, School of Dentistry, Chonnam National University) ;
  • Chung, Hyun-Ju (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Periodontology, School of Dentistry, Chonnam National University) ;
  • Jung, Ji Yeon (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University) ;
  • Kim, Won Jae (Dental Science Research Institute and Medical Research Center for Biomineralization Disorders, Department of Oral Physiology, School of Dentistry, Chonnam National University)
  • Received : 2013.09.11
  • Accepted : 2014.01.08
  • Published : 2014.02.28

Abstract

Nitric oxide (NO) is recognized as a mediator and regulator of inflammatory responses. NO is produced by nitric oxide synthase (NOS), and NOS is abundantly expressed in the human dental pulp cells (HDPCs). NO produced by NOS can be cytotoxic at higher concentrations to HDPCs. However, the mechanism by which this cytotoxic pathway is activated in cells exposed to NO is not known. The purpose of this study was to elucidate the NO-induced cytotoxic mechanism in HDPCs. Sodium nitroprusside (SNP), a NO donor, reduced the viability of HDPCs in a dose- and time-dependent manner. We investigated the in vitro effects of nitric oxide on apoptosis of cultured HDPCs. Cells showed typical apoptotic morphology after exposure to SNP. Besides, the number of Annexin V positive cells was increased among the SNP-treated HDPCs. SNP enhanced the production of reactive oxygen species (ROS), and N-acetylcysteine (NAC) ameliorated the decrement of cell viability induced by SNP. However, a soluble guanylate cyclase inhibitor (ODQ) did not inhibited the decrement of cell viability induced by SNP. SNP increased cytochrome c release from the mitochondria to the cytosol and the ratio of Bax/Bcl-2 expression levels. Moreover, SNP-treated HDPCs elevated activities of caspase-3 and caspase-9. While pretreatment with inhibitors of caspase (z-VAD-fmk, z-DEVD-fmk) reversed the NO-induced apoptosis of HDPCs. From these results, it can be suggested that NO induces apoptosis of HDPCs through the mitochondria-dependent pathway mediated by ROS and Bcl-2 family, but not by the cyclic GMP pathway.

Keywords

References

  1. Massey WL, Romberg DM, Hunter N, Hume WR. The association of carious dentin microflora with tissue changes in human pulpitis. Oral Microbiol Immunol. 1993;8:30-35. https://doi.org/10.1111/j.1399-302X.1993.tb00539.x
  2. Ohnishi T, Daikuhara Y. Hepatocyte growth factor/scatter factor in development, inflammation and carcinogenesis: its expression and role in oral tissues. Arch Oral Biol. 2003;48:797-804. https://doi.org/10.1016/S0003-9969(03)00180-8
  3. Carr AC, McCall MR, Frei B. Oxidation of LDL by myeloperoxidase and reactive nitrogen species: reaction pathways and antioxidant protection. Arterioscler Thromb Vasc Biol. 2000;20:1716-1723. https://doi.org/10.1161/01.ATV.20.7.1716
  4. Knowles RG. Nitric oxide synthases. Biochem Soc Trans. 1996;24:875-878. https://doi.org/10.1042/bst0240875
  5. Förstermann U, Closs EI, Pollock JS, Nakane M, Schwarz P, Gath I, Kleinert H. Nitric oxide synthase isozymes. Characterization, purification, molecular cloning, and functions. Hypertension. 1994;23:1121-1131. https://doi.org/10.1161/01.HYP.23.6.1121
  6. Korhonen R, Lahti A, Kankaanranta H, Moilanen E. Nitric oxide production and signaling in inflammation. Curr Drug Targets Inflamm Allergy. 2005;4:471-479. https://doi.org/10.2174/1568010054526359
  7. Boissel JP, Schwarz PM, Förstermann U. Neuronal-type NO synthase: transcript diversity and expressional regulation. Nitric Oxide. 1998;2:337-349. https://doi.org/10.1006/niox.1998.0189
  8. Shaul PW. Regulation of endothelial nitric oxide synthase: location, location, location. Annu Rev Physiol. 2002;64:749-774. https://doi.org/10.1146/annurev.physiol.64.081501.155952
  9. Shin W, Cuong TD, Lee JH, Min B, Jeon BH, Lim HK, Ryoo S. Arginase Inhibition by Ethylacetate Extract of Caesalpinia sappan Lignum Contributes to Activation of Endothelial Nitric Oxide Synthase. Korean J Physiol Pharmacol. 2011;15:123-128. https://doi.org/10.4196/kjpp.2011.15.3.123
  10. Lohinai Z, Székely AD, Benedek P, Csillag A. Nitric oxide synthase containing nerves in the cat and dog dental pulp and gingiva. Neurosci Lett. 1997;227:91-94. https://doi.org/10.1016/S0304-3940(97)00319-4
  11. Felaco M, Di Maio FD, De Fazio P, D'Arcangelo C, De Lutiis MA, Varvara G, Grilli A, Barbacane RC, Reale M, Conti P. Localization of the e-NOS enzyme in endothelial cells and odontoblasts of healthy human dental pulp. Life Sci. 2000;68:297-306. https://doi.org/10.1016/S0024-3205(00)00935-8
  12. Mei YF, Yamaza T, Atsuta I, Danjo A, Yamashita Y, Kido MA, Goto M, Akamine A, Tanaka T. Sequential expression of endothelial nitric oxide synthase, inducible nitric oxide synthase, and nitrotyrosine in odontoblasts and pulp cells during dentin repair after tooth preparation in rat molars. Cell Tissue Res. 2007;328:117-127. https://doi.org/10.1007/s00441-005-0003-5
  13. Di Nardo Di Maio F, Lohinai Z, D'Arcangelo C, De Fazio PE, Speranza L, De Lutiis MA, Patruno A, Grilli A, Felaco M. Nitric oxide synthase in healthy and inflamed human dental pulp. J Dent Res. 2004;83:312-316. https://doi.org/10.1177/154405910408300408
  14. da Silva LP, Issa JP, Del Bel EA. Action of nitric oxide on healthy and inflamed human dental pulp tissue. Micron. 2008;39:797-801. https://doi.org/10.1016/j.micron.2008.01.018
  15. Kawanishi HN, Kawashima N, Suzuki N, Suda H, Takagi M. Effects of an inducible nitric oxide synthase inhibitor on experimentally induced rat pulpitis. Eur J Oral Sci. 2004;112:332-337. https://doi.org/10.1111/j.1600-0722.2004.00139.x
  16. Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M, Krammer PH, Peter ME. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995;14:5579-5588.
  17. Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell. 1998;94:491-501. https://doi.org/10.1016/S0092-8674(00)81590-1
  18. Riedl SJ, Salvesen GS. The apoptosome: signalling platform of cell death. Nat Rev Mol Cell Biol. 2007;8:405-413. https://doi.org/10.1038/nrm2153
  19. Boulares AH, Zoltoski AJ, Sherif ZA, Yakovlev A, Smulson ME. Roles of DNA fragmentation factor and poly(ADP-ribose) polymerase-1 in sensitization of fibroblasts to tumor necrosis factor-induced apoptosis. Biochem Biophys Res Commun. 2002;290:796-801. https://doi.org/10.1006/bbrc.2001.6280
  20. Kim YM, Bombeck CA, Billiar TR. Nitric oxide as a bifunctional regulator of apoptosis. Circ Res. 1999;84:253-256. https://doi.org/10.1161/01.RES.84.3.253
  21. Dimmeler S, Haendeler J, Nehls M, Zeiher AM. Suppression of apoptosis by nitric oxide via inhibition of interleukin-1betaconverting enzyme (ICE)-like and cysteine protease protein (CPP)-32-like proteases. J Exp Med. 1997;185:601-607. https://doi.org/10.1084/jem.185.4.601
  22. Kim YM, de Vera ME, Watkins SC, Billiar TR. Nitric oxide protects cultured rat hepatocytes from tumor necrosis factor-alpha-induced apoptosis by inducing heat shock protein 70 expression. J Biol Chem. 1997;272:1402-1411. https://doi.org/10.1074/jbc.272.2.1402
  23. Hebestreit H, Dibbert B, Balatti I, Braun D, Schapowal A, Blaser K, Simon HU. Disruption of fas receptor signaling by nitric oxide in eosinophils. J Exp Med. 1998;187:415-425. https://doi.org/10.1084/jem.187.3.415
  24. Genaro AM, Hortelano S, Alvarez A, Martínez C, Boscá L. Splenic B lymphocyte programmed cell death is prevented by nitric oxide release through mechanisms involving sustained Bcl-2 levels. J Clin Invest. 1995;95:1884-1890. https://doi.org/10.1172/JCI117869
  25. Lincoln TM, Cornwell TL, Komalavilas P, Boerth N. Cyclic GMP-dependent protein kinase in nitric oxide signaling. Methods Enzymol. 1996;269:149-166. https://doi.org/10.1016/S0076-6879(96)69017-X
  26. Albina JE, Cui S, Mateo RB, Reichner JS. Nitric oxide- mediated apoptosis in murine peritoneal macrophages. J Immunol. 1993;150:5080-5085.
  27. Heneka MT, Löschmann PA, Gleichmann M, Weller M, Schulz JB, Wüllner U, Klockgether T. Induction of nitric oxide synthase and nitric oxide-mediated apoptosis in neuronal PC12 cells after stimulation with tumor necrosis factor-alpha/lipopolysaccharide. J Neurochem. 1998;71:88-94.
  28. Gross SS, Wolin MS. Nitric oxide: pathophysiological mechanisms. Annu Rev Physiol. 1995;57:737-769. https://doi.org/10.1146/annurev.ph.57.030195.003513
  29. Dawson VL, Dawson TM. Nitric oxide neurotoxicity. J Chem Neuroanat. 1996;10:179-190. https://doi.org/10.1016/0891-0618(96)00148-2
  30. Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell. 1996;86:147-157. https://doi.org/10.1016/S0092-8674(00)80085-9
  31. Jun CD, Pae HO, Kwak HJ, Yoo JC, Choi BM, Oh CD, Chun JS, Paik SG, Park YH, Chung HT. Modulation of nitric oxide-induced apoptotic death of HL-60 cells by protein kinase C and protein kinase A through mitogen-activated protein kinases and CPP32-like protease pathways. Cell Immunol. 1999;194:36-46. https://doi.org/10.1006/cimm.1999.1480
  32. Kluck RM, Bossy-Wetzel E, Green DR, Newmeyer DD. The release of cytochrome c from mitochondria: a primary site for Bcl-2 regulation of apoptosis. Science. 1997;275:1132-1136. https://doi.org/10.1126/science.275.5303.1132
  33. Gottlieb E, Vander Heiden MG, Thompson CB. Bcl-x(L) prevents the initial decrease in mitochondrial membrane potential and subsequent reactive oxygen species production during tumor necrosis factor alpha-induced apoptosis. Mol Cell Biol. 2000;20:5680-5689. https://doi.org/10.1128/MCB.20.15.5680-5689.2000
  34. Howard S, Bottino C, Brooke S, Cheng E, Giffard RG, Sapolsky R. Neuroprotective effects of bcl-2 overexpression in hippocampal cultures: interactions with pathways of oxidative damage. J Neurochem. 2002;83:914-923. https://doi.org/10.1046/j.1471-4159.2002.01198.x
  35. Hemish J, Nakaya N, Mittal V, Enikolopov G. Nitric oxide activates diverse signaling pathways to regulate gene expression. J Biol Chem. 2003;278:42321-42329. https://doi.org/10.1074/jbc.M308192200
  36. Brüne B, von Knethen A, Sandau KB. Nitric oxide and its role in apoptosis. Eur J Pharmacol. 1998;351:261-272. https://doi.org/10.1016/S0014-2999(98)00274-X
  37. López-Farré A, Rodríguez-Feo JA, Sánchez de Miguel L, Rico L, Casado S. Role of nitric oxide in the control of apoptosis in the microvasculature. Int J Biochem Cell Biol. 1998;30:1095-1106. https://doi.org/10.1016/S1357-2725(98)00071-5
  38. Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995;80:293-299. https://doi.org/10.1016/0092-8674(95)90412-3
  39. Brown GC. Nitric oxide and mitochondrial respiration. Biochim Biophys Acta. 1999;1411:351-369. https://doi.org/10.1016/S0005-2728(99)00025-0
  40. Fleury C, Mignotte B, Vayssière JL. Mitochondrial reactive oxygen species in cell death signaling. Biochimie. 2002;84:131-141. https://doi.org/10.1016/S0300-9084(02)01369-X
  41. Herrera B, Alvarez AM, Sánchez A, Fernández M, Roncero C, Benito M, Fabregat I. Reactive oxygen species (ROS) mediates the mitochondrial-dependent apoptosis induced by transforming growth factor (beta) in fetal hepatocytes. FASEB J. 2001;15:741-751. https://doi.org/10.1096/fj.00-0267com
  42. Haunstetter A, Izumo S. Apoptosis: basic mechanisms and implications for cardiovascular disease. Circ Res. 1998;82:1111-1129. https://doi.org/10.1161/01.RES.82.11.1111
  43. Green DR, Reed JC. Mitochondria and apoptosis. Science. 1998;281:1309-1312. https://doi.org/10.1126/science.281.5381.1309
  44. Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem. 1999;68:383-424. https://doi.org/10.1146/annurev.biochem.68.1.383
  45. Bal-Price A, Brown GC. Nitric-oxide-induced necrosis and apoptosis in PC12 cells mediated by mitochondria. J Neurochem. 2000;75:1455-1464.
  46. Taimor G, Hofstaetter B, Piper HM. Apoptosis induction by nitric oxide in adult cardiomyocytes via cGMP-signaling and its impairment after simulated ischemia. Cardiovasc Res. 2000;45:588-594. https://doi.org/10.1016/S0008-6363(99)00272-2
  47. Leist M, Single B, Naumann H, Fava E, Simon B, Kühnle S, Nicotera P. Nitric oxide inhibits execution of apoptosis at two distinct ATP-dependent steps upstream and downstream of mitochondrial cytochrome c release. Biochem Biophys Res Commun. 1999;258:215-221. https://doi.org/10.1006/bbrc.1999.0491
  48. Murphy MP. Nitric oxide and cell death. Biochim Biophys Acta. 1999;1411:401-414. https://doi.org/10.1016/S0005-2728(99)00029-8
  49. Yuyama K, Yamamoto H, Nishizaki I, Kato T, Sora I, Yamamoto T. Caspase-independent cell death by low concentrations of nitric oxide in PC12 cells: involvement of cytochrome C oxidase inhibition and the production of reactive oxygen species in mitochondria. J Neurosci Res. 2003;73:351-363. https://doi.org/10.1002/jnr.10669

Cited by

  1. Protective Roles of Selenium on Nitric Oxide-Mediated Apoptosis of Immune Organs Induced by Cadmium in Chickens vol.159, pp.1, 2014, https://doi.org/10.1007/s12011-014-0007-7
  2. PEG-liposomal oxaliplatin combined with nuclear factor-κB inhibitor (PDTC) induces apoptosis in human colorectal cancer cells vol.32, pp.4, 2014, https://doi.org/10.3892/or.2014.3336
  3. Cape Gooseberry [Physalis peruvianaL.] Calyces Ameliorate TNBS Acid-induced Colitis in Rats vol.9, pp.11, 2015, https://doi.org/10.1093/ecco-jcc/jjv132
  4. Mitochondrial oxidative stress-induced hepatocyte apoptosis reflects increased molybdenum intake in caprine vol.170, pp.1, 2014, https://doi.org/10.1007/s12011-015-0450-0
  5. Oxidative Stress and Cell Apoptosis in Caprine Liver Induced by Molybdenum and Cadmium in Combination vol.173, pp.1, 2014, https://doi.org/10.1007/s12011-016-0633-3
  6. Ultrasonication processed Panax ginseng berry extract induces apoptosis through an intrinsic apoptosis pathway in HepG2 cells vol.39, pp.6, 2014, https://doi.org/10.1007/s12272-016-0760-6
  7. Biological Activities and Potential Oral Applications of N-Acetylcysteine: Progress and Prospects vol.2018, pp.None, 2014, https://doi.org/10.1155/2018/2835787
  8. Acute Toxoplasma gondii infection alters the number of neurons and the proportion of enteric glial cells in the duodenum in Wistar rats vol.31, pp.3, 2014, https://doi.org/10.1111/nmo.13523
  9. Nitric Oxide in Dental Pulp Tissue: From Molecular Understanding to Clinical Application in Regenerative Endodontic Procedures vol.26, pp.4, 2014, https://doi.org/10.1089/ten.teb.2019.0316
  10. Synthesis, characterization, biological activity and electrochemistry studies of iron(III) complex with curcumin‐oxime ligand vol.47, pp.11, 2014, https://doi.org/10.1111/1440-1681.13359
  11. Physiopathology of nitric oxide in the oral environment and its biotechnological potential for new oral treatments: a literature review vol.24, pp.12, 2020, https://doi.org/10.1007/s00784-020-03629-2
  12. Antioxidant activities of inula viscosa extract and curcumin on U87 cells induced by beta-amyloid vol.46, pp.2, 2014, https://doi.org/10.17826/cumj.854213