A RAPIDLY growing body of evidence indicates that volatile anesthetics protect myocardium against reversible and irreversible ischemic injury. Identifying the mechanisms by which volatile agents mediate these antiischemic actions is the subject of intense research. This objective has been difficult to accomplish because volatile anesthetics also profoundly affect cardiovascular function. Volatile agents reduce arterial and coronary perfusion pressure, cause dose-related depression of myocardial contractility, produce coronary vasodilation, affect electrophysiologic function, and modify autonomic nervous system activity to varying degrees. Therefore, the antiischemic effects of volatile anesthetics may be mediated, at least in part, by favorable alterations in myocardial oxygen supply-demand relations, preservation of energy-dependent cellular functions, and increased coronary blood flow. However, it seems unlikely that changes in myocardial metabolism and coronary perfusion caused by volatile anesthetics are solely responsible for protection against ischemic damage. Instead, several endogenous signal transduction pathways, acting through the adenosine triphosphate (ATP)-sensitive potassium (KATP) channel and involving the generation of reactive oxygen species (ROS), have been implicated in mediating the antiischemic actions of volatile anesthetics. The experimental and clinical findings documenting the phenomenon of volatile anesthetic preconditioning against ischemic injury of myocardium are evaluated. Recent findings in vitro  and in vivo  that seek to define the intracellular mechanisms responsible for these beneficial actions are also summarized.

The antiischemic effects of volatile anesthetics were initially proposed more than 20 yr ago. Lowenstein et al.  1demonstrated that halothane reduced ST segment elevation in a canine model of brief coronary artery occlusion. These data were consistent with the hypothesis that exposure to halothane reduced acute ischemic injury. A subsequent study by this research group also demonstrated that halothane reduced myocardial infarct size when administered before prolonged coronary artery occlusion in dogs. 2Lactate production was decreased in the presence as compared with the absence of enflurane during demand-induced ischemia produced by a critical coronary artery stenosis and ventricular pacing when coronary perfusion pressure was maintained. 3These results suggested that myocardial metabolism may be improved by administration of a volatile agent during an ischemic episode independent of alterations in hemodynamics. The relative importance of these early findings was initially overshadowed 4by a series of reports published in the mid-1980s 5–8suggesting that isoflurane may be capable of producing an abnormal redistribution of coronary blood flow away from ischemic toward normal myocardium. 9–11This “coronary steal” phenomenon was attributed to the coronary vasodilating properties of isoflurane that are known to occur primarily in arterioles of less than 100 μm in diameter. 12Isoflurane was thought to be capable of directly producing myocardial ischemia in susceptible patients with “steal-prone” coronary artery anatomy under certain hemodynamic conditions 13in a fashion similar to that of potent coronary vasodilators (e.g. , adenosine, chromonar, dipyridamole).

The implication that isoflurane might produce myocardial ischemia through such a steal mechanism was subsequently dispelled by several investigations conducted in animal models 14–17and humans with coronary artery disease. 18–20For example, isoflurane did not selectively redistribute blood flow away from the collateral-dependent region in a chronically instrumented canine model of multivessel coronary artery disease. 15In contrast, adenosine produced marked coronary steal by preferentially shunting blood flow away from collateral-dependent myocardium in this model. 15Other studies 14,16–20also suggested that isoflurane-induced hypotension may reduce myocardial perfusion, but true coronary steal did not occur when coronary perfusion pressure was maintained. Subsequent investigations with the newer volatile anesthetics sevoflurane 21,22and desflurane 23showed that these drugs also did not reduce or abnormally redistribute coronary collateral blood flow. Therefore, despite initial concerns, volatile anesthetics were subsequently shown to be relatively weak coronary vasodilators that are incapable of causing coronary steal under the vast majority of clinical conditions. 24 

Contrary to the hypothesis that the use of volatile anesthetics may be potentially deleterious in some patients with coronary artery disease, many laboratory and clinical investigations conducted since the resolution of the coronary steal controversy have convincingly shown that volatile anesthetics protect  the heart against ischemia and reperfusion injury. In addition to previously cited studies suggesting that halothane 1,2and enflurane 3exerted antiischemic effects, halothane was also shown to preserve contractile function and ultrastructural integrity during cardioplegic arrest. 25This latter study was of considerable interest because these data indicated that halothane was capable of exerting a cardioprotective effect completely independent of improvements in myocardial oxygen supply-demand balance. In addition, halothane, 26enflurane, 26desflurane, 26–28and sevoflurane 26–31have been shown to reduce myocardial damage when administered during reperfusion after prolonged coronary occlusion or cardioplegic arrest. Another study showed that preservation of high-energy phosphate concentrations was coupled to the protective effects of enflurane. 32Isoflurane and desflurane did not depress but modestly enhanced left ventricular diastolic function during acute coronary occlusion in dogs. 33Halothane, 34–36enflurane, 34,37,38isoflurane, 34,35,37and sevoflurane 39were shown to improve the functional recovery of isolated hearts subjected to global ischemia and reperfusion. Halothane and isoflurane markedly augmented the recovery of regional contractile function of stunned myocardium in vivo . 40,41Halothane 2and isoflurane 42reduced myocardial infarct size in dogs, and this beneficial action was found to persist despite discontinuation of the volatile anesthetic before coronary artery occlusion. 42The myocardium acted as if it had “remembered” the previous exposure to the volatile agent. This phenomenon was termed anesthetic-induced preconditioning  (APC) 43and was characterized by a short-term memory phase similar to that observed during ischemic preconditioning (IPC).

Anesthetic-induced preconditioning has also been described in other animal species, including rats 44and rabbits. 43The efficacy of APC conferred by isoflurane to reduce infarct size has been shown to be dose dependent in rats, 44an animal model with minimal coronary collateral flow. 45High concentrations of isoflurane may also have greater efficacy to protect myocardium during conditions of low coronary collateral blood flow in dog myocardium. 46Similarly, isoflurane and sevoflurane dose-dependently preserved the viability of isolated cardiac myocytes during ischemia. 47Isoflurane has been shown to elicit cardioprotective effects after discontinuation for 15 min or 30 min before coronary artery occlusion in rats and rabbits 43,44or dogs, 42respectively. In contrast, sevoflurane did not exert antiischemic actions after a 30-min washout period. 48Taken together, these data suggest that the memory period associated with APC may differ between volatile anesthetics and among species. Interestingly, recent findings showed that isoflurane reduced myocardial damage when administered 24 h before coronary artery occlusion and reperfusion in rabbit hearts in vivo . 49Pretreatment with isoflurane also preserved endothelial and vascular smooth muscle cell viability 12–48 h after cytokine-induced injury. 50Therefore, volatile anesthetics also produce a late phase (i.e. , a second window) of myocardial protection similar to IPC. In addition, sevoflurane reduced the duration of a brief ischemic episode required to protect against infarction during IPC. 48Sevoflurane also enhanced cardioprotection when administered 24 h after an initial IPC stimulus. 51These important findings showed that administration of a volatile anesthetic combined with a brief ischemic event synergistically protects myocardium against subsequent damage as well.

Additional data about the effects of volatile agents on the coronary circulation also stand in contrast with the conclusions implicated by the coronary steal hypothesis. These results indicated that volatile agents are certainly not deleterious to but may instead exert beneficial actions on coronary collateral perfusion to ischemic myocardium. Volatile anesthetics have been shown to produce coronary vasodilation by activating KATPchannels 39,52–55or by favorably affecting intracellular Ca2+homeostasis in vascular smooth muscle. 56Halothane attenuated reductions in coronary collateral perfusion associated with acute coronary occlusion and also improved the myocardial oxygen supply-demand relation in collateral-dependent myocardium. 57In addition, halothane reduced cyclical changes in coronary blood flow and prevented the development of platelet thrombi in the presence of a critical coronary artery stenosis. 58Sevoflurane increased collateral blood flow to ischemic myocardium when perfusion pressure was maintained. 21,59Sevoflurane also improved the functional recovery of coronary vascular reactivity and nitric oxide release in isolated hearts after global ischemia. 39Lastly, volatile anesthetics attenuated neutrophil and platelet aggregation 60and also inhibited cytokine-induced cell death 50,61after ischemia-reperfusion injury in vitro . The results of these studies collectively show the protection against ischemia and reperfusion injury may be at least partially based on favorable effects of volatile agents on coronary perfusion.

The precise mechanisms responsible for volatile anesthetic-induced protection against ischemic injury remain unclear despite extensive study. Although it is clear that volatile anesthetics may indirectly improve myocardial oxygen supply-demand relations or enhance coronary collateral perfusion, it is equally clear that these actions are not entirely responsible for the antiischemic effects of these agents. This contention is emphasized by findings showing that volatile anesthetics conferred protection during cardioplegic arrest 25and during reperfusion, 26–30conditions in which myocardial oxygen supply-demand relations play little if any role. Similarly, isoflurane and sevoflurane increased the viability of isolated cardiac myocytes, 47and sevoflurane 62and desflurane 63improved contractility of isolated cardiac muscle exposed to simulated ischemia. These results were initially attributed to reductions in excessive intracellular Ca2+concentrations during ischemia and reperfusion 64produced by partial inhibition of Ca2+channel activity. 65–68However, this relatively generic Ca2+hypothesis did not address the precise mechanisms or provide deeper insight into the intracellular processes by which volatile anesthetics exert protective effects in the intact heart.

KATPChannels

The signal transduction pathways involved in APC bear striking similarity to those responsible for IPC. It is hypothesized that volatile anesthetics stimulate a trigger that initiates a cascade of events leading to activation of an end-effector that is responsible for resistance to injury. To date, adenosine type 1 (A1) receptors, 34,69,70protein kinase C (PKC), 34,71,72inhibitory guanine nucleotide binding (Gi) proteins, 73ROS, 74–76and mitochondrial and sarcolemmal KATP(mito KATPand sarc KATP, respectively) channels 42,77–79have been shown to mediate APC (fig. 1). KATPchannels are heteromultimeric complexes containing an inward-rectifying potassium (Kir) channel and a sulfonylurea receptor (SUR). 80Pharmacologic and recombinant techniques indicate that sarc KATPand mito KATPchannels 81,82are composed of the Kir6.2/SUR2A and Kir6.1/SUR1 isoforms, 83respectively. KATPchannel opening was initially implicated as the central end-effector during APC, 84similar to the findings during studies of the mechanisms responsible for IPC. 85,86Isoflurane and sevoflurane preserved myocardial viability in a cellular model of ischemia, and this protective effect was abolished by the selective mito KATPchannel antagonist 5-hydroxydecanoate (5-HD) but not the selective sarc KATPchannel antagonist HMR-1098. 47Isoflurane, 69sevoflurane, 62and desflurane 63but not halothane 69enhanced the recovery of contractile force of isolated human right atrial trabeculae after hypoxia and reoxygenation. The nonselective KATPchannel blocker glyburide (glibenclamide) or 5-HD inhibited this protective effect. HMR-1098 also attenuated the beneficial actions produced by sevoflurane in isolated human atria. 62Glyburide blocked the enhanced recovery of contractile function produced by isoflurane in stunned myocardium in vivo . 41,87Reductions in canine myocardial infarct size produced by isoflurane 42and the ATP-sparing effects of this agent 88have been shown to be blocked by glyburide as well. 5-HD also inhibited preconditioning by isoflurane in rats 44and rabbits. 77Both 5-HD and HMR-1098 abolished the protective effects of desflurane against ischemia and reperfusion injury in dogs, 78supporting a role for both mito KATPand sarc KATPchannels in APC. In contrast, another study showed that HMR-1098 did not modify desflurane-induced preconditioning in isolated human right atria in vitro . 63Therefore, some controversy continues to exist about the relative contribution of sarc KATPand mito KATPchannels in APC.

Carefully conducted in vitro  experiments suggest that volatile anesthetics are capable of modifying KATPchannel activity. Isoflurane stimulated outward K+current through sarc KATPchannels in isolated ventricular myocytes during patch clamping. 89,90Volatile anesthetics also reduced sarc KATPchannel sensitivity to inhibition by ATP, thereby increasing open state probability. 91In contrast, other patch clamp results suggested that volatile agents alone did not open KATPchannels. Isoflurane did not affect sarc KATPchannel current in human atrial cells 69and also inhibited sarc KATPchannel activity in rabbit ventricular myocytes. 91However, some volatile anesthetics were able to enhance sarc KATPchannel current by facilitating channel opening after initial activation. 89,90Isoflurane enhanced sarc KATPchannel opening in the presence of the mitochondrial uncoupler 2,4-dinitrophenol, the KATPchannel opener pinacidil, and the protein tyrosine kinase (PTK) inhibitor genistein in a whole cell patch clamp model. 89,92Activation of PKC, 90adenosine receptors, 93and phosphatidylinositol kinase 93seemed to be necessary for this process to occur. Isoflurane also directly opened sarc KATPchannels during intracellular acidosis, a condition that is known to occur during ischemia. 94These data suggest that volatile anesthetics may not directly interact with sarc KATPchannels but instead may affect other signaling elements that modulate sarc KATPchannel activity. In contrast with the findings with isoflurane, halothane had no effect on pinacidil-induced increases in sarc KATPchannel current and even inhibited KATPchannel current that had been maximally activated by 2,4-dinitrophenol. 89The anesthetic specificity for APC remains to be well characterized, although studies such as these do suggest important differences in efficacy may exist among individual agents.

The ability of volatile anesthetics to directly open mito KATPchannels has also been examined. Isoflurane and sevoflurane increased mitochondrial flavoprotein oxidation, an index of mito KATPchannel activity, in guinea pig cardiac myocytes. 95This process was inhibited by 5-HD. 95Flavoprotein fluorescence may not be entirely specific for mito KATPchannel opening, 96but isoflurane has also been shown to directly activate mito KATPchannels reconstituted in lipid bilayers. 97In contrast with these intriguing findings, 97Zaugg et al.  47recently demonstrated that although isoflurane or sevoflurane did not directly enhance flavoprotein oxidation in rat ventricular myocytes, these volatile agents did potentiate increases in fluorescence produced by the selective mito KATPchannel agonist diazoxide. These results suggested that volatile anesthetics may not directly open but instead act to prime mito KATPchannels, thus enhancing their ability to open in response to an agonist. Sarc KATPchannels may also be linked to the function of the mitochondrial inner membrane. For example, ROS generated by mitochondria may act to open sarc KATPchannels. 982,4-Dinitrophenol-induced activation of sarc KATPchannel current was reversible and accompanied by nicotinamide adenine dinucleotide oxidation, suggesting the existence of cross-talk between mito KATPand sarc KATPchannels. 99Taken as a whole, the preponderance of evidence collected to date implies that volatile anesthetics do not necessarily directly open KATPchannels but instead prime the activation of these channels in both sarcolemmal and mitochondrial membranes.

Adenosine triphosphate-sensitive potassium channels in vascular smooth muscle cells have been shown to be essential regulators of coronary vascular tone when ATP production is reduced. 100Volatile anesthetic-induced coronary vasodilation 39,52–55was attenuated by glyburide, indicating an important role for KATPchannels in this process. These data suggest that the beneficial actions of volatile agents during myocardial ischemia may be partially attributed to increased oxygen supply mediated via  KATPchannel-dependent coronary vasodilation. However, sevoflurane increased coronary collateral blood flow in the presence of glyburide in vivo , indicating that volatile anesthetics enhance collateral perfusion independent of KATPchannel activation. 59In fact, sevoflurane-induced increases in collateral perfusion were recently shown to occur as a result of Ca2+-regulated potassium and not KATPchannel activation. 101Based on these findings and results obtained in isolated cardiac myocytes where blood flow is not a factor, 47it seems highly unlikely that myocardial protection produced by volatile anesthetics is solely related to favorable alterations in coronary vascular tone mediated by KATPchannels.

G Protein-coupled Receptors

Volatile anesthetics may activate parallel or redundant signaling pathways that involve KATPchannel opening to generate a physiologically meaningful cellular response. The sequential activation of several intracellular elements within a given transduction pathway may facilitate signal amplification and interaction between other redundant signaling systems. For example, administration of isoflurane in the presence of the KATPchannel openers nicorandil 102or diazoxide 103markedly enhances protection against ischemic injury beyond that observed with either drug alone. Several receptor-mediated events and intracellular signaling elements that converge on the KATPchannel have been implicated in APC. Pretreatment with pertussis toxin abolished any reduction in infarct size produced by isoflurane, indicating that Giproteins are linked to the signal transduction pathways that mediate APC. 73In contrast, pertussis toxin did not alter the beneficial effects of direct KATPchannel opening produced by nicorandil. These data strongly support the contention that volatile anesthetics modulate KATPchannel activity through second messenger signaling.

Halothane-induced protection against infarction was completely abolished by blockade of the adenosine A1receptor. 34The nonselective adenosine receptor antagonist 8-(p-sulfophenyl)-theophylline abolished isoflurane-induced preconditioning in rabbits. 79The selective A1receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine partially attenuated the beneficial effects of isoflurane in canine stunned myocardium. 70A role for adenosine receptors in APC was also identified in isolated human right atrial myocardium in vitro . 69Isoflurane eliminated increases in interstitial adenosine during repetitive periods of coronary artery occlusion and reperfusion using a microdialysis technique. 70These findings suggest that ATP preservation and a subsequent reduction of adenosine released into the interstitium occur during isoflurane anesthesia. 70In addition, the data imply that volatile agents may either directly activate A1receptors or indirectly enhance A1receptor sensitivity to diminished endogenous adenosine concentrations. 70These results were also similar to those observed during IPC 104or bimakalim-induced pharmacologic preconditioning. 105The preservation of cardiac myocyte viability during ischemia produced by volatile anesthetics was also sensitive to adenosine receptor and Giprotein inhibition in rats. 47 

Stimulation of the δ1-opioid receptor has been shown to produce a cardioprotective effect that is abolished by selective opioid antagonists 106–110or KATPchannel blockers. 107,108,110–113The acute and delayed phases of IPC are also mediated by activation of the δ1-opioid receptor. 114Recent results indicated that the combined administration of isoflurane and selective δ1-opioid receptor agonists TAN-67 or BW373U86 potentiated KATPchannel opening and enhanced protection against myocardial ischemia and reperfusion injury. 103Combined administration of isoflurane and morphine, a μ receptor agonist with δ1receptor agonist properties, also reduced the extent of myocardial infarction to a greater degree than either drug alone. 44This beneficial effect was shown to be mediated by mito KATPchannels and opioid receptors. Interestingly, the nonselective opioid antagonist naloxone abolished isoflurane-induced preconditioning. 44These intriguing data suggest an important link between volatile anesthetics and the opioid family of G protein-coupled receptors. Another recent study also indicated that halothane competitively inhibited the ligand-binding site of G protein-coupled receptors. 115Adrenergic receptor blockade was shown to abolish desflurane-induced preconditioning in isolated human right atria 63but had no effect on the antiischemic actions of sevoflurane in isolated rat cardiac myocytes. 47Overall, APC seems to be associated with the activation of separate receptor-mediated pathways that are linked to Giproteins.

Protein Kinases

Translocation and phosphorylation of multiple protein kinases are known to be involved in signal transduction pathways involved in protecting myocardium against cell death after ischemia and reperfusion. 116–118In particular, PKC is an essential component of the signaling pathways associated with preserving cellular viability. 119The diverse PKC isoform family is a large group of serine/threonine protein kinases that are distinguished by variable regulatory domains and cofactors and also display diverse tissue and species distributions. 120Activation of G protein-coupled receptors (e.g. , A1, 121,122bradykinin, 123,124δ1opioid 125,126) stimulate PKC during IPC. Volatile anesthetics have also been shown to stimulate PKC translocation and activity, 127possibly by interacting with the regulatory domain of the enzyme. 128Inhibition of PKC attenuated isoflurane-enhanced recovery of contractile function in canine stunned myocardium. 71The antiischemic actions of halothane were abolished by selective PKC antagonism in rabbits. 34The δ and ε isoforms of PKC translocated to mitochondria and sarcolemma, respectively, 10 min after discontinuation of isoflurane in isolated rat hearts. 129In contrast, isoflurane stimulated translocation of PKC-δ and -ε to sarcolemmal and mitochondrial membranes, respectively, in the in vivo  rat heart. 72These discrepancies may be attributed to differences in experimental model or time of tissue sampling. The microtubule depolymerizing drug, colchicine, prevented isoflurane-induced reductions in myocardial infarct size in rabbits, 130suggesting that an intact cytoskeleton is essential for translocation of these protein kinases. 72,129 

Recent findings strongly suggest that volatile anesthetic-induced PKC activation is required to open KATPchannels and produce myocardial protection. For example, the nonselective PKC antagonist chelerythrine abolished sevoflurane-induced increases in mito KATPchannel activity in rat ventricular myocytes and prevented protection against simulated ischemia. 47Patch clamp experiments showed that isoflurane did not facilitate KATPchannel opening in excised membrane patches but enhanced KATPchannel current in a whole cell configuration concomitant with PKC stimulation. 90These observations were supported by other studies showing that adenosine and PKC increased KATPchannel activity. 103,131–134Specific PKC consensus sites have been identified on KATPchannels, indicating a molecular basis for phosphorylation and activation of the channel by the enzyme. 135Mito KATPchannel opening also occurred after PKC activation during IPC in isolated rabbit hearts. 136In contrast, recent evidence indicates that 5-HD inhibited PKC translocation, 72suggesting that mito KATPchannel opening may be upstream of PKC activation. Therefore, a possible feed-forward system between PKC and KATPchannel activation may occur during APC.

Protein kinase C has been shown to stimulate PTK 137,138and mitogen-activated protein kinases (MAPKs), 139and volatile anesthetics may modulate several of these critical intracellular signaling proteins independent of direct receptor activation as well. Ischemic and pharmacologic preconditioning have been shown to be mediated by activation of PKC, 34,71,106,124,140PTK, 116,137,138,141,142and MAPK. 117,118,143–145A recent investigation showed that the PTK inhibitor lavendustin A and the Src-selective inhibitor PP1 abolished isoflurane-induced preconditioning in rats. 72The MAPK family plays an important role in signal transduction from the cell surface and the nucleus and has been strongly implicated in the initiation and progression of cell death (i.e. , apoptosis). 146–148The p38 MAPK subfamily mediated IPC of myocardium in dogs, and activation of p38 MAPK was also associated with phosphorylation and translocation of heat shock protein 27 in vivo . 144In contrast with the findings during IPC, recent data suggest that p38 MAPK may not play a role in APC in isolated rat hearts. 149Nonetheless, volatile anesthetics modulate activity of one or more intracellular kinases to produce APC, and it seems that activation of PKC is critical to cardioprotection.

Reactive Oxygen Species

Large quantities of ROS are released during reperfusion of ischemic myocardium that damage proteins responsible for intracellular homeostasis, depress contractile function, and produce membrane damage. 150–152Halothane, isoflurane, and enflurane have been shown to attenuate the toxic effects of ROS on left ventricular pressure development in isolated hearts. 153Isoflurane decreased hydroxyl radical generation in the ischemic rat heart, 154and halothane had a similar effect in dogs. 155The protective effects of sevoflurane were associated with reduced dityrosine formation, an indirect marker of ROS and reactive nitrogen species. 76These results support the hypothesis that volatile anesthetics reduce the release of deleterious quantities of ROS associated with coronary artery occlusion and reperfusion. Isoflurane also inhibited superoxide anion production by activated neutrophils, an action that occurred independent of KATPchannel opening. 156In addition, isoflurane and sevoflurane have been shown to abolish activated neutrophil-induced myocardial dysfunction. 157These effects were associated with reductions in superoxide anion production and neutrophil adherence to coronary vascular endothelium. Therefore, volatile anesthetics also seem to exert beneficial actions by inhibiting neutrophil-induced injury during reperfusion.

In contrast with data implicating a pathologic role of large amounts of ROS, other findings strongly suggest that a variety of preconditioning stimuli, including brief ischemia, direct mito KATPchannel openers, opioids, and volatile anesthetics, stimulate a small burst of ROS that initiate downstream signaling events and produce protection from subsequent ischemic injury. 158For example, pretreatment with low concentrations of ROS have been shown to mimic the beneficial actions of IPC. 159,160Free radical scavengers administered before or during brief ischemia markedly attenuated the protective effect of the preconditioning stimulus on infarct size. 161,162These findings indicate that IPC is mediated by small quantities of ROS released during the preconditioning stimulus. The beneficial actions of sevoflurane against ischemic damage were abolished by scavengers of superoxide anion and inhibition of nitric oxide synthase. 76These results suggest that superoxide anion may act to trigger APC and further indicated that nitric oxide may scavenge superoxide anion on reperfusion to reduce injury. ROS scavengers attenuated isoflurane-induced reductions in myocardial infarct size in rabbits 74,75and also inhibited the beneficial effects of direct mito KATPchannel activation. 163Isoflurane has been shown to directly increase superoxide anion formation in vivo  independent of an ischemic episode by use of the fluorescent probe dihydroethidium and laser confocal microscopy. 75These data 75indicated for the first time that volatile anesthetics were capable of producing small amounts of ROS that were correlated with a reduction in myocardial infarct size after prolonged ischemia. Taken as a whole, these reports provide compelling evidence that small quantities of ROS also play a critical role in APC.

Reactive oxygen species have been shown to act as regulatory mediators in many signaling processes that protect the cells against oxidative stress. 164ROS-induced activation of PKC 159and MAPK 165–167have been implicated in both ischemic and pharmacologic preconditioning. Hydrogen peroxide activated all three MAPK subtypes in neonatal rat ventricular myocytes, but stimulation of the p38 MAPK family and the consequent phosphorylation of heat shock protein 25/27 seemed to be of critical importance during cardioprotection. 165ROS have also been shown to activate Gαiand Gαoproteins. 168,169Recent findings showed that sevoflurane-induced ROS generation was unaffected by PKC inhibition, 170but ROS scavengers inhibited isoflurane-induced PKC translocation. 72These findings provide indirect evidence linking ROS production by volatile anesthetics to subsequent activation of protein kinases implicated in the signal transduction responsible for APC.

A controversy continues to exist regarding the temporal relation between mito KATPchannel opening and ROS production during ischemic or pharmacologic preconditioning. 171Although sarc KATPchannel opening was initially assumed to be the end-effector of IPC, 81,85mito KATPchannel activation may instead trigger preconditioning by generating ROS. 163,172Mito KATPchannel opening produced by selective agonists generates ROS 163,172that seem to be essential for activation of MAPK 166and are also required for beneficial effects on myocardium. 173The mito KATPchannel agonist diazoxide caused oxidation of the ROS probe MitoTracker® orange (Molecular Probes, Eugene, OR) and enhanced cell viability after hypoxia and reoxygenation in vitro . 172These actions were attenuated by pretreatment with 5-HD or ROS scavengers. Therefore, the protective effects of mito KATPchannel agonists may occur as a consequence of triggering by ROS that subsequently reduces myocyte injury, including the release of large quantities of these reactive intermediates during reperfusion injury. Morphine increased cardiomyocyte viability and the fluorescence intensity of the hydrogen peroxide-sensitive probe, 2′7′-dichlorofluorescin. 111These actions were abolished by 5-HD pretreatment, suggesting that activation of mito KATPchannels by opioids results in ROS production. Conversely, other studies 174,175have indicated that ROS modulate mito KATPchannel activity 174to provide a beneficial effect, indirectly suggesting that mito KATPchannels may also function as an end-effector of preconditioning. For example, superoxide anion generated by xanthine oxidase activated mito KATPchannels from bovine ventricular myocardium reconstituted from lipid bilayers. 174Lebuffe et al.  175demonstrated that ROS may serve as a trigger by opening mito KATPchannels, which subsequently generates additional ROS and nitric oxide that are both required for preconditioning in isolated chick neonatal myocytes. Therefore, whether mito KATPchannel opening serves as a trigger or end-effector of ischemic or pharmacologic preconditioning remains unclear. 176Nevertheless, the apparently complimentary interaction between ROS and mito KATPchannels suggest the intriguing possibility that positive feedback loops may exist between these elements that contribute to myocardial protection.

It also remains unclear whether volatile anesthetic-induced mito KATPchannel opening precedes or follows ROS generation. Pretreatment with 5-HD or the ROS scavengers N-acetylcysteine or N-2-mercaptopropionyl glycine before administration of isoflurane abolished ROS generation in vivo . 177In contrast, administration of 5-HD after discontinuation of isoflurane but before prolonged ischemia only partially attenuated this effect. These data suggest that mito KATPchannel opening acted as a trigger of APC by generating ROS. Conversely, another recent investigation conducted in isolated guinea pig hearts showed that sevoflurane-induced generation of ROS was not inhibited by 5-HD before ischemia. 178Therefore, experimental findings remain equivocal in support of the hypothesis that mito KATPchannel opening is the major trigger of APC.

Reactive oxygen species derived from the mitochondrial respiratory chain have been shown to play important roles during IPC or pharmacologic preconditioning. 111,160,179,180The complex III inhibitor myxothiazol blocked hypoxia- 160or acetylcholine-induced 179ROS generation and abolished preconditioning in isolated chick cardiac myocytes. The precise source of volatile anesthetic-induced production of ROS has yet to be finally established, but volatile agents have been previously shown to inhibit electron transport chain complexes I and II of cardiac mitochondria. 181,182Interestingly, sevoflurane-induced complex I inhibition was attenuated by the superoxide dismutase mimetic Mn(III)tetrakis(4-benzoic acid)porphyrin chloride. 182These results suggested that ROS may inhibit mitochondrial respiration through a positive feedback mechanism to amplify the ROS signal for triggering APC. In contrast, a recent investigation showed that the complex III inhibitor myxothiazol, but not the complex I inhibitor diphenyleneiodonium, abolished isoflurane-induced reductions in myocardial infarct size and generation of ROS. 183These preliminary data indicated that mitochondrial electron transport chain complex III may be the source of ROS production induced by isoflurane during APC. Taken together, it is possible that volatile anesthetics may modulate multiple sites of the electron transport chain either directly or indirectly via  a ROS-mediated feedback mechanism. Despite these compelling results, other potential enzymatic sources of ROS (i.e. , nicotinamide adenine dinucleotide oxidase, cyclooxygenase, lipoxygenase, xanthine oxidase, nitric oxide synthase, cytochrome P450 184–188) may play a role in APC and have yet to be excluded from this process.

The precise identities of the specific ROS involved in APC have yet to be defined, and the signaling pathways that may be modulated by these ROS are also largely unknown. ROS activated PKC, restored contractility, and reduced infarct size in rabbit hearts. 159Superoxide anion also opened mito KATPchannels. 174Hydrogen peroxide stimulated PTK-dependent activation of phospholipase C in mouse embryonic fibroblasts, rendering these cells resistant to stress. 189Hydrogen peroxide has also been shown to directly activate Giand Goproteins 168,169and other protein kinases involved in reducing cellular injury. 166,190–192Hydrogen peroxide may also be converted to other more reactive species that modify cysteine residues of specific G proteins, resulting in their selective activation. 169Recent results showed that isoflurane administration produced ethidium fluorescence in rabbit myocardium. 75,177Dihydroethidium is oxidized by intracellular superoxide anion to produce ethidium that subsequently binds to DNA, further amplifying its fluorescence. 193These results strongly suggest that superoxide anion is the particular ROS involved in isoflurane-induced preconditioning. Sevoflurane also generated superoxide anion before ischemia and reperfusion in isolated hearts. 178Alternatively, different ROS may exert opposing actions on mito KATPchannel activity. Dismutation of superoxide anion leads to production of secondary ROS, including hydrogen peroxide, hydroxyl radical, and peroxynitrite, 194and these radicals may differentially alter channel activity. For example, superoxide anion and hydrogen peroxide enhanced but peroxynitrite decreased Ca2+-regulated potassium channel activity. 195Therefore, it remains possible that volatile anesthetics may also generate ROS other than superoxide that activate mito KATPchannels, or these agents may inhibit the formation of intermediates such as peroxynitrite that adversely affect mito KATPchannel function. Further research is needed to clarify this issue.

Mechanisms of Protection

Opening of sarc KATPchannels was originally implicated in IPC and pharmacologic preconditioning by shortening the action potential duration, 81,196thereby reducing intracellular Ca2+overload during ischemia. 81However, subsequent studies conducted after the discovery of mito KATPchannels 82indicated that the antiischemic actions of KATPchannel activation occurred independent of action potential duration. 197–199Nevertheless, IPC did not occur in Kir6.2-deficient mice, suggesting that the presence of the sarc KATPchannel was still required for myocardial protection to occur. 200Despite these latter data, the majority of findings accumulated indicate that preservation of mitochondrial bioenergetic function that occurs as a consequence of mito KATPchannel opening seems to be of critical importance for protection against ischemia. 201–204Selective pharmacologic openers of mito KATPchannels (e.g. , diazoxide) maintain mitochondrial Ca2+homeostasis and inhibit Ca2+overload within the organelle. 203,204Alteration of the mitochondrial oxidation-reduction balance by mito KATPchannel opening may also act to promote cellular protection. 204,205Membrane depolarization, matrix swelling, and uncoupling of ATP synthesis occur as a result of mito KATPchannel opening that may mediate cellular viability during IPC. 205Mito KATPchannel opening depolarizes the inner mitochondrial membrane and causes a transient swelling of the mitochondrial matrix, 206resulting from a shift in the ionic balance. 207These actions initially reduce ATP production 204but subsequently stimulate a compensatory increase in respiration that optimizes the efficacy of oxidative phosphorylation in part through energy-dependent matrix volume regulation. 208Therefore, the moderate disturbance of mitochondrial homeostasis caused by mito KATPchannel opening (fig. 2) may promote tolerance to subsequent ischemic damage by reducing Ca2+overload, 203,204preventing activation of necrotic or apoptotic pathways, 209,210or attenuating oxidative stress. 211 

Mitochondrial ATP synthesis has also been shown to be preserved after prolonged as compared with brief ischemia and reperfusion. 212This beneficial effect was abolished by 5-HD, suggesting that activation of mito KATPchannels improves mitochondrial energy production. 213Opening of mito KATPchannels has been hypothesized to preserve outer mitochondrial membrane permeability to ATP precursors (e.g. , adenosine, adenosine diphosphate) and cytochrome c. The structure of the intermembrane space may also be maintained as a consequence of mito KATPchannel activation despite generalized swelling of the mitochondrial matrix. 201Preservation of ATP substrates and mitochondrial structure may facilitate more efficient energy transfer between the mitochondria and the cytosol immediately after ischemia. Sevoflurane was recently shown to preserve ATP synthesis in isolated cardiac mitochondria obtained during early reperfusion after ischemia in vivo , and this beneficial effect was abolished by pretreatment with a ROS scavenger. 214Sevoflurane-induced preconditioning improved mitochondrial bioenergetics through mito KATPchannel activation in isolated guinea pig hearts as assessed using flavoprotein fluorescence. 215Therefore, it seems likely that mito KATPchannel opening by volatile anesthetics may be associated with preservation of mitochondrial function during reperfusion and, further, that this maintenance of mitochondrial performance contributes to cardioprotection.

Experiments conducted in isolated mitochondria have shown that a triggering quantity of ROS exceeding a critical threshold results in a transition in mitochondrial inner membrane permeability and the subsequent release of a burst of ROS in a process that has been termed ROS-induced ROS release . 216This mitochondrial permeability transition (MPT) has been shown to precede necrotic or apoptotic cell death, 217and glutathione is a primary defense against this event. 216,218These data suggest that volatile anesthetics or other mito KATPchannel agonists may prevent MPT in an oxidant-sensitive fashion, but this intriguing hypothesis has yet to be tested. Opening of an MPT pore using the agonist atractyloside during reperfusion was recently shown to abolish IPC and diazoxide-induced preconditioning in isolated rat hearts. 219These findings suggested that inhibition of MPT pore opening may represent a distal effector responsible for preconditioning, whereas mito KATPactivation functions as a trigger or a mediator. Most recently, rabbit hearts pretreated with desflurane before ischemia and reperfusion exhibited resistance to MPT pore opening. 220Future investigations are necessary to delineate the role of MPT during APC.

Cytosolic and mitochondrial Ca2+overload during prolonged ischemia and reperfusion have been shown to be associated with mitochondrial damage and myocardial cell death. 221–223Ischemic and sevoflurane-induced preconditioning were shown to reduce cytosolic Ca2+overload and improve the recovery of contractile function during reperfusion. 64Administration of sevoflurane after ischemia also reduced cytosolic Ca2+and myocardial damage. 31IPC and APC attenuated mitochondrial Ca2+overload during ischemia in rat and guinea pig hearts, 224,225actions that were abolished by 5-HD. These data suggested that volatile anesthetics protect against ischemia-reperfusion injury, at least in part, by attenuating cytosolic and mitochondrial Ca2+overload through a mito KATPchannel-dependent mechanism. Volatile anesthetics have also been shown to suppress sarcoplasmic reticulum Ca2+release 226,227and depress myofilament Ca2+sensitivity. 227Therefore, modulation of the sarcoplasmic reticulum to reduce cellular Ca2+overload and alterations of myofilament Ca2+sensitivity under conditions of excess Ca2+have been implicated in cardioprotection. 228,229The inhibitory actions of the volatile anesthetics on the voltage-dependent Ca2+channel are also well known, 65–68and reductions in cytosolic and mitochondrial Ca2+overload during ischemia and reperfusion injury may also occur through this mechanism.

Myocardial Protection in Clinical Conditions

Compelling experimental data in multiple animal models regarding the protective effects of volatile anesthetics remain to be translated into therapeutic approaches to reduce morbidity and mortality in patients with ischemic heart disease. However, evidence accumulated to date strongly suggests that APC occurs in human myocardium. Repetitive, brief (60- to 90-s) balloon inflations and deflations performed during percutaneous transluminal coronary angioplasty were associated with progressive reductions in the severity of chest pain and in the extent of ST segment elevation, decreases in myocardial lactate production, and declines in cardiac enzyme and troponin release. 230–233Adenosine 234or the mito KATPchannel opener nicorandil 235administered before the first balloon inflation during percutaneous transluminal coronary angioplasty was also shown to reduce the severity of ST changes during subsequent occlusions. Pretreatment with nicorandil before percutaneous transluminal coronary angioplasty also attenuated the release of troponin T, an indicator of myocyte necrosis. 236These findings suggest that both ischemic and pharmacologic preconditioning can be elicited during percutaneous transluminal coronary angioplasty. Consecutive exercise stress tests (separated by 15 min) performed in patients with critical left anterior descending coronary artery stenoses showed that anginal symptoms, ST segment depression, and myocardial oxygen consumption were reduced during the second as compared with the first exercise period for an equivalent amount of work. 237This “warm-up” phenomenon occurred independent of coronary vasodilation 237and also provides evidence of IPC in humans. 238,239 

Ischemic preconditioning has also been shown during cardiac surgery in patients with coronary artery disease. Yellon et al.  239used intermittent aortic cross clamping to produce IPC during coronary artery bypass graft surgery (CABG) and found enhanced preservation of ATP content in preconditioned hearts as compared with those that did not receive preconditioning stimuli. These investigators also showed that less troponin T was released in the presence as compared with the absence of IPC in this model. 240The incidence of ventricular tachyarrhythmias was reduced after cardiopulmonary bypass in patients undergoing CABG when cold blood cardioplegia was used for myocardial preservation. 241–243Regional IPC produced by brief coronary occlusion has also been shown to result in improved hemodynamic recovery and reduced release of cardiac troponin I during off-pump CABG. 244In contrast with the findings of these investigations, other studies have failed to show that IPC exerts beneficial effects during CABG in the presence of cardioplegia and cardiopulmonary bypass. 245–247Therefore, although the myocardial protective effects of IPC have been clearly identified in the experimental laboratory, further large scale clinical trials are needed to definitively demonstrate the beneficial actions of IPC in humans.

Documentation of APC in patients has been complicated by alterations in systemic and coronary hemodynamics; the use of other anesthetics, analgesics, or vasoactive drugs; preexisting disease states; and the acute influence of surgery on cardiovascular homeostasis. Nevertheless, isoflurane, 69desflurane, 63and sevoflurane 62enhanced the recovery of contractile function of human atrial trabeculae in vitro  by stimulation of adenosine receptors and opening of KATPchannels. Other studies have previously shown a role for adenosine receptors, MAPK, 248and ROS 172in other forms of preconditioning concomitant with opening of mito KATPchannels in human atrial myocytes. Isoflurane increased the tolerance to pacing-induced ischemia in patients with coronary artery disease. 249Isoflurane also decreased postoperative release of troponin I and creatine kinase-MB in patients undergoing CABG. 250Although the aforementioned results 250were not statistically significant, these data suggest that reduction in the extent of myocardial necrosis had occurred. Administration of isoflurane immediately before aortic cross clamping in patients undergoing CABG was shown to decrease the severity of subsequent ST segment changes and preserve cardiac index to a greater extent than that observed in patients who did not receive pretreatment with the volatile anesthetic. 251Administration of enflurane before cardioplegic arrest enhanced recovery of postischemic contractile function assessed using pressure-area relations in CABG patients. 252Sevoflurane 253–255and desflurane 253,255but not the intravenous anesthetic propofol was shown to preserve myocardial function in patients undergoing CABG as well as a reduction in troponin I release. Most recently, preconditioning with sevoflurane reduced a biochemical marker of myocardial dysfunction (i.e. , N-terminal pro-brain natriuretic peptide) in patients undergoing CABG concomitant with translocation of PKC-δ and -ε. 256This compelling evidence strongly suggests that volatile anesthetics exert beneficial effects against ischemic injury in humans.

In contrast with the aforementioned results, no differences in PKC and p38 MAPK activity or peak troponin I release were observed between patients undergoing cardioplegic arrest in the presence or absence of sevoflurane pretreatment. 257The activities of PKC, PTK, and p38 MAPK were increased equally in both groups, suggesting that cardiopulmonary bypass and cardioplegic arrest may produce a preconditioning-like effect that obscured the antiischemic actions of sevoflurane in this setting. However, sevoflurane was recently shown to reduce myocardial injury to a greater degree than propofol in patients undergoing off-pump CABG, 254a clinical setting that does not require cardiopulmonary bypass. Most investigations conducted in humans seem to indicate that volatile anesthetics represent an important therapeutic modality to reduce the sequelae of perioperative myocardial ischemia and infarction. 258,259A large-scale, randomized clinical trial is clearly needed to firmly establish this conclusion. Given the recent data suggesting that cardiopulmonary bypass and cardioplegia may exert a protective effect, 257such a clinical trial may best be conducted in patients undergoing off-pump CABG 254or those with documented coronary artery disease undergoing noncardiac surgery. 4Furthermore, no clinical study has shown that the use of volatile anesthetics in patients with coronary artery disease contributes to reduced cardiac morbidity or perioperative mortality. Additional multicenter trials also need to be conducted to identify the relative impact of APC on clinical outcome.

Experimental evidence collected indicates that volatile anesthetics exert important cardioprotective effects that reduce the consequences of reversible and irreversible ischemia and reperfusion injury. Differences in the efficacy of APC and timing of administration among volatile anesthetics alone and in combination with other cardioprotective drugs remain to be fully distinguished. Another important aspect is to determine the effect of aging on APC. 255,260Characterization of a late phase or second window of APC may be of special clinical significance to protect against ischemic events that frequently occur in the postoperative period. Several endogenous signaling elements seem to mediate APC, and mito KATPchannels, PKC, and ROS have emerged as central features in this process. Future investigations are needed to further delineate and identify essential components in these complex signal transduction cascades that mediate the early and late phases of APC. In this regard, microarray technology may prove useful in ascertaining candidate genes that are responsible for APC. 261Use of other fundamental molecular and biochemical tools is needed to determine whether preservation of mitochondrial integrity and metabolic homeostasis ultimately enhances tolerance to myocardial ischemia. Volatile anesthetics also act to elicit signaling that is probably present in many types of cells. Therefore, it is not surprising that these agents may reduce injury to other tissues. 256,262This may also be based on preventing cytokine-induced injury in endothelial and vascular smooth muscle cells. 50,61Finally, recent investigations have also strongly implied that APC occurs in humans and may represent an important therapeutic approach to reduce morbidity and mortality in patients with coronary artery disease. Nevertheless, further investigation is needed to firmly link this emerging body of clinical evidence to the already strongly established experimental data about the protective effects of volatile anesthetics in myocardium.

The authors thank David Schwabe, B.S.E.E., for technical assistance and Mary Lorence-Hanke, A.A. (Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin), for assistance in preparation of the manuscript.

1.
Bland JH, Lowenstein E: Halothane-induced decrease in experimental myocardial ischemia in the non-failing canine heart. A nesthesiology 1976; 45: 287–93
2.
Davis RF, DeBoer LW, Rude RE, Lowenstein E, Maroko PR: The effect of halothane anesthesia on myocardial necrosis, hemodynamic performance, and regional myocardial blood flow in dogs following coronary artery occlusion. A nesthesiology 1983; 59: 402–11
3.
van Ackern K, Vetter HO, Bruckner UB, Madler C, Mittman U, Peter K: Effects of enflurane on myocardial ischaemia in the dog. Br J Anaesth 1985; 57: 497–504
4.
Warltier DC, Kersten JR, Pagel PS, Gross GJ: Anesthetic preconditioning: Serendipity and science (editorial). A nesthesiology 2002; 97: 1–3
5.
Reiz S, Balfors E, Sorensen MB, Ariola S Jr, Friedman A, Truedsson H: Isoflurane: A powerful coronary vasodilator in patients with coronary artery disease. A nesthesiology 1983; 59: 91–7
6.
Becker LC: Is isoflurane dangerous for the patient with coronary artery disease? A nesthesiology 1987; 66: 259–61
7.
Buffington CW, Romson JL, Levine A, Duttlinger NC, Huang AH: Isoflurane induces coronary steal in a canine model of chronic coronary occlusion. A nesthesiology 1987; 66: 280–92
8.
Priebe HJ, Foex P: Isoflurane causes regional myocardial dysfunction in dogs with critical coronary artery stenoses. A nesthesiology 1987; 66: 293–300
9.
Gross GJ, Warltier DC: Coronary steal in four models of single or multiple vessel obstruction in dogs. Am J Cardiol 1981; 48: 84–92
10.
Warltier DC, Gross GJ, Brooks HL: Coronary steal-induced increase in myocardial infarct size after pharmacologic coronary vasodilation. Am J Cardiol 1980; 46: 83–90
11.
Becker LC: Conditions for vasodilator-induced coronary steal in experimental myocardial ischemia. Circulation 1978; 57: 1103–10
12.
Sill JC, Bove AA, Nugent M, Blaise GA, Dewey JD, Grabau C: Effects of isoflurane on coronary arteries and coronary arterioles in the intact dog. A nesthesiology 1987; 66: 273–9
13.
Buffington CW, Davis KB, Gillispie S, Pettinger M: The prevalence of steal-prone coronary anatomy in patients with coronary artery disease: An analysis of the Coronary Artery Surgery Study Registry. A nesthesiology 1988; 69: 721–7
14.
Moore PG, Kien ND, Reitan JA, White DA, Safwat AM: No evidence for blood flow redistribution with isoflurane or halothane during acute coronary artery occlusion in fentanyl-anesthetized dogs. A nesthesiology 1991; 75: 854–65
15.
Hartman JC, Kampine JP, Schmeling WT, Warltier DC: Steal-prone coronary circulation in chronically instrumented dogs: Isoflurane versus  adenosine. A nesthesiology 1991; 74: 744–56
16.
Hartman JC, Kampine JP, Schmeling WT, Warltier DC: Actions of isoflurane on myocardial perfusion in chronically instrumented dogs with poor, moderate, or well-developed coronary collaterals. J Cardiothorac Anesth 1990; 4: 715–25
17.
Hartman JC, Kampine JP, Schmeling WT, Warltier DC: Alterations in collateral blood flow produced by isoflurane in a chronically instrumented canine model of multivessel coronary artery disease. A nesthesiology 1991; 74: 120–33
18.
Leung JM, Goehner P, O’Kelly BF, Hollenberg M, Pineda N, Cason BA, Mangano DT: Isoflurane anesthesia and myocardial ischemia: Comparative risk versus  sufentanil anesthesia in patients undergoing coronary artery bypass graft surgery. The SPI (Study of Perioperative Ischemia) Research Group. A nesthesiology 1991; 74: 838–47
19.
Pulley DD, Kirvassilis GV, Kelermenos N, Kater K, Barzilai B, Genton RE, Efstathiou C, Lappas DG: Regional and global myocardial circulatory and metabolic effects of isoflurane and halothane in patients with steal-prone coronary anatomy. A nesthesiology 1991; 75: 756–66
20.
Diana P, Tullock WC, Gorcsan J III, Ferson PF, Arvan S: Myocardial ischemia: A comparison between isoflurane and enflurane in coronary artery bypass patients. Anesth Analg 1993; 77: 221–6
21.
Kersten JR, Brayer AP, Pagel PS, Tessmer JP, Warltier DC: Perfusion of ischemic myocardium during anesthesia with sevoflurane. A nesthesiology 1994; 81: 995–1004
22.
Kitahata H, Kawahito S, Nozaki J, Kimura H, Tanaka K, Kitagawa T, Oshita S: Effects of sevoflurane on regional myocardial blood flow distribution: Quantification with myocardial contrast echocardiography. A nesthesiology 1999; 90: 1436–45
23.
Hartman JC, Pagel PS, Kampine JP, Schmeling WT, Warltier DC: Influence of desflurane on regional distribution of coronary blood flow in a chronically instrumented canine model of multivessel coronary artery obstruction. Anesth Analg 1991; 72: 289–99
24.
Kenny D, Coughlan MG, Kampine JP, Montgomery RR, Bosnjak ZJ, Warltier DC: Cultured endothelial cells restore vasodilator responses to coronary arteries with impaired endothelial function and alter the response to a nitric oxide donor. Pharmacology 1994; 49: 249–56
25.
Lochner A, Harper IS, Salie R, Genade S, Coetzee AR: Halothane protects the isolated rat myocardium against excessive total intracellular calcium and structural damage during ischemia and reperfusion. Anesth Analg 1994; 79: 226–33
26.
Schlack W, Preckel B, Stunneck D, Thamer V: Effects of halothane, enflurane, isoflurane, sevoflurane and desflurane on myocardial reperfusion injury in the isolated rat heart. Br J Anaesth 1998; 81: 913–9
27.
Preckel B, Thamer V, Schlack W: Beneficial effects of sevoflurane and desflurane against myocardial reperfusion injury after cardioplegic arrest. Can J Anaesth 1999; 46: 1076–81
28.
Preckel B, Schlack W, Comfere T, Obal D, Barthel H, Thamer V: Effects of enflurane, isoflurane, sevoflurane and desflurane on reperfusion injury after regional myocardial ischaemia in the rabbit heart in vivo. Br J Anaesth 1998; 81: 905–12
29.
Ebel D, Preckel B, You A, Mullenheim J, Schlack W, Thamer V: Cardioprotection by sevoflurane against reperfusion injury after cardioplegic arrest in the rat is independent of three types of cardioplegia. Br J Anaesth 2002; 88: 828–35
30.
Obal D, Preckel B, Scharbatke H, Mullenheim J, Hoterkes F, Thamer V, Schlack W: One MAC of sevoflurane provides protection against reperfusion injury in the rat heart in vivo. Br J Anaesth 2001; 87: 905–11
31.
Varadarajan SG, An J, Novalija E, Stowe DF: Sevoflurane before or after ischemia improves contractile and metabolic function while reducing myoplasmic Ca2+loading in intact hearts. A nesthesiology 2002; 96: 125–33
32.
Kanaya N, Fujita S: The effects of isoflurane on regional myocardial contractility and metabolism in “stunned” myocardium in acutely instrumented dogs. Anesth Analg 1994; 79: 447–54
33.
Pagel PS, Hettrick DA, Lowe D, Tessmer JP, Warltier DC: Desflurane and isoflurane exert modest beneficial actions on left ventricular diastolic function during myocardial ischemia in dogs. A nesthesiology 1995; 83: 1021–35
34.
Cope DK, Impastato WK, Cohen MV, Downey JM: Volatile anesthetics protect the ischemic rabbit myocardium from infarction. A nesthesiology 1997; 86: 699–709
35.
Marijic J, Stowe DF, Turner LA, Kampine JP, Bosnjak ZJ: Differential protective effects of halothane and isoflurane against hypoxic and reoxygenation injury in the isolated guinea pig heart. A nesthesiology 1990; 73: 976–83
36.
Coetzee A, Brits W, Genade S, Lochner A: Halothane does have protective properties in the isolated ischemic rat heart. Anesth Analg 1991; 73: 711–9
37.
Coetzee A, Skein W, Genade S, Lochner A: Enflurane and isoflurane reduce reperfusion dysfunction in the isolated rat heart. Anesth Analg 1993; 76: 602–8
38.
Freedman BM, Hamm DP, Everson CT, Wechsler AS, Christian CM II: Enflurane enhances postischemic functional recovery in the isolated rat heart. A nesthesiology 1985; 62: 29–33
39.
Novalija E, Fujita S, Kampine JP, Stowe DF: Sevoflurane mimics ischemic preconditioning effects on coronary flow and nitric oxide release in isolated hearts. A nesthesiology 1999; 91: 701–12
40.
Warltier DC, al-Wathiqui MH, Kampine JP, Schmeling WT: Recovery of contractile function of stunned myocardium in chronically instrumented dogs is enhanced by halothane or isoflurane. A nesthesiology 1988; 69: 552–65
41.
Kersten JR, Lowe D, Hettrick DA, Pagel PS, Gross GJ, Warltier DC: Glyburide, a KATP channel antagonist, attenuates the cardioprotective effects of isoflurane in stunned myocardium. Anesth Analg 1996; 83: 27–33
42.
Kersten JR, Schmeling TJ, Pagel PS, Gross GJ, Warltier DC: Isoflurane mimics ischemic preconditioning via  activation of KATPchannels: Reduction of myocardial infarct size with an acute memory phase. A nesthesiology 1997; 87: 361–70
43.
Cason BA, Gamperl AK, Slocum RE, Hickey RF: Anesthetic-induced preconditioning: Previous administration of isoflurane decreases myocardial infarct size in rabbits. A nesthesiology 1997; 87: 1182–90
44.
Ludwig LM, Patel HH, Gross GJ, Kersten JR, Pagel PS, Warltier DC: Morphine enhances pharmacological preconditioning by isoflurane: Role of mitochondrial KATPchannels and opioid receptors. A nesthesiology 2003; 98: 705–11
45.
Maxwell MP, Hearse DJ, Yellon DM: Species variation in the coronary collateral circulation during regional myocardial ischaemia: A critical determinant of the rate of evolution and extent of myocardial infarction. Cardiovasc Res 1987; 21: 737–46
46.
Kehl F, Krolikowski JG, Mraovic B, Pagel PS, Warltier DC, Kersten JR: Is isoflurane-induced preconditioning dose related? A nesthesiology 2002; 96: 675–80
47.
Zaugg M, Lucchinetti E, Spahn DR, Pasch T, Schaub MC: Volatile anesthetics mimic cardiac preconditioning by priming the activation of mitochondrial KATPchannels via  multiple signaling pathways. A nesthesiology 2002; 97: 4–14
48.
Toller WG, Kersten JR, Pagel PS, Hettrick DA, Warltier DC: Sevoflurane reduces myocardial infarct size and decreases the time threshold for ischemic preconditioning in dogs. A nesthesiology 1999; 91: 1437–46
49.
Tanaka K, Ludwig LM, Krolikowski JG, Alcindor D, Pratt PF, Kersten JR, Pagel PS, Warltier DC: Isoflurane produces delayed preconditioning against myocardial ischemia and reperfusion injury: Role of cyclooxygenase-2. A nesthesiology 2004; 100: 525–31
50.
de Klaver MJ, Buckingham MG, Rich GF: Isoflurane pretreatment has immediate and delayed protective effects against cytokine-induced injury in endothelial and vascular smooth muscle cells. A nesthesiology 2003; 99: 896–903
51.
Mullenheim J, Ebel D, Bauer M, Otto F, Heinen A, Frassdorf J, Preckel B, Schlack W: Sevoflurane confers additional cardioprotection after ischemic late preconditioning in rabbits. A nesthesiology 2003; 99: 624–31
52.
Cason BA, Shubayev I, Hickey RF: Blockade of adenosine triphosphate-sensitive potassium channels eliminates isoflurane-induced coronary artery vasodilation. A nesthesiology 1994; 81: 1245–55
53.
Crystal GJ, Gurevicius J, Salem MR, Zhou X: Role of adenosine triphosphate-sensitive potassium channels in coronary vasodilation by halothane, isoflurane, and enflurane. A nesthesiology 1997; 86: 448–58
54.
Zhou X, Abboud W, Manabat NC, Salem MR, Crystal GJ: Isoflurane-induced dilation of porcine coronary arterioles is mediated by ATP-sensitive potassium channels. A nesthesiology 1998; 89: 182–9
55.
Crystal GJ, Zhou X, Gurevicius J, Czinn EA, Salem MR, Alam S, Piotrowski A, Hu G: Direct coronary vasomotor effects of sevoflurane and desflurane in in situ  canine hearts. A nesthesiology 2000; 92: 1103–13
56.
Kersten JR, Warltier DC: The coronary circulation, Anesthesia: Biologic Foundations. Edited by Yaksh TL, Lynch C III, Zapol WM, Maze M, Biebuyck JF, Saidman LJ. Philadelphia, Lippincott-Raven, 1997, pp 1169–92
57.
Smith G, Rogers K, Thorburn J: Halothane improves the balance of oxygen supply to demand in acute experimental myocardial ischaemia. Br J Anaesth 1980; 52: 577–83
58.
Bertha BG, Folts JD, Nugent M, Rusy BF: Halothane, but not isoflurane or enflurane, protects against spontaneous and epinephrine-exacerbated acute thrombus formation in stenosed dog coronary arteries. A nesthesiology 1989; 71: 96–102
59.
Kersten JR, Schmeling T, Tessmer J, Hettrick DA, Pagel PS, Warltier DC: Sevoflurane selectively increases coronary collateral blood flow independent of KATPchannels in vivo . A nesthesiology 1999; 90: 246–56
60.
Kowalski C, Zahler S, Becker BF, Flaucher A, Conzen PF, Gerlach E, Peter K: Halothane, isoflurane, and sevoflurane reduce postischemic adhesion of neutrophils in the coronary system. A nesthesiology 1997; 86: 188–95
61.
de Klaver MJ, Manning L, Palmer LA, Rich GF: Isoflurane pretreatment inhibits cytokine-induced cell death in cultured rat smooth muscle cells and human endothelial cells. A nesthesiology 2002; 97: 24–32
62.
Yvon A, Hanouz JL, Haelewyn B, Terrien X, Massetti M, Babatasi G, Khayat A, Ducouret P, Bricard H, Gerard JL: Mechanisms of sevoflurane-induced myocardial preconditioning in isolated human right atria in vitro . A nesthesiology 2003; 99: 27–33
63.
Hanouz JL, Yvon A, Massetti M, Lepage O, Babatasi G, Khayat A, Bricard H, Gerard JL: Mechanisms of desflurane-induced preconditioning in isolated human right atria in vitro . A nesthesiology 2002; 97: 33–41
64.
An J, Varadarajan SG, Novalija E, Stowe DF: Ischemic and anesthetic preconditioning reduces cytosolic [Ca2+] and improves Ca(2+) responses in intact hearts. Am J Physiol Heart Circ Physiol 2001; 281: H1508–23
65.
Eskinder H, Rusch NJ, Supan FD, Kampine JP, Bosnjak ZJ: The effects of volatile anesthetics on L- and T-type calcium channel currents in canine cardiac Purkinje cells. A nesthesiology 1991; 74: 919–26
66.
Bosnjak ZJ, Aggarwal A, Turner LA, Kampine JM, Kampine JP: Differential effects of halothane, enflurane, and isoflurane on Ca2+transients and papillary muscle tension in guinea pigs. A nesthesiology 1992; 76: 123–31
67.
Lynch C III: Effects of halothane and isoflurane on isolated human ventricular myocardium. A nesthesiology 1988; 68: 429–32
68.
Hatakeyama N, Momose Y, Ito Y: Effects of sevoflurane on contractile responses and electrophysiologic properties in canine single cardiac myocytes. A nesthesiology 1995; 82: 559–65
69.
Roscoe AK, Christensen JD, Lynch C III: Isoflurane, but not halothane, induces protection of human myocardium via  adenosine A1 receptors and adenosine triphosphate-sensitive potassium channels. A nesthesiology 2000; 92: 1692–701
70.
Kersten JR, Orth KG, Pagel PS, Mei DA, Gross GJ, Warltier DC: Role of adenosine in isoflurane-induced cardioprotection. A nesthesiology 1997; 86: 1128–39
71.
Toller WG, Montgomery MW, Pagel PS, Hettrick DA, Warltier DC, Kersten JR: Isoflurane-enhanced recovery of canine stunned myocardium: Role for protein kinase C? A nesthesiology 1999; 91: 713–22
72.
Ludwig LM, Weihrauch D, Kersten JR, Pagel PS, Warltier DC: Protein kinase C translocation and Src protein tyrosine kinase activation mediate isoflurane-induced preconditioning in vivo : Potential downstream targets of mitochondrial adenosine triphosphate-sensitive potassium channels and reactive oxygen species. A nesthesiology 2004; 100: 532–9
73.
Toller WG, Kersten JR, Gross ER, Pagel PS, Warltier DC: Isoflurane preconditions myocardium against infarction via  activation of inhibitory guanine nucleotide binding proteins. A nesthesiology 2000; 92: 1400–7
74.
Mullenheim J, Ebel D, Frassdorf J, Preckel B, Thamer V, Schlack W: Isoflurane preconditions myocardium against infarction via  release of free radicals. A nesthesiology 2002; 96: 934–40
75.
Tanaka K, Weihrauch D, Kehl F, Ludwig LM, LaDisa JF Jr, Kersten JR, Pagel PS, Warltier DC: Mechanism of preconditioning by isoflurane in rabbits: A direct role for reactive oxygen species. A nesthesiology 2002; 97: 1485–90
76.
Novalija E, Varadarajan SG, Camara AK, An J, Chen Q, Riess ML, Hogg N, Stowe DF: Anesthetic preconditioning: Triggering role of reactive oxygen and nitrogen species in isolated hearts. Am J Physiol Heart Circ Physiol 2002; 283: H44–52
77.
Piriou V, Chiari P, Knezynski S, Bastien O, Loufoua J, Lehot JJ, Foex P, Annat G, Ovize M: Prevention of isoflurane-induced preconditioning by 5-hydroxydecanoate and gadolinium: Possible involvement of mitochondrial adenosine triphosphate-sensitive potassium and stretch-activated channels. A nesthesiology 2000; 93: 756–64
78.
Toller WG, Gross ER, Kersten JR, Pagel PS, Gross GJ, Warltier DC: Sarcolemmal and mitochondrial adenosine triphosphate-dependent potassium channels: Mechanism of desflurane-induced cardioprotection. A nesthesiology 2000; 92: 1731–9
79.
Ismaeil MS, Tkachenko I, Gamperl AK, Hickey RF, Cason BA: Mechanisms of isoflurane-induced myocardial preconditioning in rabbits. A nesthesiology 1999; 90: 812–21
80.
Inagaki N, Gonoi T, Clement JP IV, Namba N, Inazawa J, Gonzalez G, Aguilar-Bryan L, Seino S, Bryan J: Reconstitution of IKATP: An inward rectifier subunit plus the sulfonylurea receptor. Science 1995; 270: 1166–70
81.
Noma A: ATP-regulated K+ channels in cardiac muscle. Nature 1983; 305: 147–8
82.
Inoue I, Nagase H, Kishi K, Higuti T: ATP-sensitive K+ channel in the mitochondrial inner membrane. Nature 1991; 352: 244–7
83.
Liu Y, Ren G, O’Rourke B, Marban E, Seharaseyon J: Pharmacological comparison of native mitochondrial K(ATP) channels with molecularly defined surface K(ATP) channels. Mol Pharmacol 2001; 59: 225–30
84.
Kersten JR, Gross GJ, Pagel PS, Warltier DC: Activation of adenosine triphosphate-regulated potassium channels: Mediation of cellular and organ protection. A nesthesiology 1998; 88: 495–513
85.
Gross GJ, Auchampach JA: Blockade of ATP-sensitive potassium channels prevents myocardial preconditioning in dogs. Circ Res 1992; 70: 223–33
86.
O’Rourke B: Myocardial K(ATP) channels in preconditioning. Circ Res 2000; 87: 845–55
87.
Kersten JR, Schmeling TJ, Hettrick DA, Pagel PS, Gross GJ, Warltier DC: Mechanism of myocardial protection by isoflurane: Role of adenosine triphosphate-regulated potassium (KATP) channels A nesthesiology 1996; 85: 794–807
88.
Nakayama M, Fujita S, Kanaya N, Tsuchida H, Namiki A: Blockade of ATP-sensitive K+ channel abolishes the anti-ischemic effects of isoflurane in dog hearts. Acta Anaesthesiol Scand 1997; 41: 531–5
89.
Kwok WM, Martinelli AT, Fujimoto K, Suzuki A, Stadnicka A, Bosnjak ZJ: Differential modulation of the cardiac adenosine triphosphate-sensitive potassium channel by isoflurane and halothane. A nesthesiology 2002; 97: 50–6
90.
Fujimoto K, Bosnjak ZJ, Kwok WM: Isoflurane-induced facilitation of the cardiac sarcolemmal KATPchannel. A nesthesiology 2002; 97: 57–65
91.
Han J, Kim E, Ho WK, Earm YE: Effects of volatile anesthetic isoflurane on ATP-sensitive K+ channels in rabbit ventricular myocytes. Biochem Biophys Res Commun 1996; 229: 852–6
92.
Stadnicka A, Kwok WM, Warltier DC, Bosnjak ZJ: Protein tyrosine kinase-dependent modulation of isoflurane effects on cardiac sarcolemmal KATPchannel. A nesthesiology 2002; 97: 1198–208
93.
Gassmayr S, Stadnicka A, Suzuki A, Kwok WM, Bosnjak ZJ: Isoflurane sensitizes the cardiac sarcolemmal adenosine triphosphate-sensitive potassium channel to pinacidil. A nesthesiology 2003; 98: 114–20
94.
Stadnicka A, Bosnjak ZJ: Isoflurane decreases ATP sensitivity of guinea pig cardiac sarcolemmal KATPchannel at reduced intracellular pH. A nesthesiology 2003; 98: 396–403
95.
Kohro S, Hogan QH, Nakae Y, Yamakage M, Bosnjak ZJ: Anesthetic effects on mitochondrial ATP-sensitive K channel. A nesthesiology 2001; 95: 1435–40
96.
Hanley PJ, Mickel M, Loffler M, Brandt U, Daut J: K(ATP) channel-independent targets of diazoxide and 5-hydroxydecanoate in the heart. J Physiol 2002; 542: 735–41
97.
Nakae Y, Kwok WM, Bosnjak ZJ, Jiang MT: Isoflurane activates rat mitochondrial ATP-sensitive K+ channels reconstituted in lipid bilayers. Am J Physiol Heart Circ Physiol 2003; 284: H1865–71
98.
Ichinari K, Kakei M, Matsuoka T, Nakashima H, Tanaka H: Direct activation of the ATP-sensitive potassium channel by oxygen free radicals in guinea-pig ventricular cells: Its potentiation by MgADP. J Mol Cell Cardiol 1996; 28: 1867–77
99.
Sasaki N, Sato T, Marban E, O’Rourke B: ATP consumption by uncoupled mitochondria activates sarcolemmal K(ATP) channels in cardiac myocytes. Am J Physiol Heart Circ Physiol 2001; 280: H1882–8
100.
Daut J, Maier-Rudolph W, von Beckerath N, Mehrke G, Gunther K, Goedel-Meinen L: Hypoxic dilation of coronary arteries is mediated by ATP-sensitive potassium channels. Science 1990; 247: 1341–4
101.
Kehl F, Krolikowski JG, Tessmer JP, Pagel PS, Warltier DC, Kersten JR: Increases in coronary collateral blood flow produced by sevoflurane are mediated by calcium-activated potassium (BKCa) channels in vivo . A nesthesiology 2002; 97: 725–31
102.
Piriou V, Ross S, Bastien O, Pigott D, Trivin F, Foex P: Cardiovascular effects of concomitant administration of isoflurane and nicorandil in dogs. Br J Anaesth 1998; 80: 481–7
103.
Patel HH, Ludwig LM, Fryer RM, Hsu AK, Warltier DC, Gross GJ: Delta opioid agonists and volatile anesthetics facilitate cardioprotection via potentiation of K(ATP) channel opening. FASEB J 2002; 16: 1468–70
104.
Van Wylen DG: Effect of ischemic preconditioning on interstitial purine metabolite and lactate accumulation during myocardial ischemia. Circulation 1994; 89: 2283–9
105.
Mizumura T, Nithipatikom K, Gross GJ: Bimakalim, an ATP-sensitive potassium channel opener, mimics the effects of ischemic preconditioning to reduce infarct size, adenosine release, and neutrophil function in dogs. Circulation 1995; 92: 1236–45
106.
Fryer RM, Wang Y, Hsu AK, Gross GJ: Essential activation of PKC-delta in opioid-initiated cardioprotection. Am J Physiol Heart Circ Physiol 2001; 280: H1346–53
107.
Fryer RM, Hsu AK, Nagase H, Gross GJ: Opioid-induced cardioprotection against myocardial infarction and arrhythmias: Mitochondrial versus sarcolemmal ATP-sensitive potassium channels. J Pharmacol Exp Ther 2000; 294: 451–7
108.
Schultz Jel-J, Hsu AK, Nagase H, Gross GJ: TAN-67, a delta 1-opioid receptor agonist, reduces infarct size via activation of Gi/o proteins and KATP channels. Am J Physiol 1998; 274: H909–14
109.
Patel HH, Hsu A, Moore J, Gross GJ: BW373U86, a delta opioid agonist, partially mediates delayed cardioprotection via a free radical mechanism that is independent of opioid receptor stimulation. J Mol Cell Cardiol 2001; 33: 1455–65
110.
Fryer RM, Hsu AK, Eells JT, Nagase H, Gross GJ: Opioid-induced second window of cardioprotection: Potential role of mitochondrial KATP channels. Circ Res 1999; 84: 846–51
111.
McPherson BC, Yao Z: Morphine mimics preconditioning via free radical signals and mitochondrial K(ATP) channels in myocytes. Circulation 2001; 103: 290–5
112.
Liang BT, Gross GJ: Direct preconditioning of cardiac myocytes via opioid receptors and KATP channels. Circ Res 1999; 84: 1396–400
113.
Schultz JE, Hsu AK, Gross GJ: Morphine mimics the cardioprotective effect of ischemic preconditioning via a glibenclamide-sensitive mechanism in the rat heart. Circ Res 1996; 78: 1100–4
114.
Schultz JE, Hsu AK, Gross GJ: Ischemic preconditioning in the intact rat heart is mediated by delta1- but not mu- or kappa-opioid receptors. Circulation 1998; 97: 1282–9
115.
Ishizawa Y, Pidikiti R, Liebman PA, Eckenhoff RG: G protein-coupled receptors as direct targets of inhaled anesthetics. Mol Pharmacol 2002; 61: 945–52
116.
Fryer RM, Schultz JE, Hsu AK, Gross GJ: Importance of PKC and tyrosine kinase in single or multiple cycles of preconditioning in rat hearts. Am J Physiol Heart Circ Physiol 1999; 276: H1229–35
117.
Fryer RM, Patel HH, Hsu AK, Gross GJ: Stress-activated protein kinase phosphorylation during cardioprotection in the ischemic myocardium. Am J Physiol Heart Circ Physiol 2001; 281: H1184–92
118.
Fryer RM, Pratt PF, Hsu AK, Gross GJ: Differential activation of extracellular signal regulated kinase isoforms in preconditioning and opioid-induced cardioprotection. J Pharmacol Exp Ther 2001; 296: 642–9
119.
Liu H, McPherson BC, Yao Z: Preconditioning attenuates apoptosis and necrosis: Role of protein kinase C epsilon and -delta isoforms. Am J Physiol Heart Circ Physiol 2001; 281: H404–10
120.
Puceat M, Vassort G: Signalling by protein kinase C isoforms in the heart. Mol Cell Biochem 1996; 157: 65–72
121.
Liu GS, Thornton J, Van Winkle DM, Stanley AW, Olsson RA, Downey JM: Protection against infarction afforded by preconditioning is mediated by A1 adenosine receptors in rabbit heart. Circulation 1991; 84: 350–6
122.
Tsuchida A, Miura T, Miki T, Shimamoto K, Iimura O: Role of adenosine receptor activation in myocardial infarct size limitation by ischaemic preconditioning. Cardiovasc Res 1992; 26: 456–61
123.
Wall TM, Sheehy R, Hartman JC: Role of bradykinin in myocardial preconditioning. J Pharmacol Exp Ther 1994; 270: 681–9
124.
Goto M, Liu Y, Yang XM, Ardell JL, Cohen MV, Downey JM: Role of bradykinin in protection of ischemic preconditioning in rabbit hearts. Circ Res 1995; 77: 611–21
125.
Schultz JE, Rose E, Yao Z, Gross GJ: Evidence for involvement of opioid receptors in ischemic preconditioning in rat hearts. Am J Physiol Heart Circ Physiol 1995; 268: H2157–61
126.
Schultz JJ, Hsu AK, Gross GJ: Ischemic preconditioning and morphine-induced cardioprotection involve the delta (delta)-opioid receptor in the intact rat heart. J Mol Cell Cardiol 1997; 29: 2187–95
127.
Hemmings HC Jr, Adamo AI: Activation of endogenous protein kinase C by halothane in synaptosomes. A nesthesiology 1996; 84: 652–62
128.
Hemmings HC Jr: General anesthetic effects on protein kinase C. Toxicol Lett 1998; 100–101: 89–95
129.
Uecker M, da Silva R, Grampp T, Pasch T, Schaub MC, Zaugg M: Translocation of protein kinase C isoforms to subcellular targets in ischemic and anesthetic preconditioning. A nesthesiology 2003; 99: 138–47
130.
Ismaeil MS, Tkachenko I, Hickey RF, Cason BA: Colchicine inhibits isoflurane-induced preconditioning. A nesthesiology 1999; 91: 1816–22
131.
Sato T, O’Rourke B, Marban E: Modulation of mitochondrial ATP-dependent K+ channels by protein kinase C. Circ Res 1998; 83: 110–4
132.
Hu K, Duan D, Li GR, Nattel S: Protein kinase C activates ATP-sensitive K+ current in human and rabbit ventricular myocytes. Circ Res 1996; 78: 492–8
133.
Liu Y, Gao WD, O’Rourke B, Marban E: Synergistic modulation of ATP-sensitive K+ currents by protein kinase C and adenosine: Implications for ischemic preconditioning. Circ Res 1996; 78: 443–54
134.
Sato T, Sasaki N, O’Rourke B, Marban E: Adenosine primes the opening of mitochondrial ATP-sensitive potassium channels: A key step in ischemic preconditioning? Circulation 2000; 102: 800–5
135.
Light PE, Bladen C, Winkfein RJ, Walsh MP, French RJ: Molecular basis of protein kinase C-induced activation of ATP-sensitive potassium channels. Proc Natl Acad Sci U S A 2000; 97: 9058–63
136.
Ohnuma Y, Miura T, Miki T, Tanno M, Kuno A, Tsuchida A, Shimamoto K: Opening of mitochondrial K(ATP) channel occurs downstream of PKC-epsilon activation in the mechanism of preconditioning. Am J Physiol Heart Circ Physiol 2002; 283: H440–7
137.
Baines CP, Wang L, Cohen MV, Downey JM: Protein tyrosine kinase is downstream of protein kinase C for ischemic preconditioning’s anti-infarct effect in the rabbit heart. J Mol Cell Cardiol 1998; 30: 383–92
138.
Ping P, Zhang J, Zheng YT, Li RC, Dawn B, Tang XL, Takano H, Balafanova Z, Bolli R: Demonstration of selective protein kinase C-dependent activation of Src and Lck tyrosine kinases during ischemic preconditioning in conscious rabbits. Circ Res 1999; 85: 542–50
139.
Ping P, Zhang J, Cao X, Li RC, Kong D, Tang XL, Qiu Y, Manchikalapudi S, Auchampach JA, Black RG, Bolli R: PKC-dependent activation of p44/p42 MAPKs during myocardial ischemia-reperfusion in conscious rabbits. Am J Physiol Heart Circ Physiol 1999; 276: H1468–81
140.
Ytrehus K, Liu Y, Downey JM: Preconditioning protects ischemic rabbit heart by protein kinase C activation. Am J Physiol Heart Circ Physiol 1994; 266: H1145–52
141.
Tanno M, Tsuchida A, Nozawa Y, Matsumoto T, Hasegawa T, Miura T, Shimamoto K: Roles of tyrosine kinase and protein kinase C in infarct size limitation by repetitive ischemic preconditioning in the rat. J Cardiovasc Pharmacol 2000; 35: 345–52
142.
Hattori R, Otani H, Uchiyama T, Imamura H, Cui J, Maulik N, Cordis GA, Zhu L, Das DK: Src tyrosine kinase is the trigger but not the mediator of ischemic preconditioning. Am J Physiol Heart Circ Physiol 2001; 281: H1066–74
143.
Fryer RM, Hsu AK, Gross GJ: ERK and p38 MAP kinase activation are components of opioid-induced delayed cardioprotection. Basic Res Cardiol 2001; 96: 136–42
144.
Sanada S, Kitakaze M, Papst PJ, Hatanaka K, Asanuma H, Aki T, Shinozaki Y, Ogita H, Node K, Takashima S, Asakura M, Yamada J, Fukushima T, Ogai A, Kuzuya T, Mori H, Terada N, Yoshida K, Hori M: Role of phasic dynamism of p38 mitogen-activated protein kinase activation in ischemic preconditioning of the canine heart. Circ Res 2001; 88: 175–80
145.
Mocanu MM, Baxter GF, Yue Y, Critz SD, Yellon DM: The p38 MAPK inhibitor, SB203580, abrogates ischaemic preconditioning in rat heart but timing of administration is critical. Basic Res Cardiol 2000; 95: 472–8
146.
Canman CE, Kastan MB: Signal transduction: Three paths to stress relief. Nature 1996; 384: 213–4
147.
Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, Takagi M, Matsumoto K, Miyazono K, Gotoh Y: Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 1997; 275: 90–4
148.
Xia Z, Dickens M, Raingeaud J, Davis RJ, Greenberg ME: Opposing effects of ERK and JNK-p38 MAP kinases on apoptosis. Science 1995; 270: 1326–31
149.
da Silva R, Grampp T, Pasch T, Schaub MC, Zaugg M: Differential activation of mitogen-activated protein kinases in ischemic and anesthetic preconditioning. A nesthesiology 2004; 100: 59–69
150.
Zweier JL, Flaherty JT, Weisfeldt ML: Direct measurement of free radical generation following reperfusion of ischemic myocardium. Proc Natl Acad Sci U S A 1987; 84: 1404–7
151.
Bolli R, Patel BS, Jeroudi MO, Lai EK, McCay PB: Demonstration of free radical generation in “stunned” myocardium of intact dogs with the use of the spin trap alpha-phenyl N-tert-butyl nitrone. J Clin Invest 1988; 82: 476–85
152.
Ambrosio G, Zweier JL, Duilio C, Kuppusamy P, Santoro G, Elia PP, Tritto I, Cirillo P, Condorelli M, Chiariello M: Evidence that mitochondrial respiration is a source of potentially toxic oxygen free radicals in intact rabbit hearts subjected to ischemia and reflow. J Biol Chem 1993; 268: 18532–41
153.
Tanguay M, Blaise G, Dumont L, Beique G, Hollmann C: Beneficial effects of volatile anesthetics on decrease in coronary flow and myocardial contractility induced by oxygen-derived free radicals in isolated rabbit hearts. J Cardiovasc Pharmacol 1991; 18: 863–70
154.
Nakamura T, Kashimoto S, Oguchi T, Kumazawa T: Hydroxyl radical formation during inhalation anesthesia in the reperfused working rat heart. Can J Anaesth 1999; 46: 470–5
155.
Glantz L, Ginosar Y, Chevion M, Gozal Y, Elami A, Navot N, Kitrossky N, Drenger B: Halothane prevents postischemic production of hydroxyl radicals in the canine heart. A nesthesiology 1997; 86: 440–7
156.
Hu G, Vinten-Johansen J, Salem MR, Zhao ZQ, Crystal GJ: Isoflurane inhibits neutrophil-endothelium interactions in the coronary circulation: Lack of a role for adenosine triphosphate-sensitive potassium channels. Anesth Analg 2002; 94: 849–56
157.
Hu G, Vasiliauskas T, Salem MR, Rhone DP, Crystal GJ: Neutrophils pretreated with volatile anesthetics lose ability to cause cardiac dysfunction. A nesthesiology 2003; 98: 712–8
158.
Ambrosio G, Tritto I, Chiariello M: The role of oxygen free radicals in preconditioning. J Mol Cell Cardiol 1995; 27: 1035–9
159.
Tritto I, D’Andrea D, Eramo N, Scognamiglio A, De Simone C, Violante A, Esposito A, Chiariello M, Ambrosio G: Oxygen radicals can induce preconditioning in rabbit hearts. Circ Res 1997; 80: 743–8
160.
Vanden Hoek TL, Becker LB, Shao Z, Li C, Schumacker PT: Reactive oxygen species released from mitochondria during brief hypoxia induce preconditioning in cardiomyocytes. J Biol Chem 1998; 273: 18092–8
161.
Baines CP, Goto M, Downey JM: Oxygen radicals released during ischemic preconditioning contribute to cardioprotection in the rabbit myocardium. J Mol Cell Cardiol 1997; 29: 207–16
162.
Tanaka M, Fujiwara H, Yamasaki K, Sasayama S: Superoxide dismutase and N-2-mercaptopropionyl glycine attenuate infarct size limitation effect of ischaemic preconditioning in the rabbit. Cardiovasc Res 1994; 28: 980–6
163.
Pain T, Yang XM, Critz SD, Yue Y, Nakano A, Liu GS, Heusch G, Cohen MV, Downey JM: Opening of mitochondrial K(ATP) channels triggers the preconditioned state by generating free radicals. Circ Res 2000; 87: 460–6
164.
Droge W: Free radicals in the physiological control of cell function. Physiol Rev 2002; 82: 47–95
165.
Clerk A, Michael A, Sugden PH: Stimulation of multiple mitogen-activated protein kinase sub-families by oxidative stress and phosphorylation of the small heat shock protein, HSP25/27, in neonatal ventricular myocytes. Biochem J 1998; 333: 581–9
166.
Samavati L, Monick MM, Sanlioglu S, Buettner GR, Oberley LW, Hunninghake GW: Mitochondrial K(ATP) channel openers activate the ERK kinase by an oxidant-dependent mechanism. Am J Physiol Cell Physiol 2002; 283: C273–81
167.
Kulisz A, Chen N, Chandel NS, Shao Z, Schumacker PT: Mitochondrial ROS initiate phosphorylation of p38 MAP kinase during hypoxia in cardiomyocytes. Am J Physiol Lung Cell Mol Physiol 2002; 282: L1324–9
168.
Nishida M, Maruyama Y, Tanaka R, Kontani K, Nagao T, Kurose H: G alpha(i) and G alpha(o) are target proteins of reactive oxygen species. Nature 2000; 408: 492–5
169.
Nishida M, Schey KL, Takagahara S, Kontani K, Katada T, Urano Y, Nagano T, Nagao T, Kurose H: Activation mechanism of Gi and Go by reactive oxygen species. J Biol Chem 2002; 277: 9036–42
170.
Novalija E, Kevin LG, Camara AK, Bosnjak ZJ, Kampine JP, Stowe DF: Reactive oxygen species precede the epsilon isoform of protein kinase C in the anesthetic preconditioning signaling cascade. A nesthesiology 2003; 99: 421–8
171.
Patel HH, Gross GJ: Diazoxide induced cardioprotection: What comes first, K(ATP) channels or reactive oxygen species? Cardiovasc Res 2001; 51: 633–6
172.
Carroll R, Gant VA, Yellon DM: Mitochondrial K(ATP) channel opening protects a human atrial-derived cell line by a mechanism involving free radical generation. Cardiovasc Res 2001; 51: 691–700
173.
Forbes RA, Steenbergen C, Murphy E: Diazoxide-induced cardioprotection requires signaling through a redox-sensitive mechanism. Circ Res 2001; 88: 802–9
174.
Zhang DX, Chen YF, Campbell WB, Zou AP, Gross GJ, Li PL: Characteristics and superoxide-induced activation of reconstituted myocardial mitochondrial ATP-sensitive potassium channels. Circ Res 2001; 89: 1177–83
175.
Lebuffe G, Schumacker PT, Shao ZH, Anderson T, Iwase H, Vanden Hoek TL: ROS and NO trigger early preconditioning: Relationship to mitochondrial KATP channel. Am J Physiol Heart Circ Physiol 2003; 284: H299–308
176.
Gross GJ, Fryer RM: Mitochondrial K(ATP) channels: Triggers or distal effectors of ischemic or pharmacological preconditioning? Circ Res 2000; 87: 431–3
177.
Tanaka K, Weihrauch D, Ludwig LM, Kersten JR, Pagel PS, Warltier DC: Mitochondrial adenosine triphosphate-regulated potassium channel opening acts as a trigger for isoflurane-induced preconditioning by generating reactive oxygen species. A nesthesiology 2003; 98: 935–43
178.
Kevin LG, Novalija E, Riess ML, Camara AK, Rhodes SS, Stowe DF: Sevoflurane exposure generates superoxide but leads to decreased superoxide during ischemia and reperfusion in isolated hearts. Anesth Analg 2003; 96: 949–55
179.
Yao Z, Tong J, Tan X, Li C, Shao Z, Kim WC, vanden Hoek TL, Becker LB, Head CA, Schumacker PT: Role of reactive oxygen species in acetylcholine-induced preconditioning in cardiomyocytes. Am J Physiol Heart Circ Physiol 1999; 277: H2504–9
180.
Vanden Hoek TL, Shao Z, Li C, Schumacker PT, Becker LB: Mitochondrial electron transport can become a significant source of oxidative injury in cardiomyocytes. J Mol Cell Cardiol 1997; 29: 2441–50
181.
Hanley PJ, Ray J, Brandt U, Daut J: Halothane, isoflurane and sevoflurane inhibit NADH: ubiquinone oxidoreductase (complex I) of cardiac mitochondria. J Physiol 2002; 544: 687–93
182.
Riess ML, Stowe DF, Henry MM, Camara AK, Eells JT, Kevin LG: Concentration dependent attenuation of mitochondrial respiration by sevoflurane in isolated cardiac mitochondria is mediated in part by reactive oxygen species (on-line abstract available at:www.anesthesiology.org). A nesthesiology 2003; 99:A1556
183.
Tanaka K, Ludwig LM, Kersten JR, Pagel PS, Warltier DC: Reactive oxygen species released from mitochondrial electron transport chain complex III are involved in isoflurane-induced preconditioning in vivo  (on-line abstract available at:www.anesthesiology.org). A nesthesiology 2003; 99:A735
184.
Mohazzab KM, Kaminski PM, Wolin MS: NADH oxidoreductase is a major source of superoxide anion in bovine coronary artery endothelium. Am J Physiol Heart Circ Physiol 1994; 266: H2568–72
185.
Xia Y, Zweier JL: Superoxide and peroxynitrite generation from inducible nitric oxide synthase in macrophages. Proc Natl Acad Sci U S A 1997; 94: 6954–8
186.
Turrens JF: Superoxide production by the mitochondrial respiratory chain. Biosci Rep 1997; 17: 3–8
187.
Cai H, Harrison DG: Endothelial dysfunction in cardiovascular diseases: The role of oxidant stress. Circ Res 2000; 87: 840–4
188.
Abe J, Berk BC: Reactive oxygen species as mediators of signal transduction in cardiovascular disease. Trends Cardiovasc Med 1998; 8: 59–64
189.
Wang XT, McCullough KD, Wang XJ, Carpenter G, Holbrook NJ: Oxidative stress-induced phospholipase C-gamma 1 activation enhances cell survival. J Biol Chem 2001; 276: 28364–71
190.
Maulik N, Watanabe M, Zu YL, Huang CK, Cordis GA, Schley JA, Das DK: Ischemic preconditioning triggers the activation of MAP kinases and MAPKAP kinase 2 in rat hearts. FEBS Lett 1996; 396: 233–7
191.
Aikawa R, Komuro I, Yamazaki T, Zou Y, Kudoh S, Tanaka M, Shiojima I, Hiroi Y, Yazaki Y: Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J Clin Invest 1997; 100: 1813–21
192.
Dabrowski A, Boguslowicz C, Dabrowska M, Tribillo I, Gabryelewicz A: Reactive oxygen species activate mitogen-activated protein kinases in pancreatic acinar cells. Pancreas 2000; 21: 376–84
193.
Carter WO, Narayanan PK, Robinson JP: Intracellular hydrogen peroxide and superoxide anion detection in endothelial cells. J Leukoc Biol 1994; 55: 253–8
194.
Cuzzocrea S, Riley DP, Caputi AP, Salvemini D: Antioxidant therapy: A new pharmacological approach in shock, inflammation, and ischemia/reperfusion injury. Pharmacol Rev 2001; 53: 135–59
195.
Liu Y, Gutterman DD: Oxidative stress and potassium channel function. Clin Exp Pharmacol Physiol 2002; 29: 305–11
196.
Nichols CG, Lederer WJ: The regulation of ATP-sensitive K+ channel activity in intact and permeabilized rat ventricular myocytes. J Physiol 1990; 423: 91–110
197.
Yao Z, Gross GJ: Effects of the KATP channel opener bimakalim on coronary blood flow, monophasic action potential duration, and infarct size in dogs. Circulation 1994; 89: 1769–75
198.
Munch-Ellingsen J, Lokebo JE, Bugge E, Jonassen AK, Ravingerova T, Ytrehus K: 5-HD abolishes ischemic preconditioning independently of monophasic action potential duration in the heart. Basic Res Cardiol 2000; 95: 228–34
199.
Hamada K, Yamazaki J, Nagao T: Shortening of action potential duration is not prerequisite for cardiac protection by ischemic preconditioning or a KATP channel opener. J Mol Cell Cardiol 1998; 30: 1369–79
200.
Suzuki M, Sasaki N, Miki T, Sakamoto N, Ohmoto-Sekine Y, Tamagawa M, Seino S, Marban E, Nakaya H: Role of sarcolemmal K(ATP) channels in cardioprotection against ischemia/reperfusion injury in mice. J Clin Invest 2002; 109: 509–16
201.
Dos Santos P, Kowaltowski AJ, Laclau MN, Seetharaman S, Paucek P, Boudina S, Thambo JB, Tariosse L, Garlid KD: Mechanisms by which opening the mitochondrial ATP- sensitive K(+) channel protects the ischemic heart. Am J Physiol Heart Circ Physiol 2002; 283: H284–95
202.
Dzeja PP, Holmuhamedov EL, Ozcan C, Pucar D, Jahangir A, Terzic A: Mitochondria: Gateway for cytoprotection. Circ Res 2001; 89: 744–6
203.
Holmuhamedov EL, Wang L, Terzic A: ATP-sensitive K+ channel openers prevent Ca2+ overload in rat cardiac mitochondria. J Physiol 1999; 519: 347–60
204.
Holmuhamedov EL, Jovanovic S, Dzeja PP, Jovanovic A, Terzic A: Mitochondrial ATP-sensitive K+ channels modulate cardiac mitochondrial function. Am J Physiol Heart Circ Physiol 1998; 275: H1567–76
205.
Minners J, Lacerda L, McCarthy J, Meiring JJ, Yellon DM, Sack MN: Ischemic and pharmacological preconditioning in Girardi cells and C2C12 myotubes induce mitochondrial uncoupling. Circ Res 2001; 89: 787–92
206.
Halestrap AP: The regulation of the matrix volume of mammalian mitochondria in vivo and in vitro and its role in the control of mitochondrial metabolism. Biochim Biophys Acta 1989; 973: 355–82
207.
Garlid KD: Cation transport in mitochondria: The potassium cycle. Biochim Biophys Acta 1996; 1275: 123–6
208.
Garlid KD: On the mechanism of regulation of the mitochondrial K+/H+ exchanger. J Biol Chem 1980; 255: 11273–9
209.
Green DR, Reed JC: Mitochondria and apoptosis. Science 1998; 281: 1309–12
210.
Akao M, Ohler A, O’Rourke B, Marban E: Mitochondrial ATP-sensitive potassium channels inhibit apoptosis induced by oxidative stress in cardiac cells. Circ Res 2001; 88: 1267–75
211.
Ozcan C, Bienengraeber M, Dzeja PP, Terzic A: Potassium channel openers protect cardiac mitochondria by attenuating oxidant stress at reoxygenation. Am J Physiol Heart Circ Physiol 2002; 282: H531–9
212.
Fryer RM, Eells JT, Hsu AK, Henry MM, Gross GJ: Ischemic preconditioning in rats: Role of mitochondrial K(ATP) channel in preservation of mitochondrial function. Am J Physiol Heart Circ Physiol 2000; 278: H305–12
213.
Tanonaka K, Taguchi T, Koshimizu M, Ando T, Morinaka T, Yogo T, Konishi F, Takeo S: Role of an ATP-sensitive potassium channel opener, YM934, in mitochondrial energy production in ischemic/reperfused heart. J Pharmacol Exp Ther 1999; 291: 710–6
214.
Novalija E, Kevin LG, Eells JT, Henry MM, Stowe DF: Anesthetic preconditioning improves adenosine triphosphate synthesis and reduces reactive oxygen species formation in mitochondria after ischemia by a redox dependent mechanism. A nesthesiology 2003; 98: 1155–63
215.
Riess ML, Novalija E, Camara AK, Eells JT, Chen Q, Stowe DF: Preconditioning with sevoflurane reduces changes in nicotinamide adenine dinucleotide during ischemia-reperfusion in isolated hearts: Reversal by 5-hydroxydecanoic acid. A nesthesiology 2003; 98: 387–95
216.
Zorov DB, Filburn CR, Klotz LO, Zweier JL, Sollott SJ: Reactive oxygen species (ROS)-induced ROS release: A new phenomenon accompanying induction of the mitochondrial permeability transition in cardiac myocytes. J Exp Med 2000; 192: 1001–14
217.
Kroemer G, Reed JC: Mitochondrial control of cell death. Nat Med 2000; 6: 513–9
218.
Kowaltowski AJ, Castilho RF, Vercesi AE: Mitochondrial permeability transition and oxidative stress. FEBS Lett 2001; 495: 12–5
219.
Hausenloy DJ, Maddock HL, Baxter GF, Yellon DM: Inhibiting mitochondrial permeability transition pore opening: A new paradigm for myocardial preconditioning? Cardiovasc Res 2002; 55: 534–43
220.
Piriou V, Chiari P, Roesch OG, Lehot JJ, Ovize M: Effect of desflurane-induced preconditioning on mitochondrial transition pore opening (on-line abstract available at:www.anesthesiology.org). A nesthesiology 2003; 99:A1538
221.
Allard MF, Flint JD, English JC, Henning SL, Salamanca MC, Kamimura CT, English DR: Calcium overload during reperfusion is accelerated in isolated hypertrophied rat hearts. J Mol Cell Cardiol 1994; 26: 1551–63
222.
Miyamae M, Camacho SA, Weiner MW, Figueredo VM: Attenuation of postischemic reperfusion injury is related to prevention of [Ca2+]m overload in rat hearts. Am J Physiol Heart Circ Physiol 1996; 271: H2145–53
223.
Di Lisa F, Bernardi P: Mitochondrial function as a determinant of recovery or death in cell response to injury. Mol Cell Biochem 1998; 184: 379–91
224.
Wang L, Cherednichenko G, Hernandez L, Halow J, Camacho SA, Figueredo V, Schaefer S: Preconditioning limits mitochondrial Ca(2+) during ischemia in rat hearts: Role of K(ATP) channels. Am J Physiol Heart Circ Physiol 2001; 280: H2321–8
225.
Riess ML, Camara AK, Novalija E, Chen Q, Rhodes SS, Stowe DF: Anesthetic preconditioning attenuates mitochondrial Ca2+ overload during ischemia in guinea pig intact hearts: Reversal by 5-hydroxydecanoic acid. Anesth Analg 2002; 95: 1540–6
226.
Siegmund B, Schlack W, Ladilov YV, Balser C, Piper HM: Halothane protects cardiomyocytes against reoxygenation-induced hypercontracture. Circulation 1997; 96: 4372–9
227.
Davies LA, Gibson CN, Boyett MR, Hopkins PM, Harrison SM: Effects of isoflurane, sevoflurane, and halothane on myofilament Ca2+sensitivity and sarcoplasmic reticulum Ca2+release in rat ventricular myocytes. A nesthesiology 2000; 93: 1034–44
228.
Zucchi R, Ronca F, Ronca-Testoni S: Modulation of sarcoplasmic reticulum function: A new strategy in cardioprotection? Pharmacol Ther 2001; 89: 47–65
229.
Piper HM, Meuter K, Schafer C: Cellular mechanisms of ischemia-reperfusion injury. Ann Thorac Surg 2003; 75: S644–8
230.
Yellon DM, Dana A: The preconditioning phenomenon: A tool for the scientist or a clinical reality? Circ Res 2000; 87: 543–50
231.
Kloner RA, Jennings RB: Consequences of brief ischemia: Stunning, preconditioning, and their clinical implications: II. Circulation 2001; 104: 3158–67
232.
Deutsch E, Berger M, Kussmaul WG, Hirshfeld JW Jr, Herrmann HC, Laskey WK: Adaptation to ischemia during percutaneous transluminal coronary angioplasty: Clinical, hemodynamic, and metabolic features. Circulation 1990; 82: 2044–51
233.
Laskey WK: Beneficial impact of preconditioning during PTCA on creatine kinase release. Circulation 1999; 99: 2085–9
234.
Leesar MA, Stoddard M, Ahmed M, Broadbent J, Bolli R: Preconditioning of human myocardium with adenosine during coronary angioplasty. Circulation 1997; 95: 2500–7
235.
Saito S, Mizumura T, Takayama T, Honye J, Fukui T, Kamata T, Moriuchi M, Hibiya K, Tamura Y, Ozawa Y: Antiischemic effects of nicorandil during coronary angioplasty in humans. Cardiovasc Drugs Ther 1995; 9( suppl 2): 257–63
236.
Sakai K, Yamagata T, Teragawa H, Matsuura H, Chayama K: Nicorandil-induced preconditioning as evidenced by troponin T measurements after coronary angioplasty in patients with stable angina pectoris. Jpn Heart J 2002; 43: 443–53
237.
Okazaki Y, Kodama K, Sato H, Kitakaze M, Hirayama A, Mishima M, Hori M, Inoue M: Attenuation of increased regional myocardial oxygen consumption during exercise as a major cause of warm-up phenomenon. J Am Coll Cardiol 1993; 21: 1597–604
238.
Marber MS, Joy MD, Yellon DM: Is warm-up in angina ischaemic preconditioning? Br Heart J 1994; 72: 213–5
239.
Yellon DM, Alkhulaifi AM, Pugsley WB: Preconditioning the human myocardium. Lancet 1993; 342: 276–7
240.
Jenkins DP, Pugsley WB, Alkhulaifi AM, Kemp M, Hooper J, Yellon DM: Ischaemic preconditioning reduces troponin T release in patients undergoing coronary artery bypass surgery. Heart 1997; 77: 314–8
241.
Wu ZK, Tarkka MR, Pehkonen E, Kaukinen L, Honkonen EL, Kaukinen S: Ischaemic preconditioning has a beneficial effect on left ventricular haemodynamic function after a coronary artery biopass grafting operation. Scand Cardiovasc J 2000; 34: 247–53
242.
Wu ZK, Tarkka MR, Pehkonen E, Kaukinen L, Honkonen EL, Kaukinen S: Beneficial effects of ischemic preconditioning on right ventricular function after coronary artery bypass grafting. Ann Thorac Surg 2000; 70: 1551–7
243.
Wu ZK, Iivainen T, Pehkonen E, Laurikka J, Tarkka MR: Ischemic preconditioning suppresses ventricular tachyarrhythmias after myocardial revascularization. Circulation 2002; 106: 3091–6
244.
Laurikka J, Wu ZK, Iisalo P, Kaukinen L, Honkonen EL, Kaukinen S, Tarkka MR: Regional ischemic preconditioning enhances myocardial performance in off-pump coronary artery bypass grafting. Chest 2002; 121: 1183–9
245.
Kaukoranta PK, Lepojarvi MP, Ylitalo KV, Kiviluoma KT, Peuhkurinen KJ: Normothermic retrograde blood cardioplegia with or without preceding ischemic preconditioning. Ann Thorac Surg 1997; 63: 1268–74
246.
Cremer J, Steinhoff G, Karck M, Ahnsell T, Brandt M, Teebken OE, Hollander D, Haverich A: Ischemic preconditioning prior to myocardial protection with cold blood cardioplegia in coronary surgery. Eur J Cardiothorac Surg 1997; 12: 753–8
247.
Cremer J, Karck M, Ahnsel T, Steinhoff G, Brandt M, Hollander D, Teebken O, Zick G, Haverich A: Ischemic preconditioning as an adjunct to crystalloid or blood cardioplegia for myocardial protection in routine coronary surgery. Thorac Cardiovasc Surg 1998; 46( suppl 2): 298–301
248.
Carroll R, Yellon DM: Delayed cardioprotection in a human cardiomyocyte-derived cell line: The role of adenosine, p38MAP kinase and mitochondrial KATP. Basic Res Cardiol 2000; 95: 243–9
249.
Tarnow J, Markschies-Hornung A, Schulte-Sasse U: Isoflurane improves the tolerance to pacing-induced myocardial ischemia. A nesthesiology 1986; 64: 147–56
250.
Belhomme D, Peynet J, Louzy M, Launay JM, Kitakaze M, Menasche P: Evidence for preconditioning by isoflurane in coronary artery bypass graft surgery. Circulation 1999; 100( suppl 19): II340–4
251.
Haroun-Bizri S, Khoury SS, Chehab IR, Kassas CM, Baraka A: Does isoflurane optimize myocardial protection during cardiopulmonary bypass? J Cardiothorac Vasc Anesth 2001; 15: 418–21
252.
Penta de Peppo A, Polisca P, Tomai F, De Paulis R, Turani F, Zupancich E, Sommariva L, Pasqualetti P, Chiariello L: Recovery of LV contractility in man is enhanced by preischemic administration of enflurane. Ann Thorac Surg 1999; 68: 112–8
253.
De Hert SG, ten Broecke PW, Mertens E, Van Sommeren EW, De Blier IG, Stockman BA, Rodrigus IE: Sevoflurane but not propofol preserves myocardial function in coronary surgery patients. A nesthesiology 2002; 97: 42–9
254.
Conzen PF, Fischer S, Detter C, Peter K: Sevoflurane provides greater protection of the myocardium than propofol in patients undergoing off-pump coronary artery bypass surgery. A nesthesiology 2003; 99: 826–33
255.
De Hert SG, Cromheecke S, ten Broecke PW, Mertens E, De Blier IG, Stockman BA, Rodrigus IE, Van der Linden PJ: Effects of propofol, desflurane, and sevoflurane on recovery of myocardial function after coronary surgery in elderly high-risk patients. A nesthesiology 2003; 99: 314–23
256.
Julier K, da Silva R, Garcia C, Bestmann L, Frascarolo P, Zollinger A, Chassot PG, Schmid ER, Turina MI, von Segesser LK, Pasch T, Spahn DR, Zaugg M: Preconditioning by sevoflurane decreases biochemical markers for myocardial and renal dysfunction in coronary artery bypass graft surgery: A double-blinded, placebo-controlled, multicenter study. A nesthesiology 2003; 98: 1315–27
257.
Pouzet B, Lecharny JB, Dehoux M, Paquin S, Kitakaze M, Mantz J, Menasche P: Is there a place for preconditioning during cardiac operations in humans? Ann Thorac Surg 2002; 73: 843–8
258.
Warltier DC, Pagel PS, Kersten JR: Approaches to the prevention of perioperative myocardial ischemia. A nesthesiology 2000; 92: 253–9
259.
Van Der Linden PJ, Daper A, Trenchant A, De Hert SG: Cardioprotective effects of volatile anesthetics in cardiac surgery. A nesthesiology 2003; 99: 516–7
260.
Sniecinski R, Liu H: The effect of myocardial function protection of sevoflurane preconditioning in senescent rats (on-line abstract available at:www.anesthesiology.org). A nesthesiology 2003; 99:A1548
261.
Zaugg M, da Silva R, Sergeev PV, Pasch T, Schaub MC: Unraveling the gene expression patterns in ischemic and anesthetic preconditioning by gene chip analysis (on-line abstract available at:www.anesthesiology.org). A nesthesiology 2003; 99:A1544
262.
Homi HM, Mixco JM, Sheng H, Grocott HP, Pearlstein RD, Warner DS: Severe hypotension is not essential for isoflurane neuroprotection against forebrain ischemia in mice. A nesthesiology 2003; 99: 1145–51