Cite

Jones DP. Redefining oxidative stress. Antioxid Redox Signal 2006;8:1865–79. doi: 10.1089/ars.2006.8.1865 Jones DP Redefining oxidative stress Antioxid Redox Signal 200681865 79 10.1089/ars.2006.8.1865Open DOISearch in Google Scholar

Liguori I, Russo G, Curcio F, Bulli G, Aran L, Della-Morte D, Gargiulo G, Testa G, Cacciatore F, Bonaduce D, Abete P. Oxidative stress, aging, and diseases. Clin Interv Aging 2018;13:757–72. doi: 10.2147/CIA.S158513 Liguori I Russo G Curcio F Bulli G Aran L Della-Morte D Gargiulo G Testa G Cacciatore F Bonaduce D Abete P Oxidative stress, aging, and diseases Clin Interv Aging 201813757 72 10.2147/CIA.S158513Open DOISearch in Google Scholar

Lee Y-M, Song C, Yeum K-J. Impact of volatile anesthetics on oxidative stress and inflammation. Biomed Res Int 2015;2015:242709. doi: 10.1155/2015/242709 Lee Y-M Song C Yeum K-J Impact of volatile anesthetics on oxidative stress and inflammation Biomed Res Int 20152015242709 10.1155/2015/242709Open DOISearch in Google Scholar

Pisoschi AM, Pop A. The role of antioxidants in the chemistry of oxidative stress: A review. Eur J Med Chem 2015;97:55–74. doi: 10.1016/j.ejmech.2015.04.040 Pisoschi AM Pop A The role of antioxidants in the chemistry of oxidative stress: A review Eur J Med Chem 20159755 74 10.1016/j.ejmech.2015.04.040Open DOISearch in Google Scholar

Milne GL, Yin H, Brooks JD, Sanchez S, Roberts LJ, Morrow JD. Quantification of F2-isoprostanes in biological fluids and tissues as a measure of oxidant stress. Methods Enzymol 2007;433:113–26. doi: 10.1016/S0076-6879(07)33006-1 Milne GL Yin H Brooks JD Sanchez S Roberts LJ Morrow JD Quantification of F2-isoprostanes in biological fluids and tissues as a measure of oxidant stress Methods Enzymol 2007433113 26 10.1016/S0076-6879(07)33006-1Open DOISearch in Google Scholar

Del Rio D, Stewart AJ, Pellegrini N. A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress. Nutr Metab Cardiovasc Dis 2005;15:316–28. doi: 10.1016/j.numecd.2005.05.003 Del RioD Stewart AJ Pellegrini N A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress Nutr Metab Cardiovasc Dis 200515316 28 10.1016/j.numecd.2005.05.003Open DOISearch in Google Scholar

Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and antioxidant defense. World Allergy Organ J 2012;5:9–19. doi: 10.1097/WOX.0b013e3182439613 Birben E Sahiner UM Sackesen C Erzurum S Kalayci O Oxidative stress and antioxidant defense World Allergy Organ J 201259 19 10.1097/WOX.0b013e3182439613Open DOISearch in Google Scholar

Davies MJ. Singlet oxygen-mediated damage to proteins and its consequences. Biochem Biophys Res Commun 2003;305:761–70. doi: 10.1016/S0006-291X(03)00817-9 Davies MJ Singlet oxygen-mediated damage to proteins and its consequences Biochem Biophys Res Commun 2003305761 70 10.1016/S0006-291X(03)00817-9Open DOISearch in Google Scholar

Ozdogan M, Devay AO, Gurer A, Ersoy E, Devay SD, Kulacoglu H, Gundogdu H. Plasma total anti-oxidant capacity correlates inversely with the extent of acute appendicitis: A case control study. World J Emerg Surg 2006;1:6. doi: 10.1186/1749-7922-1-6 Ozdogan M Devay AO Gurer A Ersoy E Devay SD Kulacoglu H Gundogdu H Plasma total anti-oxidant capacity correlates inversely with the extent of acute appendicitis: A case control study World J Emerg Surg 200616 10.1186/1749-7922-1-6147583916759345Open DOISearch in Google Scholar

Baysal Z, Togrul T, Aksoy N, Cengiz M, Çelik H, Boleken ME, Kaya M, Yavuz G. Evaluation of total oxidative and antioxidative status in pediatric patients undergoing laparoscopic surgery. J Pediatr Surg 2009;44:1367–70. doi: 10.1016/j.jpedsurg.2008.11.031 Baysal Z Togrul T Aksoy N Cengiz M Çelik H Boleken ME Kaya M Yavuz G Evaluation of total oxidative and antioxidative status in pediatric patients undergoing laparoscopic surgery J Pediatr Surg 2009441367 70 10.1016/j.jpedsurg.2008.11.03119573663Open DOISearch in Google Scholar

Glantzounis GK, Tselepis AD, Tambaki AP, Trikalinos TA, Manataki AD, Galaris DA, Tsimoyiannis EC, Kappas AM. Laparoscopic surgery-induced changes in oxidative stress markers in human plasma. Surg Endosc 2001;15:1315–9. doi: 10.1007/s00464-001-0034-2 Glantzounis GK Tselepis AD Tambaki AP Trikalinos TA Manataki AD Galaris DA Tsimoyiannis EC Kappas AM Laparoscopic surgery-induced changes in oxidative stress markers in human plasma Surg Endosc 2001151315 9 10.1007/s00464-001-0034-2Open DOISearch in Google Scholar

Marik PE. Propofol: An immunomodulating agent. Pharmacotherapy 2005;25:28S–33S. doi: 10.1592/phco.2005.25.5_Part_2.28S Marik PE Propofol: An immunomodulating agent Pharmacotherapy 20052528S 33S 10.1592/phco.2005.25.5_Part_2.28SOpen DOISearch in Google Scholar

Kanto J, Gepts E. Pharmacokinetic implications for the clinical use of propofol. Clin Pharmacokinet 1989;17:308–26. doi: 10.2165/00003088-198917050-00002 Kanto J Gepts E Pharmacokinetic implications for the clinical use of propofol Clin Pharmacokinet 198917308 26 10.2165/00003088-198917050-00002Open DOISearch in Google Scholar

Rigby-Jones AE, Sneyd JR. Propofol and children – What we know and what we do not know. Paediatr Anaesth 2011;21:247–54. doi: 10.1111/j.1460-9592.2010.03454.x Rigby-Jones AE Sneyd JR Propofol and children – What we know and what we do not know Paediatr Anaesth 201121247 54 10.1111/j.1460-9592.2010.03454.xOpen DOISearch in Google Scholar

Boisset S, Steghens JP, Favetta P, Terreux R, Guitton J. Relative antioxidant capacities of propofol and its main metabolites. Arch Toxicol 2004;78:635–42. doi: 10.1007/s00204-004-0585-9 Boisset S Steghens JP Favetta P Terreux R Guitton J Relative antioxidant capacities of propofol and its main metabolites Arch Toxicol 200478635 42 10.1007/s00204-004-0585-9Open DOISearch in Google Scholar

Aarts L, van der Hee R, Dekker I, de Jong J, Langemeijer H, Bast A. The widely used anesthetic agent propofol can replace α-tocopherol as an antioxidant. FEBS Lett 1995;357:83–5. doi: 10.1016/0014-5793(94)01337-Z Aarts L van der HeeR Dekker I de JongJ Langemeijer H Bast A The widely used anesthetic agent propofol can replace α-tocopherol as an antioxidant FEBS Lett 199535783 5 10.1016/0014-5793(94)01337-ZOpen DOISearch in Google Scholar

De La Cruz JP, Sedeno G, Carmona JA, de la Cuesta FS. The in vitro effects of propofol on tissular oxidative stress in the rat. Anesth Analg 1998;87:1141–6. doi: 10.1213/00000539-199811000-00031 De La CruzJP Sedeno G Carmona JA de la CuestaFS The in vitro effects of propofol on tissular oxidative stress in the rat Anesth Analg 1998871141 6 10.1213/00000539-199811000-00031Open DOISearch in Google Scholar

Acquaviva R, Campisi A, Murabito P, Raciti G, Avola R, Mangiameli S, Musumeci I, Barcelona ML, Vanella A, Volti GL. Propofol attenuates peroxynitrite-mediated DNA damage and apoptosis in cultured astrocytes: An alternative protective mechanism. Anesthesiology 2004;101:1363–71. doi: 10.1097/00000542-200412000-00017 Acquaviva R Campisi A Murabito P Raciti G Avola R Mangiameli S Musumeci I Barcelona ML Vanella A Volti GL Propofol attenuates peroxynitrite-mediated DNA damage and apoptosis in cultured astrocytes: An alternative protective mechanism Anesthesiology 20041011363 71 10.1097/00000542-200412000-0001715564944Open DOISearch in Google Scholar

Braz MG, Braz LG, Freire CMM, Lucio LMC, Braz JRC, Tang G, Salvadori DMF, Yeum KJ, Amornyotin S. Isoflurane and propofol contribute to increasing the antioxidant status of patients during minor elective surgery a randomized clinical study. Medicine (Baltimore) 2015;94(31):e1266. doi: 10.1097/MD.0000000000001266 Braz MG Braz LG Freire CMM Lucio LMC Braz JRC Tang G Salvadori DMF Yeum KJ Amornyotin S Isoflurane and propofol contribute to increasing the antioxidant status of patients during minor elective surgery a randomized clinical study Medicine (Baltimore) 20159431e1266 10.1097/MD.0000000000001266461661226252290Open DOISearch in Google Scholar

Braz MG, Braz LG, Barbosa BS, Giacobino J, Orosz JEB, Salvadori DMF, Braz JRC. DNA damage in patients who underwent minimally invasive surgery under inhalation or intravenous anesthesia. Mutat Res 2011;726:251–4. doi: 10.1016/j.mrgentox.2011.09.007 Braz MG Braz LG Barbosa BS Giacobino J Orosz JEB Salvadori DMF Braz JRC DNA damage in patients who underwent minimally invasive surgery under inhalation or intravenous anesthesia Mutat Res 2011726251 4 10.1016/j.mrgentox.2011.09.00721944903Open DOISearch in Google Scholar

Karahalil B, Yağar S, Bahadir G, Durak P, Şardaş S. Diazepam and propofol used as anesthetics during open-heart surgery do not cause chromosomal aberrations in peripheral blood lymphocytes. Mutat Res 2005;581:181–6. doi: 10.1016/j.mrgentox.2004.10.021 Karahalil B Yağar S Bahadir G Durak P Şardaş S. Diazepam and propofol used as anesthetics during open-heart surgery do not cause chromosomal aberrations in peripheral blood lymphocytes Mutat Res 2005581181 6 10.1016/j.mrgentox.2004.10.02115725617Open DOISearch in Google Scholar

Li Y, Zhong D, Lei L, Jia Y, Zhou H, Yang B. Propofol prevents renal ischemia-reperfusion injury via inhibiting the oxidative stress pathways. Cell Physiol Biochem 2015;37:14–26. doi: 10.1159/000430329 Li Y Zhong D Lei L Jia Y Zhou H Yang B Propofol prevents renal ischemia-reperfusion injury via inhibiting the oxidative stress pathways Cell Physiol Biochem 20153714 26 10.1159/00043032926277932Open DOISearch in Google Scholar

Xie L-J, Zhao S, Zhang J-X, Li L. Propofol protects hearts from ischemia-reperfusion injury through interfering with mitochondria-dependent apoptotic pathway. Chinese J Pharmacol Toxicol 2007;21:247–54. Xie L-J Zhao S Zhang J-X Li L Propofol protects hearts from ischemia-reperfusion injury through interfering with mitochondria-dependent apoptotic pathway Chinese J Pharmacol Toxicol 200721247 54Search in Google Scholar

Bellanti F, Mirabella L, Mitarotonda D, Blonda M, Tamborra R, Cinnella G, Fersini A, Ambrosi A, Dambrosio M, Vendemiale G, Serviddio G. Propofol but not sevoflurane prevents mitochondrial dysfunction and oxidative stress by limiting HIF-1α activation in hepatic ischemia/reperfusion injury. Free Radic Biol Med 2016;96:323–33. doi: 10.1016/j.freeradbiomed.2016.05.002 Bellanti F Mirabella L Mitarotonda D Blonda M Tamborra R Cinnella G Fersini A Ambrosi A Dambrosio M Vendemiale G Serviddio G Propofol but not sevoflurane prevents mitochondrial dysfunction and oxidative stress by limiting HIF-1α activation in hepatic ischemia/reperfusion injury Free Radic Biol Med 201696323 33 10.1016/j.freeradbiomed.2016.05.00227154980Open DOISearch in Google Scholar

Kaptanoglu E, Sen S, Beskonakli E, Surucu HS, Tuncel M, Kilinc K, Taskin Y. Antioxidant actions and early ultrastructural findings of thiopental and propofol in experimental spinal cord injury. J Neurosurg Anesthesiol 2002;14:114–22. doi: 10.1097/00008506-200204000-00005 Kaptanoglu E Sen S Beskonakli E Surucu HS Tuncel M Kilinc K Taskin Y Antioxidant actions and early ultrastructural findings of thiopental and propofol in experimental spinal cord injury J Neurosurg Anesthesiol 200214114 22 10.1097/00008506-200204000-0000511907391Open DOISearch in Google Scholar

Yuan D, Su G, Liu Y, Chi X, Feng J, Zhu Q, Cai J, Luo G, Hei Z. Propofol attenuated liver transplantation-induced acute lung injury via connexin43 gap junction inhibition. J Transl Med 2016;14:194. doi: 10.1186/s12967-016-0954-1 Yuan D Su G Liu Y Chi X Feng J Zhu Q Cai J Luo G Hei Z Propofol attenuated liver transplantation-induced acute lung injury via connexin43 gap junction inhibition J Transl Med 201614194 10.1186/s12967-016-0954-1492977427364362Open DOISearch in Google Scholar

Yao W, Han X, Zhang Y, Guan J, Ge M, Chen C, Wu S, Chen J, Luo G, Huang P, Hei Z. Intravenous anesthetic protects hepatocyte from reactive oxygen species-induced cellular apoptosis during liver transplantation in vivo. Oxid Med Cell Longev 2018;2018:4780615. doi: 10.1155/2018/4780615 Yao W Han X Zhang Y Guan J Ge M Chen C Wu S Chen J Luo G Huang P Hei Z Intravenous anesthetic protects hepatocyte from reactive oxygen species-induced cellular apoptosis during liver transplantation in vivo Oxid Med Cell Longev 201820184780615 10.1155/2018/4780615623039230510620Open DOISearch in Google Scholar

Yildiz H, Coskuner I, Bulbuloglu E, Silay E, Kurutas EB, Dogan Z, Kantarceken B, Oksuz H, Senoglu N, Yuzbasioglu MF, Cetinkaya A, Ciralik H. The protective effects of ketamine and propofol in obstructive jaundice: an experimental study. Bratisl Med J 2012;113:139–44. doi: 10.4149/BLL_2012_034 Yildiz H Coskuner I Bulbuloglu E Silay E Kurutas EB Dogan Z Kantarceken B Oksuz H Senoglu N Yuzbasioglu MF Cetinkaya A Ciralik H The protective effects of ketamine and propofol in obstructive jaundice: an experimental study Bratisl Med J 2012113139 44 10.4149/BLL_2012_03422428761Open DOISearch in Google Scholar

Brasil LJ, San-Miguel B, Kretzmann NA, Amaral JLG Do, Zettler CG, Marroni N, González-Gallego J, Tuñón MJ. Halothane induces oxidative stress and NF-κB activation in rat liver: Protective effect of propofol. Toxicology 2006;227:53–61. doi: 10.1016/j.tox.2006.07.013 Brasil LJ San-Miguel B Kretzmann NA AmaralJLG Do Zettler CG Marroni N González-Gallego J Tuñón MJ Halothane induces oxidative stress and NF-κB activation in rat liver: Protective effect of propofol Toxicology 200622753 61 10.1016/j.tox.2006.07.01316965849Open DOISearch in Google Scholar

Alipour F, Emami MR, Mohri M. Endocrine and oxidative stress characteristics in different anesthetic methods during pneumoperitoneum in dogs. Comp Clin Pathol 2018;27:1667–73. doi: 10.1007/s00580-018-2792-4 Alipour F Emami MR Mohri M Endocrine and oxidative stress characteristics in different anesthetic methods during pneumoperitoneum in dogs Comp Clin Pathol 2018271667 73 10.1007/s00580-018-2792-4Open DOISearch in Google Scholar

Lee JY. Oxidative stress due to anesthesia and surgical trauma and comparison of the effects of propofol and thiopental in dogs. J Vet Med Sci 2012;74:663–5. doi: 10.1292/jvms.11-0221 Lee JY Oxidative stress due to anesthesia and surgical trauma and comparison of the effects of propofol and thiopental in dogs J Vet Med Sci 201274663 5 10.1292/jvms.11-022122198056Open DOISearch in Google Scholar

Félix LM, Correia F, Pinto PA, Campos SP, Fernandes T, Videira R, Oliveira MM, Peixoto FP, Antunes LM. Propofol affinity to mitochondrial membranes does not alter mitochondrial function. Eur J Pharmacol 2017;803:48–56. doi: 10.1016/j.ejphar.2017.03.044 Félix LM Correia F Pinto PA Campos SP Fernandes T Videira R Oliveira MM Peixoto FP Antunes LM Propofol affinity to mitochondrial membranes does not alter mitochondrial function Eur J Pharmacol 201780348 56 10.1016/j.ejphar.2017.03.04428341344Open DOISearch in Google Scholar

Ucar M, Ozgül U, Polat A, Toprak HI, Erdogan MA, Aydogan MS, Durmus M, Ersoy MO. Comparison of antioxidant effects of isoflurane and propofol in patients undergoing donor hepatectomy. Transplant Proc 2015;47:469–72. doi: 10.1016/j.transproceed.2014.11.043 Ucar M Ozgül U Polat A Toprak HI Erdogan MA Aydogan MS Durmus M Ersoy MO Comparison of antioxidant effects of isoflurane and propofol in patients undergoing donor hepatectomy Transplant Proc 201547469 72 10.1016/j.transproceed.2014.11.04325769593Open DOISearch in Google Scholar

Khoshraftar E, Ranjbar A, Kharkhane B, Heidary ST, Gharebaghi Z, Zadkhosh N. Antioxidative effects of propofol vs. Ketamin in individuals undergoing surgery. Arch Iran Med 2014;17:486–9. doi: 0141707/AIM.008 Khoshraftar E Ranjbar A Kharkhane B Heidary ST Gharebaghi Z Zadkhosh N Antioxidative effects of propofol vs. Ketamin in individuals undergoing surgery Arch Iran Med 201417486 9 0141707/AIM.00824979561Open DOISearch in Google Scholar

Dumaresq DMH, de Vasconcelos RC, Guimarães SB, Cavalcante SL, Garcia JHP, de Vasconcelos PRL. Metabolic and oxidative effects of sevoflurane and propofol in children undergoing surgery for congenital heart disease. Acta Cir Bras 2011;26:66–71. doi: 10.1590/S0102-86502011000700014 Dumaresq DMH de VasconcelosRC Guimarães SB Cavalcante SL Garcia JHP de VasconcelosPRL Metabolic and oxidative effects of sevoflurane and propofol in children undergoing surgery for congenital heart disease Acta Cir Bras 20112666 71 10.1590/S0102-8650201100070001421971661Open DOISearch in Google Scholar

Yagmurdur H, Cakan T, Bayrak A, Arslan M, Baltaci B, Inan N, Kilinc K. The effects of etomidate, thiopental, and propofol in induction on hypoperfusion-reperfusion phenomenon during laparoscopic cholecystectomy. Acta Anaesthesiol Scand 2004;48:772–7. doi: 10.1111/j.0001-5172.2004.00417.x Yagmurdur H Cakan T Bayrak A Arslan M Baltaci B Inan N Kilinc K The effects of etomidate, thiopental, and propofol in induction on hypoperfusion-reperfusion phenomenon during laparoscopic cholecystectomy Acta Anaesthesiol Scand 200448772 7 10.1111/j.0001-5172.2004.00417.x15196111Open DOISearch in Google Scholar

Laviolle B, Basquin C, Aguillon D, Compagnon P, Morel I, Turmel V, Seguin P, Boudjema K, Bellissant E, Mallédant Y. Effect of an anesthesia with propofol compared with desflurane on free radical production and liver function after partial hepatectomy. Fundam Clin Pharmacol 2012;26:735–42. doi: 10.1111/j.1472-8206.2011.00958.x Laviolle B Basquin C Aguillon D Compagnon P Morel I Turmel V Seguin P Boudjema K Bellissant E Mallédant Y Effect of an anesthesia with propofol compared with desflurane on free radical production and liver function after partial hepatectomy Fundam Clin Pharmacol 201226735 42 10.1111/j.1472-8206.2011.00958.x21692846Open DOISearch in Google Scholar

Tsuchiya M, Shiomoto K, Mizutani K, Fujioka K, Suehiro K, Yamada T, Sato EF, Nishikawa K. Reduction of oxidative stress a key for enhanced postoperative recovery with fewer complications in esophageal surgery patients: Randomized control trial to investigate therapeutic impact of anesthesia management and usefulness of simple blood test for pre. Medicine (Baltimore) 2018;97(47):e12845. doi: 10.1097/MD.0000000000012845 Tsuchiya M Shiomoto K Mizutani K Fujioka K Suehiro K Yamada T Sato EF Nishikawa K Reduction of oxidative stress a key for enhanced postoperative recovery with fewer complications in esophageal surgery patients: Randomized control trial to investigate therapeutic impact of anesthesia management and usefulness of simple blood test for pre Medicine (Baltimore) 20189747e12845 10.1097/MD.0000000000012845639272530461602Open DOISearch in Google Scholar

Lai HC, Yeh YC, Wang LC, Ting CT, Lee WL, Lee HW, Wang KY, Wu A, Su CS, Liu TJ. Propofol ameliorates doxorubicin-induced oxidative stress and cellular apoptosis in rat cardiomyocytes. Toxicol Appl Pharmacol 2011;257:437–48. doi: 10.1016/j.taap.2011.10.001 Lai HC Yeh YC Wang LC Ting CT Lee WL Lee HW Wang KY Wu A Su CS Liu TJ Propofol ameliorates doxorubicin-induced oxidative stress and cellular apoptosis in rat cardiomyocytes Toxicol Appl Pharmacol 2011257437 48 10.1016/j.taap.2011.10.00122015447Open DOISearch in Google Scholar

Hsu SS, Jan CR, Liang WZ. Evaluation of cytotoxicity of propofol and its related mechanism in glioblastoma cells and astrocytes. Environ Toxicol 2017;32:2440–54. doi: 10.1002/tox.22458 Hsu SS Jan CR Liang WZ Evaluation of cytotoxicity of propofol and its related mechanism in glioblastoma cells and astrocytes Environ Toxicol 2017322440 54 10.1002/tox.2245828804952Open DOISearch in Google Scholar

Baronica R, Tonković D, Bačić G, Karadjole T, Šuran J, Bačić Baronica K, editors. Croatian International Symposium on Intensive Care Medicine / Simpozij intenzivne medicine s međunarodnim sudjelovanjem. Neurol Croat 2013;62(Suppl 2):1–170. Baronica R Tonković D Bačić G Karadjole T Šuran J Bačić Baronica K editors Croatian International Symposium on Intensive Care Medicine / Simpozij intenzivne medicine s međunarodnim sudjelovanjem Neurol Croat 201362Suppl 21 170Search in Google Scholar

Wittmann S, Daniels S, Ittner KP, Fröhlich D. Thiopentone and methohexitone enantiomers do not act stereoselectively on the oxidative response in human neutrophils in vitro. Pharmacology 2004;72:12–9. doi: 10.1159/000078627 Wittmann S Daniels S Ittner KP Fröhlich D Thiopentone and methohexitone enantiomers do not act stereoselectively on the oxidative response in human neutrophils in vitro Pharmacology 20047212 9 10.1159/00007862715292650Open DOISearch in Google Scholar

Nishina K, Akamatsu H, Mikawa K, Shiga M, Maekawa N, Obara H, Niwa Y. The inhibitory effects of thiopental, midazolam, and ketamine on human neutrophil functions. Anesth Analg 1998;86:159–65. doi: 10.1097/00000539-199801000-00032 Nishina K Akamatsu H Mikawa K Shiga M Maekawa N Obara H Niwa Y The inhibitory effects of thiopental, midazolam, and ketamine on human neutrophil functions Anesth Analg 199886159 65 10.1097/00000539-199801000-000329428872Open DOISearch in Google Scholar

Ahiskalioglu EO, Aydin P, Ahiskalioglu A, Suleyman B, Kuyrukluyildiz U, Kurt N, Altuner D, Coskun R, Suleyman H. The effects of ketamine and thiopental used alone or in combination on the brain, heart, and bronchial tissues of rats. Arch Med Sci 2018;14:645–54. doi: 10.5114/aoms.2016.59508 Ahiskalioglu EO Aydin P Ahiskalioglu A Suleyman B Kuyrukluyildiz U Kurt N Altuner D Coskun R Suleyman H The effects of ketamine and thiopental used alone or in combination on the brain, heart, and bronchial tissues of rats Arch Med Sci 201814645 54 10.5114/aoms.2016.59508594990429765454Open DOISearch in Google Scholar

Dinis-Oliveira RJ. Metabolism and metabolomics of ketamine: a toxicological approach. Forensic Sci Res 2017;2:2–10. doi: 10.1080/20961790.2017.1285219 Dinis-Oliveira RJ. Metabolism and metabolomics of ketamine: a toxicological approach Forensic Sci Res 201722 10 10.1080/20961790.2017.1285219619710730483613Open DOISearch in Google Scholar

Bai X, Yan Y, Canfield S, Muravyeva MY, Kikuchi C, Zaja I, Corbett JA, Bosnjak ZJ. Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway. Anesth Analg 2013;116:869–80. doi: 10.1213/ANE.0b013e3182860fc9 Bai X Yan Y Canfield S Muravyeva MY Kikuchi C Zaja I Corbett JA Bosnjak ZJ Ketamine enhances human neural stem cell proliferation and induces neuronal apoptosis via reactive oxygen species-mediated mitochondrial pathway Anesth Analg 2013116869 80 10.1213/ANE.0b013e3182860fc9360667723460563Open DOISearch in Google Scholar

Ito H, Uchida T, Makita K. Ketamine causes mitochondrial dysfunction in human induced pluripotent stem cell: Derived neurons. PLoS One 2015;10(5):e0128445. doi: 10.1371/journal.pone.0128445 Ito H Uchida T Makita K Ketamine causes mitochondrial dysfunction in human induced pluripotent stem cell: Derived neurons PLoS One 2015105e0128445 10.1371/journal.pone.0128445444738226020236Open DOISearch in Google Scholar

Hatipoglu S, Yildiz H, Bulbuloglu E, Coskuner I, Kurutas EB, Hatipoglu F, Ciralik H, Berhuni MS. Protective effects of intravenous anesthetics on kidney tissue in obstructive jaundice. World J Gastroenterol 2014;20:3320–6. doi: 10.3748/wjg.v20.i12.3320 Hatipoglu S Yildiz H Bulbuloglu E Coskuner I Kurutas EB Hatipoglu F Ciralik H Berhuni MS Protective effects of intravenous anesthetics on kidney tissue in obstructive jaundice World J Gastroenterol 2014203320 6 10.3748/wjg.v20.i12.3320396440224695809Open DOISearch in Google Scholar

Parvin R, Akhter N. Protective effect of tomato against adrenaline-induced myocardial infarction in rats. Bangladesh Med Res Counc Bull 2008;34:104–8. doi: 10.3329/bmrcb.v34i3.1974 Parvin R Akhter N Protective effect of tomato against adrenaline-induced myocardial infarction in rats Bangladesh Med Res Counc Bull 200834104 8 10.3329/bmrcb.v34i3.197419476257Open DOISearch in Google Scholar

Liang J, Wu S, Xie W, He H. Ketamine ameliorates oxidative stress-induced apoptosis in experimental traumatic brain injury via the Nrf2 pathway. Drug Des Devel Ther 2018;12:845–53. doi: 10.2147/DDDT.S160046 Liang J Wu S Xie W He H Ketamine ameliorates oxidative stress-induced apoptosis in experimental traumatic brain injury via the Nrf2 pathway Drug Des Devel Ther 201812845 53 10.2147/DDDT.S160046590778529713142Open DOISearch in Google Scholar

Welters ID, Feurer M-K, Preiss V, Mller M, Scholz S, Kwapisz M, Mogk M, Neuhuser C. Continuous S-(+)-ketamine administration during elective coronary artery bypass graft surgery attenuates pro-inflammatory cytokine response during and after cardiopulmonary bypass. Br J Anaesth 2011;106:172–9. doi: 10.1093/bja/aeq341 Welters ID Feurer M-K Preiss V Mller M Scholz S Kwapisz M Mogk M Neuhuser C Continuous S-(+)-ketamine administration during elective coronary artery bypass graft surgery attenuates pro-inflammatory cytokine response during and after cardiopulmonary bypass Br J Anaesth 2011106172 9 10.1093/bja/aeq34121138901Open DOISearch in Google Scholar

Dale O, Somogyi AA, Li Y, Sullivan T, Shavit Y. Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and meta-analysis. Anesth Analg 2012;115:934–43. doi: 10.1213/ANE.0b013e3182662e30 Dale O Somogyi AA Li Y Sullivan T Shavit Y Does intraoperative ketamine attenuate inflammatory reactivity following surgery? A systematic review and meta-analysis Anesth Analg 2012115934 43 10.1213/ANE.0b013e3182662e3022826531Open DOISearch in Google Scholar

Durandy Y. Minimizing systemic inflammation during cardiopulmonary bypass in the pediatric population. Artif Organs 2014;38:11–8. doi: 10.1111/aor.12195 Durandy Y Minimizing systemic inflammation during cardiopulmonary bypass in the pediatric population Artif Organs 20143811 8 10.1111/aor.1219524392866Open DOISearch in Google Scholar

Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology 2011;114:695–707. doi: 10.1097/ALN.0b013e3181ff72b5 Forman SA Clinical and molecular pharmacology of etomidate Anesthesiology 2011114695 707 10.1097/ALN.0b013e3181ff72b5310815221263301Open DOISearch in Google Scholar

Raines DE. The Pharmacology of Etomidate and Etomidate Derivatives. Int Anesthesiol Clin 2015;53:63–75. doi:10.1097/AIA.0000000000000050. Raines DE The Pharmacology of Etomidate and Etomidate Derivatives Int Anesthesiol Clin 20155363 75 10.1097/AIA.000000000000005025807019Open DOISearch in Google Scholar

Allen C, Washington S. The role of etomidate as an anaesthetic induction agent for critically ill patients. Br J Hosp Med 2016;77:282–6. doi: 10.12968/hmed.2016.77.5.282 Allen C Washington S The role of etomidate as an anaesthetic induction agent for critically ill patients Br J Hosp Med 201677282 6 10.12968/hmed.2016.77.5.28227166106Open DOISearch in Google Scholar

Li X, Lu F, Li W, Xu J, Sun XJ, Qin LZ, Zhang QL, Yao Y, Yu QK, Liang XL. Underlying mechanisms of memory deficits induced by etomidate anesthesia in aged rat model: Critical role of immediate early genes. Chin Med J (Engl) 2016;129:48–53. doi: 10.4103/0366-6999.172570 Li X Lu F Li W Xu J Sun XJ Qin LZ Zhang QL Yao Y Yu QK Liang XL Underlying mechanisms of memory deficits induced by etomidate anesthesia in aged rat model: Critical role of immediate early genes Chin Med J (Engl) 201612948 53 10.4103/0366-6999.172570479754226712432Open DOISearch in Google Scholar

Cayli SR, Ates O, Karadag N, Altinoz E, Yucel N, Yologlu S, Kocak A, Cakir CO. Neuroprotective effect of etomidate on functional recovery in experimental spinal cord injury. Int J Dev Neurosci 2006;24:233–9. doi: 10.1016/j.ijdevneu.2006.04.003 Cayli SR Ates O Karadag N Altinoz E Yucel N Yologlu S Kocak A Cakir CO Neuroprotective effect of etomidate on functional recovery in experimental spinal cord injury Int J Dev Neurosci 200624233 9 10.1016/j.ijdevneu.2006.04.00316701976Open DOISearch in Google Scholar

Li R, Fan L, Ma F, Cao Y, Gao J, Liu H, Li Y. Effect of etomidate on the oxidative stress response and levels of inflammatory factors from ischemia-reperfusion injury after tibial fracture surgery. Exp Ther Med 2017;13:971–5. doi: 10.3892/etm.2017.4037 Li R Fan L Ma F Cao Y Gao J Liu H Li Y Effect of etomidate on the oxidative stress response and levels of inflammatory factors from ischemia-reperfusion injury after tibial fracture surgery Exp Ther Med 201713971 5 10.3892/etm.2017.4037540351928450928Open DOISearch in Google Scholar

Lv X, Yan J, Jiang H. Inhaled anesthetic sevoflurane: neurotoxicity or neuroprotection in the developing brain. Int J Clin Exp Med 2017;10:9930–8. Lv X Yan J Jiang H Inhaled anesthetic sevoflurane: neurotoxicity or neuroprotection in the developing brain Int J Clin Exp Med 2017109930 8Search in Google Scholar

Kaye AD, Fox CJ, Padnos IW, Ehrhardt KP, Diaz JH, Cornett EM, Chandler D, Sen S, Patil S. Pharmacologic considerations of anesthetic agents in pediatric patients: A comprehensive review. Anesthesiol Clin 2017;35(2):e73–e94. doi: 10.1016/j.anclin.2017.01.012 Kaye AD Fox CJ Padnos IW Ehrhardt KP Diaz JH Cornett EM Chandler D Sen S Patil S Pharmacologic considerations of anesthetic agents in pediatric patients: A comprehensive review Anesthesiol Clin 2017352e73 e94 10.1016/j.anclin.2017.01.01228526162Open DOISearch in Google Scholar

Szyfter K, Szulc R, Mikstacki A, Stachecki I, Rydzanicz M, Jałoszyński P. Genotoxicity of inhalation anaesthetics: DNA lesions generated by sevoflurane in vitro and in vivo. J Appl Genet 2004;45:369–74. PMID: 15306730 Szyfter K Szulc R Mikstacki A Stachecki I Rydzanicz M Jałoszyński P Genotoxicity of inhalation anaesthetics: DNA lesions generated by sevoflurane in vitro and in vivo J Appl Genet 200445369 74 PMID: 15306730Search in Google Scholar

Brozović G, Oršolić N, Rozgaj R, Knežević F, Knežević AH, Maričić M, Krsnik D, Benković V. Sevoflurane and isoflurane genotoxicity in kidney cells of mice. Arh Hig Rada Toksikol 2017;68:228–35. doi: 10.1515/aiht-2017-68-2941 Brozović G Oršolić N Rozgaj R Knežević F Knežević AH Maričić M Krsnik D Benković V Sevoflurane and isoflurane genotoxicity in kidney cells of mice Arh Hig Rada Toksikol 201768228 35 10.1515/aiht-2017-68-294128976881Open DOISearch in Google Scholar

Krause T, Scholz J, Jansen L, Boettcher H, Koch C, Wappler F, Schulte am Esch J. Sevoflurane anaesthesia does not induce the formation of sister chromatid exchanges in peripheral blood lymphocytes of children. Br J Anaesth 2003;90:233–5. doi: 10.1093/bja/aeg030 Krause T Scholz J Jansen L Boettcher H Koch C Wappler F Schulte am Esch J. Sevoflurane anaesthesia does not induce the formation of sister chromatid exchanges in peripheral blood lymphocytes of children Br J Anaesth 200390233 5 10.1093/bja/aeg03012538381Open DOISearch in Google Scholar

Karabiyik L, Şardaş S, Polat U, Kocabaş NA, Karakaya AE. Comparison of genotoxicity of sevoflurane and isoflurane in human lymphocytes studied in vivo using the comet assay. Mutat Res 2001;492:99–107. doi: 10.1016/S1383-5718(01)00159-0 Karabiyik L Şardaş S Polat U Kocabaş NA Karakaya AE Comparison of genotoxicity of sevoflurane and isoflurane in human lymphocytes studied in vivo using the comet assay Mutat Res 200149299 107 10.1016/S1383-5718(01)00159-0Open DOISearch in Google Scholar

Arslan M, Isik B, Kavutcu M, Kurtipek O. Oxidative stress and antioxidant activity of female rat liver tissue after sevoflurane anaesthesia: young versus old. Bratisl Med J 2012;113:702–6. doi: 10.4149/bll_2012_159 Arslan M Isik B Kavutcu M Kurtipek O Oxidative stress and antioxidant activity of female rat liver tissue after sevoflurane anaesthesia: young versus old Bratisl Med J 2012113702 6 10.4149/bll_2012_15923173627Open DOISearch in Google Scholar

Türkan H, Aydin A, Sayal A, Eken A, Akay C, Karahalil B. Oxidative and antioxidative effects of desflurane and sevoflurane on rat tissue in vivo. Arh Hig Rada Toksikol 2011;62:113–9. doi: 10.2478/10004-1254-62-2011-2096 Türkan H Aydin A Sayal A Eken A Akay C Karahalil B Oxidative and antioxidative effects of desflurane and sevoflurane on rat tissue in vivo Arh Hig Rada Toksikol 201162113 9 10.2478/10004-1254-62-2011-209621705298Open DOISearch in Google Scholar

Molin SZFD, Kruel CRP, de Fraga RS, Alboim C, de Oliveira JR, Alvares-da-Silva MR. Differential protective effects of anaesthesia with sevoflurane or isoflurane. Eur J Anaesthesiol 2014;31:695–700. doi: 10.1097/EJA.0000000000000127 Molin SZFD Kruel CRP de FragaRS Alboim C de OliveiraJR Alvares-da-Silva MR Differential protective effects of anaesthesia with sevoflurane or isoflurane Eur J Anaesthesiol 201431695 700 10.1097/EJA.000000000000012725105848Open DOISearch in Google Scholar

Sivaci R, Kahraman A, Serteser M, Sahin DA, Dilek ON. Cytotoxic effects of volatile anesthetics with free radicals undergoing laparoscopic surgery. Clin Biochem 2006;39:293–8. doi: 10.1016/j.clinbiochem.2006.01.001 Sivaci R Kahraman A Serteser M Sahin DA Dilek ON Cytotoxic effects of volatile anesthetics with free radicals undergoing laparoscopic surgery Clin Biochem 200639293 8 10.1016/j.clinbiochem.2006.01.00116494857Open DOISearch in Google Scholar

Erbas M, Demiraran Y, Ak Yildirim H, Sezen G, Iskender A, Karagoz I, Kandis H. Comparison of effects on the oxidant/antioxidant system of sevoflurane, desflurane and propofol infusion during general anesthesia. Brazilian J Anesthesiol 2015;65:68–72. doi: 10.1016/j.bjane.2014.05.004 Erbas M Demiraran Y Ak YildirimH Sezen G Iskender A Karagoz I Kandis H Comparison of effects on the oxidant/antioxidant system of sevoflurane, desflurane and propofol infusion during general anesthesia Brazilian J Anesthesiol 20156568 72 10.1016/j.bjane.2014.05.004Open DOISearch in Google Scholar

Zhou X, Lu D, Li W da, Chen X hui, Yang X yu, Chen X, Zhou Z bin, Ye JH, Feng X. Sevoflurane affects oxidative stress and alters apoptosis status in children and cultured neural stem cells. Neurotox Res 2018;33:790–800. doi: 10.1007/s12640-017-9827-5 Zhou X Lu D daLi W huiChen X yuYang X Chen X binZhou Z Ye JH Feng X Sevoflurane affects oxidative stress and alters apoptosis status in children and cultured neural stem cells Neurotox Res 201833790 800 10.1007/s12640-017-9827-529071560Open DOISearch in Google Scholar

Allaouchiche B, Debon R, Goudable J, Chassard D, Duflo F. Oxidative stress status during exposure to propofol, sevoflurane and desflurane. Anesth Analg 2001;93:981–5. doi: 10.1097/00000539-200110000-00036 Allaouchiche B Debon R Goudable J Chassard D Duflo F Oxidative stress status during exposure to propofol, sevoflurane and desflurane Anesth Analg 200193981 5 10.1097/00000539-200110000-0003611574369Open DOISearch in Google Scholar

Shen X, Dong Y, Xu Z, Wang H, Miao C, Soriano SG, Sun D, Baxter MG, Zhang Y, Xie Z. Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment. Anesthesiology 2013;118:502–15. doi: 10.1097/ALN.0b013e3182834d77 Shen X Dong Y Xu Z Wang H Miao C Soriano SG Sun D Baxter MG Zhang Y Xie Z Selective anesthesia-induced neuroinflammation in developing mouse brain and cognitive impairment Anesthesiology 2013118502 15 10.1097/ALN.0b013e3182834d77358000223314110Open DOISearch in Google Scholar

Yalcin S, Aydoğan H, Yuce HH, Kucuk A, Karahan MA, Vural M, Camuzcuoğlu A, Aksoy N. Effects of sevoflurane and desflurane on oxidative stress during general anesthesia for elective cesarean section. Wien Klin Wochenschr 2013;125:467–73. doi: 10.1007/s00508-013-0397-0 Yalcin S Aydoğan H Yuce HH Kucuk A Karahan MA Vural M Camuzcuoğlu A Aksoy N Effects of sevoflurane and desflurane on oxidative stress during general anesthesia for elective cesarean section Wien Klin Wochenschr 2013125467 73 10.1007/s00508-013-0397-023860695Open DOISearch in Google Scholar

Eroglu F, Yavuz L, Ceylan BG, Yílmaz F, Eroglu E, Delibas N, Nazíroğlu M. New volatile anesthetic, desflurane, reduces vitamin e level in blood of operative patients via oxidative stress. Cell Biochem Funct 2010;28:211–6. doi: 10.1002/cbf.1641 Eroglu F Yavuz L Ceylan BG Yílmaz F Eroglu E Delibas N Nazíroğlu M New volatile anesthetic, desflurane, reduces vitamin e level in blood of operative patients via oxidative stress Cell Biochem Funct 201028211 6 10.1002/cbf.1641Open DOISearch in Google Scholar

Nogueira FR, Braz LG, Souza KM, Aun AG, Arruda NM, Carvalho LR, Chen CYO, Braz JRC, Braz MG. Comparison of DNA damage and oxidative stress in patients anesthetized with desflurane associated or not with nitrous oxide: A prospective randomized clinical trial. Anesth Analg 2018;126:1198–205. doi: 10.1213/ANE.0000000000002729 Nogueira FR Braz LG Souza KM Aun AG Arruda NM Carvalho LR Chen CYO Braz JRC Braz MG Comparison of DNA damage and oxidative stress in patients anesthetized with desflurane associated or not with nitrous oxide: A prospective randomized clinical trial Anesth Analg 20181261198 205 10.1213/ANE.0000000000002729Open DOISearch in Google Scholar

Cukurova Z, Cetingok H, Ozturk S, Gedikbasi A, Hergunsel O, Ozturk D, Don B, Cefle K, Palanduz S, Ertem DH, Tarantino G. DNA damage effects of inhalation anesthetics in human bronchoalveolar cells. Medicine (Baltimore) 2019;98:e16518. doi: 10.1097/MD.0000000000016518 Cukurova Z Cetingok H Ozturk S Gedikbasi A Hergunsel O Ozturk D Don B Cefle K Palanduz S Ertem DH Tarantino G DNA damage effects of inhalation anesthetics in human bronchoalveolar cells Medicine (Baltimore) 201998e16518 10.1097/MD.0000000000016518Open DOISearch in Google Scholar

Schallner N, Ulbrich F, Engelstaedter H, Biermann J, Auwaerter V, Loop T, Goebel U. Isoflurane but not sevoflurane or desflurane aggravates injury to neurons in vitro and in vivo via p75NTR-NF-κB activation. Anesth Analg 2014;119:1429–41. doi: 10.1213/ANE.0000000000000488 Schallner N Ulbrich F Engelstaedter H Biermann J Auwaerter V Loop T Goebel U Isoflurane but not sevoflurane or desflurane aggravates injury to neurons in vitro and in vivo via p75NTR-NF-κB activation Anesth Analg 20141191429 41 10.1213/ANE.0000000000000488Open DOISearch in Google Scholar

Türkan H, Bukan N, Sayal A, Aydin A, Bukan MH. Effects of halothane, enflurane, and isoflurane on plasma and erythrocyte antioxidant enzymes and trace elements. Biol Trace Elem Res 2004;102:105–12. doi: 10.1385/BTER:102:1-3:105 Türkan H Bukan N Sayal A Aydin A Bukan MH Effects of halothane, enflurane, and isoflurane on plasma and erythrocyte antioxidant enzymes and trace elements Biol Trace Elem Res 2004102105 12 10.1385/BTER:102:1-3:105Open DOISearch in Google Scholar

Husum B, Wulf HC, Neebuhr E, Kyst A, Valentin N. Sister chromatid exchanges in lymphocytes of humans anaesthetized with isoflurane. Br J Anaesth 1984;56:559–64. doi: 10.1093/bja/56.6.559 Husum B Wulf HC Neebuhr E Kyst A Valentin N Sister chromatid exchanges in lymphocytes of humans anaesthetized with isoflurane Br J Anaesth 198456559 64 10.1093/bja/56.6.5596721966Open DOISearch in Google Scholar

Sanders RD, Weimann J, Maze M. Biologic effects of nitrous oxide: A mechanistic and toxicologic review. Anesthesiology 2008;109:707–22. doi: 10.1097/ALN.0b013e3181870a17 Sanders RD Weimann J Maze M Biologic effects of nitrous oxide: A mechanistic and toxicologic review Anesthesiology 2008109707 22 10.1097/ALN.0b013e3181870a1718813051Open DOISearch in Google Scholar

van Amsterdam J, Nabben T, van den Brink W. Recreational nitrous oxide use: Prevalence and risks. Regul Toxicol Pharmacol 2015;73:790–6. doi: 10.1016/j.yrtph.2015.10.017 van AmsterdamJ Nabben T van den BrinkW Recreational nitrous oxide use: Prevalence and risks Regul Toxicol Pharmacol 201573790 6 10.1016/j.yrtph.2015.10.01726496821Open DOISearch in Google Scholar

Hert SGD. The current place of nitrous oxide in clinical practice. Eur J Anaesthesiol 2015;32:517–20. doi: 10.1097/EJA.0000000000000264 Hert SGD The current place of nitrous oxide in clinical practice Eur J Anaesthesiol 201532517 20 10.1097/EJA.000000000000026426244467Open DOISearch in Google Scholar

American Society of Anesthesiologists. Anesthetic Gases: Information for Management in Anesthetizing Areas and Postanesthesia Care Unit. Park Ridge (IL): ASA; 1999. American Society of Anesthesiologists Anesthetic Gases: Information for Management in Anesthetizing Areas and Postanesthesia Care Unit Park Ridge (IL) ASA 1999Search in Google Scholar

Wrońska-Nofer T, Nofer JR, Jajte J, Dziubałtowska E, Szymczak W, Krajewski W, Waogoneksowicz W, Rydzyński K. Oxidative DNA damage and oxidative stress in subjects occupationally exposed to nitrous oxide (N2O). Mutat Res 2012;731:58–63. doi: 10.1016/j.mrfmmm.2011.10.010 Wrońska-Nofer T Nofer JR Jajte J Dziubałtowska E Szymczak W Krajewski W Waogoneksowicz W Rydzyński K Oxidative DNA damage and oxidative stress in subjects occupationally exposed to nitrous oxide (N2O) Mutat Res 201273158 63 10.1016/j.mrfmmm.2011.10.01022085808Open DOISearch in Google Scholar

Badner NH, Drader K, Freeman D, Spence JD. The use of intraoperative nitrous oxide leads to postoperative increases in plasma homocysteine. Anesth Analg 1998;87:711–3. doi: 10.1213/00000539-199809000-00041 Badner NH Drader K Freeman D Spence JD The use of intraoperative nitrous oxide leads to postoperative increases in plasma homocysteine Anesth Analg 199887711 3 10.1213/00000539-199809000-00041Open DOISearch in Google Scholar

Myles PS, Chan MTV, Kaye DM, McIlroy DR, Lau CW, Symons JA, Chen S. Effect of nitrous oxide anesthesia on plasma homocysteine and endothelial function. Anesthesiology 2008;109:657–63. doi: 10.1097/ALN.0b013e31818629db Myles PS Chan MTV Kaye DM McIlroy DR Lau CW Symons JA Chen S Effect of nitrous oxide anesthesia on plasma homocysteine and endothelial function Anesthesiology 2008109657 63 10.1097/ALN.0b013e31818629dbOpen DOISearch in Google Scholar

Badner NH, Freeman D, Spence JD. Preoperative oral B vitamins prevent nitrous oxide-induced postoperative plasma homocysteine increases. Anesth Analg 2001;93:1507–10. doi: 10.1097/00000539-200112000-00034 Badner NH Freeman D Spence JD Preoperative oral B vitamins prevent nitrous oxide-induced postoperative plasma homocysteine increases Anesth Analg 2001931507 10 10.1097/00000539-200112000-00034Open DOISearch in Google Scholar

Chai D, Cheng Y, Jiang H. Fundamentals of fetal toxicity relevant to sevoflurane exposures during pregnancy. Int J Dev Neurosci 2019;72:31–5. doi: 10.1016/j.ijdevneu.2018.11.001 Chai D Cheng Y Jiang H Fundamentals of fetal toxicity relevant to sevoflurane exposures during pregnancy Int J Dev Neurosci 20197231 5 10.1016/j.ijdevneu.2018.11.001Open DOISearch in Google Scholar

Tian Y, Wu X, Guo S, Ma L, Wei H, Zhao X. Minocycline attenuates sevoflurane-induced cell injury via activation of Nrf2. Int J Mol Med 2017;39:869–78. doi: 10.3892/ijmm.2017.2908 Tian Y Wu X Guo S Ma L Wei H Zhao X Minocycline attenuates sevoflurane-induced cell injury via activation of Nrf2 Int J Mol Med 201739869 78 10.3892/ijmm.2017.2908Open DOISearch in Google Scholar

Johnson SC, Pan A, Li L, Sedensky M, Morgan P. Neurotoxicity of anesthetics: Mechanisms and meaning from mouse intervention studies. Neurotoxicol Teratol 2019;71:22–31. doi: 10.1016/j.ntt.2018.11.004 Johnson SC Pan A Li L Sedensky M Morgan P Neurotoxicity of anesthetics: Mechanisms and meaning from mouse intervention studies Neurotoxicol Teratol 20197122 31 10.1016/j.ntt.2018.11.004Open DOISearch in Google Scholar

Olsen EA, Brambrink AM. Anesthesia for the young child undergoing ambulatory procedures: Current concerns regarding harm to the developing brain. Curr Opin Anaesthesiol 2013;26:677–84. doi: 10.1097/ACO.0000000000000016 Olsen EA Brambrink AM Anesthesia for the young child undergoing ambulatory procedures: Current concerns regarding harm to the developing brain Curr Opin Anaesthesiol 201326677 84 10.1097/ACO.0000000000000016Open DOISearch in Google Scholar

McCann ME, Berde C, Soriano S, Marmor J, Bellinger D, de Graaff JC, de Graaff JC, Dorris L, Bell G, Morton N, Dorris L, Morton N, Disma N, Giribaldi G, Withington D, Withington D, Grobler A, Stargatt R, Hunt RW, Sheppard SJ, Marmor J, Giribaldi G, Bellinger DC, Hartmann PL, Hardy P, Frawley G, Izzo F, Sternberg BS, Lynn A, Wilton N, Mueller M, Polaner DM, Absalom AR, Szmuk P, Morton N, Berde C, Soriano S, Davidson AJ, for the GAS Consortium. Neurodevelopmental outcome at 5 years of age after general anaesthesia or awake-regional anaesthesia in infancy (GAS): an international, multicentre, randomised, controlled equivalence trial. Lancet 2019;393:664–77. doi: 10.1016/S0140-6736(18)32485-1 McCann ME Berde C Soriano S Marmor J Bellinger D de GraaffJC de Graaff JC Dorris L Bell G Morton N Dorris L Morton N Disma N Giribaldi G Withington D Withington D Grobler A Stargatt R Hunt RW Sheppard SJ Marmor J Giribaldi G Bellinger DC Hartmann PL Hardy P Frawley G Izzo F Sternberg BS Lynn A Wilton N Mueller M Polaner DM Absalom AR Szmuk P Morton N Berde C Soriano S Davidson AJ for the GAS Consortium Neurodevelopmental outcome at 5 years of age after general anaesthesia or awake-regional anaesthesia in infancy (GAS): an international, multicentre, randomised, controlled equivalence trial Lancet 2019393664 77 10.1016/S0140-6736(18)32485-1Open DOISearch in Google Scholar

eISSN:
1848-6312
Languages:
English, Slovenian
Publication timeframe:
4 times per year
Journal Subjects:
Medicine, Basic Medical Science, other