Abstract
Oxidative stress occurs when reactive oxygen species (ROS) production overwhelms cell protection by antioxidants. This review is focused on general anaesthesia-induced oxidative stress because it increases the rate of complications and delays recovery after surgery. It is important to know what effects of anaesthetics to expect in terms of oxidative stress, particularly in surgical procedures with high ROS production, because their either additive or antagonistic effect may be pivotal for the outcome of surgery. In vitro and animal studies on this topic are numerous but show large variability. There are not many human studies and what we know has been learned from different surgical procedures measuring different endpoints in blood samples taken mostly before and after surgery. In these studies most intravenous anaesthetics have antioxidative properties, while volatile anaesthetics temporarily increase oxidative stress in longer surgical procedures.
Keywords: glutathione, malondialdehyde, reactive oxygen species, superoxide dismutase, TBARS
Abstract
Ovo je pregledni rad o oksidacijskom stresu uzrokovanom općim anesteticima zbog toga što uzrokuje komplikacije nakon operacije i usporava oporavak. Oksidacijski stres nastaje kada je stvaranje reaktivnih kisikovih spojeva (ROS) veće od stanične zaštite antioksidansima. Poznavanje učinka anestetika na oksidacijski stres posebice je važno pri planiranju kirurških zahvata kod kojih je poznato da uzrokuju nastanak veće količine ROS-a. Naime, antagonistički ili aditivni učinak anestetika na oksidacijski stres može kod takvih kirurških zahvata biti ključan za ishod operacije. Brojna su istraživanja o toj temi, no protokoli pokusa na staničnim kulturama i pokusnim životinjama vrlo su različiti. U istraživanjima na ljudima anestetici su upotrebljavani tijekom vrlo raznorodnih kirurških zahvata, često uz primjenu više anestetika, mjereni su različiti parametri oksidacijskoga stresa, a uzorci (najčešće krvi) uzimani su obično prije zahvata i u različitim intervalima nakon njega. Zbog svega je toga ponekad teško razlučiti kakav je učinak nekog anestetika na oksidacijski stres. Općenito intravenozni anestetici imaju antioksidacijsko djelovanje, imaju ga i plinoviti anestetici – uglavnom nakon kratkotrajne anestezije, a dugotrajna anestezija uzrokuje privremeni porast oksidacijskoga stresa.
Ključne riječi: glutation, malondialdehid, reaktivni kisikovi spojevi, SOD, TBARS
Oxidative stress is the imbalance of reactive oxygen species (ROS) production and antioxidative cell defence, which disrupts redox signalling and control and causes molecular damage (1). It is considered to be involved in the process of ageing, inflammations, cancers, degenerative diseases (2), and exposure to xenobiotics and drugs, such as anaesthetics (3). Anaesthetics-induced oxidative stress may affect lipids, proteins, and DNA. Among them, the most susceptible to oxidation are lipids (4). ROS interact with polyunsaturated fatty acids (PUFA) to form highly reactive lipoperoxides. Oxidative attack on lipids may result in the production of aldehydes such as 4-hydroxynonenal (HNE) and malondialdehyde (MDA) or prostaglandin-like compounds called F2-isoprostanes (F2-IsoPs) (5). Anaesthetics-related oxidative stress is most often measured as plasma MDA levels or thiobarbituric acid reacting substances (TBARS) (6).
Oxidative damage to proteins results in their functional impairment or loss and in increased susceptibility to proteases (7, 8). Biomarkers of oxidative stress of proteins are carbonyl groups introduced into amino acid side chains, but they are not measured frequently in studies on anaesthetics-induced oxidative stress.
DNA damage is established by upregulation of 8-hydroxydeoxyguanosine (8-OHdG) and migration of broken DNA measured either by single-cell gel electrophoresis (comet assay) or more specific comet assays such as formamidopyridine DNA N-glycolase (Fpg) or enzyme-modified and human 8-oxoguanine DNA glycosylase (hOGG1) comet assay. In human studies DNA damage is measured in peripheral blood lymphocytes. In animal studies measurements may involve other organs as well.
Oxidative stress can be prevented or decreased by natural cell antioxidant defence, which transforms highly reactive oxygen species to less reactive compounds. Among numerous antioxidants the most frequently measured are glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GSHR), and glutathione transferase (GST).
Other biomarkers of oxidative stress include total antioxidant status (TAS), total oxidative status (TOS), and oxidative stress index (OSI). OSI is the ratio between TOS and TAS and indicates the level of oxidative stress.
Surgery patients undergo procedures that can cause surgical trauma, inflammation, and ischaemia-reperfusion injury, all of which cause oxidative stress (9, 10, 11). To avoid further tissue injury caused by anaesthetics, it is very important to choose the ones that would minimise oxidative stress. This review seeks to summarise the effects of the most common intravenous and inhalation (volatile) anaesthetics in terms of oxidative stress and related genotoxic potential. We relied on preclinical (in vitro and animal) and available clinical data. Even though they are often inconclusive and even controversial, because the experiments greatly vary in cell cultures, animal species, concentrations, times of exposure, and different end-points, our goal was to find common grounds that could provide some informed advice to anaesthetists and surgeons.
Intravenous anaesthetics
Propofol
Propofol (2,6-diisopropylphenol) is a widely used anaesthetic with many favourable properties. It reduces intracranial pressure and prevents inflammation and convulsions (12). Its pharmacokinetics in adults has been aptly described by Kanto and Gepts (13) and in children by Rigby-Jones and Sneyd (14). Children have higher clearance than adults and need higher induction and maintenance doses to achieve the same propofol blood concentration.
As propofol contains a phenolic OH-group, in vitro studies suggest that it has a high potential to prevent lipid peroxidation (15) by scavenging lipidperoxyl radicals (LOO˙) and could replace α-tocopherol in that respect (16). It also seems to strengthen the GSH system in rat cells by inhibiting GPx and increasing GSHR and GST activity (17).
Genotoxicity studies seem to agree that propofol does not damage DNA. In primary rat astroglial cells it has been reported to protect DNA from lesions by scavenging peroxynitrite (18). In patients who underwent ear surgery propofol did not cause DNA damage in peripheral blood lymphocytes (19, 20). Similar was reported for 21 patients who underwent open-heart surgery and showed no increase in chromosomal aberrations of peripheral blood lymphocytes (21).
In rat studies in vivo, propofol has been reported to protect from kidney (22), heart (23), and liver ischaemia/reperfusion injury (24), decrease lipid peroxidation in the spinal cord after injury (25), attenuate postoperative signs of organ injury after liver transplantation (26, 27), protect from obstructive jaundice (28), hepatocyte necrosis, and infiltration of inflammatory cells, and restore liver architecture after halothane-induced intoxication (29).
In dogs anaesthetised with propofol, however, findings are less straightforward regarding total antioxidant capacity (TAC). One study reported unchanged TAC up to 48 h after gas evacuation for pneumoperitoneum (30), and another study a significant decrease in TAS with unchanged TOS at the end of anaesthesia for laparotomy and gastrotomy, which resulted with significant increase in OSI (TOS and TAS ratio) (31). A study on rats did not give consistent results regarding plasma TBARS, GSH/GSSG, SOD, CAT, and GSHR (32).
Findings in humans are generally favourable. One study (33) showed no significant plasma MDA changes in 29 liver donors anaesthetised with propofol before and after surgery. Another study (19) in 15 patients who underwent minor surgery and were receiving propofol anaesthesia for two hours showed increased plasma TAS and tocopherol concentrations, which confirmed its antioxidative properties. One clinical trial (34) comparing 50 patients under propofol anaesthesia and 50 patients on ketamine reported lower blood lipid peroxidation, GPx, and SOD activity in the first group. In 10 children with acyanotic heart disease, plasma MDA, GSH, lactate, and pyruvate concentrations remained unchanged after surgery (35). One study with 12 patients who underwent laparoscopic cholecystectomy (36) showed a significant drop in plasma MDA 1 min before desufflation and 20 min after desufflation when compared with concentrations before insufflation. In contrast, another study (37) reported higher plasma MDA concentrations in patients (N=17) undergoing partial hepatectomy after 60 min of propofol anaesthesia. These returned to preoperative values 24 h after surgery. A comparative study in patients with radical oesophagectomy showed significantly lower incidence of severe postoperative complications with propofol (7 of 92) than with sevoflurane anaesthesia (18 of 94) (P=0.03) (38).
Because of its antioxidative properties, propofol has, in fact, been proposed for application other than anaesthetic and sedative. In cultured neonatal rat cardiomyocytes it reduced doxorubicin-induced stress, and the authors of the study suggested that it could be used as adjuvant in doxorubicin therapy (39). It was also suggested for the treatment of glioblastoma, as it triggered apoptosis in human brain glioblastoma multiforme cells by decreasing Bcl-2 and increasing Bax and caspase activity (40). It also showed beneficial effects in critically ill patients with systemic inflammatory response and sepsis (41).
Thiopental
Thiopental or penthiobarbital (2-thio-5-ethyl-5-sec-pentylbarbituric acid) is a rapid-onset short-acting general anaesthetic. Even though it has largely been replaced by propofol, it is still used for rapid induction of anaesthesia in emergency cases. Research of its antioxidative and immunomodulating properties in human neutrophils showed a significant drop in O2-, H 2O 2, and OH levels in the presence of clinically relevant thiopental plasma concentrations (30 μg/L) (42). Thiopental also showed anti-inflammatory properties by significantly reducing chemotaxis and phagocytosis (43).
Animal studies of thiopental effects on oxidative stress report uneven findings. In rats with spinal cord contusion injury it showed beneficial effects, because spinal cord MDA was significantly lower than in control rats not treated with thiopental (25). In another study (44), in contrast, rats showed higher MDA and lower GSH, GPx, and GSHR levels in brain, heart, and bronchial tissues. In dogs with surgical trauma thiopental significantly increased plasma TOS, while TAS remained unchanged, which resulted with increased OSI (31).
In humans, the study with patients who underwent laparoscopic cholecystectomy (36) showed no changes in plasma MDA throughout the whole procedure, which suggests antioxidative effects of thiopental.
Ketamine
Ketamine (C13H16ClNO) is an N-methyl-D-aspartate (NMDA) receptor antagonist (45).
In vitro, ketamine was reported to produce toxic effects only at levels much higher than those corresponding to anaesthetic concentrations (46, 47). When they did correspond to plasma concentrations in anaesthesia (30 μg/mL), human neutrophil ROS (O2-, H2O2, and OH) production did not differ from controls (43).
In rats, ketamine showed protective effects against oxidative stress. In fact, ketamine anaesthesia resulted in significantly lower kidney and liver tissue MDA than propofol, thiopental, and fentanyl (28, 48). In another study (44), the authors attributed significantly higher MDA and lower GSHR and GPx levels to ketamine-triggered boost of adrenaline, which is known to induce oxidative stress (49). In mice, ketamine also turned out to mitigate (secondary) brain injury which occurs when ROS production overwhelms the antioxidant system (50). It lowered brain MDA content in these mice and increased GPx, SOD, and NRf2 activity. These effects were accompanied by a significant reduction of brain water content and improved brain function (grip test) score.
Reports of ketamine effects in humans are positive. In adult and paediatric patients with cardiopulmonary bypass (CPB) ketamine mitigated the systemic inflammatory response syndrome (SIRS) by reducing proinflammatory IL-6 and IL-8, and/or increasing anti-inflammatory IL-10 (51, 52, 53).
Etomidate
Etomidate (R-1-(1-ethylphenyl)imidazole-5-ethyl ester) is a short-acting intravenous anaesthetic, whose R (+) enantiomer has a much higher hypnotic activity than the S (-) enantiomer (54). It has been in clinical use since 1972 thanks to high therapeutic index and minimal effects on blood pressure and breathing. However, its use as anaesthetic has been limited since reports of serious adrenocortical suppression and myoclonus were published (55). It is no longer used to induce anaesthesia in critically ill patients, because adrenal suppression decreases cortisol release and thus weakens anti-inflammatory response. This means longer stays at intensive care units and higher mortality rate (56).
In vitro, etomidate does not affect TBARS production and the glutathione defence system (GPx, GSHR, GST) (17).
Similar was observed in animal studies. It does not seem to affect MDA levels or change SOD activity (57) in rat brain, while it significantly decreased MDA and increased GSH levels in the spinal cord after injury (58).
In human studies, its effect on plasma MDA was compared with that of thiopental and propofol. In patients who underwent laparoscopic cholecystectomy it fared worse than either (36), as it increased MDA levels before and after desufflation compared to other anaesthetics. In another study with 60 patients who underwent tibial fracture surgery (59) etomidate did not affect SOD activity, and patients receiving it had shorter hospitalisation time because of fewer postoperative complications such as numbness, lower limb pain, and coldness than patients on propofol.
Inhalational (volatile) anaesthetics
Sevoflurane
Sevoflurane [fluoromethyl-2,2,2trifluoro-1-(trifluoromethyl) ethyl ether] has been used for general anaesthesia since the 1990s. It has a lower blood-gas partition coefficient than other volatile anaesthetics, which allows rapid induction of anaesthesia and quick reawakening. Because it does not cause irritation, acts as bronchodilator, and barely has any smell, it is the anaesthetic of choice in children (60), especially those with respiratory infections, airway hyper-reactivity, and seasonal allergies (61).
Genotoxicity studies point to none to low DNA damage. In one study (62), alkaline comet assay showed no sevoflurane genotoxicity in human peripheral lymphocytes, but in another (63) DNA damage was found in kidney homogenates of rats exposed for two hours over three consecutive days. Excessive exposure in this experiment may explain why the comet tail length and intensity increased gradually until the end of experiment (24 h). In peripheral lymphocytes of patients who underwent minimally invasive surgery that lasted two hours, the assay revealed no DNA damage (20), and no damage was reported in peripheral lymphocytes of children under sevoflurane anaesthesia for about one hour (64). In patients with lower abdominal surgery DNA damage was short-lived and tail parameters started to return to normal on the third day, and repair was complete five days after anaesthesia (65).
In animal studies, findings of its effects on oxidative stress parameters vary, however. One study (66) reported higher GST and SOD activity and TBARS level in the liver of young and adult female rats after sevoflurane anaesthesia. In adult male rats it also increased MDA in the lungs, but decreased it in the kidney and brain (67). It also significantly reduced liver MDA levels in rats with ischaemia/reperfusion injury (57). Compared to isoflurane in another study (68), it produced significantly lower TBARS levels.
Several human studies suggest that sevoflurane does not increase oxidative stress, especially in minor surgeries (3), and where it does (as in major surgeries such as orthopaedic, cholecystectomy, hysterectomy, and alike) oxidative stress parameters return to normal 24–48 h after exposure has stopped. In patients who underwent laparoscopic surgery it did not increase plasma MDA and protein carbonyls (69) nor did it affect MDA, GSH, lactate, and pyruvate levels in peripheral blood of ten acyanotic children who underwent elective cardiac surgery (35) or GPx activity and TOS in 15–50-year-old patients who underwent laparoscopic cholecystectomy (70), while their TAS significantly increased. In a study of 12–36 months old children undergoing hypospadias repair surgery (71) sevoflurane increased SOD, GPx, and caspase-3 mRNA levels two hours after surgery, but these returned to normal three days later. The authors conclude that sevoflurane temporarily increases oxidative stress in children.
Desflurane
Desflurane (1,2,2,2-tetrafluoroethyl difluoromethyl ether) has the blood-gas partition coefficient lower than any other anaesthetic, which grants rapid recovery from general anaesthesia. It is contraindicated in children because of its pungent smell, which makes children hold their breath, cough, and salivate profusely (61).
Animal studies report controversial findings. One study comparing the effects of desflurane on oxidative stress in pig peripheral blood and bronchoalveolar lavage (BAL) fluid with those of sevoflurane and propofol (72) reported significantly higher MDA levels and lower GPx activity. Another, comparing it with sevoflurane, reported no brain accumulation of IL-6 and TNF-a or cognitive impairment in six days old mice (73).
Clinical studies also report inconclusive findings. In mothers with elective Caesarean section serum TOS and OSI were significantly lower after surgery than before it (74), but compared to sevoflurane, these parameters were significantly higher, especially in umbilical artery blood, which also had higher LOOH. In patients with laparoscopic cholecystectomy (70) it increased total oxidant capacity, but did not affect TAS and GPx activity. Similar were the findings reported for Turkish patients who underwent unspecified elective surgery (75): while serum GPx and erythrocyte SOD activity did not change, MDA levels soared, and α-tocopherol plummeted in the first hour after surgery, but both returned to baseline values 12 h later. This transitory increase in MDA and protein carbonyl content was also reported six hours after desflurane anaesthesia (given together either with fentanyl or N2O), which started to returned to normal 24 h later (69).
As for its genotoxic potential, one study reports that desflurane does not cause DNA damage if used for minor surgeries for at least 90 minutes (76), while another study (77) revealed significantly higher 8-OHdg concentrations in plasma samples in addition higher comet tail length and intensity in BAL samples taken after anaesthesia, which points to oxidative stress as the mechanism of genotoxicity (77).
Isoflurane
Isoflurane (2-chloro-2-(difluoromethoxy)1,1,1-trifluoro-ethane) has been used as anaesthetic since the 1980s (3, 20) thanks to particularly (s)low metabolism and solubility, which results in very short induction and recovery time.
Isoflurane has antioxidative properties in vitro (3, 78), but in vivo it induces oxidative stress by increasing lipid peroxidation (68). In an animal experimental model simulating liver transplantation isoflurane induced significantly higher TBARS levels than sevoflurane.
Clinical studies evidence some oxidative stress. In one, isoflurane increased plasma MDA in patients undergoing donor hepatectomy, but it did not affect TOS, TAC, and SOD activity (33). Plasma SOD and GPx activities were also not affected in another study (79).
Genotoxicity/mutagenicity studies generally suggest no effects (77, 80), especially not after a minor surgery (19, 20). However, after a major lower abdominal surgery, one study (65) reported significantly increased comet assay parameters in peripheral blood lymphocytes two hours after the beginning of isoflurane anaesthesia. These parameters returned to normal three days later.
Nitrous oxide
Having the lowest potency among inhalational anaesthetics, nitrous oxide (N2O) is often combined with more powerful ones (81) or used as anxiolytic. As it does not cause addiction but causes brief (several minutes long) bursts of euphoria, dissociation, and excitement, it is not prohibited by law, and people have also been using it for recreational purposes ever since 1775 (laughing gas parties) (82).
Even so, its use as anaesthetic has been in decline over postoperative nausea and vomiting, which are more pronounced if used for more than one hour (83). However, the European Society of Anaesthesiology believes that its benefits outweigh the adverse effects and find no real arguments to abandon it.
N2O is not mutagenic in bacterial and mammalian cell systems (84). It is teratogenic to embryos of pregnant animals exposed to high doses of N2O for 24 h during organogenesis but is not carcinogenic (76, 84). However, one study in nurses showed a positive correlation between the level of exposure to N2O and oxidative DNA damage measured with the Fpg-modified comet assay (85). These nurses also had significantly higher lymphocyte ROS and plasma and urine TBARS levels and lower GPx activity than unexposed healthcare workers.
It is known that N2O irreversibly inactivates methionine synthase (86), which leads to increased concentrations of homocysteine in plasma (87), which, in turn, inhibits the expression of antioxidant enzymes, including GPx. This effect can be counteracted by oral vitamin therapy (folate 2.5 mg, B6 25 mg and B12 500 mg) for one week before surgery (88). In addition, N2O causes endothelial dysfunction in patients with cardiovascular disease undergoing non-cardiac surgery, which, in turn, increases the risk of postoperative cardiac events and mortality (87). One clinical study in patients anaesthetised with desflurane with or without N2O (76) found no difference in lipid peroxidation, protein carbonyls, DNA damage, and ferric-reducing antioxidant power, which indicates that N2O does not increase oxidative stress.
Volatile anaesthetic developmental neurotoxicity
One specific aspect of concern with anaesthetics – and particularly with sevoflurane, which is the most frequently used anaesthetic in paediatric surgery – is their neurotoxic potential. Considering that anaesthetics are lipophilic and readily pass the placental barrier, they may cause brain injury in foetuses and children. Judging by animal model studies (reviewed in ref. 89), exposure to sevoflurane during uterine quiescence can induce neuroinflammation, neuroapoptosis, and eventually memory impairment. Cognitive impairment was also reported in young mice repeatedly exposed to sevoflurane, which caused an increase in proinflammatory proteins IL-6 and TNF-α (73). IL-6 and Nrf2 were also increased in rats exposed to sevoflurane for five hours (90). Another review article (91) of preclinical intervention studies in neonatal mice suggests that volatile anaesthetics can lead to oxidative stress-related acute neurotoxicity and learning and memory deficits later in life. The risks of attention deficit, hyperactivity disorder, disabilities in language acquisition, and disabilities in abstract reasoning after prolonged anaesthesia have also been highlighted in a review article by Olsen and Brambrink (92). However, a randomised multicentre controlled trial following up 722 children for two and five years (93) after their one-hour general anaesthesia reported no significant neurocognitive or behavioural deficits.
Conclusions
Research of oxidative/antioxidative effects of anaesthetics has produced ambiguous findings so far, which stem from differences in exposure related to specific surgical procedures, length of anaesthesia, and timing of blood and tissue sampling. Even so, intravenous anaesthetics generally have antioxidant properties and short exposure to volatile anaesthetics does not seem to increase oxidative stress, while the effects of longer exposure are transient. Transient anaesthetics-induced oxidative stress may be the most relevant for the outcome of surgery with procedures that cause oxidative stress themselves. In such cases anaesthetics with antioxidative properties are drugs of choice.
Abbreviations
- CPB
cardiopulmonary bypass
- F2-IsoPs
F2- isoprostanes
- CAT
catalase
- GPx
glutathione peroxidase
- GSH
glutathione
- GSHR
glutathione reductase
- GST
glutathione transferase
- HNE
4-hydroxynonenal
- LOO˙
lipidperoxyl radicals
- MDA
malondialdehyde
- 8-OHdG
8-hydroxydeoxyguanosine
- OSI
oxidative stress index
- PUFA
polyunsaturated fatty acids
- ROS
reactive oxygen species
- SIRS
systematic inflammatory response syndrome
- SOD
superoxide dismutase
- TAC
total antioxidative capacity
- TAS
total antioxidative status
- TBARS
thiobarbituric acid reacting substances
- TOS
total oxidative status
References
- 1.Jones DP. Redefining oxidative stress. Antioxid Redox Signal. 2006;8:1865–79. doi: 10.1089/ars.2006.8.1865. [DOI] [PubMed] [Google Scholar]
- 2.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.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. [DOI] [PubMed] [Google Scholar]
- 5.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. [DOI] [PubMed] [Google Scholar]
- 6.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. [DOI] [PubMed] [Google Scholar]
- 7.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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. [DOI] [PubMed] [Google Scholar]
- 9.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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. [DOI] [PubMed] [Google Scholar]
- 11.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. [DOI] [PubMed] [Google Scholar]
- 12.Marik PE. Propofol: An immunomodulating agent. Pharmacotherapy. 2005;25:28S–33S. doi: 10.1592/phco.2005.25.5_Part_2.28S. [DOI] [PubMed] [Google Scholar]
- 13.Kanto J, Gepts E. Pharmacokinetic implications for the clinical use of propofol. Clin Pharmacokinet. 1989;17:308–26. doi: 10.2165/00003088-198917050-00002. [DOI] [PubMed] [Google Scholar]
- 14.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. [DOI] [PubMed] [Google Scholar]
- 15.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. [DOI] [PubMed] [Google Scholar]
- 16.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. [DOI] [PubMed] [Google Scholar]
- 17.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. [DOI] [PubMed] [Google Scholar]
- 18.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. [DOI] [PubMed] [Google Scholar]
- 19.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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. [DOI] [PubMed] [Google Scholar]
- 21.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. [DOI] [PubMed] [Google Scholar]
- 22.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. [DOI] [PubMed] [Google Scholar]
- 23.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. [Google Scholar]
- 24.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. [DOI] [PubMed] [Google Scholar]
- 25.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. [DOI] [PubMed] [Google Scholar]
- 26.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.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. [DOI] [PubMed] [Google Scholar]
- 29.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. [DOI] [PubMed] [Google Scholar]
- 30.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. [DOI] [Google Scholar]
- 31.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. [DOI] [PubMed] [Google Scholar]
- 32.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. [DOI] [PubMed] [Google Scholar]
- 33.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. [DOI] [PubMed] [Google Scholar]
- 34.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. [PubMed] [Google Scholar]
- 35.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. [DOI] [PubMed] [Google Scholar]
- 36.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. [DOI] [PubMed] [Google Scholar]
- 37.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. [DOI] [PubMed] [Google Scholar]
- 38.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.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. [DOI] [PubMed] [Google Scholar]
- 40.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. [DOI] [PubMed] [Google Scholar]
- 41.Baronica R, Tonković D, Bačić G, Karadjole T, Šuran J, Bačić Baronica K. Croatian International Symposium on Intensive Care Medicine / Simpozij intenzivne medicine s međunarodnim sudjelovanjem. Neurol Croat. 2013;62(Suppl 2):1–170. editors. [Google Scholar]
- 42.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. [DOI] [PubMed] [Google Scholar]
- 43.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. [DOI] [PubMed] [Google Scholar]
- 44.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Dinis-Oliveira RJ.. Metabolism and metabolomics of ketamine: a toxicological approach. Forensic Sci Res. 2017;2:2–10. doi: 10.1080/20961790.2017.1285219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.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. [DOI] [PubMed] [Google Scholar]
- 50.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.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. [DOI] [PubMed] [Google Scholar]
- 52.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. [DOI] [PubMed] [Google Scholar]
- 53.Durandy Y. Minimizing systemic inflammation during cardiopulmonary bypass in the pediatric population. Artif Organs. 2014;38:11–8. doi: 10.1111/aor.12195. [DOI] [PubMed] [Google Scholar]
- 54.Forman SA. Clinical and molecular pharmacology of etomidate. Anesthesiology. 2011;114:695–707. doi: 10.1097/ALN.0b013e3181ff72b5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Raines DE. The Pharmacology of Etomidate and Etomidate Derivatives. Int Anesthesiol Clin. 2015;53:63–75. doi: 10.1097/AIA.0000000000000050. [DOI] [PubMed] [Google Scholar]
- 56.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. [DOI] [PubMed] [Google Scholar]
- 57.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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. [DOI] [PubMed] [Google Scholar]
- 59.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.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. [Google Scholar]
- 61.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. [DOI] [PubMed] [Google Scholar]
- 62.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. [PubMed] [Google Scholar]
- 63.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. [DOI] [PubMed] [Google Scholar]
- 64.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. [DOI] [PubMed] [Google Scholar]
- 65.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. [DOI] [PubMed] [Google Scholar]
- 66.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. [DOI] [PubMed] [Google Scholar]
- 67.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. [DOI] [PubMed] [Google Scholar]
- 68.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. [DOI] [PubMed] [Google Scholar]
- 69.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. [DOI] [PubMed] [Google Scholar]
- 70.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. [DOI] [PubMed] [Google Scholar]
- 71.Zhou X, Lu D, da Li W, hui Chen X, yu Yang X, Chen X, bin Zhou Z, 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. [DOI] [PubMed] [Google Scholar]
- 72.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. [DOI] [PubMed] [Google Scholar]
- 73.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.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. [DOI] [PubMed] [Google Scholar]
- 75.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. [DOI] [PubMed] [Google Scholar]
- 76.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. [DOI] [PubMed] [Google Scholar]
- 77.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.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. [DOI] [PubMed] [Google Scholar]
- 79.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. [DOI] [PubMed] [Google Scholar]
- 80.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. [DOI] [PubMed] [Google Scholar]
- 81.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. [DOI] [PubMed] [Google Scholar]
- 82.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. [DOI] [PubMed] [Google Scholar]
- 83.Hert SGD. The current place of nitrous oxide in clinical practice. Eur J Anaesthesiol. 2015;32:517–20. doi: 10.1097/EJA.0000000000000264. [DOI] [PubMed] [Google Scholar]
- 84.American Society of Anesthesiologists. Anesthetic Gases: Information for Management in Anesthetizing Areas and Postanesthesia Care Unit. Park Ridge (IL): ASA; 1999. [Google Scholar]
- 85.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. [DOI] [PubMed] [Google Scholar]
- 86.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. [DOI] [PubMed] [Google Scholar]
- 87.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. [DOI] [PubMed] [Google Scholar]
- 88.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. [DOI] [PubMed] [Google Scholar]
- 89.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. [DOI] [PubMed] [Google Scholar]
- 90.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.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. [DOI] [PubMed] [Google Scholar]
- 93.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. 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. for the GAS Consortium. [DOI] [PMC free article] [PubMed] [Google Scholar]
