DOI QR코드

DOI QR Code

Protective Effect of Propofol against Hypoxia-reoxygenation Injury in HaCaT Human Keratinocytes

  • Kim, Yong-Ho (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Kang, Jin-Mo (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Kim, In-Ryoung (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Lee, Bo-Young (Department of Oral Anatomy, School of Dentistry, Pusan National University) ;
  • Yoon, Ji-Young (Department of Anesthesia and Pain Medicine, School of Dentistry, Pusan National University) ;
  • Kim, Cheul-Hong (Department of Anesthesia and Pain Medicine, School of Dentistry, Pusan National University) ;
  • Park, Bong-Soo (Department of Oral Anatomy, School of Dentistry, Pusan National University)
  • 투고 : 2014.03.10
  • 심사 : 2014.06.11
  • 발행 : 2014.06.30

초록

The aim of this study was to determine the beneficial effect of propofol on human keratinocytes that have undergone hypoxia reoxygenation (H/R) injury and to investigate whether autophagy is associated with the protective mechanism. Thus, we evaluated how propofol influences the intracellular autophagy and apoptosis during the H/R process in the HaCaT cells. The cultured human keratinocyte cells were exposed to 24 h of hypoxia (5% $CO_2$, 1% $O_2$, 94% $N_2$) followed by 12 h of reoxygenation (5% $CO_2$, 21% $O_2$, 74% $N_2$). The experiment was divided into 4 groups: (1) Control=Normoxia ; (2) H/R=Hypoxia Reoxygenation ; (3) PPC+H/R=Propofol Preconditioning+Hypoxia Reoxygenation; (4) 3-MA+PPC+ H/R=3-MA-Methyladenine+Propofol Preconditioning+ Hypoxia Reoxygenation. In addition, Western blot analysis was performed to identify the expression of apoptotic pathway parameters, including Bcl-2, Bax, and caspase 3 involved in mitochondrial-dependent pathway. Autophagy was determined by fluorescence microscopy, MDC staining, AO staining, and western blot. The H/R produced dramatic injuries in keratinocyte cells. In our study, the viability of Propofol in H/R induced HaCaT cells was first studied by MTT assay. The treatment with 25, 50, and $100{\mu}M$ Propofol in H/R induced HaCaT cells enhanced cell viability in a dose-dependent manner and $100{\mu}M$ was the most effective dose. The Atg5, Becline-1, LC3-II, and p62 were elevated in PPC group cells, but H/R-induced group showed significant reduction in HaCaT cells. The Atg5 were increased when autophagy was induced by Propofol, and they were decreased when autophagy was suppressed by 3-MA. These data provided evidence that propofol preconditioning induced autophagy and reduced apoptotic cell death in an H/R model of HaCaT cells, which was in agreement with autophagy playing a very important role in cell protection.

키워드

참고문헌

  1. Anaya-Prado R, Toledo-Pereyra LH. The molecular events underlying ischemia/reperfusion injury. Transplant Proc. 2002;34:2518-2519. https://doi.org/10.1016/S0041-1345(02)03471-1
  2. Wang Y, Zhang SX, Gozal D. Reactive oxygen species and the brain in sleep apnea. Respir Physiol Neurobiol. 2010;174:307-316. https://doi.org/10.1016/j.resp.2010.09.001
  3. Elahi MM, Kong YX, Matata BM. Oxidative stress as a mediator of cardiovascular disease. Oxid Med Cell Longev. 2009;2:259-269. https://doi.org/10.4161/oxim.2.5.9441
  4. Harrison EM, Sharpe E, Bellamy CO, McNally SJ, Devey L, Garden OJ, et al. Heat shock protein 90-binding agents protect renal cells from oxidative stress and reduce kidney ischemia-reperfusion injury. Am J Physiol Renal Physiol.2008;295:F397-405. https://doi.org/10.1152/ajprenal.00361.2007
  5. Chien CT, Chang TC, Tsai CY, Shyue SK, Lai MK. Adenovirus-mediated bcl-2 gene transfer inhibits renal ischemia/reperfusion induced tubular oxidative stress and apoptosis. Am J Transplant. 2005;5:1194-1203. https://doi.org/10.1111/j.1600-6143.2005.00826.x
  6. Ergun R, Akdemir G, Sen S, Tasci A, Ergungor F. Neuroprotective effects of propofol following global cerebral ischemia in rats. Neurosurg Rev. 2002;25:95-98. https://doi.org/10.1007/s101430100171
  7. Ito H, Watanabe Y, Isshiki A, Uchino H. Neuroprotective properties of propofol and midazolam, but not pentobarbital, on neuronal damage induced by forebrain ischemia, based on the GABAA receptors. Acta Anaesthesiol Scand. 1999;43:153-162. https://doi.org/10.1034/j.1399-6576.1999.430206.x
  8. Yano T, Nakayama R, Ushijima K. Intracerebroventricular propofol is neuroprotective against transient global ischemia in rats: extracellular glutamate level is not a major determinant. Brain Res. 2000;883:69-76. https://doi.org/10.1016/S0006-8993(00)02889-4
  9. Bayona NA, Gelb AW, Jiang Z, Wilson JX, Urquhart BL, Cechetto DF. Propofol neuroprotection in cerebral ischemia and its effects on low-molecular-weight antioxidants and skilled motor tasks. Anesthesiology 2004;100:1151-1159. https://doi.org/10.1097/00000542-200405000-00017
  10. Rossaint J, Rossaint R, Weis J, Fries M, Rex S, Coburn M. Propofol: neuroprotection in an in vitro model of traumatic brain injury. Crit Care 2009;13:R61. https://doi.org/10.1186/cc7795
  11. Velly LJ, Guillet BA, Masmejean FM, Nieoullon AL, Bruder NJ, Gouin FM, et al. Neuroprotective effects of propofol in a model of ischemic cortical cell cultures: role of glutamate and its transporters. Anesthesiology. 2003;99:368-375. https://doi.org/10.1097/00000542-200308000-00018
  12. Wu GJ, Chen WF, Hung HC, Jean YH, Sung CS, Chakraborty C, et al. Effects of propofol on proliferation and anti-apoptosis of neuroblastoma SH-SY5Y cell line: new insights into neuroprotection. Brain Res. 2011;1384:42-50. https://doi.org/10.1016/j.brainres.2011.02.004
  13. Gulcin I, Alici HA, Cesur M. Determination of in vitro antioxidant and radical scavenging activities of propofol. Chem Pharm Bull (Tokyo). 2005;53:281-285. https://doi.org/10.1248/cpb.53.281
  14. Schultz JE, Gross GJ. Opioids and cardioprotection. Pharmacol Ther. 2001;89:123-137. https://doi.org/10.1016/S0163-7258(00)00106-6
  15. Tanaka K, Ludwig LM, Kersten JR, Pagel PS, Warltier DC. Mechanisms of cardioprotection by volatile anesthetics. Anesthesiology 2004;100:707-721. https://doi.org/10.1097/00000542-200403000-00035
  16. Kroemer G, Marino G, Levine B. Autophagy and the integrated stress response. Mol Cell. 2010;40:280-293. https://doi.org/10.1016/j.molcel.2010.09.023
  17. Melendez A, Talloczy Z, Seaman M, Eskelinen EL, Hall DH, Levine B. Autophagy genes are essential for dauer development and life-span extension in C. elegans. Science 2003;301:1387-1391. https://doi.org/10.1126/science.1087782
  18. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci. 2011;36:30-38. https://doi.org/10.1016/j.tibs.2010.07.007
  19. Yan L, Sadoshima J, Vatner DE, Vatner SF. Autophagy: a novel protective mechanism in chronic ischemia. Cell Cycle 2006;5:1175-1177. https://doi.org/10.4161/cc.5.11.2787
  20. Baehrecke EH. Autophagy: dual roles in life and death? Nat Rev Mol Cell Biol. 2005;6:505-510.
  21. Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8:741-752.
  22. Javadov SA, Lim KH, Kerr PM, Suleiman MS, Angelini GD, Halestrap AP. Protection of hearts from reperfusion injury by propofol is associated with inhibition of the mitochondrial permeability transition. Cardiovasc Res. 2000;45:360-369. https://doi.org/10.1016/S0008-6363(99)00365-X
  23. Tao X, Lu LQ, Xu Q, Li SR, Lin MT. Cardioprotective effects of anesthetic preconditioning in rats with ischemia-reperfusion injury: propofol versus isoflurane. J Zhejiang Univ Sci B. 2009;10:740-747. https://doi.org/10.1631/jzus.B0920119
  24. Wickley PJ, Shiga T, Murray PA, Damron DS. Propofol decreases myofilament Ca2+ sensitivity via a protein kinase C-, nitric oxide synthase-dependent pathway in diabetic cardiomyocytes. Anesthesiology 2006;104: 978-987. https://doi.org/10.1097/00000542-200605000-00014
  25. Chen Y, Gibson SB. Is mitochondrial generation of reactive oxygen species a trigger for autophagy? Autophagy 2008;4:246-248. https://doi.org/10.4161/auto.5432
  26. Levine B, Yuan J. Autophagy in cell death: an innocent convict? J Clin Invest. 2005;115:2679-2688. https://doi.org/10.1172/JCI26390
  27. Kroemer G, Jaattela M. Lysosomes and autophagy in cell death control. Nat Rev Cancer 2005;5:886-897. https://doi.org/10.1038/nrc1738
  28. Kondo Y, Kanzawa T, Sawaya R, Kondo S. The role of autophagy in cancer development and response to therapy. Nat Rev Cancer. 2005;5:726-734. https://doi.org/10.1038/nrc1692
  29. Hamacher-Brady A, Brady NR, Gottlieb RA. Enhancing macroautophagy protects against ischemia/reperfusion injury in cardiac myocytes. J Biol Chem. 2006;281: 29776-29787. https://doi.org/10.1074/jbc.M603783200
  30. Akazawa H, Komazaki S, Shimomura H, Terasaki F, Zou Y, Takano H, et al. Diphtheria toxin-induced autophagic cardiomyocyte death plays a pathogenic role in mouse model of heart failure. J Biol Chem. 2004;279: 41095-41103. https://doi.org/10.1074/jbc.M313084200