Abstract
Ischaemic pre- and postconditioning are potent cardioprotective interventions that spare ischaemic myocardium and decrease infarct size after periods of myocardial ischaemia/reperfusion. They are dependent on complex signalling pathways involving ligands released from ischaemic myocardium, G-protein-linked receptors, membrane growth factor receptors, phospholipids, signalling kinases, NO, PKC and PKG, mitochondrial ATP-sensitive potassium channels, reactive oxygen species, TNF-α and sphingosine-1-phosphate. The final effector is probably the mitochondrial permeability transition pore and the signalling produces protection by preventing pore formation. Many investigators have worked to produce a roadmap of this signalling with the hope that it would reveal where one could intervene to therapeutically protect patients with acute myocardial infarction whose hearts are being reperfused. However, attempts to date to show efficacy of such an intervention in large clinical trials have been unsuccessful. Reasons for this inability to translate successes in the experimental laboratory to the clinical arena are evaluated in this review. It is suggested that all patients with acute coronary syndromes currently presenting to the hospital and being treated with platelet P2Y12 receptor antagonists, the current standard of care, are indeed already benefiting from protection from the conditioning pathways outlined earlier. If that proves to be the case, then future attempts to further decrease infarction will have to rely on interventions which protect by a different mechanism.
Tables of Links
TARGETS | |
---|---|
GPCRsa | Transportersd |
β-adrenoceptor | Na+/Ca2+ exchangers |
A1 receptor | Na+/H+ exchangers (NHE) |
A2A receptor | Enzymese |
A2B receptor | Akt |
A3 receptor | COX |
S1P1 receptor | eNOS |
S1P2 receptor | ERK |
S1P3 receptor | GSK-3β |
Ion channelsb | Guanylyl cyclase |
Connexin 43 (Cx43) | PI3K |
KATP channel | PKCδ |
Catalytic receptorsc | PKCε |
EGFR | PKG |
GPIIa | MMP |
GPIIb | SPHK1 |
PAF receptor | Src kinase |
P2Y12 receptor | |
TNFR1 | |
TNFR2 |
LIGANDS | |
---|---|
Adenosine | Isoflurane |
AG490 | L-NAME |
Aspirin | Metoprolol |
Atenolol | Nitric oxide (NO) |
BAY 58-2667 | PAF |
Bradykinin | PD98059 |
Cangrelor | Sevoflurane |
cGMP | Sphingosine |
Chelerythrine | Sphingosine 1-phosphate |
Clopidogrel | TGFβ1 |
Cyclosporin A | Ticagrelor |
Desflurane | Tirofiban |
Erythropoietin | Tyrosine |
Exenatide | Urocortin |
Glibenclamide | Wortmannin |
Insulin |
These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (a,b,c,d,eAlexander et al., 2013a,b,c,d,e).
Introduction
With the exception of revascularization, ischaemic preconditioning (IPC) is undeniably the most powerful cardioprotective intervention targeting ischaemia/reperfusion injury yet to be identified. All scientists agree that this intervention can salvage ischaemic myocardium following a period of ischaemia and reperfusion and reduce infarct size, the original observation and time-honored parameter of cardioprotection. Yet this success in the experimental laboratory has yet to be translated into a clinical procedure that has produced equally satisfying results. Thus, both scientists and clinicians continue to search for the miraculous intervention that can be applied to the patient with an acute myocardial infarction (AMI) to decrease infarct size and diminish the clinical sequelae of coronary artery occlusion and reperfusion. The closest we have come to this is revascularization therapy. In patients with coronary occlusion and AMI, current standards demand that the coronary artery be opened to reperfuse the ischaemic myocardium. Although tissue salvage understandably is dependent on reflow, this revascularization paradoxically creates injury of its own, so-called ‘reperfusion injury’. It is the latter which most recent cardioprotective interventions purport to target. To appreciate why identification of an appropriate cardioprotective agent has largely failed to date and to provide hope that we may be close to developing an effective approach, it is necessary to understand the history and science of cardioprotection.
Pioneering studies of infarct size modification
ST-segment shifts as a marker of infarct size
In 1971, Maroko, working in Braunwald's laboratory, proposed strategies for limiting necrosis following acute coronary occlusion (Maroko et al., 1971). Maroko et al. recognized the importance of infarct size on outcomes following infarction and were the first to suggest that infarction might be therapeutically reduced. These investigators mapped the degree of ST-segment shifts on the anterior myocardial surface at the end of 10-min coronary occlusions in dogs. After the heart was reperfused and had recovered, the occlusion and mapping were repeated after an experimental intervention. The sum of the ST-segment shifts was thought to reflect the severity of the ischaemia. If the sum of ST-segment shifts was attenuated, it was concluded that the intervention had protected the heart. The concept was brilliant in that it allowed each animal to serve as its own control, but it relied on the assumption that the ST-segment sum represented true infarct size which, unfortunately, turned out to not always be the case. Maroko et al. as well as subsequent investigators were also greatly handicapped by a lack of scientific information as to how ischaemia/reperfusion actually kills heart tissue. The Braunwald laboratory focused on a supply–demand relationship. Demand could be reduced by β-blockers and supply could be increased by interventions thought to promote oxygen delivery such as hyaluronidase (Maroko et al., 1972). As it turned out, the supply–demand relationship was but only one determinant of cell death. Yet their pioneering efforts started a field of research that still thrives today.
Reperfusion injury: a paradox
Hearse et al. (1973) introduced the ‘oxygen paradox’. Perfusion of a rat heart with hypoxic buffer for a prolonged period seemed to have little consequence, but switching back to oxygenated perfusate caused immediate cell destruction. While the reintroduction of oxygen was needed for recovery, at the same time it was associated with an injury. That was the paradox. The concept of reperfusion injury was very attractive because at that time it was recognized that AMI was caused by a coronary thrombus that could be dissolved with a thrombolytic agent. If much of the injury occurred at reperfusion, it would not be too late to prevent it with some intervention despite presentation of the patients with ischaemia in progress. It was hypothesized that reintroduction of oxygen produced a burst of free radicals that in turn led to membrane damage, interference with ion pumps and volume dysregulation. A closely associated hypothesis was that leukocytes would invade reperfused tissue and attack viable myocytes by releasing free radicals. Personnel in Lucchesi's laboratory concentrated on the role of free radicals in myocardial infarction (Jolly et al., 1984). Thus, free radical scavengers appeared to decrease infarction in a canine model of ischaemia/reperfusion. Although these studies were championed by local advocates, the inconsistent results obtained in other independent laboratories suggested problems with this approach (Reimer et al., 1989). The same held for investigations of anti-inflammatory agents (Tissier et al., 2007a). The reason for divergent results among the many studies of antioxidant and anti-inflammatory agents have never been resolved, but even in the most supportive studies salvage was hardly greater than 10%, probably too modest to have meaningful clinical impact. One began to wonder if it was even possible to alter the vulnerability of ischaemic myocardium to infarction.
IPC
Then, in 1986, Charles Murry, in the laboratory of Reimer and Jennings, made a seminal observation (Murry et al., 1986). It was reported that preceding a 40 min coronary occlusion in dogs with four cycles of 5 min coronary occlusion/5 min reperfusion would decrease the amount of infarction of the risk area subtended by the occluded vessel from 28 to 7%. That was a 75% reduction in infarct size despite the fact that those hearts endured an additional 20 min of ischaemia. They called this phenomenon IPC. Perhaps because of Murry's frankly antithetical observation that more ischaemia was better, confirmation of the observation was not immediate. Three years passed before scientific papers dealing with IPC began to appear. But when they did, confirmation was overwhelming. Those studies noted that the intervention uniformly protected canine (Murry et al., 1986; Gross and Auchampach, 1992), rodent (Liu and Downey, 1992; Yellon et al., 1992), porcine (Schott et al., 1990), rabbit (Van Winkle et al., 1991; Toombs et al., 1993), primate (Yang et al., 2010) and even avian (Rischard and McKean, 1998) hearts from myocardial infarction. At last there was conclusive proof that infarct size could be modified, at least by this singular intervention of IPC. Of course, therapeutic IPC of a heart would be impossible to implement clinically in any setting except, perhaps, open heart surgery. Translation of IPC into clinical practice would have to wait until its mechanism was better understood before a treatment could be identified that could be administered after ischaemia had begun.
Mechanism of IPC: triggering phase
Surface receptors trigger IPC
The first insight into IPC's mechanism was reported by Liu et al. (1991). They announced that IPC is triggered by receptor occupancy. Activation of the Gi-coupled adenosine A1 receptor in rabbits triggered IPC's protection. Thus, an adenosine receptor antagonist blocked IPC's protection, while infusion of adenosine or an A1-selective agonist in lieu of brief ischaemia duplicated IPC's protection. Liu et al. proposed that net dephosphorylation of ATP during ischaemia results in production and release of adenosine which then would bind to A1 adenosine receptors leading to a preconditioned phenotype. So had these investigators defined IPC's mechanism and were they ready to propose an intervention that could be used clinically? Hardly. They had identified a pharmacological trigger, but unfortunately the trigger, like IPC, had to be given prior to the onset of ischaemia. Identification of more parts of IPC's signal transduction pathway and of the overall mechanism would be required.
Opioid and bradykinin's signalling
Two other endogenously released trigger substances, bradykinin (Wall et al., 1994) and opioids (Schultz et al., 1995), were also found to be involved in IPC's protective action. Inhibition of any of these three receptors aborted protection from a single preconditioning cycle. However, simply increasing the number of preconditioning cycles could restore protection suggesting that the three receptors had an additive effect which was required to reach a protective threshold (Goto et al., 1995). Thus, the additional cycles of ischaemia/reperfusion produced increased stimulation of the two uninhibited receptors so that the protective threshold could finally be reached.
All three of these triggers, adenosine, bradykinin and opioids, bind to Gi-coupled receptors. The proposed multiple trigger theory implies that all triggers converge on a common target. Ytrehus et al. (1994) reported that PKC seemed to play a major role in IPC and it was found that protection triggered by any of the three receptors could be blocked by PKC inhibitors (Goto et al., 1995; Sakamoto et al., 1995; Baines et al., 1997; Miki et al., 1998a). Thus, PKC is believed to be this common target. Hence, adenosine, bradykinin and opioids bind to their respective receptors and the second messenger G-protein is cleaved into active α and βγ subunits which then result in activation of PKC. It would seem intuitive that the various agonists coupled to common Gi-proteins should trigger identical signalling. However, mysteriously this is not the case. Adenosine, bradykinin and opioids activate very divergent pathways; however, all three pathways eventually converge on PKC.
Opioid cardioprotection is dependent on downstream metalloproteinase and EGF receptor (Cohen et al., 2007a). This part of the signalling pathway was first mapped by studying ACh-stimulated receptors, Gi-coupled receptors, whose downstream protection is governed by signalling similar to that of opioids (Krieg et al., 2002; 2004,; Oldenburg et al., 2002; 2003,). While ACh is a potent trigger of preconditioning's protection, it is not a physiological trigger as transient ischaemia does not cause its release. ACh binds to its receptor resulting in cleavage of Gi and subsequent metalloproteinase-dependent cleavage of heparin-binding EGF-like growth factor (HB-EGF) from membrane-associated pro-HB-EGF (Figure 1). The liberated HB-EGF then activates membrane-bound EGFR by binding to its ectodomain resulting in EGFR dimerization which in turn leads to autophosphorylation of tyrosine residues on both EGFRs and binding of sarcoma (src) tyrosine kinase to form a signalling module. The latter attracts and activates PI3K.
Figure 1.
Proposed signalling scheme for conditioning. Abbreviations: Brady, bradykinin; eNOS, endothelial NOS; KATP, ATP-dependent potassium channel; MEK, MAPK kinase; MMP, matrix metalloproteinase; p70S6K, p70S6 kinase; PDK1/2, 3′-phosphoinositide-dependent kinase-1/-2; PI3,4,5P3, phosphatidylinositol trisphosphate; PI4,5P2, phosphatidylinositol bisphosphate; Pro, pro-HB-EGF; Sphingo 1-P, sphingosine 1-phosphate; Src, sarcoma tyrosine kinase; TNF, TNF-α; Tyr, tyrosine. Modified from Tissier et al. (2007a).
Bradykinin's signalling is comparable, although a different metalloproteinase is involved. Methner et al. (2009) demonstrated involvement of metalloproteinase-8 and the EGFR. Downstream steps are similar to those for ACh and opioids (Cohen et al., 2007a).
NO
PI3K-produced phosphorylated lipid metabolites phosphatidylinositol 3,4,5-trisphosphate and phosphatidylinositol 3,4-bisphosphate induce Akt to translocate to the plasma membrane (Andjelković et al., 1997) where it is phosphorylated by PDK1 and 2 (Stephens et al., 1998) and this initiates a signalling cascade. Akt activates ERK and endothelial NOS (Dimmeler et al., 1999). The latter enzyme catalyzes production of NO which stimulates guanylyl cyclase (GC). GC catalyzes the production of cGMP which itself activates PKG (Figure 1).
NO is a gaseous free radical and important biological regulator and cellular signalling molecule. In 1992, Vegh et al. (1992) proposed that endogenous NO might be involved in preconditioning. This announcement triggered a significant controversy regarding the precise role of NO in IPC (Weselcouch et al., 1995) to which we unwittingly contributed. In a study in in vitro rabbit hearts published in 2000, we noted that Nω-nitro-L-arginine methyl ester (L-NAME), a NOS inhibitor, had no effect on the dramatic protection induced by IPC, whereas the NO donor S-nitroso-N-acetylpenicillamine administered before the index ischaemia in lieu of the repeated brief 5 min coronary occlusions mimicked IPC and protected hearts (Nakano et al., 2000). We concluded that exogenously administered NO could trigger the preconditioned state, but that endogenous production of NO was not involved in IPC. This conundrum was not resolved for several years until we repeated studies with L-NAME in IPC in in vivo rabbit hearts (Cohen et al., 2006). L-NAME blocked the protection of IPC. Our earlier observations, although accurate, were dependent on the in vitro model used. As seen in Figure 1, bradykinin, opioids and adenosine are released by the ischaemic heart. But in the isolated, buffer-perfused heart, the absence of circulating kininogens would minimize release of bradykinin. In addition, opioid release would be attenuated because of the absence of cardiac innervation. Therefore, virtually all triggering would be the result of adenosine release which bypasses the NO-dependent trigger pathway (Figure 1).
As noted earlier, classical signalling dogma indicates that NO stimulates GC leading to generation of cGMP which in turn activates PKG (Figure 1). Studies with activators and inhibitors of PKG and cGMP analogues (Han et al., 2002; Oldenburg et al., 2004; Qin et al., 2004; Kuno et al., 2008) clearly demonstrated the involvement of PKG in IPC, and Baxter's laboratory (D'Souza et al., 2003; Burley et al., 2007) demonstrated increased myocardial levels of cGMP after protection by B-type natriuretic peptide. Additionally, BAY 58-2667, a NO-independent GC activator, conditioned rat and rabbit hearts (Krieg et al., 2009). Thus, in addition to proven involvement of endogenous NO, there is much evidence to support participation of GC and PKG in conditioning's protection.
Several investigators have also demonstrated involvement of a NO-mediated, PKG-independent signalling pathway (Sun et al., 2013; Penna et al., 2014). NO can directly modify sulfhydryl residues by S-nitrosylation. The latter is an important post-translational protein modification in signalling. IPC increases S-nitrosylation and IPC cardioprotection can be aborted by treatment with ascorbate which is a reducing agent resulting in specific degradation of S-nitrosylated compounds. Additionally, in isolated mouse hearts, pharmacologic inhibition of the soluble GC/cGMP/PKG pathway failed to block IPC-induced cardioprotection. Thus, in at least some models, this alternative pathway of NO signalling may be important and it is possible that each pathway may contribute to cardioprotection and the ischaemic stimulus itself or other unidentified factors may determine whether one or the other pathway is utilized. Once again, there is redundancy to the response to ischaemia which may be an adaptive change insuring a maximal cardioprotective result.
ATP-sensitive potassium channels and redox signalling
The next critical step in this signalling cascade is opening of an ATP-sensitive K+ channel (KATP). At the same time that we were uncovering the importance of adenosine in IPC, Garret Gross' laboratory was doing studies with KATP channels. Those investigators found that glibenclamide, a blocker of the channel, could also selectively block IPC's protection (Gross and Auchampach, 1992). Similarly, pretreatment with a KATP channel opener mimicked the protection (Gross and Auchampach, 1992). For a while it seemed that it had to be either adenosine or potassium channels which governed protection, but it turned out that they were simply two links in the same chain; both were involved. Several subsequent studies revealed that the KATP channel in question was located not on the sarcolemma but in the inner mitochondrial membrane (Garlid et al., 1997; Liu et al., 1998).
Mitochondrial KATP channels (mtKATP) are not triggers for IPC but rather are a critical link in the signalling pathway between surface receptors and PKC (Pain et al., 2000). Opening of mtKATP is PKG-dependent (Costa et al., 2005; 2008,), but the channels are obviously not accessible to cytosolic PKG. There are intermediate steps involving PKCε in the mitochondria which transmits the signal from cytosolic PKG to the mtKATP channel (Costa et al., 2005; 2008,; Jabůrek et al., 2006). One theory proposes that PKG reaches the mitochondria via signalosomes that bud off of sarcolemmal caveolae and contain critical signalling enzymes (Garlid et al., 2009). Channel opening permits K+ to enter the matrix along its electrochemical gradient. K+ influx is balanced by electrogenic H+ efflux driven by the respiratory chain.
An important link in this signalling is the redox coupling of mtKATP channel opening and PKC activation. Forbes et al. (2001) were the first to recognize this link when they noticed that either of the antioxidants N-acetylcysteine or N-2-mercaptopropionylglycine could block the protection from the mtKATP opener, diazoxide. It is not known exactly how mtKATP opening causes production of free radicals, but one theory is that mtKATP-dependent matrix alkalinization affects complex I and/or III which are poised to generate increased amounts of superoxide and its products H2O2 and hydroxyl radical (Costa and Garlid, 2008). All of the signalling steps to this point occur in ischaemic cells. However, generation of this burst of reactive oxygen species (ROS) must await reintroduction of oxygen into the myocardium which occurs during the reflow phase of the preconditioning cycle of ischaemia/reflow. There are many PKC isozymes. It appears that activation of PKCε is necessary and sufficient to achieve cardioprotection, while activation of PKCδ specifically blocks protection (Dorn et al., 1999; Ping et al., 2002; Inagaki et al., 2003a,b,). Thus, activation of PKC continues the signalling cascade.
The relationship among mtKATP, ROS and PKC is poorly understood. While ROS can directly activate PKC by causing release of Zn++ from the regulatory domain (Korichneva et al., 2002), connexin 43 (Cx43) appears to be a vital link in redox signalling. Cx43 which makes most of the gap junctions between cardiomyocytes was noted to be necessary for preconditioning's protection (Schwanke et al., 2002). It was later noted that protection depended on a mitochondrial population of Cx43 hemichannels located on the inner membrane. Depletion of these channels attenuates both protection and ROS production from an mtKATP opener (Heinzel et al., 2005). Most recently, it was shown that an mtKATP opener causes phosphorylation of Cx43 by PKC and that phosphorylation is required for protection (Srisakuldee et al., 2009). This suggests some sort of circular signalling circuit as phosphoCx43 is needed for ROS production and ROS cause PKC activation, but PKC phosphorylates Cx43. The role actually played by Cx43 in the protective process (e.g. a channel, a signalling molecule or a scaffold) is still a mystery.
The redox signalling step explains one of the mysteries of IPC. Why is the heart protected when a prolonged ischaemic period is preceded by a short coronary occlusion followed by reperfusion but yet is not protected during a single prolonged insult? All of the trigger receptors are activated during the single prolonged ischaemic insult, but signalling stops at the step requiring redox coupling to PKC because of the lack of oxygen which is supplied during IPC's short reperfusion. A ROS scavenger blocks protection from IPC and it can easily be seen that the critical time for that blockade is during IPC's reperfusion phase (Dost et al., 2008). While ROS-sensitive dyes indicate that radical production can occur during ischaemia (Becker et al., 1999), apparently the ROS species generated is not one capable of the redox signalling. We also have found that reperfusing with hypoxic perfusate during the preconditioning protocol abrogates IPC's protection (Dost et al., 2008). The identity of the ROS species involved has not been positively identified but seems to be a downstream product of HO· and is likely a product of phospholipid oxidation (Garlid et al., 2013).
Adenosine signalling
Signalling initiated by the third endogenous agonist that triggers IPC, adenosine, is different. Adenosine's cardioprotective effect is not dependent on Src tyrosine kinase or PI3K (Qin et al., 2003). Adenosine signalling seems to completely bypass mtKATP and ROS production (Cohen et al., 2001) and it more directly activates PKC (see Figure 1) which is where all of the trigger signalling converges. The adenosine A1 receptor is coupled through Gi to PLC and PLD. After the ligand binds to the receptor, Gi is cleaved into α and βγ moieties which activate PLC in the sarcolemma. This enzyme catalyzes the hydrolysis of membrane inositol-containing phospholipids, including phosphatidylinositol 4,5-bisphosphate. The resulting DAG stimulates translocation and activation of PKC. PLD also increases DAG levels by degrading phosphatidylcholine into choline and phosphatidic acid and the latter is transformed by a phosphohydrolase into DAG. These phospholipid activators of PKC also trigger release of zinc from PKC's regulatory domain (Korichneva et al., 2002). The diversity of signalling among the triggers is confusing, but also reassuring. The redundancy ensures cardioprotection even if one or more elements in the triggering cascade are blocked.
The mediator phase
All signalling to this point occurs during the preconditioning cycles of ischaemia and reflow. These steps are collectively called the trigger phase. Subsequent steps, and there are several, are part of the mediator phase which occurs following termination of the prolonged period of ischaemia (the index ischaemia) with reperfusion (Figure 1).
A2B receptors
It had been noted that adenosine receptors were required for IPC's protection in the mediator phase. One hypothesis suggested that preconditioning is protective by increasing tissue adenosine levels through activation of ecto-5′-nucleotidase (Kitakaze et al., 1993). However, measurements of myocardial adenosine levels revealed that tissue adenosine concentration actually falls in IPC hearts (Goto et al., 1996; Martin et al., 1997). Our studies have indicated that the initial step of the mediator phase is activation of adenosine A2B receptors (Philipp et al., 2006). This receptor has a very low affinity for adenosine such that even during ischaemia when tissue adenosine levels reach 1–4 μM, this level would still be well below the A2B adenosine receptor's KD of 5–15 μM. However, PKC activation appears to raise the affinity of the A2B receptor permitting the adenosine concentration in ischaemic myocardium to be sufficient for occupation of this receptor (Kuno et al., 2007). It had already been shown that PKC activity can sensitize A2B signalling, although no physiologic significance was attributed to the observation (Nordstedt et al., 1989; Nash et al., 1997; Trincavelli et al., 2004). Although the details of this sensitization are still unknown, it would appear A2B receptors can respond to the heart's endogenous adenosine only after this sensitization. Thus, we proposed that the affinity state of the A2B receptor is the determinant that distinguishes the preconditioned from the non-preconditioned phenotype. Our observation of involvement of the A2B receptor in IPC was supported by Eckle et al. (2007) who studied mice genetically modified to lack one of the four adenosine receptor subtypes. While A1, A2A and A3 adenosine receptor knockout mice could be preconditioned, A2B knockout mice could not. However, there is evidence suggesting a cooperative role of A2A and A2B adenosine receptors in some forms of cardioprotection (Xi et al., 2009; Methner et al., 2010).
The reperfusion injury survival kinases (RISK) pathway
A kinase cascade involving PI3K, Akt and ERK has been proposed to occur in the first minutes of reperfusion following the index ischaemia (Hausenloy and Yellon, 2004; Hausenloy et al., 2005). These kinases have collectively been termed RISK (Hausenloy and Yellon, 2004). Although RISK are clearly involved in cardioprotection in rat (Hausenloy et al., 2005) and rabbit (Yang et al., 2004b; 2005,) hearts, their involvement may not be universal. In pig hearts, RISK are less important (Skyschally et al., 2009a). A distinct alternate pathway utilizing membrane TNF-α receptors and cytoplasmic JAK and STAT has been proposed (see succeeding text), although the end-effector for this and the RISK pathways appears to be identical.
IPC's end-effector
IPC's end-effector appears to be the mitochondrial permeability transition pore (mPTP) and its inhibition is considered to be the final step in the protective signal transduction pathway (Griffiths and Halestrap, 1993; 1995,; Squadrito et al., 1999; Di Lisa et al., 2001; Hausenloy et al., 2002). Although the molecular structure of mPTP is controversial, when formed it is a high conductance pore in the inner mitochondrial membrane that dissipates the transmembrane proton/electrochemical gradient that drives ATP generation. The presence of the pore would logically lead to ATP depletion, enhanced ROS production, failure of membrane ion pumps, solute entry, organelle swelling and ultimate mitochondrial rupture. Destruction of large numbers of mitochondria will result in necrosis of the cardiomyocyte. Importantly, mPTP formation is inhibited by acidosis and promoted by calcium and ROS. The low pH during ischaemia inhibits transition pore formation. But restoration of pH coupled with rapid elevation in mitochondrial calcium and ROS cause the pores to form soon after reperfusion.
The cardioprotective signalling pathways keep mPTP closed. In the RISK pathway, there is involvement of an additional intervening kinase, glycogen synthase kinase-3β (GSK-3β) (Tong et al., 2002; Gross et al., 2004; Juhaszova et al., 2004). This kinase is likely the final cytoplasmic kinase in IPC's signal transduction pathway. Interestingly, preconditioning leads to Ser9 phosphorylation and inhibition, not activation, of this kinase. Thus, GSK-3β inhibition blocks mPTP formation. Accordingly, GSK-3β inhibitors given at reperfusion mimic preconditioning (Förster et al., 2006).
Alternative signalling pathways
As already noted, the importance of RISK has been clearly demonstrated in rat (Hausenloy et al., 2005) and rabbit (Yang et al., 2004b; 2005,) hearts, but their involvement may not be required in all species. In a well-established pig heart model, activation of RISK was not increased by ischaemic postconditioning (IPoC), protection mediated by several brief reocclusions after release of the prolonged index coronary occlusion, over that seen in control hearts without postconditioning (Skyschally et al., 2009a). Furthermore, wortmannin, a potent antagonist of PI3K, could not abort postconditioning's protection (Skyschally et al., 2009a). In response to this conundrum, additional investigations uncovered another signalling pathway not dependent on RISK.
Survivor activating factor enhancement (SAFE) pathway
The SAFE pathway has been established, at least in rodent (Lecour et al., 2005b; Lacerda et al., 2009; Lecour, 2009) and porcine (Bhatt et al., 2013) hearts. However, a caveat is important. Some individual signalling steps have been identified, but a roadmap or signal transduction pathway as developed for the RISK pathway (Figure 1) is not yet available. So evidence supporting involvement of the SAFE pathway is more fragmentary, even if compelling.
TNF-α signalling
The cytokine TNF-α is an important endogenous cardioprotectant released by IPC (Smith et al., 2002; Lecour, 2009) and IPoC (Lacerda et al., 2009), possibly as part of the myocardial inflammatory response during reperfusion. TNF-α knockout mice cannot be protected by either IPC (Smith et al., 2002; Lecour, 2009) or IPoC (Lacerda et al., 2009; Lecour, 2009), whereas low-dose exogenous TNF-α in lieu of ischaemia can both precondition (Smith et al., 2002; Lecour et al., 2005b; Lecour, 2009) and postcondition (Lacerda et al., 2009; Lecour, 2009) hearts. As seen with the Gi-coupled receptor triggering described earlier for IPC, TNF-α preconditioning of rat hearts can be abolished by the antioxidant N-2-mercaptopropionyl glycine (Lecour et al., 2005a), a ROS scavenger, 5-hydroxydecanoate (Lecour et al., 2002), an antagonist of mtKATP, and chelerythrine (Lecour, 2009), a PKC antagonist, implying free radicals, mtKATP channels, and PKC play important roles. However, further information about the downstream effect of ROS, opening of mtKATP, or PKC or their targets is not available. Interestingly, TNF-α's effect on ischaemic myocardium is concentration-dependent. High doses of TNF-α are not protective and may actually increase infarct size (Lecour et al., 2002; Lecour, 2009).
There are two TNF receptor isoforms, TNFR1 and TNFR2. Exogenous TNF-α confers cardioprotection in TNFR1 (also known as TNFRSF1A) but not TNFR2 (also known as TNFRSF1B) knockout mice, thereby implying it is TNFR2 which is responsible for the ligand's cardioprotective effect (Lacerda et al., 2009). TNF-α administered as either a pre- or postconditioning-mimetic does not lead to phosphorylation of either Akt or ERK, and neither PD98059 nor wortmannin, antagonists of the ERK and PI3K pathways, respectively, can abort the protection of exogenous TNF-α. The SAFE pathway's downstream signalling is, therefore, not dependent on these traditional RISK (Lecour et al., 2005b; Lacerda et al., 2009). In contrast, TNF-α, IPC and IPoC all phosphorylate STAT3 and the protective effect of pharmacological pre- and postconditioning with TNF-α is abolished by AG490, an inhibitor of STAT3 (Lecour et al., 2005b; Lacerda et al., 2009). JAKs are a family of tyrosine kinases associated with the cytoplasmic domains of cytokine and growth factor receptors, for example, IL-6, growth hormone and TNFR2. After the TNF-α ligand binds to its receptor, two adjacent JAKs are transphosphorylated and subsequently activate STAT proteins by phosphorylation. Tyrosine-phosphorylated STAT proteins form homo- and heterodimers that translocate to the nucleus where they influence gene transcription, especially of stress-responsive genes (Levy and Lee, 2002; Myers, 2009). Serine-phosphorylated STAT translocates to mitochondria to regulate electron transport (Myers, 2009; Wegrzyn et al., 2009). Although STAT3 is by definition a transcription factor, its effects in ischaemia/reperfusion are much too rapid to assume that it is working by modulating gene transcription. Therefore, it must have additional direct effects. It appears to protect by phosphorylating and, therefore, inactivating GSK-3β (Lacerda et al., 2009; Pedretti and Raddatz, 2011), also a downstream target in the RISK pathway. Thus, the RISK and SAFE pathways appear to converge on the same targets. In fact there is some evidence of cross-talk between these two pathways, so they may not be totally independent (Lecour, 2009; Somers et al., 2012). Additionally, IPoC in pigs increased tyrosine phosphorylation of mitochondrial STAT3 which improves complex I respiration and calcium retention capacity (Heusch et al., 2011). Inhibition of JAK/STAT blocks both increased phosphorylation of mitochondrial STAT3 and the cardioprotective effect of IPoC. Mitochondrial STAT3 co-immunoprecipitates with cyclophilin D, the target of the mPTP inhibitor cyclosporin A and, therefore, could inhibit pore formation (Boengler et al., 2010).
Sphingosine is a trigger for RISK and SAFE
Sphingosine is a membrane sphingolipid which is catalyzed to sphingosine 1-phosphate (S1P) by two sphingosine kinase (SPHK) isoforms, SPHK1 and SPHK2. It is the former that is associated with cell survival. S1P is released in both IPC and IPoC (Karliner, 2013). Many S1P actions are mediated through S1P GPCR subtypes. The S1P1 receptor is most prominently expressed in cardiomyocytes. The S1P1 receptor couples to Gαi. S1P2 and S1P3 receptors are also present on cardiomyocytes and couple to both Gαq and Gαi. Binding of the ligand S1P to the S1P1 receptor leads to downstream activation of ERK1/2 and S1P3 receptor binding results in activation of PI3K and Akt. Thus, S1P cardioprotection in part depends on RISK (Knapp, 2011; Somers et al., 2012). However, S1P cardioprotection is also dependent on the SAFE pathway through the S1P2 receptor which activates ERK1/2 and subsequently STAT3 (Knapp, 2011; Somers et al., 2012). As already noted, multiple pathways provide potential for robust protection.
Sphingosine intermediates are also involved in cardioprotection mediated by TNF-α. The latter's protective effect is attenuated in the presence of inhibitors of the sphingolipid pathway (Lecour et al., 2002). TNF receptor-associated factor 2 (TRAF2) is a downstream target of TNFR2. TRAF2 can activate intracellular formation of S1P by up-regulating SPHK1 (Frias et al., 2012). Also, S1P activates STAT3.
This redundancy of pathways probably enhances the potential survival of the cell. Blockade of any one pathway does not lead to inevitable death of the cell. It is still possible for an alternate pathway to provide some protection. Alternatively, we may simply be looking at isolated sections of a larger complex integrated system that we still do not fully appreciate. This may be analogous to the fable of the blind men describing an elephant based only on the part of the animal they were touching.
Genesis of reperfusion therapy
Hence, IPC is a potent cardioprotective intervention that is the result of a complex, two-phase signalling pathway leading to inhibition of mPTP formation. However, the obvious drawback is that IPC by definition must be instituted prior to the onset of ischaemia. In patients presenting to the hospital with an AMI, ischaemia is already ongoing and preconditioning is not possible. However, Hausenloy et al. (2005) proposed that if IPC, an intervention introduced before the onset of ischaemia, protects by inducing activation of the RISK pathway at reperfusion, then it should still be possible to activate this pathway during ischaemia and still effect salvage of myocardium. This revolutionary paradigm shift provided hope that IPC could be translated into a meaningful clinical intervention by focusing on early reperfusion. Indeed, multiple reagents were found to protect the myocardium when given in the first minutes of reperfusion, for example, insulin (Baines et al., 1999), the adenosine A1/A2 agonist AMP579 (Xu et al., 2000), the A2B adenosine receptor-selective agonist BAY 60-6583 (Albrecht et al., 2006), TGFβ1 (Baxter et al., 2001), urocortin (Schulman et al., 2002), cardiotrophin-1 (Liao et al., 2002), adenosine agonist 5′-(N-ethylcarboxamido) adenosine (Yang et al., 2004a), bradykinin (Yang et al., 2004a), natriuretic peptides (Baxter, 2004; Yang et al., 2006a), erythropoietin (Cai and Semenza, 2004; Parsa et al., 2004) and cyclosporin A (Hausenloy et al., 2009). All depend on activation of PI3K and/or ERK except for cyclosporin A which is a direct inhibitor of mPTP formation.
Clinical trials
Not surprisingly, several clinical trials of proposed IPC-mimetics have been completed, although none has been greatly successful. These trials require study of many patients and are expensive. Repeated failures have left the pharmaceutical companies quite leery. Two large clinical trials, Amistad I (Mahaffey et al., 1999) and II (Ross et al., 2005), were organized to evaluate the effectiveness of adenosine. In the first trial, all patients with AMI were evaluated, whereas in the second trial, patients with only higher risk anterior infarcts were included. In Amistad I, there was no difference between the control and adenosine-treated subjects, although a post hoc analysis suggested that data in the subgroup with anterior infarcts looked promising (Birnbaum et al., 2002). In Amistad II, results were again disappointing. Smaller infarcts were noted in only a high-dose subgroup, but clinical outcomes were not improved. Although adenosine plays an important role in preconditioning as both a trigger and a mediator, the Amistad trials used low-dose i.v. infusion of adenosine which in preclinical studies had clearly not been universally successful at protecting ischaemic myocardium (Olafsson et al., 1987; Goto et al., 1991; Norton et al., 1991; 1992,; Pitarys et al., 1991; Velasco et al., 1991; Vander Heide and Reimer, 1996; Budde et al., 2000). It was probably not advisable to undertake such large and expensive trials until the cause of the discrepant data had been identified and resolved. Adenosine's hypotensive side effect limits the concentration that can be administered parentally and we were unable to precondition rabbit hearts with the highest dose of i.v. adenosine they would tolerate (Liu et al., 1991). We could, however, condition them with receptor-selective adenosine analogs such as AMP579 (Xu et al., 2003).
Atrial natriuretic peptide activates PKG in cardiomyocytes and mimics IPC when injected just prior to reperfusion in animals (Yang et al., 2006a). It produced a statistically significant, but very modest, reduction in infarct size and a similarly modest increase in ejection fraction (Kitakaze et al., 2007). The disappointingly modest effect might be explained by failure to stratify patients into low- and high-risk groups. The size of a patient's ischaemic zone is dependent on the location in the coronary artery where the thrombus forms. Studies from Ovize's laboratory (Staat et al., 2005) indicate that in patients with AMI who are reperfused with primary angioplasty, those with small ischaemic zones have very small infarcts and virtually complete recovery regardless of treatment. Including these patients in the analysis greatly dilutes the potential significance of any intervention. Therapeutic benefit can best be appreciated in the subgroup of high-risk patients presenting with large ischaemic zones. Other possible reasons for the modest result are discussed in the succeeding text.
During ischaemia, pH falls as H+ accumulates. As a result, the Na+/H+ exchanger (NHE) is activated and Na+ exchanges for H+ in a 1:1 stoichiometric manner. In turn, the Na+/Ca2+ exchanger will transport Na+ out of the cell in favour of Ca2+. These ionic movements are magnified during reperfusion when restored blood flow quickly normalizes the pH of the extracellular fluid. The resulting high intracellular Ca2+ concentration should encourage mPTP formation. It was decided to evaluate NHE blockers cariporide (Théroux et al., 2000; Mentzer et al., 2008) and eniporide (Zeymer et al., 2001) in large clinical trials despite preclinical investigations which demonstrated efficacy only when the drug was administered before ischaemia (Miura et al., 1997). In the first trial of cariporide, GUARDIAN, patients studied had unstable angina pectoris, non-ST-segment elevation myocardial infarction, angioplasty or coronary revascularization surgery (Théroux et al., 2000). Only those that had pretreatment, the surgical group, showed any benefit. A second trial, EXPEDITION, concentrated on coronary bypass patients, but the new study design inexplicably included prolonged infusions of cariporide which were associated with more strokes (Mentzer et al., 2008). The increased stroke risk doomed further consideration of cariporide. Although eniporide, another NHE blocker, was evaluated in patients with acute ST-segment-elevation myocardial infarction (STEMI), no difference was observed (Zeymer et al., 2001).
Investigations of other interventions which may have shown some promise in preclinical evaluations have also not been very successful. Thus, pexelizumab (APEX AMI Investigators et al., 2007), an antibody to a complement component, erythropoietin (Cleland et al., 2010), a stimulator of haematopoiesis in response to hypoxia, and delcasertib (Lincoff et al., 2014), a selective inhibitor of PKCδ, all failed to meet the primary objectives of the trials, reduction of infarct size and improvement of the clinical status of the subjects. There is a lesson: clinical trials should probably not be undertaken until multiple preclinical laboratories have confirmed salutary effects of the intervention and until practical information about dosing and timing of administration has been established (Downey and Cohen, 2009).
A small proof-of-concept study of cyclosporin A in patients with AMI produced very encouraging results (Piot et al., 2008). Patients were stratified by the size of their ischaemic zone and each received a bolus of cyclosporin A before recanalization. Those with the highest risk benefited the most from exposure to the drug. There are plans to repeat this investigation in a much larger cohort in Europe. However, as explained in the succeeding text, a second study may be problematic because concomitant use of antiplatelet drugs dictated by current standard of care considerations may mask cyclosporin's protection.
There have been other proof-of-concept studies examining commonly used agents. Van de Werf et al. (1993) studied the effect of atenolol administered prior to thrombolysis in patients with AMI. This β-blocker had no impact on infarct size, a result echoing that of a study in dogs by Reimer and Jennings (1984). Nonetheless, a very recent examination of i.v. metoprolol shortly before percutaneous coronary intervention (PCI) in patients with STEMI was encouraging (Ibanez et al., 2013). This β-blocker modestly decreased infarcted myocardium as a percentage of risk zone from approximately 78% in untreated hearts to 68%. Although there was protection, small group sizes limit the significance of the conclusion and require conduct of a large, expensive clinical trial for confirmation. There is no evidence that β-adrenoceptor blockade triggers IPC signalling as detailed earlier. The same is true for exenatide, a glucagon-like peptide-1 (Lønborg et al., 2012). IPC's signalling is not the only way to protect the heart against infarction, and hypothermia and early reperfusion are obvious examples.
IPoC
It was clearly understood that preconditioning cycles must be completed before initiation of ischaemia. Yet Hausenloy et al. (Hausenloy and Yellon, 2004; Hausenloy et al., 2005) found that IPC actually exerted its protection at reperfusion. This finding led Vinten-Johansen et al. to test whether serial coronary occlusions after the index coronary occlusion/reperfusion might also protect the ischaemic heart. After many tries, they found that several (three) short (30 s) cycles of reperfusion/occlusion immediately after the initial reperfusion were almost as protective as IPC in an open-chest dog model (Zhao et al., 2003). They called this IPoC. This seemingly improbable observation has been reproduced in many laboratories (Skyschally et al., 2009b) and the ensuing protection was shown to be dependent on the same signals as IPC (Yang et al., 2004b; 2005,).
Again, the final effector for IPoC appeared to be prevention of mPTP formation (Argaud et al., 2005; Gateau-Roesch et al., 2006). Because of the widespread success in the experimental laboratory, a leap was made to the clinical arena. Patients with acute STEMI have thrombotic occlusion of a coronary artery. Standard treatment is recanalization, usually by mechanical aspiration or pulverization of the thrombus by percutaneous transluminal coronary angioplasty (PTCA). Opening of the occluded coronary artery by PTCA is equivalent to removing the ligature around the snared coronary artery in the experimental animal. For patients treated with primary angioplasty, IPoC could be accomplished by repeated balloon inflations to interrupt reflow for the postconditioning cycles. The initial report of IPoC in the cardiac catheterization laboratory was very encouraging (Staat et al., 2005). Using a risk stratification design, they showed a highly significant reduction of infarct size in IPoC patients with large ischaemic zones. But why does staccato reperfusion lead to myocardial salvage?
In the non-conditioned naïve heart following coronary occlusion, mtKATP open during ischaemia, but there is no oxygen so the pathway is blocked at the redox signalling step. During ischaemia, mPTPs are inhibited by acidosis in the tissue probably by blocking calcium binding to cyclophilin and displacing the latter which is required for mPTP formation. Upon reperfusion, acids quickly wash away restoring pH to 7.4 and mPTP forms before PKC can be activated to trigger the remainder of the signalling pathway thus resulting in necrosis of the tissue. On the other hand, IPoC maintains some acidosis in the reperfused tissue because of the repeated occlusion periods while still allowing oxygenation during the reperfusion periods (Cohen et al., 2007b; 2008,). Reintroduction of oxygen while the tissue pH is still acidic allows the tissue to activate PKC through redox signalling while mPTP formation is still inhibited (Cohen et al., 2007b; 2008,). Once the PKC pathway is activated, the cell is able to inhibit mPTP formation through the conditioning pathway that IPC uses even after pH is normalized, and hence necrosis is reduced.
Thus, there is a race between ROS-mediated activation of PKC leading to subsequent triggering of the remainder of the signal cascade and washout of mPTP-inhibiting H+. Figure 2 summarizes the pH hypothesis of IPoC's protection. mPTPs in the naïve, non-conditioned heart (Figure 2, upper panel) are inhibited by the low pH during the ischaemic period. But as soon as reperfusion is permitted, H+ is washed out and mPTPs open leading to tissue necrosis. In IPC (Figure 2, middle panel), signalling up to the opening of mtKATP occurs during the first brief ischaemic period. During the brief reperfusion oxygen is resupplied which leads to ROS generation and activation of PKC which then can sensitize adenosine receptors. Thus, at the beginning of reperfusion following the prolonged index coronary occlusion, RISK are activated and mPTPs are inhibited. The bottom panel of Figure 2 depicts events in IPoC. The initial signalling up to opening of mtKATP occurs during the prolonged period of ischaemia, but signalling cannot proceed until reperfusion when oxygen is reintroduced into the ischaemic tissue leading to generation of ROS. Because of the limited reflow, pH only partially recovers. The low pH inhibits mPTP formation long enough until redox signalling can lead to adenosine receptor population with sensitization and subsequent RISK signalling that results in inhibition of mPTP formation even after the muscle is fully reperfused.
Figure 2.
Signalling during ischaemic pre- and postconditioning and effect of pH and transient reoxygenation on that signalling and mPTP formation. See Figure 1 for abbreviations. Modified from Cohen et al. (2007b; 2008).
The pH hypothesis is consistent with other observations made in experimental animals. Reperfusion interventions must be applied in the first minutes of reperfusion. Thus, delayed IPoC (Yang et al., 2004b; Philipp et al., 2005) or late infusion of the cardioprotective AMP579 (Xu et al., 2003) leads to loss of the cardioprotective effects. Presumably, such delay would permit pH normalization before initiation of the intervention, thus allowing mPTP formation. Once this occurs, no intervention dependent for its success on keeping mPTP closed would be expected to salvage myocardium in the risk zone. Also, simply reperfusing the heart for several minutes with low pH buffer mimics IPoC (Cohen et al., 2007b).
Conditioning with volatile anaesthetic agents
Shortly after IPC was recognized as a universally acknowledged cardioprotective strategy, volatile anaesthetic gases, principally sevoflurane, isoflurane and desflurane were noted to also have cardioprotective abilities when applied in lieu of the brief cycles of ischaemia/reperfusion either before the prolonged index coronary occlusion (preconditioning) (Cope et al., 1997) or following it (postconditioning) (Chiari et al., 2005). The signalling steps are not as clearly defined as in IPC and IPoC, but it is fair to say there are many parallels. Basically, the volatile gases signal through adenosine and opioid receptors, modulate G proteins, stimulate PKC and other intracellular kinases, open mtKATP channels leading to ROS generation and activate RISK to keep mPTP from forming (Tanaka et al., 2004; Chiari et al., 2005; Feng et al., 2005; Pravdic et al., 2009). The gases may also have more direct effects on mtKATP. The potential clinical impact is obvious, although utility is limited to the surgical suite, either cardiac or non-cardiac (Swyers et al., 2014).
Platelets and cardioprotection
As noted earlier, one of the earliest cardioprotective interventions applied clinically was IPoC. The initial clinical report was very encouraging (Staat et al., 2005) and the intervention was adopted by many cardiac catheterization laboratories, partly because of the ease of application and partly because of the anticipated small likelihood of complications. Despite this early enthusiasm, other clinicians who attempted to replicate the positive results could not (Sörensson et al., 2010; Freixa et al., 2012; Tarantini et al., 2012; Hahn et al., 2013; Limalanathan et al., 2014). Would this intervention have to join all of the other interventions which initially showed great promise but which did not live up to expectations and which failed to produce a significant and consistent beneficial clinical effect? Could there be a logical explanation for the inability of these later studies of IPoC to reproduce the early encouraging data?
Platelets are important cellular elements for initiation and propagation of a thrombus. It is now universally accepted that STEMI is caused by thrombosis and coronary occlusion following rupture of a plaque. Exposure of circulating platelets to collagen leads to their activation and accumulation. Platelet-activating factor (PAF) is a phospholipid which is released by neutrophils and monocytes during oxidative stress or ischaemia/reperfusion (Penna et al., 2011). PAF is a chemoattractant for platelets and neutrophils and predisposes to capillary plugging and release of proteolytic enzymes and inflammatory mediators. Additionally, it causes coronary vasoconstriction. Cardiomyocytes also produce PAF and the latter's synthesis is triggered by ROS generation and oxidative stress at the beginning of reperfusion. PAF binds to receptors located on various cell types including smooth muscle cells, cardiomyocytes and endothelial cells. PAF receptor antagonists limit infarction in models of ischaemia/reperfusion (Montrucchio et al., 1990; Ma et al., 1992). Curiously, low doses of PAF are paradoxically cardioprotective (Penna et al., 2005; 2011,). The PAF receptor is a Gi protein-coupled receptor that triggers signalling similar to that seen in IPC (Figure 1).
Because the major risk of intracoronary dilatation and stenting is stent thrombosis and occlusion, antiplatelet drugs were tested as anticoagulants in patients undergoing primary angioplasty for AMI. Many clinical studies have demonstrated that antiplatelet agents do indeed improve prognosis of patients after AMI and minimize complications of stenting (Antiplatelet Trialists' Collaboration, 1994; Yusuf et al., 2001; Sabatine et al., 2005a,b; Wiviott et al., 2007; Wallentin et al., 2009; Bhatt et al., 2013). This protection is particularly evident when the antiplatelet drug is given as a loading dose prior to the recanalization procedure. Thus, the COX antagonist aspirin, the thienopyridines clopidogrel and prasugrel and the triazolopyrimidines ticagrelor and cangrelor are very effective agents and all except cangrelor have won regulatory approval and have become standard of care in the treatment of patients with AMI or stenting. Aspirin blocks production of thromboxane by the platelet, while the thienopyridines and triazolopyrimidines block the platelet P2Y12 ADP receptor and all effectively attenuate platelet aggregation. It has been assumed that it is the anti-aggregatory effect of these agents on platelets to minimize intravascular thrombosis that is responsible for their well-documented clinical benefits. However, that mechanism of action has not been unequivocally determined. In this regard, it is instructive to review some of the preclinical data.
Preclinical studies of antiplatelet drugs
Barrabés et al. (2010) noted that activated platelets from patients with AMI infused into isolated rat hearts before onset of ischaemia/reperfusion increased infarct size, whereas platelets from healthy volunteers had no effect. Furthermore, perfusion of previously ischaemic hearts with platelets activated in a second animal following ischaemia/reperfusion led to deterioration of left ventricular function and larger myocardial infarcts (Knight et al., 2001; Mirabet et al., 2002). Aggregation of platelets from mice with deficiencies of either platelet receptor glycoprotein (GP) VI (Takaya et al., 2005; Li et al., 2007) or signalling protein Gq (Weig et al., 2008) is diminished, and infarcts are smaller following ischaemia/reperfusion. These observations suggest activated platelets have deleterious effects on ischaemic myocardium and provide some support for the hypothesis that there is a relationship between platelet aggregation and consequences of myocardial ischaemia/reperfusion, for example, infarct size.
Preclinical investigations of blockade of platelet aggregation in ischaemia/reperfusion have mostly studied effects of GPIIb/IIIa antagonists. An experimental GPIIb/IIIa inhibitor which abolished in vitro platelet aggregation decreased infarction in dogs undergoing ischaemia/reperfusion when it was administered before reperfusion (Kingma et al., 2000). However, Kingma et al. (2000) also noted that the platelet antagonist had no effect on myocardial blood flow during reperfusion, and therefore, postulated that this infarct-sparing action was not related to blood flow but rather was the result of a direct protective effect on heart muscle. This was the first suggestion of a direct cardioprotective effect by an inhibitor of platelet aggregation.
Kunichika et al. (2004) made similar observations in dogs treated with tirofiban, a GPIIb/IIIa antagonist. However, this agent which decreased infarct size also increased myocardial blood flow within the risk area. Consequently, the investigators attributed the agent's cardioprotection to improvement in microvascular flow. Tirofiban also decreased the area of no-reflow in pigs during reperfusion and decreased infarct size (Yang et al., 2006b). In dogs with coronary thrombosis treated with angioplasty, tirofiban improved myocardial blood flow following reperfusion, decreased the size of the no-reflow zone and made infarcts smaller (Sakuma et al., 2005). It was assumed that inhibition of platelet aggregation protected by preventing microthromboembolism.
Additional studies failed to corroborate this hypothesis. The deleterious effect of the addition of activated pig platelets to perfused, isolated rat hearts subjected to ischaemia/reperfusion was not blocked by tirofiban (Mirabet et al., 2002). A second GPIIb/IIIa inhibitor had no effect on infarct size in a porcine model of ischaemia/reperfusion (Barrabés et al., 2002). In both studies, platelet aggregation was blocked by the GPIIb/IIIa antagonists. It is not known why these latter studies differed from the former.
P2Y12 receptor inhibitors may be postconditioning agents
Because of this confusion, we evaluated a variety of platelet inhibitors in rabbits undergoing 30 min coronary occlusion/3 h reperfusion (Yang et al., 2013c). For most of the studies, we examined the effects of cangrelor, an i.v. agent which could be administered minutes before reperfusion and which would have immediate effects. Oral agents suffer from the limitations imposed by intestinal absorption and the requirement for conversion of the administered pro-drug clopidogrel or prasugrel to active metabolites. This delay causes an uncertainty of timing of onset of biological effect. A cangrelor bolus of 60 μg·kg−1 followed by an infusion of 6 μg·kg−1·min−1 attenuated platelet aggregation by more than 94%. Cangrelor resulted in an impressive decrease in infarct size from 38% of the risk zone in control rabbits to 19%, similar to the degree of protection seen after IPoC. Delay in cangrelor administration until 10 min after release of the coronary occlusion led to abrogation of protection, similar to that seen with delayed IPoC (Yang et al., 2004b; Philipp et al., 2005).
As described earlier, IPoC's protection is known to depend on a complex signal transduction pathway. Accordingly, we tested seven inhibitors of IPoC's signalling and protection: wortmannin and LY294002 (PI3K/Akt antagonists), PD98059 (antagonist of MAPK kinase 1/2 and therefore, ERK 1/2), 5-hydroxydecanoic acid (putative blocker of mtKATP), 8-(p-sulfophenyl) theophylline (non-selective antagonist of adenosine receptors), MRS 1754 (selective antagonist of adenosine A2B receptors) and N-2-mercaptopropionylglycine (scavenger of ROS and blocker of redox signalling). All abolished cangrelor's protection. However, importantly, none restored platelet reactivity. Therefore, cangrelor's anti-aggregatory effect was still intact, but its cardioprotective action was totally blocked.
Hence, IPoC and cangrelor have identical kinase and receptor ‘fingerprints’ and this conclusion strongly supports the contention that the signalling and mechanism of protection of the two interventions are the same. We also determined whether the combination of cangrelor and IPoC would have an additive protective effect. It did not, further supporting the assumption that both induce protection by the same mechanism (Yang et al., 2013c). Of course, to make this conclusion, it is critical that the effect of each individual intervention is maximized. Obviously, there would be an additive effect of two agents using the same pathway if one or both was used at a submaximal concentration. Hence, we propose that cangrelor is a bona fide conditioning agent and signal transduction rather than any effect on thrombosis causes the protection. Yet, we found that the magnitude of protection was correlated with the degree of suppression of aggregation indicating that P2Y12 blockade was common to both processes.
Cangrelor in both primate (Yang et al., 2013b) and rodent (Yang et al., 2013a) hearts was also very protective when administered just before reperfusion. Cangrelor's protection was equivalent to that of IPoC in macaque hearts (Yang et al., 2013b) and IPC in rat hearts (Yang et al., 2013a). In monkeys, an antibody to platelet GPVI also decreased infarct size, indicating another intervention which decreased platelet aggregation and infarction (Yang et al., 2013b).
Clopidogrel, a widely used P2Y12 antagonist in patients with AMI and PTCA, was fed to rabbits for 2 days and it blocked platelet aggregation by 78% (Yang et al., 2013c). Clopidogrel-treated animals also had significantly smaller infarcts than untreated rabbits. Wortmannin and MRS 1754 each abolished the protective effect, but the drug's anti-aggregatory action remained intact.
Finally, ticagrelor, a third platelet P2Y12 receptor antagonist, administered by gavage to rats 2 h before coronary occlusion, decreased infarction from 45% of the risk zone in control animals to 26% (Yang et al., 2013a). This protective effect was predictably blocked by wortmannin and PKC antagonist chelerythrine. Neither blocker interfered with ticagrelor's inhibitory effect on platelet reactivity.
Therefore, several pharmacologic and biologic interventions that blocked platelet aggregation also spared ischaemic myocardium from infarcting. It is critical to realize that although an antiplatelet effect linked all of the agents, inhibition of platelet activity was not the determining factor for cardioprotection. These appear to be true conditioning agents. Cangrelor in isolated rabbit hearts perfused with platelet-free Krebs buffer was not protective, suggesting that some blood element, presumably platelets, is somehow involved. Indeed, cangrelor's protective effect was lost in rats made thrombocytopenic with anti-thrombocyte serum (unpublished observation).
Are today's patients already postconditioned by loading doses of antiplatelet drugs?
In addition to the possible confounding effects of co-morbidities in man, generally absent in animal models, these experimental data on antiplatelet interventions, specifically P2Y12 receptor antagonists, may help to explain the observations of protection or lack of protection following clinical IPoC. Because P2Y12 antagonists are conditioning agents, they themselves would already be protecting the heart. Therefore, addition of a second intervention as IPoC which protected by the same mechanism would be expected to have little additional effect, similar to our observations in rabbits in which cangrelor and IPoC together were no more protective than either alone (Yang et al., 2013c). In the later clinical studies of IPoC, virtually, all patients had received clopidogrel before the intervention (Sörensson et al., 2010; Freixa et al., 2012; Tarantini et al., 2012; Hahn et al., 2013; Limalanathan et al., 2014), thus marking the patients as protected and perhaps dooming IPoC to be a superfluous and unneeded intervention. However, in the initial study by Ovize's group in which patients with AMI were recruited prior to 2005, only about half had been premedicated with clopidogrel (Staat et al., 2005). At our urging, these investigators went back to their database and segregated patients according to pre-IPoC administration of clopidogrel (Roubille et al., 2012). Control patients treated with clopidogrel had smaller infarcts than those who were untreated, supporting our conclusion that clopidogrel is a cardioprotective agent. But these authors also noted that the combination of clopidogrel and IPoC salvaged more tissue than IPoC, contrary to our observations in rabbits. However, it is likely that neither intervention, clopidogrel nor IPoC, was optimized to fully condition the hearts when applied individually in contrast to our ability to maximize the effect of each intervention in animal models. Most of the patients in Roubille's retrospective analysis (Roubille et al., 2012) had received only 300 mg of clopidogrel which is clearly suboptimal (Patti et al., 2011). Also, the optimal postconditioning protocol for human hearts is unknown and only one protocol was tested. If neither intervention was optimal, an additive effect would be expected.
Piot et al. (2008) reported a significant reduction in infarct size by cyclosporin A in patients undergoing PCI. However, those patients were recruited by the same investigators as noted earlier (Staat et al., 2005) and around the same time. The authors have not disclosed the clopidogrel usage in that cohort.
These observations on the effect of clopidogrel and other P2Y12 receptor inhibitors are more than academic. Use of these agents in patients with AMI is now standard of care and their dosing has been optimized so that most of today's patients may already be postconditioned at the time of PTCA. Hence, any additional intervention using the same protective mechanism as the P2Y12 receptor inhibitor would add little to the protection. This problem is compounded by many recent clinical trials that have led to incremental improvements in antiplatelet treatment. New blockers have greatly shortened absorption times and their dosing is being optimized so that eventually all patients presenting with AMI are likely to be optimally postconditioned. To obtain additional cardioprotection, an adjunct intervention must have a different mechanism of action.
Additional cardioprotection in the presence of an antiplatelet drug
We have found that some protective interventions can be combined to produce more potent protection. Unlike IPoC and cangrelor, mild hypothermia (Miki et al., 1998b; Tissier et al., 2007b) and cariporide (Miura et al., 1997) are most protective when applied during ischaemia rather than reperfusion. Thus, protection from IPC which protects against a reperfusion injury could be added to that of cooling which protects against an ischaemic injury (Miki et al., 1998b). Similarly, the protective effect of the pharmacological postconditioning agent AMP579 which protects against a reperfusion injury (Xu et al., 2003) could be added to that of cariporide that protects against an ischaemic injury (Xu et al., 2002).
We tested whether any interventions could be additive with cangrelor treatment (Yang et al., 2013a). In rats, peritoneal lavage with ice-cold saline 10 min before coronary occlusion lowered blood temperature to 32–33°C and decreased infarction to 25% of the risk zone, equivalent to that seen with cangrelor treatment just before reperfusion. The two interventions together halved infarction to 14% of myocardium at risk. Cariporide's protective effect (27% infarction) is comparable with cangrelor's. The combination of cangrelor and cariporide nearly halved infarction to 16% of the risk zone. When all three interventions were combined, infarction again halved to only 6% of the risk zone. Obviously, treatment during the ischaemic period is logistically problematic. However, most patients spend an extended period of time with healthcare professionals before recanalization has been accomplished during which time these interventions could be effectively implemented.
We suggest that future animal studies of cardioprotective interventions be conducted on a background of a P2Y12 inhibitor to provide a more clinically relevant model. Unless an agent can provide additive protection in that model, it would be of little clinical value. Although we have tested IPC and IPoC combined with the P2Y12 inhibitor cangrelor and have found no additional protection, it is unknown whether interventions such as remote conditioning (Lim and Hausenloy, 2012) or promoters of autophagy (Sala-Mercado et al., 2010) whose mechanisms are less well understood might have additive effects.
Concluding remarks
Our 43-year journey has brought us to this point where we have a good understanding of the type of intervention that must be introduced to spare ischaemic myocardium. Now that conditioning's protection is being applied to patients routinely with the antiplatelet drugs, we should look elsewhere for the next generation of cardioprotective drugs. We propose that an intervention not based on the signalling of IPC or IPoC would be most likely to add protection to that already resulting from treatment with standard antiplatelet agents. Thus, IPoC or even cyclosporin which prevents formation of mPTP would be expected to have only small, if any, effect in this setting. Infarct size reduction clearly reduces mortality and morbidity in AMI as clinical trials with reperfusion therapy and P2Y12 inhibitors have proven. Yet further protection against infarction is still indicated as AMI continues to be a deadly and debilitating disease. The preclinical studies completed by many investigators have established the foundation for cardioprotective strategies and we must continue to search for new strategies to preserve myocardium from a transient ischaemic insult. In the past, the improvements in outcome in AMI have been incremental rather than revolutionary. By using relevant animal models, we can hopefully identify candidates for future clinical testing and someday make myocardial infarction a mere historical medical footnote.
Conflicts of interest
None.
Glossary
- AMI
acute myocardial infarction
- Cx43
connexin 43
- GP
glycoprotein
- GSK-3β
glycogen synthase kinase-3β
- HB-EGF
heparin-binding EGF-like growth factor
- IPC
ischaemic preconditioning
- IPoC
ischaemic postconditioning
- KATP
ATP-sensitive K+ channel
- mPTP
mitochondrial permeability transition pore
- mtKATP
mitochondrial KATP
- NHE
Na+/H+ exchanger
- PAF
platelet-activating factor
- PCI
percutaneous coronary intervention
- PDK
3′-phosphoinositide-dependent kinase
- PTCA
percutaneous transluminal coronary angioplasty
- RISK
reperfusion injury survival kinases
- ROS
reactive oxygen species
- S1P
sphingosine-1-phosphate
- SAFE
survivor activating factor enhancement
- SPHK1
sphingosine kinase
- Src
sarcoma
- STEMI
ST-segment-elevation myocardial infarction
- TRAF-2
TNF receptor-associated factor 2
References
- Albrecht B, Krahn T, Philipp S, Rosentreter U, Cohen MV, Downey JM. Selective adenosine A2b receptor activation mimics postconditioning in a rabbit infarct model. Circulation. 2006;114(Suppl. II):II-14–II-15. [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: G Protein-Coupled Receptors. Br J Pharmacol. 2013a;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Ion channels. Br J Pharmacol. 2013b;170:1607–1651. doi: 10.1111/bph.12447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Catalytic Receptors. Br J Pharmacol. 2013c;170:1676–1705. doi: 10.1111/bph.12449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Transporters. Br J Pharmacol. 2013d;170:1706–1796. doi: 10.1111/bph.12450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Enzymes. Br J Pharmacol. 2013e;170:1797–1867. doi: 10.1111/bph.12451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andjelković M, Alessi DR, Meier R, Fernandez A, Lamb NJC, Frech M, et al. Role of translocation in the activation and function of protein kinase B. J Biol Chem. 1997;272:31515–31524. doi: 10.1074/jbc.272.50.31515. [DOI] [PubMed] [Google Scholar]
- Antiplatelet Trialists' Collaboration. Collaborative overview of randomised trials of antiplatelet therapy–I: Prevention of death, myocardial infarction, and stroke by prolonged antiplatelet therapy in various categories of patients. BMJ. 1994;308:81–106. [PMC free article] [PubMed] [Google Scholar]
- APEX AMI Investigators. Armstrong PW, Granger CB, Adams PX, Hamm C, Holmes D, Jr, et al. Pexelizumab for acute ST-elevation myocardial infarction in patients undergoing primary percutaneous coronary intervention: a randomized controlled trial. JAMA. 2007;297:43–51. doi: 10.1001/jama.297.1.43. [DOI] [PubMed] [Google Scholar]
- Argaud L, Gateau-Roesch O, Raisky O, Loufouat J, Robert D, Ovize M. Postconditioning inhibits mitochondrial permeability transition. Circulation. 2005;111:194–197. doi: 10.1161/01.CIR.0000151290.04952.3B. [DOI] [PubMed] [Google Scholar]
- Baines CP, Goto M, Downey JM. Oxygen radicals released during ischemic preconditioning contribute to cardioprotection in the rabbit myocardium. J Mol Cell Cardiol. 1997;29:207–216. doi: 10.1006/jmcc.1996.0265. [DOI] [PubMed] [Google Scholar]
- Baines CP, Wang L, Cohen MV, Downey JM. Myocardial protection by insulin is dependent on phosphatidylinositol 3-kinase but not protein kinase C or KATP channels in the isolated rabbit heart. Basic Res Cardiol. 1999;94:188–198. doi: 10.1007/s003950050142. [DOI] [PubMed] [Google Scholar]
- Barrabés JA, Garcia-Dorado D, Mirabet M, Lidón R-M, Soriano B, Ruiz-Meana M, et al. Lack of effect of glycoprotein IIb/IIIa blockade on myocardial platelet or polymorphonuclear leukocyte accumulation and on infarct size after transient coronary occlusion in pigs. J Am Coll Cardiol. 2002;39:157–165. doi: 10.1016/s0735-1097(01)01712-0. [DOI] [PubMed] [Google Scholar]
- Barrabés JA, Inserte J, Mirabet M, Quiroga A, Hernando V, Figueras J, et al. Antagonism of P2Y12 or GPIIb/IIIa receptors reduces platelet-mediated myocardial injury after ischaemia and reperfusion in isolated rat hearts. Thromb Haemost. 2010;104:128–135. doi: 10.1160/TH09-07-0440. [DOI] [PubMed] [Google Scholar]
- Baxter GF. Natriuretic peptides and myocardial ischaemia. Basic Res Cardiol. 2004;99:90–93. doi: 10.1007/s00395-004-0458-7. [DOI] [PubMed] [Google Scholar]
- Baxter GF, Mocanu MM, Brar BK, Latchman DS, Yellon DM. Cardioprotective effects of transforming growth factor-β1 during early reoxygenation or reperfusion are mediated by p42/p44 MAPK. J Cardiovasc Pharmacol. 2001;38:930–939. doi: 10.1097/00005344-200112000-00015. [DOI] [PubMed] [Google Scholar]
- Becker LB, Vanden Hoek TL, Shao Z-H, Li C-Q, Schumacker PT. Generation of superoxide in cardiomyocytes during ischemia before reperfusion. Am J Physiol Heart Circ Physiol. 1999;277:H2240–H2246. doi: 10.1152/ajpheart.1999.277.6.H2240. [DOI] [PubMed] [Google Scholar]
- Bhatt DL, Stone GW, Mahaffey KW, Gibson CM, Steg PG, Hamm CW, et al. Effect of platelet inhibition with cangrelor during PCI on ischemic events. N Engl J Med. 2013;368:1303–1313. doi: 10.1056/NEJMoa1300815. [DOI] [PubMed] [Google Scholar]
- Birnbaum Y, Mahaffey KW, Criger DA, Gates KB, Barbash GI, Barbagelata A, et al. Grade III ischemia on presentation with acute myocardial infarction predicts rapid progression of necrosis and less myocardial salvage with thrombolysis. Cardiology. 2002;97:166–174. doi: 10.1159/000063334. [DOI] [PubMed] [Google Scholar]
- Boengler K, Hilfiker-Kleiner D, Heusch G, Schulz R. Inhibition of permeability transition pore opening by mitochondrial STAT3 and its role in myocardial ischemia/reperfusion. Basic Res Cardiol. 2010;105:771–785. doi: 10.1007/s00395-010-0124-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Budde JM, Velez DA, Zhao Z-Q, Clark KL, Morris CD, Muraki S, et al. Comparative study of AMP579 and adenosine in inhibition of neutrophil-mediated vascular and myocardial injury during 24 h of reperfusion. Cardiovasc Res. 2000;47:294–305. doi: 10.1016/s0008-6363(00)00115-2. [DOI] [PubMed] [Google Scholar]
- Burley DS, Ferdinandy P, Baxter GF. Cyclic GMP and protein kinase-G in myocardial ischaemia-reperfusion: opportunities and obstacles for survival signaling. Br J Pharmacol. 2007;152:855–869. doi: 10.1038/sj.bjp.0707409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cai Z, Semenza GL. Phosphatidylinositol-3-kinase signaling is required for erythropoietin-mediated acute protection against myocardial ischemia/reperfusion injury. Circulation. 2004;109:2050–2053. doi: 10.1161/01.CIR.0000127954.98131.23. [DOI] [PubMed] [Google Scholar]
- Chiari PC, Bienengraeber MW, Pagel PS, Krolikowski JG, Kersten JR, Warltier DC. Isoflurane protects against myocardial infarction during early reperfusion by activation of phosphatidylinositol-3-kinase signal transduction: evidence for anesthetic-induced postconditioning in rabbits. Anesthesiology. 2005;102:102–109. doi: 10.1097/00000542-200501000-00018. [DOI] [PubMed] [Google Scholar]
- Cleland JGF, Coletta AP, Torabi A, Ahmed D, Clark AL. Clinical trials update from the European Society of Cardiology Meeting 2010: SHIFT, PEARL-HF, STAR-heart, and HEBE-III. Eur J Heart Fail. 2010;12:1261–1264. doi: 10.1093/eurjhf/hfq186. [DOI] [PubMed] [Google Scholar]
- Cohen MV, Yang X-M, Liu GS, Heusch G, Downey JM. Acetylcholine, bradykinin, opioids, and phenylephrine, but not adenosine, trigger preconditioning by generating free radicals and opening mitochondrial KATP channels. Circ Res. 2001;89:273–278. doi: 10.1161/hh1501.094266. [DOI] [PubMed] [Google Scholar]
- Cohen MV, Yang X-M, Downey JM. Nitric oxide is a preconditioning mimetic and cardioprotectant and is the basis of many available infarct-sparing strategies. Cardiovasc Res. 2006;70:231–239. doi: 10.1016/j.cardiores.2005.10.021. [DOI] [PubMed] [Google Scholar]
- Cohen MV, Philipp S, Krieg T, Cui L, Kuno A, Solodushko V, et al. Preconditioning-mimetics bradykinin and DADLE activate PI3-kinase through divergent pathways. J Mol Cell Cardiol. 2007a;42:842–851. doi: 10.1016/j.yjmcc.2007.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cohen MV, Yang X-M, Downey JM. The pH hypothesis of postconditioning: staccato reperfusion reintroduces oxygen and perpetuates myocardial acidosis. Circulation. 2007b;115:1895–1903. doi: 10.1161/CIRCULATIONAHA.106.675710. [DOI] [PubMed] [Google Scholar]
- Cohen MV, Yang X-M, Downey JM. Acidosis, oxygen, and interference with mitochondrial permeability transition pore formation in the early minutes of reperfusion are critical to postconditioning's success. Basic Res Cardiol. 2008;103:464–471. doi: 10.1007/s00395-008-0737-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cope DK, Impastato WK, Cohen MV, Downey JM. Volatile anesthetics protect the ischemic rabbit myocardium from infarction. Anesthesiology. 1997;86:699–709. doi: 10.1097/00000542-199703000-00023. [DOI] [PubMed] [Google Scholar]
- Costa ADT, Garlid KD. Intramitochondrial signaling: interactions among mitoKATP, PKCε, ROS, and MPT. Am J Physiol Heart Circ Physiol. 2008;295:H874–H882. doi: 10.1152/ajpheart.01189.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Costa ADT, Garlid KD, West IC, Lincoln TM, Downey JM, Cohen MV, et al. Protein kinase G transmits the cardioprotective signal from cytosol to mitochondria. Circ Res. 2005;97:329–336. doi: 10.1161/01.RES.0000178451.08719.5b. [DOI] [PubMed] [Google Scholar]
- Costa ADT, Pierre SV, Cohen MV, Downey JM, Garlid KD. cGMP signalling in pre- and post-conditioning: the role of mitochondria. Cardiovasc Res. 2008;77:344–352. doi: 10.1093/cvr/cvm050. [DOI] [PubMed] [Google Scholar]
- Di Lisa F, Menabò R, Canton M, Barile M, Bernardi P. Opening of the mitochondrial permeability transition pore causes depletion of mitochondrial and cytosolic NAD+ and is a causative event in the death of myocytes in postischemic reperfusion of the heart. J Biol Chem. 2001;276:2571–2575. doi: 10.1074/jbc.M006825200. [DOI] [PubMed] [Google Scholar]
- Dimmeler S, Fleming I, Fisslthaler B, Hermann C, Busse R, Zeiher AM. Activation of nitric oxide synthase in endothelial cells by Akt-dependent phosphorylation. Nature. 1999;399:601–605. doi: 10.1038/21224. [DOI] [PubMed] [Google Scholar]
- Dorn GW, Souroujon MC, Liron T, Chen C-H, Gray MO, Zhou HZ, et al. Sustained in vivo cardiac protection by a rationally designed peptide that causes ε protein kinase C translocation. Proc Natl Acad Sci U S A. 1999;96:12798–12803. doi: 10.1073/pnas.96.22.12798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dost T, Cohen MV, Downey JM. Redox signaling triggers protection during the reperfusion rather than the ischemic phase of preconditioning. Basic Res Cardiol. 2008;103:378–384. doi: 10.1007/s00395-008-0718-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Downey JM, Cohen MV. Why do we still not have cardioprotective drugs? Circ J. 2009;73:1171–1177. doi: 10.1253/circj.cj-09-0338. [DOI] [PubMed] [Google Scholar]
- D'Souza SP, Yellon DM, Martin C, Schulz R, Heusch G, Onody A, et al. B-type natriuretic peptide limits infarct size in rat isolated hearts via KATP channel opening. Am J Physiol Heart Circ Physiol. 2003;284:H1592–H1600. doi: 10.1152/ajpheart.00902.2002. [DOI] [PubMed] [Google Scholar]
- Eckle T, Krahn T, Grenz A, Köhler D, Mittelbronn M, Ledent C, et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation. 2007;115:1581–1590. doi: 10.1161/CIRCULATIONAHA.106.669697. [DOI] [PubMed] [Google Scholar]
- Feng J, Lucchinetti E, Ahuja P, Pasch T, Perriard J-C, Zaugg M. Isoflurane postconditioning prevents opening of the mitochondrial permeability transition pore through inhibition of glycogen synthase kinase 3β. Anesthesiology. 2005;103:987–995. doi: 10.1097/00000542-200511000-00013. [DOI] [PubMed] [Google Scholar]
- Forbes RA, Steenbergen C, Murphy E. Diazoxide-induced cardioprotection requires signaling through a redox-sensitive mechanism. Circ Res. 2001;88:802–809. doi: 10.1161/hh0801.089342. [DOI] [PubMed] [Google Scholar]
- Förster K, Paul I, Solenkova N, Staudt A, Cohen MV, Downey JM, et al. NECA at reperfusion limits infarction and inhibits formation of the mitochondrial permeability transition pore by activating p70S6 kinase. Basic Res Cardiol. 2006;101:319–326. doi: 10.1007/s00395-006-0593-4. [DOI] [PubMed] [Google Scholar]
- Freixa X, Bellera N, Ortiz-Pérez JT, Jiménez M, Paré C, Bosch X, et al. Ischaemic postconditioning revisited: lack of effects on infarct size following primary percutaneous coronary intervention. Eur Heart J. 2012;33:103–112. doi: 10.1093/eurheartj/ehr297. [DOI] [PubMed] [Google Scholar]
- Frias MA, Lecour S, James RW, Pedretti S. High density lipoprotein/sphingosine-1-phosphate-induced cardioprotection: role of STAT3 as part of the SAFE pathway. JAKSTAT. 2012;1:92–100. doi: 10.4161/jkst.19754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garlid AO, Jaburek M, Jacobs JP, Garlid KD. Mitochondrial reactive oxygen species: which ROS signals cardioprotection? Am J Physiol Heart Circ Physiol. 2013;305:H960–H968. doi: 10.1152/ajpheart.00858.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garlid KD, Paucek P, Yarov-Yarovoy V, Murray HN, Darbenzio RB, D'Alonzo AJ, et al. Cardioprotective effect of diazoxide and its interaction with mitochondrial ATP-sensitive K+ channels: possible mechanism of cardioprotection. Circ Res. 1997;81:1072–1082. doi: 10.1161/01.res.81.6.1072. [DOI] [PubMed] [Google Scholar]
- Garlid KD, Costa ADT, Quinlan CL, Pierre SV, Dos Santos P. Cardioprotective signaling to mitochondria. J Mol Cell Cardiol. 2009;46:858–866. doi: 10.1016/j.yjmcc.2008.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gateau-Roesch O, Argaud L, Ovize M. Mitochondrial permeability transition pore and postconditioning. Cardiovasc Res. 2006;70:264–273. doi: 10.1016/j.cardiores.2006.02.024. [DOI] [PubMed] [Google Scholar]
- Goto M, Miura T, Iliodoromitis EK, O'Leary EL, Ishimoto R, Yellon DM, et al. Adenosine infusion during early reperfusion failed to limit myocardial infarct size in a collateral deficient species. Cardiovasc Res. 1991;25:943–949. doi: 10.1093/cvr/25.11.943. [DOI] [PubMed] [Google Scholar]
- Goto M, Liu Y, Yang X-M, Ardell JL, Cohen MV, Downey JM. Role of bradykinin in protection of ischemic preconditioning in rabbit hearts. Circ Res. 1995;77:611–621. doi: 10.1161/01.res.77.3.611. [DOI] [PubMed] [Google Scholar]
- Goto M, Cohen MV, Van Wylen DGL, Downey JM. Attenuated purine production during subsequent ischemia in preconditioned rabbit myocardium is unrelated to the mechanism of protection. J Mol Cell Cardiol. 1996;28:447–454. doi: 10.1006/jmcc.1996.0041. [DOI] [PubMed] [Google Scholar]
- Griffiths EJ, Halestrap AP. Protection by cyclosporin A of ischemia/reperfusion-induced damage in isolated rat hearts. J Mol Cell Cardiol. 1993;25:1461–1469. doi: 10.1006/jmcc.1993.1162. [DOI] [PubMed] [Google Scholar]
- Griffiths EJ, Halestrap AP. Mitochondrial non-specific pores remain closed during cardiac ischaemia, but open upon reperfusion. Biochem J. 1995;307:93–98. doi: 10.1042/bj3070093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection occurs via glycogen synthase kinase β inhibition during reperfusion in intact rat hearts. Circ Res. 2004;94:960–966. doi: 10.1161/01.RES.0000122392.33172.09. [DOI] [PubMed] [Google Scholar]
- Gross GJ, Auchampach JA. Blockade of ATP-sensitive potassium channels prevents myocardial preconditioning in dogs. Circ Res. 1992;70:223–233. doi: 10.1161/01.res.70.2.223. [DOI] [PubMed] [Google Scholar]
- Hahn J-Y, Song YB, Kim EK, Yu CW, Bae J-W, Chung W-Y, et al. Ischemic postconditioning during primary percutaneous coronary intervention: the effects of postconditioning on myocardial reperfusion in patients with ST-segment elevation myocardial infarction (POST) randomized trial. Circulation. 2013;128:1889–1896. doi: 10.1161/CIRCULATIONAHA.113.001690. [DOI] [PubMed] [Google Scholar]
- Han J, Kim N, Joo H, Kim E, Earm YE. ATP-sensitive K+ channel activation by nitric oxide and protein kinase G in rabbit ventricular myocytes. Am J Physiol Heart Circ Physiol. 2002;283:H1545–H1554. doi: 10.1152/ajpheart.01052.2001. [DOI] [PubMed] [Google Scholar]
- Hausenloy DJ, Yellon DM. New directions for protecting the heart against ischaemia-reperfusion injury: targeting the Reperfusion Injury Salvage Kinase (RISK)-pathway. Cardiovasc Res. 2004;61:448–460. doi: 10.1016/j.cardiores.2003.09.024. [DOI] [PubMed] [Google Scholar]
- Hausenloy DJ, Maddock HL, Baxter GF, Yellon DM. Inhibiting mitochondrial permeability transition pore opening: a new paradigm for myocardial preconditioning? Cardiovasc Res. 2002;55:534–543. doi: 10.1016/s0008-6363(02)00455-8. [DOI] [PubMed] [Google Scholar]
- Hausenloy DJ, Tsang A, Mocanu MM, Yellon DM. Ischemic preconditioning protects by activating prosurvival kinases at reperfusion. Am J Physiol Heart Circ Physiol. 2005;288:H971–H976. doi: 10.1152/ajpheart.00374.2004. [DOI] [PubMed] [Google Scholar]
- Hausenloy DJ, Ong S-B, Yellon DM. The mitochondrial permeability transition pore as a target for preconditioning and postconditioning. Basic Res Cardiol. 2009;104:189–202. doi: 10.1007/s00395-009-0010-x. [DOI] [PubMed] [Google Scholar]
- Hearse DJ, Humphrey SM, Chain EB. Abrupt reoxygenation of the anoxic potassium-arrested perfused rat heart: a study of myocardial enzyme release. J Mol Cell Cardiol. 1973;5:395–407. doi: 10.1016/0022-2828(73)90030-8. [DOI] [PubMed] [Google Scholar]
- Heinzel FR, Luo Y, Li X, Boengler K, Buechert A, García-Dorado D, et al. Impairment of diazoxide-induced formation of reactive oxygen species and loss of cardioprotection in connexin 43 deficient mice. Circ Res. 2005;97:583–586. doi: 10.1161/01.RES.0000181171.65293.65. [DOI] [PubMed] [Google Scholar]
- Heusch G, Musiolik J, Gedik N, Skyschally A. Mitochondrial STAT3 activation and cardioprotection by ischemic postconditioning in pigs with regional myocardial ischemia/reperfusion. Circ Res. 2011;109:1302–1308. doi: 10.1161/CIRCRESAHA.111.255604. [DOI] [PubMed] [Google Scholar]
- Ibanez B, Macaya C, Sánchez-Brunete V, Pizarro G, Fernández-Friera L, Mateos A, et al. Effect of early metoprolol on infarct size in ST-segment-elevation myocardial infarction patients undergoing primary percutaneous coronary intervention: the Effect of Metoprolol in Cardioprotection During an Acute Myocardial Infarction (METOCARD-CNIC) trial. Circulation. 2013;128:1495–1503. doi: 10.1161/CIRCULATIONAHA.113.003653. [DOI] [PubMed] [Google Scholar]
- Inagaki K, Chen L, Ikeno F, Lee FH, Imahashi K-i, Bouley DM, et al. Inhibition of δ-protein kinase C protects against reperfusion injury of the ischemic heart in vivo. Circulation. 2003a;108:2304–2307. doi: 10.1161/01.CIR.0000101682.24138.36. [DOI] [PubMed] [Google Scholar]
- Inagaki K, Hahn HS, Dorn GW, II, Mochly-Rosen D. Additive protection of the ischemic heart ex vivo by combined treatment with δ-protein kinase C inhibitor and ε-protein kinase C activator. Circulation. 2003b;108:869–875. doi: 10.1161/01.CIR.0000081943.93653.73. [DOI] [PubMed] [Google Scholar]
- Jabůrek M, Costa ADT, Burton JR, Costa CL, Garlid KD. Mitochondrial PKCε and mitochondrial ATP-sensitive K+ channel copurify and coreconstitute to form a functioning signaling module in proteoliposomes. Circ Res. 2006;99:878–883. doi: 10.1161/01.RES.0000245106.80628.d3. [DOI] [PubMed] [Google Scholar]
- Jolly SR, Kane WJ, Bailie MB, Abrams GD, Lucchesi BR. Canine myocardial reperfusion injury: its reduction by the combined administration of superoxide dismutase and catalase. Circ Res. 1984;54:277–285. doi: 10.1161/01.res.54.3.277. [DOI] [PubMed] [Google Scholar]
- Juhaszova M, Zorov DB, Kim S-H, Pepe S, Fu Q, Fishbein KW, et al. Glycogen synthase kinase-3β mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. J Clin Invest. 2004;113:1535–1549. doi: 10.1172/JCI19906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Karliner JS. Sphingosine kinase and sphingosine 1-phosphate in the heart: a decade of progress. Biochim Biophys Acta. 2013;1831:203–212. doi: 10.1016/j.bbalip.2012.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kingma JG, Jr, Plante S, Bogaty P. Platelet GPIIb/IIIa receptor blockade reduces infarct size in a canine model of ischemia-reperfusion. J Am Coll Cardiol. 2000;36:2317–2324. doi: 10.1016/s0735-1097(00)01016-0. [DOI] [PubMed] [Google Scholar]
- Kitakaze M, Hori M, Takashima S, Sato H, Inoue M, Kamada T. Ischemic preconditioning increases adenosine release and 5′-nucleotidase activity during myocardial ischemia and reperfusion in dogs: implications for myocardial salvage. Circulation. 1993;87:208–215. doi: 10.1161/01.cir.87.1.208. [DOI] [PubMed] [Google Scholar]
- Kitakaze M, Asakura M, Kim J, Shintani Y, Asanuma H, Hamasaki T, et al. Human atrial natriuretic peptide and nicorandil as adjuncts to reperfusion treatment for acute myocardial infarction (J-WIND): two randomised trials. Lancet. 2007;370:1483–1493. doi: 10.1016/S0140-6736(07)61634-1. [DOI] [PubMed] [Google Scholar]
- Knapp M. Cardioprotective role of sphingosine-1-phosphate. J Physiol Pharmacol. 2011;62:601–607. [PubMed] [Google Scholar]
- Knight DR, Smith AH, Flynn DM, MacAndrew JT, Ellery SS, Kong JX, et al. A novel sodium-hydrogen exchanger isoform-1 inhibitor, zoniporide, reduces ischemic myocardial injury in vitro and in vivo. J Pharmacol Exp Ther. 2001;297:254–259. [PubMed] [Google Scholar]
- Korichneva I, Hoyos B, Chua R, Levi E, Hammerling U. Zinc release from protein kinase C as the common event during activation by lipid second messenger or reactive oxygen. J Biol Chem. 2002;277:44327–44331. doi: 10.1074/jbc.M205634200. [DOI] [PubMed] [Google Scholar]
- Krieg T, Qin Q, McIntosh EC, Cohen MV, Downey JM. ACh and adenosine activate PI3-kinase in rabbit hearts through transactivation of receptor tyrosine kinases. Am J Physiol Heart Circ Physiol. 2002;283:H2322–H2330. doi: 10.1152/ajpheart.00474.2002. [DOI] [PubMed] [Google Scholar]
- Krieg T, Cui L, Qin Q, Cohen MV, Downey JM. Mitochondrial ROS generation following acetylcholine-induced EGF receptor transactivation requires metalloproteinase cleavage of proHB-EGF. J Mol Cell Cardiol. 2004;36:435–443. doi: 10.1016/j.yjmcc.2003.12.013. [DOI] [PubMed] [Google Scholar]
- Krieg T, Liu Y, Rütz T, Methner C, Yang X-M, Dost T, et al. BAY 58-2667, a nitric oxide-independent guanylyl cyclase activator, pharmacologically post-conditions rabbit and rat hearts. Eur Heart J. 2009;30:1607–1613. doi: 10.1093/eurheartj/ehp143. [DOI] [PubMed] [Google Scholar]
- Kunichika H, Ben-Yehuda O, Lafitte S, Kunichika N, Peters B, DeMaria AN. Effects of glycoprotein IIb/IIIa inhibition on microvascular flow after coronary reperfusion. A quantitative myocardial contrast echocardiography study. J Am Coll Cardiol. 2004;43:276–283. doi: 10.1016/j.jacc.2003.08.040. [DOI] [PubMed] [Google Scholar]
- Kuno A, Critz SD, Cui L, Solodushko V, Yang X-M, Krahn T, et al. Protein kinase C protects preconditioned rabbit hearts by increasing sensitivity of adenosine A2b-dependent signaling during early reperfusion. J Mol Cell Cardiol. 2007;43:262–271. doi: 10.1016/j.yjmcc.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuno A, Solenkova NV, Solodushko V, Dost T, Liu Y, Yang X-M, et al. Infarct limitation by a protein kinase G activator at reperfusion in rabbit hearts is dependent on sensitizing the heart to A2b agonists by protein kinase C. Am J Physiol Heart Circ Physiol. 2008;295:H1288–H1295. doi: 10.1152/ajpheart.00209.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lacerda L, Somers S, Opie LH, Lecour S. Ischaemic postconditioning protects against reperfusion injury via the SAFE pathway. Cardiovasc Res. 2009;84:201–208. doi: 10.1093/cvr/cvp274. [DOI] [PubMed] [Google Scholar]
- Lecour S. Activation of the protective survivor activating factor enhancement (SAFE) pathway against reperfusion injury: does it go beyond the RISK pathway? J Mol Cell Cardiol. 2009;47:32–40. doi: 10.1016/j.yjmcc.2009.03.019. [DOI] [PubMed] [Google Scholar]
- Lecour S, Smith RM, Woodward B, Opie LH, Rochette L, Sack MN. Identification of a novel role for sphingolipid signaling in TNFα and ischemic preconditioning mediated cardioprotection. J Mol Cell Cardiol. 2002;34:509–518. doi: 10.1006/jmcc.2002.1533. [DOI] [PubMed] [Google Scholar]
- Lecour S, Rochette L, Opie L. Free radicals trigger TNFα-induced cardioprotection. Cardiovasc Res. 2005a;65:239–243. doi: 10.1016/j.cardiores.2004.10.003. [DOI] [PubMed] [Google Scholar]
- Lecour S, Suleman N, Deuchar GA, Somers S, Lacerda L, Huisamen B, et al. Pharmacological preconditioning with tumor necrosis factor-α activates signal transducer and activator of transcription-3 at reperfusion without involving classic prosurvival kinases (Akt and extracellular signal-regulated kinase) Circulation. 2005b;112:3911–3918. doi: 10.1161/CIRCULATIONAHA.105.581058. [DOI] [PubMed] [Google Scholar]
- Levy DE, Lee C-k. What does Stat3 do? J Clin Invest. 2002;109:1143–1148. doi: 10.1172/JCI15650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li H, Takizawa H, Gong X, Concepcion A, Kambayashi J, Tandon N, et al. Platelet glycoprotein VI (GPVI) deletion reduces myocardial infarction in mice. Circulation. 2007;116(Suppl. II):II–200. [Google Scholar]
- Liao Z, Brar BK, Cai Q, Stephanou A, O'Leary RM, Pennica D, et al. Cardiotrophin-1 (CT-1) can protect the adult heart from injury when added both prior to ischaemia and at reperfusion. Cardiovasc Res. 2002;53:902–910. doi: 10.1016/s0008-6363(01)00531-4. [DOI] [PubMed] [Google Scholar]
- Lim SY, Hausenloy DJ. Remote ischemic conditioning: from bench to bedside. Front Physiol. 2012;3:27. doi: 10.3389/fphys.2012.00027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Limalanathan S, Andersen GØ, Kløw N-E, Abdelnoor M, Hoffmann P, Eritsland J. Effect of ischemic postconditioning on infarct size in patients with ST-elevation myocardial infarction treated by primary PCI: results of the POSTEMI (POstconditioning in ST-Elevation Myocardial Infarction) randomized trial. J Am Heart Assoc. 2014;3:e000679. doi: 10.1161/JAHA.113.000679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lincoff AM, Roe M, Aylward P, Galla J, Rynkiewicz A, Guetta V, et al. Inhibition of delta-protein kinase C by delcasertib as an adjunct to primary percutaneous coronary intervention for acute anterior ST-segment elevation myocardial infarction: results of the PROTECTION AMI Randomized Controlled Trial. Eur Heart J. 2014;35:2516–2523. doi: 10.1093/eurheartj/ehu177. [DOI] [PubMed] [Google Scholar]
- Liu GS, Thornton J, Van Winkle DM, Stanley AWH, Olsson RA, Downey JM. Protection against infarction afforded by preconditioning is mediated by A1 adenosine receptors in rabbit heart. Circulation. 1991;84:350–356. doi: 10.1161/01.cir.84.1.350. [DOI] [PubMed] [Google Scholar]
- Liu Y, Downey JM. Ischemic preconditioning protects against infarction in rat heart. Am J Physiol Heart Circ Physiol. 1992;263:H1107–H1112. doi: 10.1152/ajpheart.1992.263.4.H1107. [DOI] [PubMed] [Google Scholar]
- Liu Y, Sato T, O'Rourke B, Marban E. Mitochondrial ATP-dependent potassium channels: novel effectors of cardioprotection? Circulation. 1998;97:2463–2469. doi: 10.1161/01.cir.97.24.2463. [DOI] [PubMed] [Google Scholar]
- Lønborg J, Kelbæk H, Vejlstrup N, Bøtker HE, Kim WY, Holmvang L, et al. Exenatide reduces final infarct size in patients with ST-segment-elevation myocardial infarction and short-duration of ischemia. Circ Cardiovasc Interv. 2012;5:288–295. doi: 10.1161/CIRCINTERVENTIONS.112.968388. [DOI] [PubMed] [Google Scholar]
- Ma X-L, Weyrich AS, Krantz S, Lefer AM. Mechanisms of the cardioprotective actions of WEB-2170, bepafant, a platelet activating factor antagonist, in myocardial ischemia and reperfusion. J Pharmacol Exp Ther. 1992;260:1229–1236. [PubMed] [Google Scholar]
- Mahaffey KW, Puma JA, Barbagelata NA, DiCarli MF, Leesar MA, Browne KF, et al. Adenosine as an adjunct to thrombolytic therapy for acute myocardial infarction. Results of a multicenter, randomized, placebo-controlled trial: the Acute Myocardial Infarction STudy of ADenosine (AMISTAD) Trial. J Am Coll Cardiol. 1999;34:1711–1720. doi: 10.1016/s0735-1097(99)00418-0. [DOI] [PubMed] [Google Scholar]
- Maroko PR, Kjekshus JK, Sobel BE, Watanabe T, Covell JW, Ross J, Jr, et al. Factors influencing infarct size following experimental coronary artery occlusions. Circulation. 1971;43:67–82. doi: 10.1161/01.cir.43.1.67. [DOI] [PubMed] [Google Scholar]
- Maroko PR, Libby P, Bloor CM, Sobel BE, Braunwald E. Reduction by hyaluronidase of myocardial necrosis following coronary artery occlusion. Circulation. 1972;46:430–437. doi: 10.1161/01.cir.46.3.430. [DOI] [PubMed] [Google Scholar]
- Martin BJ, McClanahan TB, Van Wylen DGL, Gallagher KP. Effects of ischemia, preconditioning, and adenosine deaminase inhibition on interstitial adenosine levels and infarct size. Basic Res Cardiol. 1997;92:240–251. doi: 10.1007/BF00788519. [DOI] [PubMed] [Google Scholar]
- Mentzer RM, Jr, Bartels C, Bolli R, Boyce S, Buckberg GD, Chaitman B, et al. Sodium-hydrogen exchange inhibition by cariporide to reduce the risk of ischemic cardiac events in patients undergoing coronary artery bypass grafting: results of the EXPEDITION study. Ann Thorac Surg. 2008;85:1261–1270. doi: 10.1016/j.athoracsur.2007.10.054. [DOI] [PubMed] [Google Scholar]
- Methner C, Donat U, Felix SB, Krieg T. Cardioprotection of bradykinin at reperfusion involves transactivation of the epidermal growth factor receptor via matrix metalloproteinase-8. Acta Physiol (Oxf) 2009;197:265–271. doi: 10.1111/j.1748-1716.2009.02018.x. [DOI] [PubMed] [Google Scholar]
- Methner C, Schmidt K, Cohen MV, Downey JM, Krieg T. Both A2a and A2b adenosine receptors at reperfusion are necessary to reduce infarct size in mouse hearts. Am J Physiol Heart Circ Physiol. 2010;299:H1262–H1264. doi: 10.1152/ajpheart.00181.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miki T, Cohen MV, Downey JM. Opioid receptor contributes to ischemic preconditioning through protein kinase C activation in rabbits. Mol Cell Biochem. 1998a;186:3–12. [PubMed] [Google Scholar]
- Miki T, Liu GS, Cohen MV, Downey JM. Mild hypothermia reduces infarct size in the beating rabbit heart: a practical intervention for acute myocardial infarction? Basic Res Cardiol. 1998b;93:372–383. doi: 10.1007/s003950050105. [DOI] [PubMed] [Google Scholar]
- Mirabet M, Garcia-Dorado D, Inserte J, Barrabés JA, Lidón R-M, Soriano B, et al. Platelets activated by transient coronary occlusion exacerbate ischemia-reperfusion injury in rat hearts. Am J Physiol Heart Circ Physiol. 2002;283:H1134–H1141. doi: 10.1152/ajpheart.00065.2002. [DOI] [PubMed] [Google Scholar]
- Miura T, Ogawa T, Suzuki K, Goto M, Shimamoto K. Infarct size limitation by a new Na+-H+ exchange inhibitor, Hoe 642: difference from preconditioning in the role of protein kinase C. J Am Coll Cardiol. 1997;29:693–701. doi: 10.1016/s0735-1097(96)00522-0. [DOI] [PubMed] [Google Scholar]
- Montrucchio G, Alloatti G, Mariano F, De Paulis R, Comino A, Emanuelli G, et al. Role of platelet-activating factor in the reperfusion injury of rabbit ischemic heart. Am J Pathol. 1990;137:71–83. [PMC free article] [PubMed] [Google Scholar]
- Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–1136. doi: 10.1161/01.cir.74.5.1124. [DOI] [PubMed] [Google Scholar]
- Myers MG., Jr Moonlighting in mitochondria. Science. 2009;323:723–724. doi: 10.1126/science.1169660. [DOI] [PubMed] [Google Scholar]
- Nakano A, Liu GS, Heusch G, Downey JM, Cohen MV. Exogenous nitric oxide can trigger a preconditioned state through a free radical mechanism, but endogenous nitric oxide is not a trigger of classical ischemic preconditioning. J Mol Cell Cardiol. 2000;32:1159–1167. doi: 10.1006/jmcc.2000.1152. [DOI] [PubMed] [Google Scholar]
- Nash MS, Wood JPM, Osborne NN. Protein kinase C activation by serotonin potentiates agonist-induced stimulation of cAMP production in cultured rat retinal pigment epithelial cells. Exp Eye Res. 1997;64:249–255. doi: 10.1006/exer.1996.0214. [DOI] [PubMed] [Google Scholar]
- Nordstedt C, Kvanta A, Van der Ploeg I, Fredholm BB. Dual effects of protein kinase-C on receptor-stimulated cAMP accumulation in a human T-cell leukemia line. Eur J Pharmacol. 1989;172:51–60. doi: 10.1016/0922-4106(89)90044-8. [DOI] [PubMed] [Google Scholar]
- Norton ED, Jackson EK, Virmani R, Forman MB. Effect of intravenous adenosine on myocardial reperfusion injury in a model with low myocardial collateral blood flow. Am Heart J. 1991;122:1283–1291. doi: 10.1016/0002-8703(91)90567-2. [DOI] [PubMed] [Google Scholar]
- Norton ED, Jackson EK, Turner MB, Virmani R, Forman MB. The effects of intravenous infusions of selective adenosine A1-receptor and A2-receptor agonists on myocardial reperfusion injury. Am Heart J. 1992;123:332–338. doi: 10.1016/0002-8703(92)90643-a. [DOI] [PubMed] [Google Scholar]
- Olafsson B, Forman MB, Puett DW, Pou A, Cates CU, Friesinger GC, et al. Reduction of reperfusion injury in the canine preparation by intracoronary adenosine: importance of the endothelium and the no-reflow phenomenon. Circulation. 1987;76:1135–1145. doi: 10.1161/01.cir.76.5.1135. [DOI] [PubMed] [Google Scholar]
- Oldenburg O, Qin Q, Sharma AR, Cohen MV, Downey JM, Benoit JN. Acetylcholine leads to free radical production dependent on KATP channels, Gi proteins, phosphatidylinositol 3-kinase and tyrosine kinase. Cardiovasc Res. 2002;55:544–552. doi: 10.1016/s0008-6363(02)00332-2. [DOI] [PubMed] [Google Scholar]
- Oldenburg O, Critz SD, Cohen MV, Downey JM. Acetylcholine-induced production of reactive oxygen species in adult rabbit ventricular myocytes is dependent on phosphatidylinositol 3- and Src-kinase activation and mitochondrial KATP channel opening. J Mol Cell Cardiol. 2003;35:653–660. doi: 10.1016/s0022-2828(03)00083-x. [DOI] [PubMed] [Google Scholar]
- Oldenburg O, Qin Q, Krieg T, Yang X-M, Philipp S, Critz SD, et al. Bradykinin induces mitochondrial ROS generation via NO, cGMP, PKG, and mitoKATP channel opening and leads to cardioprotection. Am J Physiol Heart Circ Physiol. 2004;286:H468–H476. doi: 10.1152/ajpheart.00360.2003. [DOI] [PubMed] [Google Scholar]
- Pain T, Yang X-M, Critz SD, Yue Y, Nakano A, Liu GS, et al. Opening of mitochondrial KATP channels triggers the preconditioned state by generating free radicals. Circ Res. 2000;87:460–466. doi: 10.1161/01.res.87.6.460. [DOI] [PubMed] [Google Scholar]
- Parsa CJ, Kim J, Riel RU, Pascal LS, Thompson RB, Petrofski JA, et al. Cardioprotective effects of erythropoietin in the reperfused ischemic heart: a potential role for cardiac fibroblasts. J Biol Chem. 2004;279:20655–20662. doi: 10.1074/jbc.M314099200. [DOI] [PubMed] [Google Scholar]
- Patti G, Bárczi G, Orlic D, Mangiacapra F, Colonna G, Pasceri V, et al. Outcome comparison of 600- and 300-mg loading doses of clopidogrel in patients undergoing primary percutaneous coronary intervention for ST-segment elevation myocardial infarction: results from the ARMYDA-6 MI (Antiplatelet therapy for Reduction of MYocardial Damage during Angioplasty-Myocardial Infarction) randomized study. J Am Coll Cardiol. 2011;58:1592–1599. doi: 10.1016/j.jacc.2011.06.044. [DOI] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SPH, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucl Acids Res. 2014;42(Database Issue):D1098–D1106. doi: 10.1093/nar/gkt1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pedretti S, Raddatz E. STAT3α interacts with nuclear GSK3beta and cytoplasmic RISK pathway and stabilizes rhythm in the anoxic-reoxygenated embryonic heart. Basic Res Cardiol. 2011;106:355–369. doi: 10.1007/s00395-011-0152-5. [DOI] [PubMed] [Google Scholar]
- Penna C, Alloatti G, Cappello S, Gattullo D, Berta G, Mognetti B, et al. Platelet-activating factor induces cardioprotection in isolated rat heart akin to ischemic preconditioning: role of phosphoinositide 3-kinase and protein kinase C activation. Am J Physiol Heart Circ Physiol. 2005;288:H2512–H2520. doi: 10.1152/ajpheart.00599.2004. [DOI] [PubMed] [Google Scholar]
- Penna C, Bassino E, Alloatti G. Platelet activating factor: the good and the bad in the ischemic/reperfused heart. Exp Biol Med. 2011;236:390–401. doi: 10.1258/ebm.2011.010316. [DOI] [PubMed] [Google Scholar]
- Penna C, Angotti C, Pagliaro P. Protein S-nitrosylation in preconditioning and postconditioning. Exp Biol Med. 2014;239:647–662. doi: 10.1177/1535370214522935. [DOI] [PubMed] [Google Scholar]
- Philipp S, Yang X-M, Willenbrock R, Downey JM, Cohen MV. Postconditioning must be initiated in less than 1 minute following reperfusion and is dependent on adenosine 2b receptors and PI3-kinase. Z Kardiol. 2005;94(Suppl. 1):V803. [Google Scholar]
- Philipp S, Yang X-M, Cui L, Davis AM, Downey JM, Cohen MV. Postconditioning protects rabbit hearts through a protein kinase C-adenosine A2b receptor cascade. Cardiovasc Res. 2006;70:308–314. doi: 10.1016/j.cardiores.2006.02.014. [DOI] [PubMed] [Google Scholar]
- Ping P, Song C, Zhang J, Guo Y, Cao X, Li RCX, et al. Formation of protein kinase Cε-Lck signaling modules confers cardioprotection. J Clin Invest. 2002;109:499–507. doi: 10.1172/JCI13200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Piot C, Croisille P, Staat P, Thibault H, Rioufol G, Mewton N, et al. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med. 2008;359:473–481. doi: 10.1056/NEJMoa071142. [DOI] [PubMed] [Google Scholar]
- Pitarys CJ, II, Virmani R, Vildibill HD, Jr, Jackson EK, Forman MB. Reduction of myocardial reperfusion injury by intravenous adenosine administered during the early reperfusion period. Circulation. 1991;83:237–247. doi: 10.1161/01.cir.83.1.237. [DOI] [PubMed] [Google Scholar]
- Pravdic D, Sedlic F, Mio Y, Vladic N, Bienengraeber M, Bosnjak ZJ. Anesthetic-induced preconditioning delays opening of mitochondrial permeability transition pore via protein kinase C-ε-mediated pathway. Anesthesiology. 2009;111:267–274. doi: 10.1097/ALN.0b013e3181a91957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin Q, Downey JM, Cohen MV. Acetylcholine but not adenosine triggers preconditioning through PI3-kinase and a tyrosine kinase. Am J Physiol Heart Circ Physiol. 2003;284:H727–H734. doi: 10.1152/ajpheart.00476.2002. [DOI] [PubMed] [Google Scholar]
- Qin Q, Yang X-M, Cui L, Critz SD, Cohen MV, Browner NC, et al. Exogenous NO triggers preconditioning via a cGMP- and mitoKATP-dependent mechanism. Am J Physiol Heart Circ Physiol. 2004;287:H712–H718. doi: 10.1152/ajpheart.00954.2003. [DOI] [PubMed] [Google Scholar]
- Reimer KA, Jennings RB. Can we really quantitate myocardial cell injury? In: Hearse DJ, Yellon DM, editors. Therapeutic Approaches to Myocardial Infarct Size Limitation. New York: Raven Press; 1984. pp. 163–184. [Google Scholar]
- Reimer KA, Murry CE, Richard VJ. The role of neutrophils and free radicals in the ischemic-reperfused heart: why the confusion and controversy? J Mol Cell Cardiol. 1989;21:1225–1239. doi: 10.1016/0022-2828(89)90669-x. [DOI] [PubMed] [Google Scholar]
- Rischard F, McKean T. Ischemia and ischemic preconditioning in the buffer-perfused pigeon heart. Comp Biochem Physiol C Pharmacol Toxicol Endocrinol. 1998;119:59–65. doi: 10.1016/s0742-8413(97)00182-5. [DOI] [PubMed] [Google Scholar]
- Ross AM, Gibbons RJ, Stone GW, Kloner RA, Alexander RW. A randomized, double-blinded, placebo-controlled multicenter trial of adenosine as an adjunct to reperfusion in the treatment of acute myocardial infarction (AMISTAD-II) J Am Coll Cardiol. 2005;45:1775–1780. doi: 10.1016/j.jacc.2005.02.061. [DOI] [PubMed] [Google Scholar]
- Roubille F, Lairez O, Mewton N, Rioufol G, Ranc S, Sanchez I, et al. Cardioprotection by clopidogrel in acute ST-elevated myocardial infarction patients: a retrospective analysis. Basic Res Cardiol. 2012;107:275. doi: 10.1007/s00395-012-0275-3. [DOI] [PubMed] [Google Scholar]
- Sabatine MS, Cannon CP, Gibson CM, López-Sendón JL, Montalescot G, Theroux P, et al. Addition of clopidogrel to aspirin and fibrinolytic therapy for myocardial infarction with ST-segment elevation. N Engl J Med. 2005a;352:1179–1189. doi: 10.1056/NEJMoa050522. [DOI] [PubMed] [Google Scholar]
- Sabatine MS, Cannon CP, Gibson CM, López-Sendón JL, Montalescot G, Theroux P, et al. Effect of clopidogrel pretreatment before percutaneous coronary intervention in patients with ST-elevation myocardial infarction treated with fibrinolytics: the PCI-CLARITY study. JAMA. 2005b;294:1224–1232. doi: 10.1001/jama.294.10.1224. [DOI] [PubMed] [Google Scholar]
- Sakamoto J, Miura T, Goto M, Iimura O. Limitation of myocardial infarct size by adenosine A1 receptor activation is abolished by protein kinase C inhibitors in the rabbit. Cardiovasc Res. 1995;29:682–688. [PubMed] [Google Scholar]
- Sakuma T, Sari I, Goodman CN, Lindner JR, Klibanov AL, Kaul S. Simultaneous integrin αvβ3 and glycoprotein IIb/IIIa inhibition causes reduction in infarct size in a model of acute coronary thrombosis and primary angioplasty. Cardiovasc Res. 2005;66:552–561. doi: 10.1016/j.cardiores.2005.01.016. [DOI] [PubMed] [Google Scholar]
- Sala-Mercado JA, Wider J, Undyala VVR, Jahania S, Yoo W, Mentzer RM, Jr, et al. Profound cardioprotection with chloramphenicol succinate in the swine model of myocardial ischemia-reperfusion injury. Circulation. 2010;122(Suppl. 1):S179–S184. doi: 10.1161/CIRCULATIONAHA.109.928242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schott RJ, Rohmann S, Braun ER, Schaper W. Ischemic preconditioning reduces infarct size in swine myocardium. Circ Res. 1990;66:1133–1142. doi: 10.1161/01.res.66.4.1133. [DOI] [PubMed] [Google Scholar]
- Schulman D, Latchman DS, Yellon DM. Urocortin protects the heart from reperfusion injury via upregulation of p42/p44 MAPK signaling pathway. Am J Physiol Heart Circ Physiol. 2002;283:H1481–H1488. doi: 10.1152/ajpheart.01089.2001. [DOI] [PubMed] [Google Scholar]
- Schultz JE, Rose E, Yao Z, Gross GJ. Evidence for involvement of opioid receptors in ischemic preconditioning in rat hearts. Am J Physiol. 1995;268:H2157–H2161. doi: 10.1152/ajpheart.1995.268.5.H2157. [DOI] [PubMed] [Google Scholar]
- Schwanke U, Konietzka I, Duschin A, Li X, Schulz R, Heusch G. No ischemic preconditioning in heterozygous connexin43-deficient mice. Am J Physiol Heart Circ Physiol. 2002;283:H1740–H1742. doi: 10.1152/ajpheart.00442.2002. [DOI] [PubMed] [Google Scholar]
- Skyschally A, van Caster P, Boengler K, Gres P, Musiolik J, Schilawa D, et al. Ischemic postconditioning in pigs: no causal role for RISK activation. Circ Res. 2009a;104:15–18. doi: 10.1161/CIRCRESAHA.108.186429. [DOI] [PubMed] [Google Scholar]
- Skyschally A, van Caster P, Iliodromitis EK, Schulz R, Kremastinos DT, Heusch G. Ischemic postconditioning: experimental models and protocol algorithms. Basic Res Cardiol. 2009b;104:469–483. doi: 10.1007/s00395-009-0040-4. [DOI] [PubMed] [Google Scholar]
- Smith RM, Suleman N, McCarthy J, Sack MN. Classic ischemic but not pharmacologic preconditioning is abrogated following genetic ablation of the TNFα gene. Cardiovasc Res. 2002;55:553–560. doi: 10.1016/s0008-6363(02)00283-3. [DOI] [PubMed] [Google Scholar]
- Somers SJ, Frias M, Lacerda L, Opie LH, Lecour S. Interplay between SAFE and RISK pathways in sphingosine-1-phosphate-induced cardioprotection. Cardiovasc Drugs Ther. 2012;26:227–237. doi: 10.1007/s10557-012-6376-2. [DOI] [PubMed] [Google Scholar]
- Sörensson P, Saleh N, Bouvier F, Böhm F, Settergren M, Caidahl K, et al. Effect of postconditioning on infarct size in patients with ST elevation myocardial infarction. Heart. 2010;96:1710–1715. doi: 10.1136/hrt.2010.199430. [DOI] [PubMed] [Google Scholar]
- Squadrito F, Altavilla D, Squadrito G, Saitta A, Campo GM, Arlotta M, et al. Cyclosporin-A reduces leukocyte accumulation and protects against myocardial ischaemia reperfusion injury in rats. Eur J Pharmacol. 1999;364:159–168. doi: 10.1016/s0014-2999(98)00823-1. [DOI] [PubMed] [Google Scholar]
- Srisakuldee W, Jeyaraman MM, Nickel BE, Tanguy S, Jiang Z-S, Kardami E. Phosphorylation of connexin-43 at serine 262 promotes a cardiac injury-resistant state. Cardiovasc Res. 2009;83:672–681. doi: 10.1093/cvr/cvp142. [DOI] [PubMed] [Google Scholar]
- Staat P, Rioufol G, Piot C, Cottin Y, Cung TT, L'Huillier I, et al. Postconditioning the human heart. Circulation. 2005;112:2143–2148. doi: 10.1161/CIRCULATIONAHA.105.558122. [DOI] [PubMed] [Google Scholar]
- Stephens L, Anderson K, Stokoe D, Erdjument-Bromage H, Painter GF, Holmes AB, et al. Protein kinase B kinases that mediate phosphatidylinositol 3,4,5-trisphosphate-dependent activation of protein kinase B. Science. 1998;279:710–714. doi: 10.1126/science.279.5351.710. [DOI] [PubMed] [Google Scholar]
- Sun J, Aponte AM, Kohr MJ, Tong G, Steenbergen C, Murphy E. Essential role of nitric oxide in acute ischemic preconditioning: S-nitros(yl)ation versus sGC/cGMP/PKG signaling? Free Radic Biol Med. 2013;54:105–112. doi: 10.1016/j.freeradbiomed.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Swyers T, Redford D, Larson DF. Volatile anesthetic-induced preconditioning. Perfusion. 2014;29:10–15. doi: 10.1177/0267659113503975. [DOI] [PubMed] [Google Scholar]
- Takaya N, Katoh Y, Iwabuchi K, Hayashi I, Konishi H, Itoh S, et al. Platelets activated by collagen through the immunoreceptor tyrosine-based activation motif in the Fc receptor γ-chain play a pivotal role in the development of myocardial ischemia-reperfusion injury. J Mol Cell Cardiol. 2005;39:856–864. doi: 10.1016/j.yjmcc.2005.07.006. [DOI] [PubMed] [Google Scholar]
- Tanaka K, Ludwig LM, Kersten JR, Pagel PS, Warltier DC. Mechanisms of cardioprotection by volatile anesthetics. Anesthesiology. 2004;100:707–721. doi: 10.1097/00000542-200403000-00035. [DOI] [PubMed] [Google Scholar]
- Tarantini G, Favaretto E, Marra MP, Frigo AC, Napodano M, Cacciavillani L, et al. Postconditioning during coronary angioplasty in acute myocardial infarction: the POST-AMI trial. Int J Cardiol. 2012;162:33–38. doi: 10.1016/j.ijcard.2012.03.136. [DOI] [PubMed] [Google Scholar]
- Théroux P, Chaitman BR, Danchin N, Erhardt L, Meinertz T, Schroeder JS, et al. Inhibition of the sodium-hydrogen exchanger with cariporide to prevent myocardial infarction in high-risk ischemic situations. Main results of the GUARDIAN trial. Circulation. 2000;102:3032–3038. doi: 10.1161/01.cir.102.25.3032. [DOI] [PubMed] [Google Scholar]
- Tissier R, Cohen MV, Downey JM. Protecting the acutely ischemic myocardium beyond reperfusion therapies: are we any closer to realizing the dream of infarct size elimination? Arch Mal Coeur Vaiss. 2007a;100:794–802. [PubMed] [Google Scholar]
- Tissier R, Hamanaka K, Kuno A, Parker JC, Cohen MV, Downey JM. Total liquid ventilation provides ultra-fast cardioprotective cooling. J Am Coll Cardiol. 2007b;49:601–605. doi: 10.1016/j.jacc.2006.09.041. [DOI] [PubMed] [Google Scholar]
- Tong H, Imahashi K, Steenbergen C, Murphy E. Phosphorylation of glycogen synthase kinase-3β during preconditioning through a phosphatidylinositol-3-kinase-dependent pathway is cardioprotective. Circ Res. 2002;90:377–379. doi: 10.1161/01.res.0000012567.95445.55. [DOI] [PubMed] [Google Scholar]
- Toombs CF, Wiltse AL, Shebuski RJ. Ischemic preconditioning fails to limit infarct size in reserpinized rabbit myocardium. Implication of norepinephrine release in the preconditioning effect. Circulation. 1993;88:2351–2358. doi: 10.1161/01.cir.88.5.2351. [DOI] [PubMed] [Google Scholar]
- Trincavelli ML, Marroni M, Tuscano D, Ceruti S, Mazzola A, Mitro N, et al. Regulation of A2B adenosine receptor functioning by tumour necrosis factor a in human astroglial cells. J Neurochem. 2004;91:1180–1190. doi: 10.1111/j.1471-4159.2004.02793.x. [DOI] [PubMed] [Google Scholar]
- Van de Werf F, Janssens L, Brzostek T, Mortelmans L, Wackers FJT, Willems GM, et al. Short-term effects of early intravenous treatment with a beta-adrenergic blocking agent or a specific bradycardiac agent in patients with acute myocardial infarction receiving thrombolytic therapy. J Am Coll Cardiol. 1993;22:407–416. doi: 10.1016/0735-1097(93)90044-2. [DOI] [PubMed] [Google Scholar]
- Van Winkle DM, Thornton JD, Downey DM, Downey JM. The natural history of preconditioning: cardioprotection depends on duration of transient ischemia and time to subsequent ischemia. Coron Artery Dis. 1991;2:613–619. [Google Scholar]
- Vander Heide RS, Reimer KA. Effect of adenosine therapy at reperfusion on myocardial infarct size in dogs. Cardiovasc Res. 1996;31:711–718. doi: 10.1016/0008-6363(95)00235-9. [DOI] [PubMed] [Google Scholar]
- Vegh A, Szekeres L, Parratt J. Preconditioning of the ischaemic myocardium; involvement of the L-arginine nitric oxide pathway. Br J Pharmacol. 1992;107:648–652. doi: 10.1111/j.1476-5381.1992.tb14501.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Velasco CE, Turner M, Cobb MA, Virmani R, Forman MB. Myocardial reperfusion injury in the canine model after 40 minutes of ischemia: effect of intracoronary adenosine. Am Heart J. 1991;122:1561–1570. doi: 10.1016/0002-8703(91)90272-j. [DOI] [PubMed] [Google Scholar]
- Wall TM, Sheehy R, Hartman JC. Role of bradykinin in myocardial preconditioning. J Pharmacol Exp Ther. 1994;270:681–689. [PubMed] [Google Scholar]
- Wallentin L, Becker RC, Budaj A, Cannon CP, Emanuelsson H, Held C, et al. Ticagrelor versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2009;361:1045–1057. doi: 10.1056/NEJMoa0904327. [DOI] [PubMed] [Google Scholar]
- Wegrzyn J, Potla R, Chwae Y-J, Sepuri NBV, Zhang Q, Koeck T, et al. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;323:793–797. doi: 10.1126/science.1164551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weig H-J, Bott-Flügel L, Städele C, Winter K, Schmidt R, Gawaz M, et al. Impaired platelet function reduces myocardial infarct size in Gαq knock-out mice in vivo. J Mol Cell Cardiol. 2008;44:143–150. doi: 10.1016/j.yjmcc.2007.09.018. [DOI] [PubMed] [Google Scholar]
- Weselcouch EO, Baird AJ, Sleph P, Grover GJ. Inhibition of nitric oxide synthesis does not affect ischemic preconditioning in isolated perfused rat hearts. Am J Physiol Heart Circ Physiol. 1995;268:H242–H249. doi: 10.1152/ajpheart.1995.268.1.H242. [DOI] [PubMed] [Google Scholar]
- Wiviott SD, Braunwald E, McCabe CH, Montalescot G, Ruzyllo W, Gottlieb S, et al. Prasugrel versus clopidogrel in patients with acute coronary syndromes. N Engl J Med. 2007;357:2001–2015. doi: 10.1056/NEJMoa0706482. [DOI] [PubMed] [Google Scholar]
- Xi J, McIntosh R, Shen X, Lee S, Chanoit G, Criswell H, et al. Adenosine A2A and A2B receptors work in concert to induce a strong protection against reperfusion injury in rat hearts. J Mol Cell Cardiol. 2009;47:684–690. doi: 10.1016/j.yjmcc.2009.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu Z, Yang X-M, Cohen MV, Neumann T, Heusch G, Downey JM. Limitation of infarct size in rabbit hearts by the novel adenosine receptor agonist AMP 579 administered at reperfusion. J Mol Cell Cardiol. 2000;32:2339–2347. doi: 10.1006/jmcc.2000.1264. [DOI] [PubMed] [Google Scholar]
- Xu Z, Jiao Z, Cohen MV, Downey JM. Protection from AMP 579 can be added to that from either cariporide or ischemic preconditioning in ischemic rabbit heart. J Cardiovasc Pharmacol. 2002;40:510–518. doi: 10.1097/00005344-200210000-00003. [DOI] [PubMed] [Google Scholar]
- Xu Z, Downey JM, Cohen MV. Timing and duration of administration are crucial for antiinfarct effect of AMP 579 infused at reperfusion in rabbit heart. Heart Dis. 2003;5:368–371. doi: 10.1097/01.hdx.0000098614.29006.a7. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Krieg T, Cui L, Downey JM, Cohen MV. NECA and bradykinin at reperfusion reduce infarction in rabbit hearts by signaling through PI3K, ERK, and NO. J Mol Cell Cardiol. 2004a;36:411–421. doi: 10.1016/j.yjmcc.2003.12.008. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Proctor JB, Cui L, Krieg T, Downey JM, Cohen MV. Multiple, brief coronary occlusions during early reperfusion protect rabbit hearts by targeting cell signaling pathways. J Am Coll Cardiol. 2004b;44:1103–1110. doi: 10.1016/j.jacc.2004.05.060. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Philipp S, Downey JM, Cohen MV. Postconditioning's protection is not dependent on circulating blood factors or cells but involves adenosine receptors and requires PI3-kinase and guanylyl cyclase activation. Basic Res Cardiol. 2005;100:57–63. doi: 10.1007/s00395-004-0498-4. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Philipp S, Downey JM, Cohen MV. Atrial natriuretic peptide administered just prior to reperfusion limits infarction in rabbit hearts. Basic Res Cardiol. 2006a;101:311–318. doi: 10.1007/s00395-006-0587-2. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Liu Y, Liu Y, Tandon N, Kambayashi J, Downey JM, et al. Attenuation of infarction in cynomolgus monkeys: preconditioning and postconditioning. Basic Res Cardiol. 2010;105:119–128. doi: 10.1007/s00395-009-0050-2. [DOI] [PubMed] [Google Scholar]
- Yang X-M, Cui L, Alhammouri A, Downey JM, Cohen MV. Triple therapy greatly increases myocardial salvage during ischemia/reperfusion in the in situ rat heart. Cardiovasc Drugs Ther. 2013a;27:403–412. doi: 10.1007/s10557-013-6474-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X-M, Liu Y, Cui L, Yang X, Liu Y, Tandon N, et al. Two classes of anti-platelet drugs reduce anatomical infarct size in monkey hearts. Cardiovasc Drugs Ther. 2013b;27:109–115. doi: 10.1007/s10557-012-6436-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang X-M, Liu Y, Cui L, Yang X, Liu Y, Tandon N, et al. Platelet P2Y12 blockers confer direct postconditioning-like protection in reperfused rabbit hearts. J Cardiovasc Pharmacol Ther. 2013c;18:251–262. doi: 10.1177/1074248412467692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang Y-J, Zhao J-L, You S-J, Wu Y-J, Jing Z-C, Yang W-X, et al. Different effects of tirofiban and aspirin plus clopidogrel on myocardial no-reflow in a mini-swine model of acute myocardial infarction and reperfusion. Heart. 2006b;92:1131–1137. doi: 10.1136/hrt.2005.077164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yellon DM, Alkhulaifi AM, Browne EE, Pugsley WB. Ischaemic preconditioning limits infarct size in the rat heart. Cardiovasc Res. 1992;26:983–987. doi: 10.1093/cvr/26.10.983. [DOI] [PubMed] [Google Scholar]
- 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–H1152. doi: 10.1152/ajpheart.1994.266.3.H1145. [DOI] [PubMed] [Google Scholar]
- Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK. Effects of clopidogrel in addition to aspirin in patients with acute coronary syndromes without ST-segment elevation. N Engl J Med. 2001;345:494–502. doi: 10.1056/NEJMoa010746. [DOI] [PubMed] [Google Scholar]
- Zeymer U, Suryapranata H, Monassier JP, Opolski G, Davies J, Rasmanis G, et al. The Na+/H+ exchange inhibitor eniporide as an adjunct to early reperfusion therapy for acute myocardial infarction. Results of the Evaluation of the Safety and Cardioprotective effects of eniporide in Acute Myocardial Infarction (ESCAMI) trial. J Am Coll Cardiol. 2001;38:1644–1650. doi: 10.1016/s0735-1097(01)01608-4. [DOI] [PubMed] [Google Scholar]
- Zhao Z-Q, Corvera JS, Halkos ME, Kerendi F, Wang N-P, Guyton RA, et al. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol Heart Circ Physiol. 2003;285:H579–H588. doi: 10.1152/ajpheart.01064.2002. [DOI] [PubMed] [Google Scholar]