J Dent Anesth Pain Med.  2016 Sep;16(3):175-184. 10.17245/jdapm.2016.16.3.175.

Remifentanil induces autophagy and prevents hydrogen peroxide-induced apoptosis in Cos-7 cells

Affiliations
  • 1Department of Dental Anesthesia and Pain Medicine, Pusan National University Dental Hospital, Yangsan, Korea. wjdental@hanmail.net
  • 2Department of Anesthesia and Pain Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea.

Abstract

BACKGROUND
This study investigated the effect of remifentanil pretreatment on Cos-7 cells exposed to oxidative stress, and the influence of remifentanil on intracellular autophagy and apoptotic cell death.
METHODS
Cells were divided into 4 groups: (1) Control: non-pretreated cells were incubated in normoxia (5% COâ‚‚, 21% Oâ‚‚, and 74% Nâ‚‚). (2) Hâ‚‚Oâ‚‚: non-pretreated cells were exposed to Hâ‚‚Oâ‚‚ for 24 h. (3) RPC+Hâ‚‚Oâ‚‚: cells pretreated with remifentanil were exposed to Hâ‚‚Oâ‚‚ for 24 h. (4) 3-MA+RPC+Hâ‚‚Oâ‚‚: cells pretreated with 3-Methyladenine (3-MA) and remifentanil were exposed to Hâ‚‚Oâ‚‚ for 24 h. We determined the cell viability of each group using an MTT assay. Hoechst staining and FACS analysis of Cos-7 cells were performed to observe the effect of remifentanil on apoptosis. Autophagy activation was determined by fluorescence microscopy, MDC staining, and AO staining. The expression of autophagy-related proteins was observed using western blotting.
RESULTS
Remifentanil pretreatment increased the viability of Cos-7 cells exposed to oxidative stress. Hoechst staining and FACS analysis revealed that oxidative stress-dependent apoptosis was suppressed by the pretreatment. Additionally, fluorescence microscopy showed that remifentanil pretreatment led to autophagy-induction in Cos-7 cells, and the expression of autophagy-related proteins was increased in the RPC+Hâ‚‚Oâ‚‚ group.
CONCLUSIONS
The study showed that remifentanil pretreatment stimulated autophagy and increased viability in an oxidative stress model of Cos-7 cells. Therefore, we suggest that apoptosis was activated upon oxidative stress, and remifentanil preconditioning increased the survival rate of the cells by activating autophagy.

Keyword

Apoptosis; Autophagy; Cos-7 cells; Oxidative stress; Remifentanil

MeSH Terms

Animals
Apoptosis*
Autophagy*
Blotting, Western
Cell Death
Cell Survival
COS Cells*
Hydrogen*
Microscopy, Fluorescence
Oxidative Stress
Survival Rate
Hydrogen

Figure

  • Fig. 1 Cell viability of Cos-7 cells assessed by MTT assay. (A) The effect of remifentanil and 3-MA + remifentanil preconditioning on Cos-7 cell viability at various concentrations (0, 0.1, 0.5, 1, 2 ng/ml) without exposure to H2O2, (B) Cell viability of Cos-7 cells upon exposure to various concentrations (0, 50, 100, 200, 400) of H2O2.

  • Fig. 2 The effect of remifentanil pretreatment on Cos-7 cell viability with or without autophagy inhibition, as examined by MTT assay. *P < 0.05 as compared with control group. Control: no H2O2, remifentanil treatment group, H2O2: hydrogen peroxide exposure only, RPC + H2O2: remifentanil pretreatment group plus hydrogen peroxide exposure, 3-MA + RPC + H2O2: pretreatment with 3-MA and remifentanil plus hydrogen peroxide exposure.

  • Fig. 3 Hoechst staining: Morphological changes in Cos-7 cells upon pretreatment with remifentanil alone or in combination with 3-MA before exposure to oxidative stress. Cos-7 cells were pretreated with remifentanil or remifentanil and 3-MA, then exposed to oxidative stress. Cells were stained with Hoechst stain and observed by fluorescence microscopy. Apoptotic bodies were seen in H2O2 and 3-MA + RPC + H2O2 group cells. In contrast, they were remarkably reduced in RPC + H2O2 group cells. Control: no H2O2, remifentanil treatment group, H2O2: hydrogen peroxide exposure only, RPC + H2O2: remifentanil pretreatment group plus hydrogen peroxide exposure, 3-MA + RPC + H2O2: pretreatment with 3-MA and remifentanil plus hydrogen peroxide exposure.

  • Fig. 4 Fluorescence activated cell sorting (FACS) analysis of Cos-7 cells to determine apoptotic cell populations of each experimental group. Control: no H2O2, remifentanil treatment group, H2O2: hydrogen peroxide exposure only, RPC + H2O2: remifentanil pretreatment group plus hydrogen peroxide exposure, 3-MA + RPC + H2O2: pretreatment with 3-MA and remifentanil plus hydrogen peroxide exposure.

  • Fig. 5 Fluorescence microscopic analysis of autophagosomes in Cos-7 cells under oxidative stress to observe autophagy activation upon different pretreatment conditions. (A) MDC staining of cytoplasmic vacuoles after preconditioning with remifentanil in Cos-7 cells followed by exposure to oxidative stress. Cellular oxidative stress resulted in greater accumulation of autophagosomes containing partially digested cytoplasmic contents compared to the control group. The remifentanil pretreatment group showed increased formation of autophagosomes. Compared with the RPC + H2O2 group, the autophagy pathway inhibitor, 3-MA, blocked formation of autophagosomes. Magnification: 400 ×, (B) AO staining of autophagosomes after remifentanil pretreatment in Cos-7 cells followed by exposure to oxidative stress. AO staining indicated by red fluorescent spots appeared in RPC + H2O2 group cells, while the control, H2O2, and 3-MA + RPC + H2O2 groups showed mainly green cytoplasmic fluorescence. The green shows where the dye has stained the nucleus and the red indicates autophagosome formation. Magnification: 400 ×. Control: no H2O2, remifentanil treatment group, H2O2: hydrogen peroxide exposure only, RPC + H2O2: remifentanil pretreatment group plus hydrogen peroxide exposure, 3-MA + RPC + H2O2: pretreatment with 3-MA and remifentanil plus hydrogen peroxide exposure.

  • Fig. 6 Western blot analysis and densitometry to determine the effects of remifentanil treatment ATG5, Beclin-1, LC3-II, and p62 protein levels in each experimental group of Cos-7 cells. In RPC + H2O2 groups, cellular expression of ATG5, Beclin-1, LC3-II and p62 were upregulated, while these proteins were downregulated in 3-MA+RPC+H2O2 groups. Control: no H2O2, remifentanil treatment group, H2O2: hydrogen peroxide exposure only, RPC+H2O2: remifentanil pretreatment group plus hydrogen peroxide exposure, 3-MA+RPC+H2O2: pretreatment with 3-MA and remifentanil plus hydrogen peroxide exposure. *P < 0.05 as compared with the control group.


Reference

1. Elahi MM, Kong YX, Matata BM. Oxidative stress as a mediator of cardiovascular disease. Oxid Med Cell Longev. 2009; 2:259–269.
Article
2. Droge W. Free radicals in the physiological control of cell function. Physiol Rev. 2002; 82:47–95.
Article
3. Halliwell B. Reactive species and antioxidants. Redox biology is a fundamental theme of aerobic life. Plant Physiol. 2006; 141:312–322.
Article
4. Valko M, Rhodes C, Moncol J, Izakovic M, Mazur M. Free radicals, metals and antioxidants in oxidative stress-induced cancer. Chem Biol Interact. 2006; 160:1–40.
Article
5. Zhao Y, Zhao B. Oxidative stress and the pathogenesis of alzheimer's disease. Oxid Med Cell Longev. 2013; 2013:316523.
Article
6. Singh DK, Winocour P, Farrington K. Oxidative stress in early diabetic nephropathy: fueling the fire. Nat Rev Endocrinol. 2011; 7:176–184.
Article
7. Giordano FJ. Oxygen, oxidative stress, hypoxia, and heart failure. J Clin Invest. 2005; 115:500–508.
Article
8. Ryter SW, Kim HP, Hoetzel A, Park JW, Nakahira K, Wang X, et al. Mechanisms of cell death in oxidative stress. Antioxid Redox Signal. 2007; 9:49–89.
9. Mizushima N. The pleiotropic role of autophagy: From protein metabolism to bactericide. Cell Death Differ. 2005; 12:1535–1541.
10. Guan X, Qian Y, Shen Y, Zhang L, Du Y, Dai H, et al. Autophagy protects renal tubular cells against ischemia/reperfusion injury in a time-dependent manner. Cell Physiol Biochem. 2015; 36:285–298.
Article
11. Kim YH, Kang JM, Kim IR, Lee BY, Yoon JY, Kim CH, et al. Protective effect of propofol against hypoxia-reoxygenation injury in HaCaT human keratinocytes. Int J Oral Biol. 2014; 39:97–105.
Article
12. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem Sci. 2011; 36:30–38.
Article
13. Kwon JY, Park BS, Kim YH, Kim YD, Kim CH, Yoon JY, et al. Remifentanil protects human keratinocytes against hypoxia-reoxygenation injury through activation of autophagy. PloS One. 2015; 10:e0116982.
Article
14. Scott LJ, Perry CM. Spotlight on remifentanil for general anaesthesia. CNS drugs. 2005; 19:1069–1074.
Article
15. Komatsu R, Turan A, Orhan-Sungur M, McGuire J, Radke O, Apfel C. Remifentanil for general anaesthesia: A systematic review. Anaesthesia. 2007; 62:1266–1280.
Article
16. Marsh D, Hodkinson B. Remifentanil in paediatric anaesthetic practice. Anaesthesia. 2009; 64:301–308.
Article
17. Cho SS, Rudloff I, Berger PJ, Irwin MG, Nold MF, Cheng W, et al. Remifentanil ameliorates intestinal ischemia-reperfusion injury. BMC Gastroenterol. 2013; 13:69.
Article
18. Zhang Y, Irwin MG, Wong TM, Chen M, Cao CM. Remifentanil preconditioning confers cardioprotection via cardiac kappa- and delta-opioid receptors. Anesthesiology. 2005; 102:371–378.
Article
19. Kim H, Cho J, Hong S, Kim S, Shim J, Kwak Y. Remifentanil protects myocardium through activation of anti-apoptotic pathways of survival in ischemia-reperfused rat heart. Physiol Res. 2010; 59:347–356.
Article
20. Zhang TZ, Zhou J, Jin Q, Sun YJ, Diao YG, Zhang YN, et al. Protective effects of remifentanil preconditioning on cerebral injury during pump-assisted coronary artery bypass graft. Genet Mol Res. 2014; 13:7658–7665.
Article
21. Chang C, Chen P, Lu S, Hsieh M, Lin C, Lee H, et al. Propofol-enhanced autophagy increases motility and angiogenic capacity of cultured human umbilical vascular endothelial cells. Life Sci. 2015; 142:49–59.
Article
22. Anding AL, Baehrecke EH. Chapter three-autophagy in cell life and cell death. Curr Top Dev Biol. 2015; 114:67–91.
Article
23. Baehrecke EH. Autophagy: Dual roles in life and death? Nat Rev Mol Cell Biol. 2005; 6:505–510.
Article
24. Fitzwalter BE, Thorburn A. Recent insights into cell death and autophagy. FEBS J. 2015; 282:4279–4288.
Article
25. Levine B, Yuan J. Autophagy in cell death: An innocent convict? J Clin Invest. 2005; 115:2679–2688.
Article
26. Huang C, Lee C, Lin H, Chen M, Lin C, Chang J. Autophagy-regulated ROS from xanthine oxidase acts as an early effector for triggering late mitochondria-dependent apoptosis in cathepsin S-targeted tumor cells. PloS one. 2015; 10:e0128045.
Article
27. Elahi M, Matata B. Free radicals in blood: Evolving concepts in the mechanism of ischemic heart disease. Arch Biochem Biophys. 2006; 450:78–88.
Article
28. Finkel T, Holbrook NJ. Oxidants, oxidative stress and the biology of ageing. Nature. 2000; 408:239–247.
Article
29. Yue Z, Friedman L, Komatsu M, Tanaka K. The cellular pathways of neuronal autophagy and their implication in neurodegenerative diseases. Biochim Biophys Acta. 2009; 1793:1496–1507.
Article
30. Yan L, Sadoshima J, Vatner DE, Vatner SF. Autophagy: A novel protective mechanism in chronic ischemia. Cell Cycle. 2006; 5:1175–1177.
Article
31. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: Physiology and pathology. Trends Biochem Sci. 2011; 36:30–38.
Article
32. Zhao G, Shen X, Nan H, Yan L, Zhao H, Yu J, et al. Remifentanil protects liver against ischemia/reperfusion injury through activation of anti-apoptotic pathways. J Surg Res. 2013; 183:827–834.
Article
33. Peart JN, Gross ER, Gross GJ. Opioid-induced preconditioning: Recent advances and future perspectives. Vascul Pharmacol. 2005; 42:211–218.
Article
34. Headrick JP, See Hoe LE, Du Toit EF, Peart JN. Opioid receptors and cardioprotection–'opioidergic conditioning' of the heart. Br J Pharmacol. 2015; 172:2026–2050.
Article
35. Costa-Malaquias A, Almeida MB, Monteiro JRS, de Matos Macchi B, do Nascimento JLM, Crespo-Lopez ME. Morphine protects against methylmercury intoxication: A role for opioid receptors in oxidative stress? PloS One. 2014; 9:e110815.
Article
Full Text Links
  • JDAPM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr