DOI QR코드

DOI QR Code

Propofol protects against oxidative-stress-induced COS-7 cell apoptosis by inducing autophagy

  • Yoon, Ji-Young (Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute) ;
  • Baek, Chul-Woo (Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute) ;
  • Kim, Eun-Jung (Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute) ;
  • Park, Bong-Soo (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Yu, Su-Bin (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Yoon, Ji-Uk (Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University) ;
  • Kim, Eok-Nyun (Department of Dental Anesthesia and Pain Medicine, School of Dentistry, Pusan National University, Dental Research Institute)
  • Received : 2017.01.24
  • Accepted : 2017.03.07
  • Published : 2017.03.30

Abstract

Background: In oxidative stress, reactive oxygen species (ROS) production contributes to cellular dysfunction and initiates the apoptotic cascade. Autophagy is considered the mechanism that decreases ROS concentration and oxidative damage. Propofol shows antioxidant properties, but the mechanisms underlying the effect of propofol preconditioning (PPC) on oxidative injury remain unclear. Therefore, we investigated whether PPC protects against cell damage from hydrogen peroxide ($H_2O_2$)-induced oxidative stress and influences cellular autophagy. Method: COS-7 cells were randomly divided into the following groups: control, cells were incubated in normoxia (5% $CO_2$, 21% $O_2$, and 74% $N_2$) for 24 h without propofol; $H_2O_2$, cells were exposed to $H_2O_2$ ($400{\mu}M$) for 2 h; $PPC+H_2O_2$, cells pretreated with propofol were exposed to $H_2O_2$; and 3-methyladenine $(3-MA)+PPC+H_2O_2$, cells pretreated with 3-MA (1 mM) for 1 h and propofol were exposed to $H_2O_2$. Cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide thiazolyl blue (MTT) reduction. Apoptosis was determined using Hoechst 33342 staining and fluorescence microscopy. The relationship between PPC and autophagy was detected using western blot analysis. Results: Cell viability decreased more significantly in the $H_2O_2$ group than in the control group, but it was improved by PPC ($100{\mu}M$). Pretreatment with propofol effectively decreased $H_2O_2$-induced COS-7 cell apoptosis. However, pretreatment with 3-MA inhibited the protective effect of propofol during apoptosis. Western blot analysis showed that the level of autophagy-related proteins was higher in the $PPC+H_2O_2$ group than that in the $H_2O_2$ group. Conclusion: PPC has a protective effect on $H_2O_2$-induced COS-7 cell apoptosis, which is mediated by autophagy activation.

Keywords

References

  1. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature 2000; 408: 239-47. https://doi.org/10.1038/35041687
  2. Geiszt M, Kopp JB, Varnai P, Leto TL. Identification of renox, an NAD(P)H oxidase in kidney. Proc Natl Acad Sci U S A 2000; 97: 8010-4. https://doi.org/10.1073/pnas.130135897
  3. Boveris A, Chance B. The mitochondrial generation of hydrogen peroxide. general properties and effect of hyperbaric oxygen. Biochem J 1973; 134: 707-16. https://doi.org/10.1042/bj1340707
  4. Chien CT, Lee PH, Chen CF, Ma MC, Lai MK, Hsu SM. De novo demonstration and co-localization of freeradical production and apoptosis formation in rat kidney subjected to ischemia/reperfusion. J Am Soc Nephrol 2001; 12: 973-82.
  5. Filomeni G, De Zio D, Cecconi F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ 2015; 22: 377-88. https://doi.org/10.1038/cdd.2014.150
  6. Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Mol Cell 2010; 40: 280-93. https://doi.org/10.1016/j.molcel.2010.09.023
  7. Hamacher-Brady A, Brady NR, Logue SE, Sayen MR, Jinno M, Kirshenbaum LA, et al. Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy. Cell Death Differ 2007; 14: 146-57. https://doi.org/10.1038/sj.cdd.4401936
  8. Filomeni G, Desideri E, Cardaci S, Rotilio G, Ciriolo MR. Under the ROS...thiol network is the principal suspect for autophagy commitment. Autophagy 2010; 6: 999-1005. https://doi.org/10.4161/auto.6.7.12754
  9. Murphy MP. How mitochondria produce reactive oxygen species. Biochem J 2009; 417: 1-13. https://doi.org/10.1042/BJ20081386
  10. Tesauro M, Thompson WC, Moss J. Effect of staurosporineinduced apoptosis on endothelial nitric oxide synthase in transfected COS-7 cells and primary endothelial cells. Cell Death Differ 2006; 13: 597-606. https://doi.org/10.1038/sj.cdd.4401770
  11. Xu JJ, Wang YL. Propofol attenuation of hydrogen peroxide-mediated oxidative stress and apoptosis in cultured cardiomyocytes involves haeme oxygenase-1. Eur J Anaesthesiol 2008; 25: 395-402. https://doi.org/10.1017/S0265021508003542
  12. Kobayashi K, Yoshino F, Takahashi SS, Todoki K, Maehata Y, Komatsu T, et al. Direct assessments of the antioxidant effects of propofol medium chain triglyceride/long chain triglyceride on the brain of stroke-prone spontaneously hypertensive rats using electron spin resonance spectroscopy. Anesthesiology 2008; 109: 426-35. https://doi.org/10.1097/ALN.0b013e318182a903
  13. Ansley DM, Lee J, Godin DV, Garnett ME, Qayumi AK. Propofol enhances red cell antioxidant capacity in swine and humans. Can J Anaesth 1998; 45: 233-9. https://doi.org/10.1007/BF03012908
  14. Stadnicka A, Marinovic J, Ljubkovic M, Bienengraeber MW, Bosnjak ZJ. Volatile anesthetic-induced cardiac preconditioning. J Anesth 2007; 21: 212-9. https://doi.org/10.1007/s00540-006-0486-6
  15. Liu KX, Rinne T, He W, Wang F, Xia Z. Propofol attenuates intestinal mucosa injury induced by intestinal ischemia-reperfusion in the rat. Can J Anaesth 2007; 54: 366-74. https://doi.org/10.1007/BF03022659
  16. Kamada N, Kanaya N, Hirata N, Kimura S, Namiki A. Cardioprotective effects of propofol in isolated ischemiareperfused guinea pig hearts: Role of KATP channels and GSK-3beta. Can J Anaesth 2008; 55: 595-605. https://doi.org/10.1007/BF03021433
  17. Zaugg M, Lucchinetti E, Uecker M, Pasch T, Schaub MC. Anaesthetics and cardiac preconditioning. part I. signalling and cytoprotective mechanisms. Br J Anaesth 2003; 91: 551-65. https://doi.org/10.1093/bja/aeg205
  18. Das M, Das DK. Molecular mechanism of preconditioning. IUBMB Life 2008; 60: 199-203. https://doi.org/10.1002/iub.31
  19. Assad AR, Delou JM, Fonseca LM, Villela NR, Nascimento JH, Vercosa N, et al. The role of KATP channels on propofol preconditioning in a cellular model of renal ischemia-reperfusion. Anesth Analg 2009; 109: 1486-92. https://doi.org/10.1213/ANE.0b013e3181b76396
  20. Imlay JA, Linn S. DNA damage and oxygen radical toxicity. Science 1988; 240: 1302-9. https://doi.org/10.1126/science.3287616
  21. Navarro A, Boveris A. The mitochondrial energy transduction system and the aging process. Am J Physiol Cell Physiol 2007; 292: C670-86. https://doi.org/10.1152/ajpcell.00213.2006
  22. Cooke MS, Evans MD, Dizdaroglu M, Lunec J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J 2003; 17: 1195-214. https://doi.org/10.1096/fj.02-0752rev
  23. Cadet J, Delatour T, Douki T, Gasparutto D, Pouget JP, Ravanat JL, et al. Hydroxyl radicals and DNA base damage. Mutat Res 1999; 424: 9-21. https://doi.org/10.1016/S0027-5107(99)00004-4
  24. Wang H, Xue Z, Wang Q, Feng X, Shen Z. Propofol protects hepatic L02 cells from hydrogen peroxide-induced apoptosis via activation of extracellular signal-regulated kinases pathway. Anesth Analg 2008; 107: 534-40. https://doi.org/10.1213/ane.0b013e3181770be9
  25. Wu XJ, Zheng YJ, Cui YY, Zhu L, Lu Y, Chen HZ. Propofol attenuates oxidative stress-induced PC12 cell injury via p38 MAP kinase dependent pathway. Acta Pharmacol Sin 2007; 28: 1123-8. https://doi.org/10.1111/j.1745-7254.2007.00610.x
  26. Li Volti G, Basile F, Murabito P, Galvano F, Di Giacomo C, Gazzolo D, et al. Antioxidant properties of anesthetics: The biochemist, the surgeon and the anesthetist. Clin Ter 2008; 159: 463-9.
  27. Li Volti G, Sorrenti V, Murabito P, Galvano F, Veroux M, Gullo A, et al. Pharmacological induction of heme oxygenase-1 inhibits iNOS and oxidative stress in renal ischemia-reperfusion injury. Transplant Proc 2007; 39: 2986-91. https://doi.org/10.1016/j.transproceed.2007.09.047
  28. Scapagnini G, Foresti R, Calabrese V, Giuffrida Stella AM, Green CJ, Motterlini R. Caffeic acid phenethyl ester and curcumin: A novel class of heme oxygenase-1 inducers. Mol Pharmacol 2002; 61: 554-61. https://doi.org/10.1124/mol.61.3.554
  29. Acquaviva R, Campisi A, Murabito P, Raciti G, Avola R, Mangiameli S, et al. Propofol attenuates peroxynitritemediated DNA damage and apoptosis in cultured astrocytes: An alternative protective mechanism. Anesthesiology 2004; 101: 1363-71.
  30. Hayashi K, Dan K, Goto F, Tshuchihashi N, Nomura Y, Fujioka M, et al. The autophagy pathway maintained signaling crosstalk with the Keap1-Nrf2 system through p62 in auditory cells under oxidative stress. Cell Signal 2015; 27: 382-93. https://doi.org/10.1016/j.cellsig.2014.11.024
  31. Huang Q, Wu YT, Tan HL, Ong CN, Shen HM. A novel function of poly(ADP-ribose) polymerase-1 in modulation of autophagy and necrosis under oxidative stress. Cell Death Differ 2009; 16: 264-77. https://doi.org/10.1038/cdd.2008.151
  32. Huang Q, Shen HM. To die or to live: The dual role of poly(ADP-ribose) polymerase-1 in autophagy and necrosis under oxidative stress and DNA damage. Autophagy 2009; 5: 273-6. https://doi.org/10.4161/auto.5.2.7640
  33. Glick D, Barth S, Macleod KF. Autophagy: Cellular and molecular mechanisms. J Pathol 2010; 221: 3-12. https://doi.org/10.1002/path.2697
  34. Mori F, Tanji K, Odagiri S, Toyoshima Y, Yoshida M, Kakita A, et al. Autophagy-related proteins (p62, NBR1 and LC3) in intranuclear inclusions in neurodegenerative diseases. Neurosci Lett 2012; 522: 134-8. https://doi.org/10.1016/j.neulet.2012.06.026
  35. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, et al. The role of autophagy during the early neonatal starvation period. Nature 2004; 432: 1032-6. https://doi.org/10.1038/nature03029
  36. Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 2005; 169: 425-34. https://doi.org/10.1083/jcb.200412022
  37. Nishida Y, Arakawa S, Fujitani K, Yamaguchi H, Mizuta T, Kanaseki T, et al. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 2009; 461: 654-8. https://doi.org/10.1038/nature08455

Cited by

  1. The protective effect of propofol on ionizing radiation-induced hematopoietic system damage in mice vol.9, pp.62, 2017, https://doi.org/10.1039/c9ra07262d
  2. Unusual mtDNA Control Region Length Heteroplasmy in the COS-7 Cell Line vol.11, pp.6, 2020, https://doi.org/10.3390/genes11060607
  3. Propofol protects against oxygen/glucose deprivation-induced cell injury via gap junction inhibition in astrocytes vol.22, pp.4, 2017, https://doi.org/10.3892/mmr.2020.11357