Abstract
Nitric oxide (NO) plays an important role in the regulation of cardiovascular function. In addition to the classic NO activation of the cyclic guanosine monophosphate (cGMP)-dependent pathway, NO can also regulate cell function through protein S-nitrosylation, a redox dependent, thiol-based, reversible post-translational protein modification that involves attachment of an NO moiety to a nucleophilic protein sulfhydryl group. There are emerging data suggesting that S-nitrosylation of proteins plays an important role in cardioprotection. Protein S-nitrosylation not only leads to changes in protein structure and function, but also prevents these thiol(s) from further irreversible oxidative/nitrosative modification. A better understanding of the mechanism regulating protein S-nitrosylation and its role in cardioprotection will provide us new therapeutic opportunities and targets for interventions in cardiovascular diseases.
Keywords: cardioprotection, mitochondria, nitric oxide, nitrosative and oxidative stress, S-nitrosylation
I. Introduction
Nitric oxide (NO) plays an important role in the regulation of redox signaling and cellular function1-3. NO can be generated by NO synthase (NOS) or by the breakdown of nitrite or other compounds to NO4-6. NOS catalyzes the synthesis of NO by the conversion of L-arginine and oxygen to L-citrulline and NO, in the presence of nicotinamide adenine dinucleotide phosphate (NADPH) and tetrahydrobiopterin. Both endothelial NOS (eNOS) and neuronal NOS (nNOS) are constitutively expressed in distinct subcellular locations within cardiomyocytes: eNOS is generally thought to be predominantly localized to invaginations of the sarcolemma called caveolae7, 8 and nNOS is mostly found in the sarcoplasmic reticulum (SR)8, 9. However, changes of expression and redistribution of these two constitutive isoforms of NOS have been found in some disease conditions such as myocardial infarction10, 11 and heart failure12. The third isoform of NOS, the inducible NOS (iNOS), is barely detected in the myocardium under normal conditions, but expression of iNOS is stimulated by inflammatory mediators13, 14. A mitochondrial NOS has been reported to be present in the inner mitochondrial membrane or matrix15, although there are conflicting results regarding mitochondrial NOS16-19. An alternative source of mitochondrial NO could be nNOS associated with the SR, as the SR membrane has been shown to be attached to the outer mitochondrial membrane20-22. This differential NOS localization provides organelle or subcellular generation of NO, which in turn provides a localized signal for protein S-nitrosylation23, 24.
In addition to activating cyclic guanosine monophosphate (cGMP)-dependent signaling pathways, recent studies suggest that NO, by attachment of an NO moiety to a nucleophilic protein sulfhydryl resulting in S-nitrosothiol (SNO) formation, generates a post-translational modification known as protein S-nitrosylation, which plays an important role in biology25, 26. Recent data suggests that protein SNO is important in cardioprotection. This review will focus on the role of S-nitrosylation in cardioprotection and the potential mechanism(s) involved.
II. NO and Ischemia/Reperfusion (I/R) Injury
NO is generated during I/R
A number of studies have shown that NO is produced during ischemia4, 27-29. The NO that is generated appears to be due to both NOS dependent and NOS-independent pathways. During ischemiawhen oxygen is low, nitrite conversion into NO is likely to be an important source of NO. However there is likely to be some residual oxygen, particularly at the beginning of ischemia, and Zweier et al have shown that with short durations of ischemia (30 minutes) N-nitro-L-arginine methyl ester (L-NAME), a NOS inhibitor, blocks approximately 60-80% of NO generation27. Thus it appears that both NOS dependent and independent (nitrite) mechanisms are involved in the generation of NO during ischemia. The exact proportion of NOS dependent and independent NO generation may depend on the model (the duration of ischemia, the diet, and the species). These data show that NO is generated during ischemia, which raised the question of whether the NO generated during ischemia is beneficial or detrimental?
Increasing NO protects
Addition of NO donors, particularly at concentrations that release NO at physiological (sub-micromolar) levels, has been shown to reduce I/R injury30, 31. Similarly, cardiac specific overexpression of eNOS32, 33 or iNOS34 has been reported to reduce I/R injury. Thus, there is reasonable agreement that a slight increase in NO is cardioprotective.
Consistent with a protective role for NO, a number of cardioprotective models have been shown to involve an increase in NOS35. For example, protection in females36, 37 and erythropoietin-mediated protection38, 39 require NOS activation. Also in models of I/R injury in various tissues such as heart and liver, nitrite has been shown to reduce infarct size and to reduce apoptosis and necrosis5, 6. The mechanism of nitrite-mediated cytoprotection appears to be NO-dependent, as a loss of cytoprotection occurred when animals were treated with a NO scavenger5, 6.
Reducing NO below basal levels has no consistent effect on protection
Whether loss or inhibition of NOS exacerbates I/R injury has been controversial, especially the data involving eNOS knockout (eNOS-KO) mice. Studies comparing infarct size after I/R in wild type (WT) and eNOS-KO hearts report no difference40-42, a reduction43 and an increase in infarct size43, 44. Part, but not all, of the discrepancy appears to be related to two different models of eNOS-KO mice. Lefer and coworkers43 studied I/R in the eNOS-KO mice generated at Harvard compared to those generated at UNC. They found an 84% increase in infarct size in the Harvard eNOS-KO mice, but a decrease in infarct size in the UNC eNOS-KO mice. This difference was attributed to a compensatory increase in iNOS in the UNC eNOS-KO mice which resulted in protection in these mice43. However, Guo et al42 also examined I/R injury in both of these mouse models and in contrast to the study by Lefer's group, reported no difference in infarct size in either UNC or Harvard eNOS-KO hearts compared to WT hearts. It is possible that the induction of iNOS in the UNC mice might depend on the housing and feeding conditions of the mice, and this might account for some of the differences observed. Most studies of infarct size show no difference between WT and nNOS knockout (nNOS-KO) hearts44, 45. Most recent studies find that NOS inhibition does not alter infarct size or recovery of function following I/R42, 46, 47. However, there are some older reports showing that NOS inhibition can be protective27, 48, 49. NO can combine with reactive oxygen species (ROS) to generate peroxynitrite which can be detrimental particularly at high levels50, 51. This discrepancy between the beneficial and detrimental effects of NO might depend on the relative availability of NO versus ROS. NOS can also become “uncoupled” resulting in the generation of ROS and this might also complicate the interpretation of the effects of NOS inhibitors. Thus the effect of NOS inhibition is variable and might depend on the level of oxidative stress. Taken together, the data suggest that under most conditions an increase in NO above a threshold level, as might occur with NO donors or overexpression of NOS, is protective; however lowering NO below basal levels does not appear to increase injury.
III. NO and Cardioprotection
As mentioned above, a slight increase in NO has been shown to be cardioprotective. Cardioprotection can be initiated acutely by activation of signaling pathways (acute protection) or by upregulation of new proteins (delayed protection), and NO has been shown to be important in both models. For simplicity we will discuss these separately and focus on acute protection. Acute protection can be initiated by signaling pathways activated prior to and during ischemia, i.e., preconditioning (PC), as well as pathways activated at reperfusion, such as postconditioning.
A. NO and acute PC
NO increases during PC
Brief episodes of ischemia and reperfusion, i.e., PC, render the heart more resistant to subsequent prolonged periods of ischemia52. PC has been shown to lead to an increase in NO and inhibitors of PC such at inhibitors of the PI3K pathway block the increase in NO production53-55. Inhibitors of NOS have been shown to block protection in a number of different cardioprotective models. As will be discussed, inhibitors of NOS block protection associated with delayed PC and postconditioning. Also cardioprotection in females is blocked by inhibitors of NOS or loss of either eNOS or nNOS11, 56, and erythropoietin mediated protection has also been reported to be blocked by NOS inhibitors57. The ability of NOS inhibition or loss of NOS to block acute PC, however, has resulted in discrepant results. Some58, 59, but not all studies60, 61 find that NOS inhibitors block acute PC. Interestingly, although Downey's group initially reported that NOS inhibitors do not block acute PC60, Cohen, Downey and coworker54 recently reported the ability of L-NAME to block PC depends on the relative contribution of signaling through the adenosine pathways compared to the bradykinin or opioid pathways. They suggest that signaling via the adenosine pathway is not blocked by L-NAME. Downey's group also reportedthat NOS inhibitors block bradykinin mediated protection62. Studies with mice lacking eNOS have also provided mixed results. Bell and Yellon reported that in eNOS-KO hearts, PC still reduced infarct size with 4 cycles of PC, but there was no reduction in infarct size with only 2 or 3 cycles63. However, Guo et al in a very well established in vivo infarct model found that both Harvard and UNC eNOS-KO hearts exhibited a PC mediated reduction in infarct size that was comparable to that observed in WT hearts, suggesting the eNOS is not necessary for acute PC42.
So what do we conclude regarding the role of NOS and NO in acute PC? Data consistently show that PC results in an increase in NO. However, inhibition of NOS, by either pharmacological inhibitors or genetic ablation, results in inconsistent results; in some cases PC protection is blocked, but not in others. One explanation provided by the Downey, Cohen group is that L-NAME does not block under conditions where PC signaling is predominantly via adenosine, rather than bradykinin or opioids. Another possible explanation for this discrepancy might be a differential role provided by non-enzymatic production of NO during PC. NO can be generated from nitrite under acidic conditions that occur during PC64, and this non-enzymatically generated NO would not be inhibited by NOS inhibitors. The levels of nitrite can vary depending on diet, thus leading to diet dependent differences in the NO that can be generated from nitrite65. Thus, some of the differences regarding whether NOS generated NO is required for acute PC, might depend on the levels of non-enzymatically generated NO, which in turn might depend on diet. If sufficient NO is generated via non-enzymatic mechanisms, then NOS inhibitors would not be expected to block PC. This explanation might account for the more consistent role for NOS inhibitors in pharmacological PC (where there is no acidosis to generate NO nonenzymatically) and it would also be consistent with NOS inhibitors blocking after 2 or 3 cycles, but not after four cycles of PC54, 63. This hypothesis would also be consistent with a recent study reporting that dietary nitrite restores cardioprotection in eNOS-KO hearts65.
B. NO and postconditioning
It has been shown that following a sustained period of ischemia, brief intermittent periods of ischemia and reperfusion administered at the start of reperfusion (postconditioning) reduced infarct size66. Tsang et al reported that postconditioning resulted in an increase in phosphorylation of eNOS and that the protective effects of postconditioning were blocked by the inhibition of the phosphatidylinositol 3-kinase (PI3K) with wortmannin, which blocked the subsequent eNOS phosphorylation67. Consistent with a role for NOS in postconditioning, Yang et al found that the NOS inhibitor, L-NAME, administered just before reperfusion blocked the protective effects of postconditioning68. In a recent study, addition of a mitochondria-targeted SNO at the start of reperfusion has also been found to be cardioprotective69. Thus, NO appears to be an important mediator in postconditioning.
C. NO and delayed PC
As discussed, a trigger phase of brief intermittent periods of ischemia and reperfusion reduces infarct size following a subsequent sustained period of ischemia. The protection afforded by PC is lost if there is a delay between the brief intermittent episodes of ischemia and the sustained ischemia. Typically, if there is an hour or longer delay between the trigger preconditioning period and the sustained period of ischemia the protection is lost. However, a delayed protection occurs about 24 hours after the PC trigger phase. In contrast to acute PC70, delayed PC involves alterations in gene expression13, 71. Interestingly it has been shown that NO generated by eNOS during the trigger phase is required for the protection that is observed 24 hours later53, 58, 72, 73. NO generated during the trigger phase activates a signaling cascade that leads to upregulation of iNOS, which is essential for the delayed PC, and that the delayed PC was blocked in mice lacking eNOS74, 75. Furthermore, cardiomyocyte-specific overexpression of iNOS has been shown to attenuate reperfusion-induced ROS production and protect against I/R injury by preventing the mitochondrial permeability transition pore (MPTP) opening and cell death34.
III. Mechanism of NO Protection
Initial studies indicated that many NO mediated effects were due to NO activation of soluble guanylyl cyclase (sGC) and the resulting increase in cGMP76. However, NO can modify protein thiol groups resulting in the formation of protein SNO, and this protein modification can also mediate effects of NO25, 26. Both sGC and SNO pathways have been reported to be important in cardioprotection. NO activation of sGC/cGMP/protein kinase G(PKG) is reported to activate the mitochondrial KATP channel, by a mechanism that has not been elucidated, but which presumably involves phosphorylation by PKG77, 78. In support of this hypothesis, 1H-[1,2,4]-oxadiazole-[4,3-a]-quinoxalin-1-one (ODQ), an inhibitor of sGC, has been found to block the bradykinin mediated increase in ROS62. Lochner et al reported that ODQ significantly attenuated (to an intermediate level) the improved contractility that occurs in PC hearts58.
Previous studies have shown that the cardioprotection of PC could be attenuated or abolished by antioxidants79-81. Such a redox-based mechanism for cardioprotection of PC suggests that protein S-nitrosylation, a redox-reversible posttranslational protein modification, might play an important cardioprotective role in PC31, 82. Recent studies have clearly shown that in addition to NO activation of sGC, NO can mediate protection by S-nitrosylation and a number of studies have found S-nitrosylated proteins that appear to be important in cardioprotection (see Table 1). An increase of protein S-nitrosylation has also been found to be involved in nitrite-mediated cardioprotection64, 83. Before discussing the role of S-nitrosylation in cardioprotection we will first briefly discuss the mechanisms involved in regulating protein S-nitrosylation with an emphasis on mechanisms that would be important in I/R and cardioprotection.
Table 1.
Cardioprotective effects of S-nitrosylated protein
S-nitrosylated proteins | Cardioprotective effects |
---|---|
I. Mitochondrial proteins | |
Complex I | Inhibits activity, and attenuates ROS generation during I/R31, 64, 119 |
Cytochrome c oxidase | Inhibits its activity, and decrease oxygen consumption120 |
F1F0ATPase | Inhibits activity, and reduces ATP consumption during I/R31 |
Creatine kinase | Inhibits activity, and suppresses contractility under stress121 |
α-KGDH | Increases activity, might prevent oxidative inactivation upon I/R31 |
II. Ca2+ handling | |
L-type Ca2+ channel | Inhibits and reduces Ca2+ entry under oxidative stress11, 31, 122 |
RyR2 | Hyponitrosylation increases SR Ca2+ leak and arrhythmogenesis123 |
SERCA2a | Increases SR Ca2+ uptake, reduces Ca2+ overload during PC31 |
III. Anti-apoptosis and anti-oxidative stress | |
Caspase-3 | Inhibits activity, and elicits anti-apoptotic effect125 |
COX-2 | Increases activity, and elicits preconditioning effect126 |
HIF-1α | Provokes stabilization in normoxic condition, against I/R injury97 |
NADPH oxidase | Inhibits activity, and suppresses ROS production127 |
Thioredoxin | Increases reductase activity, and elicits anti-apoptosis effect128 |
IV. Protein trafficking | |
Dynamin | Promotes endocytosis, and elicits anti-apoptosis effect131, 132 |
NSF | Inhibits exocytosis, and elicits anti-inflammatory effect133 |
GRK2 | Decreases β-adrenergic receptor phosphorylation/desensitization134 |
β-arrestin 2 | Promotes endocytosis and β-adrenergic receptor internalization135 |
Only proteins that have been shown to be involved in the cardiovascular system are included in this table.
IV. Mechanisms of Regulation of S-nitrosylation
A. SNO formation and stabilization
Protein S-nitrosylation occurs by endogenous NO-mediated nitrosylating agents such as dinitrogen trioxide (N2O3), by transition metal catalyzed addition of NO, or by transnitrosylation from low-molecular-weight SNO such as S-nitrosoglutathione (GSNO) or S-nitrosocystiene (CysNO)24, 84. The S-nitrosylation of cysteine residue(s) on proteins depends on the precise conditions of NO, O2, hydrophobicity, nucleophilicity, and redox surrounding the targeted thiol(s), which could change drastically during I/R. The actual redox state85, 86 and ultrastructural accessibility87 of cysteine residue(s) under low oxygen tension such as hypoxia and ischemia might determine whether a particular thiol in a given protein is subjected to S-nitrosylation.
We are interested in mechanism(s) that might influence SNO in the setting of ischemia and PC. PC results in an increase in cytosolic Ca2+, which in turn activates constitutive NOS leading to an increase in NO for SNO formation. In addition, PC leads to formation of ROS and reactive nitrogen species (RNS), leading to a decreased intracellular reduced glutathione (GSH) pool, which might stabilize SNO formation by attenuating GSH-mediated trans-/denitrosylation88, 89. The effects of oxygen on protein S-nitrosylation are complex and difficult to predict a priori. Autoxidation of NO yields N2O3 as a nitrosylating agent. This reaction is accelerated dramatically in a hydrophobic environment such as a lipid membrane90, 91. PC involves intermittent ischemia and reperfusion, and reperfusion allows for generation of NO and nitrosylating agents such as N2O3. Furthermore, the stability of SNO appears to be favored by low ambient oxygen92. With increasing oxygen, the level of protein S-nitrosylation has been found to decrease while S-thiolation is promoted93, and the intermediate thiyl radical is proposed to be involved in the decomposition SNO94. Consistent with this concept, breathing low oxygen concentrations has been found to potentiate the ability of inhaled NO to increase cardiac albumin-SNO95. Also, an increase of protein S-nitrosylation has been recently reported in a study of endothelial cells exposed to acute hypoxia96. Taken together, the intermittent reperfusion can increase generation of NO/SNO, and the formed SNO will be stabilized by the low oxygen during the sustained ischemia. On reperfusion oxygen is restored and this would accelerate the breakdown of the SNO. This concept is supported by a study in which an increase in SNO occurred during PC while a decline in SNO occurred upon reperfusion11. Thus, PC would provide an environment that might favor S-nitrosylation, including NO/SNO formation, ion content, and redox equilibrium.
B. Reversibility of SNO: transnitrosylation and denitrosylation
If SNO has a regulatory role in cell biology, it is expected that there are regulated mechanisms for removal of SNO, similar to the role of phosphatases in proteins phosphorylation. In fact, a number of mechanisms have been described leading to the removal of protein S-nitrosylation (see97). The nitrosyl moiety could be removed by transnitrosylation (i.e., transfer of NO moiety between proteins) with low-molecular-mass thiols, such as GSH to form GSNO, or cysteine thiols to form CysNO84. However, unless the GSNO is denitrosylated by GSNO reductase, the low-molecular-mass thiol formed SNO could also transnitrosylate some other proteins, in which case the overall protein SNO will not change.
Compared to transnitrosylation, protein denitrosylation plays an important role in eliminating SNO proteins therefore regulating cellular SNO levels. Some enzymatic systems have been shown to function as denitrosylase, such as GSNO reductase (GSNOR)97, 98, the thioredoxin system99, carbonyl reductase100, Cu,Zn-superoxide dismutase101, and xanthine oxidoreductase102. Their physiological role as a critical regulator of SNO biology has been discussed in detail elsewhere103, 104. Given that these denitrosylases are indeed coupled to cellular antioxidant redox enzymatic systems, the level of protein S-nitrosylation will be regulated by cellular redox state. Cellular redox changes during ischemia and reperfusion and this is likely to have important effects on protein SNO.
V. Detection of S-nitrosylated protein
Most methods used to detect S-nitrosylated proteins are based on the biotin switch method105. Briefly in the biotin switch method, free thiol groups are blocked with a methyl methylating agent, the SNO groups are then reduced with ascorbate to free thiol groups that are then labeled with a sulfhydryl specific biotinylating agent. In this way the proteins that were originally S-nitrosylated are now labeled with a biotin and can be detected with an anti-biotin antibody. As is the case with any protein identification strategy, the set of SNO proteins that are detected depends to some extent on the details of the method employed. For example, if one were attempting to determine what proteins were phosphorylated by cardioprotection, one might employ different methods. Typically one will test for a specific pathway using a specific antibody such as an antibody that recognizes phospho-AKT for example. Therefore to use a similar approach to detect SNO proteins, following the biotin switch one can immunoprecipitate with antibodies for specific proteins and then immunoblot with anti-biotin to determine if the protein is S-nitrosylated. Another approach used to detect phosphorylated proteins in cardioprotection is to run a gel and then blot with an anti-phosphoserine/threonine or anti-phosphotyrosine antibody. Analogous to this, after the biotin switch method, one can run the entire extract on a gel and probe with an anti-biotin antibody. Anti-SNO specific antibodies (analogous to anti-phospho antibodies) are available and are currently being compared to results obtained with the biotin switch. Recently the use of two dimensional (2D) gel electrophoresis methods and mass spectrometry (MS) has been applied to identify phosphorylated proteins in cardioprotection106. A similar approach can also be used to measure protein SNO31, 47, 107. As is the case with phosphorylation, these different methods can result in the identification of many proteins. Although 2D gel methods provide unbiased information on a large number of proteins, high molecular weight and membrane proteins do not readily enter the 2D gel and are therefore often undetected with 2D methods108. Also because of dynamic range issues, most of the proteomic methods (2D gels and MS) are biased towards detection of high abundance proteins. Thus, lack of detection of a low abundance protein in a 2D gel or MS analysis does not mean that the protein is not subjected to SNO modification. To date, many of the studies identifying SNO proteins have used 2D gels and therefore there are likely to be many low abundance signaling molecules, which are S-nitrosylated, that have not been currently identified. It should also be mentioned that in assessing the effect of a treatment on SNO, it is important to normalize the change in SNO to total protein, similar to what is routinely done in the evaluation of phosphorylation. SNO content can be detected and quantified by colorimetric (HgCl2-coupled Griess reagent), chemiluminescent (Ozone-coupled photolysis), fluorescent methods (4,5-diaminofluorescein, DAN), and anti-CysNO-based methods.
VI. SNO and Cardioprotection
A. Cardioprotection results in SNO of many common proteins
Although NO is clearly established as a mediator of cardioprotection, there have been only a few of studies that have examined the role of protein S-nitrosylation in cardioprotection. PC has been shown to result in S-nitrosylation of a number of proteins31, as might be expected given the increase of NO formation in PC. Interestingly, although postconditioning and delayed PC both require NO, protein S-nitrosylation has not been examined under these conditions. NO donors such as GSNO have been shown to also increase S-nitrosylation of a number of proteins, many of which overlap with proteins undergoing S-nitrosylation during PC31. Estrogen has also been shown to be cardioprotective and it results in the S-nitrosylation of many proteins in common with PC and GSNO47. A recent study has shown that mice lacking GSNOR, an enzyme involved in removing SNO from proteins, results in increased protein SNO and cardioprotection97. Thus, considerable evidence is accumulating suggesting that an increase in SNO is associated with cardioprotection.
Table 1 lists a number of proteins that have been reported to show a change in SNO with different cardioprotective treatments. In this Table we have focused on acute protection in cardiomyocytes. An interesting general observation is that many of the proteins showing increased S-nitrosylation with cardioprotection are mitochondrial proteins. Possible reasons for this include the increased stability of N2O3 in the hydrophobic milieu of the mitochondria, which would favor S-nitrosylation, and the high level of reactive cysteines in mitochondrial proteins109, 110. As NO is labile, the high level of SNO of mitochondrial proteins suggests a nearby source of NO, such as the endoplasmic/sarcoplasmic reticulum as discussed previously. In addition, protein S-nitrosylation in mitochondria can occur as a result of transnitrosylation from low-molecular-mass SNO such as GSNO94.
As mentioned earlier, low abundance, signaling proteins are not well represented in the 2D proteomic screens that have been largely used to identify S-nitrosylated protein by cardioprotective treatments. Thus, it is very likely that there are other SNO proteins, some of which have not yet been identified, that play a role in cardioprotection. For example, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) has been reported to be inhibited by S-nitrosylation111, 112 and inhibition of PTEN would be expected to increase signaling through the PI3K pathway, a pathway that has been shown to initiate cardioprotection113-115. S-nitrosylation of PTEN has not been observed in heart in cardioprotection, but this might just reflect the low abundance of PTEN, which might be difficult to detect with 2D screening methods. Much additional work will be needed to fully elucidate the mechanism by which SNO modulates cardioprotection.
B. Is SNO an epiphenomena or involved in protection?
Consistent with a role for NO/SNO in protection, L-NAME blocks both the protection and SNO generation that are mediated by preconditioning or treatment with an estrogen agonist47. Furthermore the finding that loss of GSNOR is cardioprotective supports a protective role for SNO97. But to clearly establish the importance of S-nitrosylation of any protein or class of proteins in protection, it will be necessary to mutate the thiols that are S-nitrosylated and determine if this blocks protection. One of the challenges is that many proteins are S-nitrosylated in PC and other models of cardioprotection. Are one or a few of these proteins causally involved or are many or all important? If S-nitrosylation of multiple proteins and/or pathways is necessary, it may be difficult to sort out which proteins are important using a mutation strategy for a single protein. Also, it is likely that SNO can have beneficial and detrimental effects and that cardiorprotection depends on the balance.
C. Possible mechanism(s) for SNO mediated cardioprotection
General Mechanisms
Table 1 shows a large and growing list of proteins that are S-nitrosylated in the setting of cardioprotection. How might S-nitrosylation of these proteins lead to reduced cell death? Analogous to phosphorylation, S-nitrosylation can alter the activity of proteins. In addition to modulating the activity of proteins, it has been suggested that S-nitrosylation, either directly or by enhancing S-glutathionylation, can also protect reactive thiol groups from more irreversible oxidation, thereby allowing the activity of these enzyme to be restored more quickly following oxidative stress as might occur during I/R116, 117. S-nitrosylation not only modifies proteins, leading to altered activity and protection against further oxidation, but it can also facilitate additional posttranslational modifications such as S-glutathionylation, which can alter protein activity117, 118. As discussed by Baba et al, the S-nitrosylated form of aldose reductase, but not the reduced form can react with GSH to form S-glutathionylated aldose reductase and increase its activity118.
S-nitrosylation alters protein activity
S-nitrosylation of the mitochondrial complex I31, 64, 119, cytochrome c oxidase120, the F1F0ATPase (complex V)31, and creatine kinase121 inhibit their activities, whereas S-nitrosylation of α-ketoglutarate dehydrogenase (α-KGDH) increases its activity31. S-nitrosylation of the L-type Ca2+ channel has been reported to reduce the channel activity in the hearts under adrenergic stimulation11, under oxidative stress during PC31, and with atrial fibrillation122. S-nitrosylation of the cardiac SR calcium release channel/ryanodine receptor (RyR2) has been shown to increase its open channel probability. Also the decreased S-nitrosylation of RyR2 in nNOS-KO hearts has been suggested to cause cysteine residues of RyR2 to be more oxidized under oxidative stress, leading to SR Ca2+ leaking and arrhythmogenesis123. Cardiac SR Ca2+-ATPase (SERCA2a) has also been shown to be activated by an NO dependent modification. Adachi et al showed that NO dependent S-glutathionylation (occurring via peroxynitrite and therefore dependent on superoxide generation as well as NO and GSH) of SERCA2a resulted in an increase in SERCA2a activity124. Sun et al also found S-nitrosylation of SERCA2a was associated with increased SR Ca2+-ATPase activity in PC hearts31. However, whether S-nitrosylation of SERCA2a per se increases activity or whether S-nitrosylation increases SERCA2a activity by enhancing S-glutathionylation remains to be sorted out.
Protein S-nitrosylation has also shown to play an important cardioprotective role by modulating the activity of proteins involved in apoptosis and oxidative stress such as caspase 3125, cyclooxygenase-2 (COX-2)126, hypoxia inducible factor 1α (HIF1α)97, NADPH oxidase127, and thioredoxin128. Furthermore, recent studies suggest that NOS generates NO locally and regulates compartmentalized S-nitrosylation and protein trafficking in the cardiovascular system129, 130. Several important proteins that regulate protein trafficking, such as dynamin131, 132, N-ethylmaleimide-sensitive factor (NSF)133, G protein-coupled receptor kinase 2 (GRK-2)134, and β-arrestin 2135, have been identified to be S-nitrosylated and S-nitrosylation of these proteins play an important role in regulating protein trafficking and signaling transduction.
Figure 1 provides a potential mechanism(s) by which protein S-nitrosylation might lead to acute protection as occurs in PC. PC results in S-nitrosylation and inhibition of the L-type Ca2+ channel, which would reduce Ca2+ entry into the myocytes during ischemia and early reperfusion. S-nitrosylation (by itself or by promoting S-glutathionylation) also results in activation of SERCA2a, which would further reduce cytosolic Ca2+ during ischemia and early reperfusion. In fact, enhanced expression of SERCA2a has been shown to be cardioprotective 136. PC also results in S-nitrosylation and inhibition of the F1F0ATPase which would reduce ATP consumption by reverse mode of the F1F0 ATPase31. This would preserve ATP and would also reduce the mitochondrial membrane potential (Δψ) thereby reducing the driving force for Ca2+ uptake into the mitochondrial matrix. PC has also been shown to lead to S-nitrosylation and inhibition of complex I, which has been suggested to reduce ROS generation64, 119. Taken together, the increase of protein S-nitrosylation during PC would be expected to lead to reduced mitochondrial Ca2+ and reduced ROS generation, which together would be expected to prevent the MPTP opening and reduce cell death, as MPTP opening has been shown to be an important determinant of myocardial I/R death137, 138.
Figure 1. Potential mechanism(s) by which protein S-nitrosylation might lead to acute protection as occurs in PC.
PC results in S-nitrosylation and inhibition of the L-type Ca2+ channel, which would reduce Ca2+ entry into the myocyte during ischemia and early reperfusion. S-nitrosylation also results in activation of SERCA2a, thus further reducing cytosolic Ca2+ during ischemia and early reperfusion. PC also results in S-nitrosylation and inhibition of the F1F0ATPase which would reduce ATP consumption by reverse mode of the F1F0 ATPase. PC has also been shown to lead to S-nitrosylation and inhibition of complex I, which has been suggested to reduce ROS generation. Taken together, the increase of protein S-nitrosylation during PC would be expected to lead to reduced Ca2+ overload and reduced ROS generation, therefore preventing cell death during I/R injury.
SNO enhances S-glutathionylation
S-nitrosylation can also enhance the addition of glutathione to proteins through a modification known as S-glutathionylation117, 118. Similar to S-nitrosylation, S-glutathionylation can alter protein activity and protect cysteines from irreversible oxidation117, 139. For example, aldose reductase activity is increased by S-glutathionylation118, and S-glutathionylation of complex II has also been shown to enhance electron transfer140. Interestingly, S-glutathionylation of complex II is decreased during reperfusion following ischemia140; it might be interesting to determine if cardioprotection reverses this loss of S-glutathionylation. S-glutathionylation can be reversed, following the restoration of a reducing GSH/oxidized glutathione (GSSG) ratio. However, S-glutathionylation can also lead to irreversible protein posttranslational modification under excessive oxidative and nitrosative stress139.
SNO protects against irreversible oxidation
The modification of thiols can be regulated by redox-related signaling in the cell including ROS/RNS3. In addition to S-nitrosylation and S-glutationylation, free thiols can be oxidized to sulfenic acid (SOH), sulfinic acid (SO2H), disulfide bonds, or sulfonic acid (SO3H). With increasing oxidation states, the modifications become irreversible and this typically leads to irreversible modification of the activity of the protein24, 141. It has been suggested that S-nitrosylation and S-glutathionylation of thiol groups in proteins can protect these proteins against irreversible oxidative modifications116, 117, 142. Furthermore, irreversible oxidation of thiols can block the physiological modification by S-nitrosylation or S-glutathionylation and thereby interfere with normal physiological signaling124.
An example of how S-nitrosylation can protect against irreversible oxidation of a protein is provided by studies on protein-tyrosine phosphatase (PTP)143. The activity of PTP can be regulated by thiol redox modification at an active cysteine residue, which is also susceptible to S-nitrosylation. A recent study revealed that the active site cysteine 215 was the primary cysteine residue susceptible to S-nitrosylation, and the S-nitrosylation at cysteine 215 protects PTP from subsequent irreversible oxidation. Thus, S-nitrosylation of cysteine 215 might prevent PTP from permanent inactivation caused by oxidative stress143. It is suggested that S-nitrosylation might form a reversible “molecular shield” that prevents further thiol oxidation during excessive oxidative/nitrosative stress116.
S-nitrosylation can also control cellular redox through the regulation of some important redox-active enzymes, such as GSNOR97, 98, thioredoxin144-146, glutaredoxin147, peroxiredoxin148 and their corresponding reductase system. It has been suggested that NO can protect cells from oxidative stress, while loss or inhibition of NOS enhances oxidative stress123, 149.
Possible role of S-nitrosylation in explaining discrepant effects of NO
As mentioned earlier, with high levels of NO or an increase in the ratio of ROS to NO the beneficial effects of NO are lost and NO can have detrimental effects. The variability in the amount of ROS might explain some of the discrepant findings in the literature. As just discussed S-nitrosylation of several proteins might reduce production of ROS during ischemia and early reperfusion, which in turn would reduce peroxynitrite generation and might contribute to the beneficial effects of NO. Thus it is possible that the depending on the levels of NO generation and ROS generation different proteins might be S-nitrosylated and this might alter the balance between cell protection and cell death.
D. Timing of S-nitrosylation and denitroyslation
Protein S-nitrosylation is a very labile modification and it appears that SNO-mediated cardioprotection induced by PC has a very short duration. This would be an ideal property for a cardioprotective signal. For example, long-term inhibition of the F1F0ATPase or complex I would likely have detrimental consequences. However, it appears that SNO is reversed early during reperfusion. Thus, SNO can modify activity of these proteins during ischemia and the start of reperfusion, but then with the loss of SNO during early reperfusion, the activity of these proteins returns to normal. The lability of SNO might also account for the loss of PC when the period between the PC and the sustained ischemia is extended.
E. Does protection depend on the level of SNO?
The scenario proposed in Figure 1 and data in several studies31, 47, 97, 119 suggest that cardioprotection is associated with an increase in SNO. Although a general increase in SNO appears to be associated with cardioprotection, it is likely an over-simplification to assume that S-nitrosylation of all proteins is cardioprotective. If one considers phosphorylation, depending on the target, phoshorylation can stimulate cell death as well as protection and therefore it is likely that S-nitrosylation can mediate cell death as well as protection. For example, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a protein involved in glycolysis, which has recently been shown to have an additional function in regulating cell death, has been reported to initiate a cell death pathway in neurons150. S-nitrosylation of GAPDH enhances its binding to Siah1 (seven in absentia homolog 1, an E3 ubiquitin ligase) and mediates its translocation to the nucleus and the initiation of cell death. Interestingly Sen et al have reported that GOSPEL, a protein which competes with GAPDH for binding to Siah,is also S-nitrosylated151. S-nitrosylation of GOSPEL promotes the binding of GOSPEL to GAPDH which reduces GAPDH binding to Siah1 thus preventing neuronal cell injury and death151. Thus it is proposed that an NO/SNO signaling will lead to S-nitrosylation of different proteins depending on the level of SNO generation or the activation of other signaling pathways. It will be important in future studies to identify (as in the case of GAPDH and GOSPEL) the details of the pathways by which SNO regulates protection as well as death.
Therefore, it is likely that S-nitrosylation of some proteins will promote rather than inhibit cell death, and the outcome depends on the balance. We are just beginning to understand the complex relationship between SNO, cell death and cardioprotection. Similar to phosphorylation, SNO signaling is compartmentalized. There is also likely to be cross-talk with other signaling pathways such as redox and phosphorylation, and ROS/RNS have been suggested to alter the balance between phosphorylation and S-nitrosylation of key signaling molecules1, 3, 152. The association between increased SNO and cardioprotection31, 47, 97, 119 might suggest that under these cardioprotective conditions the beneficial effects of SNO predominate over the detrimental effects.
VII. Conclusion and Future Perspective
Protein S-nitrosylation, a reversible, redox-sensitive, thiol-based posttranslational modification, has been found to play an important role in a wide range of NO-mediated cardiovascular effects, including but not limited to mitochondrial metabolic regulation, intracellular Ca2+ handling, protein trafficking, and regulation of cellular defense against apoptosis and oxidative stress. As shown in Figure 2, S-nitrosylated proteins could elicit their regulatory effects and protect cells by (1) changing the structure and function of proteins due to SNO modification on the active thiol(s) and (2) shielding the modified cysteine residues (by S-nitrosylation or S-glutathionylation) from further irreversible modification under oxidative/nitrosative stress. Furthermore, depending on the localization of NO/SNO signaling, the level of protein S-nitrosylation, and/or interaction with other signaling pathways, the overall effect of protein S-nitrosylation can be protective or detrimental (e.g., GOSPEL/GAPDH). The feasibility of pharmacological preconditioning with SNO has provided an intriguing therapeutic strategy for protecting against myocardial I/R injury69, 97, 153-155. Taken together, protein S-nitrosylation and S-nitrosylated proteins have emerged as an important contributor to cardioprotection.
Figure 2. Possible mechanisms of S-nitrosylated protein in cardioprotection.
S-nitrosylated proteins could elicit their regulatory effects and protect cells by (A) changing the structure and function of protein due to SNO modification on the active thiol(s); (B) shielding the modified cysteine residues (by S-nitrosylation or S-glutathionylation) from further irreversible modification (indicated as “X”) under oxidative/nitrosative stress; (C) signaling transduction via protein interaction (e.g., GOSPEL/GAPDH).
There are a number of important areas for future studies. It has been proposed that S-nitrosylation of cysteine residues might protect them from oxidation. It will be important to determine exactly which proteins and cysteine residues are S-nitrosylated under cardioprotection which would otherwise be oxidized. It will also be important to better understand the relationship between redox, ROS levels, NO levels and protection. As discussed, S-nitrosylation of some proteins will be beneficial whereas S-nitrosylation of others will be detrimental. It will be important to understand how S-nitrosylation of different proteins is regulated. Is the S-nitrosylation of different proteins controls soley by varying the amount of NO (probably not this simple)? We know there is localization of NOS and this likely contributes to the differences in protein S-nitrosylation. There are also subcellular differences in enzymes that denitrosylate proteins and this will also contribute to the regulation of compartmentalized subcellular SNO signaling. The past decade has provided a wealth of new information on how SNO modifies cell biology, but we have only just begun!
Acknowledgments
Sources of Funding
This work was supported by the NHLBI, National Institutes of Health Intramural Program.
Non-standard Abbreviations and Acronyms
- α-KGDH
α-ketoglutarate dehydrogenase
- Δψ
mitochondrial membrane potential
- cGMP
cyclic guanosine monophosphate
- COX-2
cyclooxygenase-2
- CysNO
S-nitrosocysteine
- eNOS
endothelial NOS
- GAPDH
glyceraldehyde-3-phosphate dehydrogenase
- GOSPEL
GAPDH's competitor of Siah protein enhances life
- GRK-2
G protein-coupled receptor kinase 2
- GSH
reducing glutathione
- GSNO
S-nitrosoglutathione
- GSNOR
GSNO reductase
- GSSG
oxidized glutathione
- HIF1α
hypoxia inducible factor 1α
- iNOS
inducible NOS
- I/R
ischemia/reperfusion
- KO
knockout
- L-NAME
N-nitro-L-arginine methyl ester
- MPTP
mitochondrial permeability transition pore
- MS
mass spectrometry
- N2O3
dinitrogen trioxide
- NADPH
nicotinamide adenine dinucleotide phosphate
- nNOS
neuronal NOS
- NO
nitric oxide
- NOS
NO synthase
- NSF
N-ethylmaleimide-sensitive factor (NSF)
- ODQ
1H-[1,2,4]-oxadiazole-[4,3-a]-quinoxalin-1-one
- PC
preconditioning
- PI3K
phosphatidylinositol 3-kinase
- PKG
protein kinase G
- PTEN
phosphatase and tensin homolog deleted on chromosome 10
- PTP
protein tyrosine phosphatase
- RNS
reactive nitrogen species
- ROS
reactive oxygen species
- RyR2
cardiac SR calcium release channel/ryanodine receptor
- SERCA2a
cardiac SR Ca2+ ATPase
- Siah1
seven in absentia homolog 1, an ubiquitin E3 ligase
- sGC
soluble guanylyl cyclase
- SNO
S-nitrosothiol
- SR
sarcoplasmic reticulum
- WT
wild type
Footnotes
Disclosures
None.
References
- 1.Hare JM, Stamler JS. NO/redox disequilibrium in the failing heart and cardiovascular system. J Clin Invest. 2005;115:509–517. doi: 10.1172/JCI200524459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Schulz R, Rassaf T, Massion PB, Kelm M, Balligand JL. Recent advances in the understanding of the role of nitric oxide in cardiovascular homeostasis. Pharmacol Ther. 2005;108:225–256. doi: 10.1016/j.pharmthera.2005.04.005. [DOI] [PubMed] [Google Scholar]
- 3.Janssen-Heininger YMW, Mossman BT, Heintz NH, Forman HJ, Kalyanaraman B, Finkel T, Stamler JS, Rhee SG, van der Vliet A. Redox-based regulation of signal transduction: principles, pitfalls, and promises. Free Radic Biol Med. 2008;45:1–17. doi: 10.1016/j.freeradbiomed.2008.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zweier JL, Wang P, Samouilov A, Kuppusamy P. Enzyme-independent formation of nitric oxide in biological tissues. Nat Med. 1995;1:804–809. doi: 10.1038/nm0895-804. [DOI] [PubMed] [Google Scholar]
- 5.Webb A, Bond R, McLean P, Uppal R, Benjamin N, Ahluwalia A. Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia-reperfusion damage. Proc Natl Acad Sci USA. 2004;101:13683–13688. doi: 10.1073/pnas.0402927101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Duranski MR, Greer JJ, Dejam A, Jaganmohan S, Hogg N, Langston W, Patel RP, Yet SF, Wang X, Kevil CG, Gladwin MT, Lefer DJ. Cytoprotective effects of nitrite during in vivo ischemia-reperfusion of the heart and liver. J Clin Invest. 2005;115:1232–1240. doi: 10.1172/JCI22493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Feron O, Dessy C, Opel DJ, Arstall MA, Kelly RA, Michel T. Modulation of the endothelial nitric-oxide synthase-caveolin interaction in cardiac myocytes. Implications for the autonomic regulation of heart rate. J Biol Chem. 1998;273:30249–30254. doi: 10.1074/jbc.273.46.30249. [DOI] [PubMed] [Google Scholar]
- 8.Barouch LA, Harrison RW, Skaf MW, Rosas GO, Cappola TP, Kobeissi ZA, Hobai IA, Lemmon CA, Burnett AL, O'Rourke B, Rodriguez ER, Huang PL, Lima JAC, Berkowitz DE, Hare JM. Nitric oxide regulates the heart by spatial confinement of nitric oxide synthase isoforms. Nature. 2002;416:337–339. doi: 10.1038/416337a. [DOI] [PubMed] [Google Scholar]
- 9.Xu KY, Huso DL, Dawson TM, Bredt DS, Becker LC. Nitric oxide synthase in cardiac sarcoplasmic reticulum. Proc Natl Acad Sci USA. 1999;96:657–662. doi: 10.1073/pnas.96.2.657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Damy T, Ratajczak P, Robidel E, Bendall JK, Oliviero P, Boczkowski J, Ebrahimian T, Marotte F, Samuel J-L, Heymes C. Up-regulation of cardiac nitric oxide synthase 1-derived nitric oxide after myocardial infarction in senescent rats. FASEB J. 2003;17:1934–1936. doi: 10.1096/fj.02-1208fje. [DOI] [PubMed] [Google Scholar]
- 11.Sun J, Picht E, Ginsburg KS, Bers DM, Steenbergen C, Murphy E. Hypercontractile female hearts exhibit increased S-nitrosylation of the L-type Ca2+ channel α1 subunit and reduced ischemia/reperfusion injury. Circ Res. 2006;98:403–411. doi: 10.1161/01.RES.0000202707.79018.0a. [DOI] [PubMed] [Google Scholar]
- 12.Damy T, Ratajczak P, Shah AM, Camors E, Marty I, Hasenfuss G, Marotte F, Samuel JL, Heymes C. Increased neuronal nitric oxide synthase-derived NO production in the failing human heart. Lancet. 2004;363:1365–1367. doi: 10.1016/S0140-6736(04)16048-0. [DOI] [PubMed] [Google Scholar]
- 13.Bolli R. Cardioprotective function of inducible nitric oxide synthase and role of nitric oxide in myocardial ischemia and preconditioning: an overview of a decade of research. J Mol Cell Cardiol. 2001;33:1897–1918. doi: 10.1006/jmcc.2001.1462. [DOI] [PubMed] [Google Scholar]
- 14.Seddon M, Shah AM, Casadei B. Cardiomyocytes as effectors of nitric oxide signalling. Cardiovasc Res. 2007;75:315–326. doi: 10.1016/j.cardiores.2007.04.031. [DOI] [PubMed] [Google Scholar]
- 15.Kanai AJ, Pearce LL, Clemens PR, Birder LA, VanBibber MM, Choi S-Y, de Groat WC, Peterson J. Identification of a neuronal nitric oxide synthase in isolated cardiac mitochondria using electrochemical detection. Proc Natl Acad Sci USA. 2001;98:14126–14131. doi: 10.1073/pnas.241380298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.French S, Giulivi C, Balaban RS. Nitric oxide synthase in porcine heart mitochondria: evidence for low physiological activity. Am J Physiol. 2001;280:H2863–H2867. doi: 10.1152/ajpheart.2001.280.6.H2863. [DOI] [PubMed] [Google Scholar]
- 17.Brookes PS. Mitochondrial nitric oxide synthase. Mitochondrion. 2004;3:187–204. doi: 10.1016/j.mito.2003.10.001. [DOI] [PubMed] [Google Scholar]
- 18.Venkatakrishnan P, Nakayasu ES, Almeida IC, Miller RT. Absence of nitric-oxide synthase in sequentially purified rat liver mitochondria. J Biol Chem. 2009;284:19843–19855. doi: 10.1074/jbc.M109.003301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lacza Z, Pankotai E, Busija DW. Mitochondrial nitric oxide synthase: current concepts and controversies. Front Biosci. 2009;14:4436–4443. doi: 10.2741/3539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Balaban RS, Bose S, French SA, Territo PR. Role of calcium in metabolic signaling between cardiac sarcoplasmic reticulum and mitochondria in vitro. Am J Physiol. 2003;284:C285–C293. doi: 10.1152/ajpcell.00129.2002. [DOI] [PubMed] [Google Scholar]
- 21.Boncompagni S, Rossi AE, Micaroni M, Beznoussenko GV, Polishchuk RS, Dirksen RT, Protasi F. Mitochondria are linked to calcium stores in striated muscle by developmentally regulated tethering structures. Mol Biol Cell. 2009;20:1058–1067. doi: 10.1091/mbc.E08-07-0783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hayashi T, Martone ME, Yu Z, Thor A, Doi M, Holst MJ, Ellisman MH, Hoshijima M. Three-dimensional electron microscopy reveals new details of membrane systems for Ca2+ signaling in the heart. J Cell Sci. 2009;122:1005–1013. doi: 10.1242/jcs.028175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hess DT, Matsumoto A, Nudelman R, Stamler JS. S-nitrosylation: spectrum and specificity. Nat Cell Biol. 2001;3:E46–E49. doi: 10.1038/35055152. [DOI] [PubMed] [Google Scholar]
- 24.Hess DT, Matsumoto A, Kim S-O, Marshall HE, Stamler JS. Protein S-nitrosylation: purview and parameters. Nat Rev Mol Cell Biol. 2005;6:150–166. doi: 10.1038/nrm1569. [DOI] [PubMed] [Google Scholar]
- 25.Derakhshan B, Hao G, Gross SS. Balancing reactivity against selectivity: the evolution of protein S-nitrosylation as an effector of cell signaling by nitric oxide. Cardiovasc Res. 2007;75:210–219. doi: 10.1016/j.cardiores.2007.04.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Foster MW, Hess DT, Stamler JS. Protein S-nitrosylation in health and disease: a current perspective. Trends Mol Med. 2009;15:391–404. doi: 10.1016/j.molmed.2009.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zweier JL, Wang P, Kuppusamy P. Direct measurement of nitric oxide generation in the ischemic heart using electron paramagnetic resonance spectroscopy. J Biol Chem. 1995;270:304–307. doi: 10.1074/jbc.270.1.304. [DOI] [PubMed] [Google Scholar]
- 28.Depre C, Fierain L, Hue L. Activation of nitric oxide synthase by ischaemia in the perfused heart. Cardiovasc Res. 1997;33:82–87. doi: 10.1016/s0008-6363(96)00176-9. [DOI] [PubMed] [Google Scholar]
- 29.Martin C, Schulz R, Post H, Boengler K, Kelm M, Kleinbongard P, Gres P, Skyschally A, Konietzka I, Heusch G. Microdialysis-based analysis of interstitial NO in situ: NO synthase-independent NO formation during myocardial ischemia. Cardiovasc Res. 2007;74:46–55. doi: 10.1016/j.cardiores.2006.12.020. [DOI] [PubMed] [Google Scholar]
- 30.Takano H, Tang X-L, Qiu Y, Guo Y, French BA, Bolli R. Nitric oxide donors induce late preconditioning against myocardial stunning and infarction in conscious rabbits via an antioxidant-sensitive mechanism. Circ Res. 1998;83:73–84. doi: 10.1161/01.res.83.1.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sun J, Morgan M, Shen R-F, Steenbergen C, Murphy E. Preconditioning results in S-nitrosylation of proteins involved in regulation of mitochondrial energetics and calcium transport. Circ Res. 2007;101:1155–1163. doi: 10.1161/CIRCRESAHA.107.155879. [DOI] [PubMed] [Google Scholar]
- 32.Brunner F, Maier R, Andrew P, Wolkart G, Zechner R, Mayer B. Attenuation of myocardial ischemia/reperfusion injury in mice with myocyte-specific overexpression of endothelial nitric oxide synthase. Cardiovasc Res. 2003;57:55–62. doi: 10.1016/s0008-6363(02)00649-1. [DOI] [PubMed] [Google Scholar]
- 33.Elrod JW, Greer JM, Bryan NS, Langston W, Szot JF, Gebregzlabher H, Janssens S, Feelisch M, Lefer DJ. Cardiomyocyte-specific overexpression of nitric oxide synthase-3 protects against myocardial ischemia-reperfusion Injury. Arterioscler Thromb Vasc Biol. 2006;26:1517–1523. doi: 10.1161/01.ATV.0000224324.52466.e6. [DOI] [PubMed] [Google Scholar]
- 34.West MB, Rokosh G, Obal D, Velayutham M, Xuan Y-T, Hill BG, Keith RJ, Schrader J, Guo Y, Conklin DJ, Prabhu SD, Zweier JL, Bolli R, Bhatnagar A. Cardiac myocyte-specific expression of inducible nitric oxide synthase protects against ischemia/reperfusion injury by preventing mitochondrial permeability transition. Circulation. 2008;118:1970–1978. doi: 10.1161/CIRCULATIONAHA.108.791533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jones SP, Bolli R. The ubiquitous role of nitric oxide in cardioprotection. J Mol Cell Cardiol. 2006;40:16–23. doi: 10.1016/j.yjmcc.2005.09.011. [DOI] [PubMed] [Google Scholar]
- 36.Murphy E, Steenbergen C. Cardioprotection in females: a role for nitric oxide and altered gene expression. Heart Fail Rev. 2007;12:293–300. doi: 10.1007/s10741-007-9035-0. [DOI] [PubMed] [Google Scholar]
- 37.Murphy E, Steenbergen C. Gender-based differences in mechanisms of protection in myocardial ischemia-reperfusion injury. Cardiovasc Res. 2007;75:478–486. doi: 10.1016/j.cardiores.2007.03.025. [DOI] [PubMed] [Google Scholar]
- 38.Burger DE, Xiang F-L, Hammoud L, Jones DL, Feng Q. Erythropoietin protects the heart from ventricular arrhythmia during ischemia and reperfusion via neuronal nitric-oxide synthase. J Pharmacol Exp Ther. 2009;329:900–907. doi: 10.1124/jpet.109.150896. [DOI] [PubMed] [Google Scholar]
- 39.Mihov D, Bogdanov N, Grenacher B, Gassmann M, Zünd G, Bogdanova A, Tavakoli R. Erythropoietin protects from reperfusion-induced myocardial injury by enhancing coronary endothelial nitric oxide production. Eur J Cardiothorac Surg. 2009;35:839–846. doi: 10.1016/j.ejcts.2008.12.049. [DOI] [PubMed] [Google Scholar]
- 40.Yang X-P, Liu Y-H, Shesely EG, Bulagannawar M, Liu F, Carretero OA. Endothelial nitric oxide gene knockout mice: cardiac phenotypes and the effect of angiotensin-converting enzyme inhibitor on myocardial ischemia/reperfusion injury. Hypertension. 1999;34:24–30. doi: 10.1161/01.hyp.34.1.24. [DOI] [PubMed] [Google Scholar]
- 41.Bell RM, Yellon DM. Bradykinin limits infarction when administered as an adjunct to reperfusion in mouse heart: the role of PI3K, Akt and eNOS. J Mol Cell Cardiol. 2003;35:185–193. doi: 10.1016/s0022-2828(02)00310-3. [DOI] [PubMed] [Google Scholar]
- 42.Guo Y, Li Q, Wu W-J, Tan W, Zhu X, Mu J, Bolli R. Endothelial nitric oxide synthase is not necessary for the early phase of ischemic preconditioning in the mouse. J Mol Cell Cardiol. 2008;44:496–501. doi: 10.1016/j.yjmcc.2008.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Sharp BR, Jones SP, Rimmer DM, Lefer DJ. Differential response to myocardial reperfusion injury in eNOS-deficient mice. Am J Physiol. 2002;282:H2422–H2426. doi: 10.1152/ajpheart.00855.2001. [DOI] [PubMed] [Google Scholar]
- 44.Sumeray MS, Rees DD, Yellon DM. Infarct size and nitric oxide synthase in murine myocardium. J Mol Cell Cardiol. 2000;32:35–42. doi: 10.1006/jmcc.1999.1050. [DOI] [PubMed] [Google Scholar]
- 45.Jones SP, Girod WG, Huang PL, Lefer DJ. Myocardial reperfusion injury in neuronal nitric oxide synthase deficient mice. Coron Artery Dis. 2000;11:593–597. doi: 10.1097/00019501-200012000-00004. [DOI] [PubMed] [Google Scholar]
- 46.Ye Y, Lin Y, Manickavasagam S, Perez-Polo JR, Tieu BC, Birnbaum Y. Pioglitazone protects the myocardium against ischemia-reperfusion injury in eNOS and iNOS knockout mice. Am J Physiol. 2008;295:H2436–H2446. doi: 10.1152/ajpheart.00690.2008. [DOI] [PubMed] [Google Scholar]
- 47.Lin J, Steenbergen C, Murphy E, Sun J. Estrogen receptor-β activation results in S-nitrosylation of proteins involved in cardioprotection. Circulation. 2009;120:245–254. doi: 10.1161/CIRCULATIONAHA.109.868729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Naseem SA, Kontos MC, Rao PS, Jesse RL, Hess ML, Kukreja RC. Sustain inhibition of nitric oxide by NG-nitro-l-arginine improves myocardial function following ischemia/reperfusion in isolated perfused rat heart. J Mol Cell Cardiol. 1995;27:419–426. doi: 10.1016/s0022-2828(08)80038-7. [DOI] [PubMed] [Google Scholar]
- 49.Andelová EBM, Pancza D, Styk J, Ravingerová T. The role of NO in ischemia/reperfusion injury in isolated rat heart. Gen Physiol Biophys. 2005;24:411–426. [PubMed] [Google Scholar]
- 50.Ferdinandy P, Schulz R. Nitric oxide, superoxide, and peroxynitrite in myocardial ischaemia-reperfusion injury and preconditioning. Br J Pharmacol. 2003;138:532–543. doi: 10.1038/sj.bjp.0705080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zweier JL, Talukder MAH. The role of oxidants and free radicals in reperfusion injury. Cardiovasc Res. 2006;70:181–190. doi: 10.1016/j.cardiores.2006.02.025. [DOI] [PubMed] [Google Scholar]
- 52.Murry C, Jennings R, Reimer K. 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]
- 53.Xuan Y-T, Tang X-L, Qiu Y, Banerjee S, Takano H, Han H, Bolli R. Biphasic response of cardiac NO synthase isoforms to ischemic preconditioning in conscious rabbits. Am J Physiol. 2000;279:H2360–H2371. doi: 10.1152/ajpheart.2000.279.5.H2360. [DOI] [PubMed] [Google Scholar]
- 54.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]
- 55.Tong H, Chen W, Steenbergen C, Murphy E. Ischemic preconditioning activates phosphatidylinositol-3-kinase upstream of protein kinase C. Circ Res. 2000;87:309–315. doi: 10.1161/01.res.87.4.309. [DOI] [PubMed] [Google Scholar]
- 56.Cross HR, Murphy E, Steenbergen C. Ca2+ loading and adrenergic stimulation reveal male/female differences in susceptibility to ischemia-reperfusion injury. Am J Physiol. 2002;283:H481–H489. doi: 10.1152/ajpheart.00790.2001. [DOI] [PubMed] [Google Scholar]
- 57.Burger D, Lei M, Geoghegan-Morphet N, Lu X, Xenocostas A, Feng Q. Erythropoietin protects cardiomyocytes from apoptosis via up-regulation of endothelial nitric oxide synthase. Cardiovasc Res. 2006;72:51–59. doi: 10.1016/j.cardiores.2006.06.026. [DOI] [PubMed] [Google Scholar]
- 58.Lochner A, Marais E, Genade S, Moolman JA. Nitric oxide: a trigger for classic preconditioning? Am J Physiol. 2000;279:H2752–H2765. doi: 10.1152/ajpheart.2000.279.6.H2752. [DOI] [PubMed] [Google Scholar]
- 59.Marina Prendes M, Gonzalez M, Savino E, Varela A. Role of endogenous nitric oxide in classic preconditioning in rat hearts. Regul Pept. 2007;139:141–145. doi: 10.1016/j.regpep.2006.10.015. [DOI] [PubMed] [Google Scholar]
- 60.Nakano A, Liu G, Heusch G, Downey J, Cohen M. 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]
- 61.Post H, Schulz R, Behrends M, Gres P, Umschlag C, Heusch G. No involvement of endogenous nitric oxide in classical ischemic preconditioning in swine. J Mol Cell Cardiol. 2000;32:725–733. doi: 10.1006/jmcc.2000.1117. [DOI] [PubMed] [Google Scholar]
- 62.Oldenburg O, Qin Q, Krieg T, Yang X-M, Philipp S, Critz SD, Cohen MV, Downey JM. Bradykinin induces mitochondrial ROS generation via NO, cGMP, PKG, and mitoKATP channel opening and leads to cardioprotection. Am J Physiol. 2004;286:H468–H476. doi: 10.1152/ajpheart.00360.2003. [DOI] [PubMed] [Google Scholar]
- 63.Bell RM, Yellon DM. The contribution of endothelial nitric oxide synthase to early ischaemic preconditioning: the lowering of the preconditioning threshold. An investigation in eNOS knockout mice. Cardiovasc Res. 2001;52:274–280. doi: 10.1016/s0008-6363(01)00394-7. [DOI] [PubMed] [Google Scholar]
- 64.Shiva S, Sack MN, Greer JJ, Duranski M, Ringwood LA, Burwell L, Wang X, MacArthur PH, Shoja A, Raghavachari N, Calvert JW, Brookes PS, Lefer DJ, Gladwin MT. Nitrite augments tolerance to ischemia/reperfusion injury via the modulation of mitochondrial electron transfer. J Exp Med. 2007;204:2089–2102. doi: 10.1084/jem.20070198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Bryan NS, Calvert JW, Gundewar S, Lefer DJ. Dietary nitrite restores NO homeostasis and is cardioprotective in endothelial nitric oxide synthase-deficient mice. Free Radic Biol Med. 2008;45:468–474. doi: 10.1016/j.freeradbiomed.2008.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhao Z-Q, Corvera JS, Halkos ME, Kerendi F, Wang N-P, Guyton RA, Vinten-Johansen J. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol. 2003;285:H579–H588. doi: 10.1152/ajpheart.01064.2002. [DOI] [PubMed] [Google Scholar]
- 67.Tsang A, Hausenloy DJ, Mocanu MM, Yellon DM. Postconditioning: a form of “modified reperfusion” protects the myocardium by activating the phosphatidylinositol 3-kinase-Akt pathway. Circ Res. 2004;95:230–232. doi: 10.1161/01.RES.0000138303.76488.fe. [DOI] [PubMed] [Google Scholar]
- 68.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]
- 69.Prime TA, Blaikie FH, Evans C, Nadtochiy SM, James AM, Dahm CC, Vitturi DA, Patel RP, Hiley CR, Abakumova I, Requejo R, Chouchani ET, Hurd TR, Garvey JF, Taylor CT, Brookes PS, Smith RAJ, Murphy MP. A mitochondria-targeted S-nitrosothiol modulates respiration, nitrosates thiols, and protects against ischemia-reperfusion injury. Proc Natl Acad Sci USA. 2009;106:10764–10769. doi: 10.1073/pnas.0903250106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Thornton J, Striplin S, Liu GS, Swafford A, Stanley AW, Van Winkle DM, Downey JM. Inhibition of protein synthesis does not block myocardial protection afforded by preconditioning. Am J Physiol. 1990;259:H1822–H1825. doi: 10.1152/ajpheart.1990.259.6.H1822. [DOI] [PubMed] [Google Scholar]
- 71.Yin C, Salloum FN, Kukreja RC. A novel role of microRNA in late preconditioning: upregulation of endothelial nitric oxide synthase and heat shock protein 70. Circ Res. 2009;104:572–575. doi: 10.1161/CIRCRESAHA.108.193250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lochner A, Marais E, Du Toit E, Moolman J. Nitric oxide triggers classic ischemic preconditioning. Ann NY Acad Sci. 2002;962:402–414. doi: 10.1111/j.1749-6632.2002.tb04084.x. [DOI] [PubMed] [Google Scholar]
- 73.Bolli R, Dawn B, Tang XL, Qiu Y, Ping P, Xuan YT, Jones WK, Takano H, Guo Y, Zhang J. The nitric oxide hypothesis of late preconditioning. Basic Res Cardiol. 1998;93:325–338. doi: 10.1007/s003950050101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Guo Y, Jones WK, Xuan Y-T, Tang X-L, Bao W, Wu W-J, Han H, Laubach VE, Ping P, Yang Z, Qiu Y, Bolli R. The late phase of ischemic preconditioning is abrogated by targeted disruption of the inducible NO synthase gene. Proc Natl Acad Sci USA. 1999;96:11507–11512. doi: 10.1073/pnas.96.20.11507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Xuan Y-T, Guo Y, Zhu Y, Wang O-L, Rokosh G, Bolli R. Endothelial nitric oxide synthase plays an obligatory role in the late phase of ischemic preconditioning by activating the protein kinase C ε-p44/42 mitogen-activated protein kinase-pSer-signal transducers and activators of transcription1/3 pathway. Circulation. 2007;116:535–544. doi: 10.1161/CIRCULATIONAHA.107.689471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.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]
- 77.Costa ADT, Garlid KD, West IC, Lincoln TM, Downey JM, Cohen MV, Critz SD. 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]
- 78.Costa ADT, Garlid KD. Intramitochondrial signaling: interactions among mitoKATP, PKCε, ROS, and MPT. Am J Physiol. 2008;295:H874–H882. doi: 10.1152/ajpheart.01189.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Tanaka M, Fujiwara H, Yamasaki K, Sasayama S. Superoxide dismutase and N-2-mercaptopropionyl glycine attenuate infarct size limitation effect of ischaemic preconditioning in the rabbit. Cardiovasc Res. 1994;28:980–986. doi: 10.1093/cvr/28.7.980. [DOI] [PubMed] [Google Scholar]
- 80.Chen W, Gabel S, Steenbergen C, Murphy E. A redox-based mechanism for cardioprotection induced by ischemic preconditioning in perfused rat heart. Circ Res. 1995;77:424–429. doi: 10.1161/01.res.77.2.424. [DOI] [PubMed] [Google Scholar]
- 81.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]
- 82.Sun J. Protein S-nitrosylation: a role of nitric oxide signaling in cardiac ischemic preconditioning. Acta Physiol Sin. 2007;59:544–552. [PubMed] [Google Scholar]
- 83.Perlman DH, Bauer SM, Ashrafian H, Bryan NS, Garcia-Saura MF, Lim CC, Fernandez BO, Infusini G, McComb ME, Costello CE, Feelisch M. Mechanistic insights into nitrite-induced cardioprotection using an integrated metabolomic/proteomic approach. Circ Res. 2009;104:796–804. doi: 10.1161/CIRCRESAHA.108.187005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hogg N. The biochemistry and physiology of S-nitrosothiols. Annu Rev Pharmacol Toxicol. 2002;42:585–600. doi: 10.1146/annurev.pharmtox.42.092501.104328. [DOI] [PubMed] [Google Scholar]
- 85.Eu JP, Sun J, Xu L, Stamler JS, Meissner G. The skeletal muscle calcium release channel: coupled O2 sensor and NO signaling functions. Cell. 2000;102:499–509. doi: 10.1016/s0092-8674(00)00054-4. [DOI] [PubMed] [Google Scholar]
- 86.Takahashi H, Shin Y, Cho S-J, Zago WM, Nakamura T, Gu Z, Ma Y, Furukawa H, Liddington R, Zhang D, Tong G, Chen H-SV, Lipton SA. Hypoxia enhances S-nitrosylation-mediated NMDA receptor inhibition via a thiol oxygen sensor motif. Neuron. 2007;53:53–64. doi: 10.1016/j.neuron.2006.11.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Galkin A, Moncada S. S-Nitrosation of mitochondrial complex I depends on its structural conformation. J Biol Chem. 2007;282:37448–37453. doi: 10.1074/jbc.M707543200. [DOI] [PubMed] [Google Scholar]
- 88.Das DK, Maulik N. Preconditioning potentiates redox signaling and converts death signal into survival signal. Arch Biochem Biophys. 2003;420:305–311. doi: 10.1016/j.abb.2003.09.023. [DOI] [PubMed] [Google Scholar]
- 89.Saini H, Machackova J, NS D. Role of reactive oxygen species in ischemic preconditioning of subcellular organelles in the heart. Antioxid Redox Signal. 2004;6:393–404. doi: 10.1089/152308604322899468. [DOI] [PubMed] [Google Scholar]
- 90.Liu X, Miller MJS, Joshi MS, Thomas DD, Lancaster JR. Accelerated reaction of nitric oxide with O2 within the hydrophobic interior of biological membranes. Proc Natl Acad Sci USA. 1998;95:2175–2179. doi: 10.1073/pnas.95.5.2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Nedospasov A, Rafikov R, Beda N, Nudler E. An autocatalytic mechanism of protein nitrosylation. Proc Natl Acad Sci USA. 2000;97:13543–13548. doi: 10.1073/pnas.250398197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Foster MW, Liu L, Zeng M, Hess DT, Stamler JS. A genetic analysis of nitrosative stress. Biochemistry. 2009;48:792–799. doi: 10.1021/bi801813n. [DOI] [PubMed] [Google Scholar]
- 93.Koshiishi I, Takajo T, Tsuchida K. Regulation of S-thiolation and S-nitrosylation in the thiol/nitric oxide system by radical scavengers. Nitric Oxide. 2007;16:356–361. doi: 10.1016/j.niox.2006.12.002. [DOI] [PubMed] [Google Scholar]
- 94.Steffen M, Sarkela TM, Gybina AA, Steele TW, Trasseth NJ, Kuehl D, Giulivi C. Metabolism of S-nitrosoglutathione in intact mitochondria. Biochem J. 2001;356:395–402. doi: 10.1042/0264-6021:3560395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Nagasaka Y, Fernandez BO, Garcia-Saura MF, Petersen B, Ichinose F, Bloch KD, Feelisch M, Zapol WM. Brief periods of nitric oxide inhalation protect against myocardial ischemia-reperfusion injury. Anesthesiology. 2008;109:675–682. doi: 10.1097/ALN.0b013e318186316e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Chen SC, Huang B, Liu YC, Shyu KG, Lin PY, Wang DL. Acute hypoxia enhances proteins’ S-nitrosylation in endothelial cells. Biochem Biophys Res Commun. 2008;377:1274–1278. doi: 10.1016/j.bbrc.2008.10.144. [DOI] [PubMed] [Google Scholar]
- 97.Lima B, Forrester MT, Hess DT, Stamler JS. S-nitrosylation and cardiovascular signaling. Circ Res. 2009 doi: 10.1161/CIRCRESAHA.109.207381. In press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Liu L, Yan Y, Zeng M, Zhang J, Hanes MA, Ahearn G, McMahon TJ, Dickfeld T, Marshall HE, Que LG, Stamler JS. Essential roles of S-nitrosothiols in vascular homeostasis and endotoxic shock. Cell. 2004;116:617–628. doi: 10.1016/s0092-8674(04)00131-x. [DOI] [PubMed] [Google Scholar]
- 99.Benhar M, Forrester MT, Hess DT, Stamler JS. Regulated protein denitrosylation by cytosolic and mitochondrial thioredoxins. Science. 2008;320:1050–1054. doi: 10.1126/science.1158265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bateman RL, Rauh D, Tavshanjian B, Shokat KM. Human carbonyl reductase 1 is an S-nitrosoglutathione reductase. J Biol Chem. 2008;283:35756–35762. doi: 10.1074/jbc.M807125200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Okado-Matsumoto A, Fridovich I. Putative denitrosylase activity of Cu,Zn-superoxide dismutase. Free Radic Biol Med. 2007;43:830–836. doi: 10.1016/j.freeradbiomed.2007.05.038. [DOI] [PubMed] [Google Scholar]
- 102.Trujillo M, Alvarez MN, Peluffo G, Freeman BA, Radi R. Xanthine oxidase-mediated decomposition of S-nitrosothiols. J Biol Chem. 1998;273:7828–7834. doi: 10.1074/jbc.273.14.7828. [DOI] [PubMed] [Google Scholar]
- 103.Tannenbaum SR, White FM. Regulation and specificity of S-nitrosylation and denitrosylation. ACS Chem Biol. 2006;1:615–618. doi: 10.1021/cb600439h. [DOI] [PubMed] [Google Scholar]
- 104.Benhar M, Forrester MT, Stamler JS. Protein denitrosylation: enzymatic mechanisms and cellular functions. Nat Rev Mol Cell Biol. 2009;10:721–732. doi: 10.1038/nrm2764. [DOI] [PubMed] [Google Scholar]
- 105.Jaffrey SR, Snyder SH. The biotin switch method for the detection of S-nitrosylated proteins. Sci STKE. 2001;86:PL1. doi: 10.1126/stke.2001.86.pl1. [DOI] [PubMed] [Google Scholar]
- 106.Arrell DK, Elliott ST, Kane LA, Guo Y, Ko YH, Pedersen PL, Robinson J, Murata M, Murphy AM, Marban E, Van Eyk JE. Proteomic analysis of pharmacological preconditioning: novel protein targets converge to mitochondrial metabolism pathways. Circ Res. 2006;99:706–714. doi: 10.1161/01.RES.0000243995.74395.f8. [DOI] [PubMed] [Google Scholar]
- 107.Derakhshan B, Wille PC, Gross SS. Unbiased identification of cysteine S-nitrosylation sites on proteins. Nat Protoc. 2007;2:1685–1691. doi: 10.1038/nprot.2007.210. [DOI] [PubMed] [Google Scholar]
- 108.Pisitkun T, Hoffert JD, Yu M-J, Knepper MA. Tandem mass spectrometry in physiology. Physiology. 2007;22:390–400. doi: 10.1152/physiol.00025.2007. [DOI] [PubMed] [Google Scholar]
- 109.Beer SM, Taylor ER, Brown SE, Dahm CC, Costa NJ, Runswick MJ, Murphy MP. Glutaredoxin 2 catalyzes the reversible oxidation and glutathionylation of mitochondrial membrane thiol proteins: implication for mitochondrial redox regulation and antioxidant defense. J Biol Chem. 2004;279:47939–47951. doi: 10.1074/jbc.M408011200. [DOI] [PubMed] [Google Scholar]
- 110.Burwell LS, Brookes PS. Mitochondria as a target for the cardioprotective effects of nitric oxide in ischemia-reperfusion injury. Antioxid Redox Signal. 2008;10:579–600. doi: 10.1089/ars.2007.1845. [DOI] [PubMed] [Google Scholar]
- 111.Lim S, Clement M-V. Phosphorylation of the survival kinase Akt by superoxide is dependent on an ascorbate-reversible oxidation of PTEN. Free Radic Biol Med. 2007;42:1178–1192. doi: 10.1016/j.freeradbiomed.2007.01.013. [DOI] [PubMed] [Google Scholar]
- 112.Carver DJ, Gaston B, deRonde K, Palmer LA. Akt-mediated activation of HIF-1 in pulmonary vascular endothelial cells by S-nitrosoglutathione. Am J Respir Cell Mol Biol. 2007;37:255–263. doi: 10.1165/rcmb.2006-0289SM. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Cai Z, Semenza GL. PTEN activity is modulated during ischemia and reperfusion: involvement in the induction and decay of preconditioning. Circ Res. 2005;97:1351–1359. doi: 10.1161/01.RES.0000195656.52760.30. [DOI] [PubMed] [Google Scholar]
- 114.Mocanu MM, Yellon DM. PTEN, the Achilles’ heel of myocardial ischaemia/reperfusion injury? Br J Pharmacol. 2007;150:833–838. doi: 10.1038/sj.bjp.0707155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Oudit GY, Penninger JM. Cardiac regulation by phosphoinositide 3-kinases and PTEN. Cardiovasc Res. 2009;82:250–260. doi: 10.1093/cvr/cvp014. [DOI] [PubMed] [Google Scholar]
- 116.Sun J, Steenbergen C, Murphy E. S-nitrosylation: NO-related redox signaling to protect against oxidative stress. Antioxid Redox Signal. 2006;8:1693–1705. doi: 10.1089/ars.2006.8.1693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Martinez-Ruiz A, Lamas S. Signalling by NO-induced protein S-nitrosylation and S-glutathionylation: convergences and divergences. Cardiovasc Res. 2007;75:220–228. doi: 10.1016/j.cardiores.2007.03.016. [DOI] [PubMed] [Google Scholar]
- 118.Baba SP, Wetzelberger K, Hoetker JD, Bhatnagar A. Posttranslational glutathiolation of aldose reductase (AKR1B1): a possible mechanism of protein recovery from S-nitrosylation. Chem Biol Interact. 2009;178:250–258. doi: 10.1016/j.cbi.2008.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Burwell LS, Nadtochiy SM, Tompkins AJ, Young S, Brookes PS. Direct evidence for S-nitrosation of mitochondrial complex I. Biochem J. 2006;394:627–634. doi: 10.1042/BJ20051435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Zhang J, Jin B, Li L, Block E, Patel J. Nitric oxide-induced persistent inhibition and nitrosylation of active site cysteine residues of mitochondrial cytochrome-c oxidase in lung endothelial cells. Am J Physiol. 2005;288:C840–C849. doi: 10.1152/ajpcell.00325.2004. [DOI] [PubMed] [Google Scholar]
- 121.Arstall MA, Bailey C, Gross WL, Bak M, Balligand JL, Kelly RA. Reversible S-nitrosation of creatine kinase by nitric oxide in adult rat ventricular myocytes. J Mol Cell Cardiol. 1998;30:979–988. doi: 10.1006/jmcc.1998.0662. [DOI] [PubMed] [Google Scholar]
- 122.Carnes CA, Janssen PML, Ruehr ML, Nakayama H, Nakayama T, Haase H, Bauer JA, Chung MK, Fearon IM, Gillinov AM, Hamlin RL, Van Wagoner DR. Atrial glutathione content, calcium current and contractility. J Biol Chem. 2007;282:28063–28073. doi: 10.1074/jbc.M704893200. [DOI] [PubMed] [Google Scholar]
- 123.Gonzalez DR, Beigi F, Treuer AV, Hare JM. Deficient ryanodine receptor S-nitrosylation increases sarcoplasmic reticulum calcium leak and arrhythmogenesis in cardiomyocytes. Proc Natl Acad Sci USA. 2007;104:20612–20617. doi: 10.1073/pnas.0706796104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Adachi T, Weisbrod R, Pimentel D, Ying J, Sharov V, Schoneich C, Cohen R. S-Glutathiolation by peroxynitrite activates SERCA during arterial relaxation by nitric oxide. Nat Med. 2004;10:1200–1207. doi: 10.1038/nm1119. [DOI] [PubMed] [Google Scholar]
- 125.Maejima Y, Adachi S, Morikawa K, Ito H, Isobe M. Nitric oxide inhibits myocardial apoptosis by preventing caspase-3 activity via S-nitrosylation. J Mol Cell Cardiol. 2005;38:163–174. doi: 10.1016/j.yjmcc.2004.10.012. [DOI] [PubMed] [Google Scholar]
- 126.Atar S, Ye Y, Lin Y, Freeberg SY, Nishi SP, Rosanio S, Huang M-H, Uretsky BF, Perez-Polo JR, Birnbaum Y. Atorvastatin-induced cardioprotection is mediated by increasing inducible nitric oxide synthase and consequent S-nitrosylation of cyclooxygenase-2. Am J Physiol. 2006;290:H1960–H1968. doi: 10.1152/ajpheart.01137.2005. [DOI] [PubMed] [Google Scholar]
- 127.Selemidis S, Dusting GJ, Peshavariya H, Kemp-Harper BK, Drummond GR. Nitric oxide suppresses NADPH oxidase-dependent superoxide production by S-nitrosylation in human endothelial cells. Cardiovasc Res. 2007;75:349–358. doi: 10.1016/j.cardiores.2007.03.030. [DOI] [PubMed] [Google Scholar]
- 128.Tao L, Gao E, Bryan NS, Qu Y, Liu H-R, Hu A, Christopher TA, Lopez BL, Yodoi J, Koch WJ, Feelisch M, Ma XL. Cardioprotective effects of thioredoxin in myocardial ischemia and reperfusion: role of S-nitrosation. Proc Natl Acad Sci USA. 2004;101:11471–11476. doi: 10.1073/pnas.0402941101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Iwakiri Y, Satoh A, Chatterjee S, Toomre DK, Chalouni CM, Fulton D, Groszmann RJ, Shah VH, Sessa WC. Nitric oxide synthase generates nitric oxide locally to regulate compartmentalized protein S-nitrosylation and protein trafficking. Proc Natl Acad Sci USA. 2006;103:19777–19782. doi: 10.1073/pnas.0605907103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Lowenstein CJ. Nitric oxide regulation of protein trafficking in the cardiovascular system. Cardiovasc Res. 2007;75:240–246. doi: 10.1016/j.cardiores.2007.03.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Wang G, Moniri NH, Ozawa K, Stamler JS, Daaka Y. Nitric oxide regulates endocytosis by S-nitrosylation of dynamin. Proc Natl Acad Sci USA. 2006;103:1295–1300. doi: 10.1073/pnas.0508354103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kang-Decker N, Cao S, Chatterjee S, Yao J, Egan LJ, Semela D, Mukhopadhyay D, Shah V. Nitric oxide promotes endothelial cell survival signaling through S-nitrosylation and activation of dynamin-2. J Cell Sci. 2007;120:492–501. doi: 10.1242/jcs.03361. [DOI] [PubMed] [Google Scholar]
- 133.Matsushita K, Morrell C, Cambien B, Yang S, Yamakuchi M, Bao C, Hara M, Quick R, Cao W, O'Rourke B, Lowenstein J, Pevsner J, Wagner D, Lowenstein C. Nitric oxide regulates exocytosis by S-nitrosylation of N-ethylmaleimide-sensitive factor. Cell. 2003;115:139–150. doi: 10.1016/s0092-8674(03)00803-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Whalen EJ, Foster MW, Matsumoto A, Ozawa K, Violin JD, Que LG, Nelson CD, Benhar M, Keys JR, Rockman HA, Koch WJ, Daaka Y, Lefkowitz RJ, Stamler JS. Regulation of β-adrenergic receptor signaling by S-nitrosylation of G-protein-coupled receptor kinase 2. Cell. 2007;129:511–522. doi: 10.1016/j.cell.2007.02.046. [DOI] [PubMed] [Google Scholar]
- 135.Ozawa K, Whalen EJ, Nelson CD, Mu Y, Hess DT, Lefkowitz RJ, Stamler JS. S-nitrosylation of β-arrestin regulates β-adrenergic receptor trafficking. Mol Cell. 2008;31:395–405. doi: 10.1016/j.molcel.2008.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.del Monte F, Lebeche D, Guerrero JL, Tsuji T, Doye AA, Gwathmey JK, Hajjar RJ. Abrogation of ventricular arrhythmias in a model of ischemia and reperfusion by targeting myocardial calcium cycling. Proc Natl Acad Sci USA. 2004;101:5622–5627. doi: 10.1073/pnas.0305778101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Murphy E, Steenbergen C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol Rev. 2008;88:581–609. doi: 10.1152/physrev.00024.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Hausenloy D, Ong S-B, Yellon D. 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]
- 139.Dalle-Donne I, Milzani A, Gagliano N, Colombo R, Giustarini D, Rossi R. Molecular mechanisms and potential clinical significance of S-glutathionylation. Antioxid Redox Signal. 2008;10:445–474. doi: 10.1089/ars.2007.1716. [DOI] [PubMed] [Google Scholar]
- 140.Chen Y-R, Chen C-L, Pfeiffer DR, Zweier JL. Mitochondrial complex II in the post-ischemic heart: oxidative injury and the role of protein S-glutathionylation. J Biol Chem. 2007;282:32640–32654. doi: 10.1074/jbc.M702294200. [DOI] [PubMed] [Google Scholar]
- 141.Hare JM, Beigi F, Tziomalos K. Nitric oxide and cardiobiology: methods for intact hearts and isolated myocytes. Methods Enzymol. 2008;441:369–392. doi: 10.1016/S0076-6879(08)01221-4. [DOI] [PubMed] [Google Scholar]
- 142.Whiteman M, Chua YL, Zhang D, Duan W, Liou Y-C, Armstrong JS. Nitric oxide protects against mitochondrial permeabilization induced by glutathione depletion: role of S-nitrosylation? Biochem Biophys Res Commun. 2006;339:255–262. doi: 10.1016/j.bbrc.2005.10.200. [DOI] [PubMed] [Google Scholar]
- 143.Chen Y-Y, Chu H-M, Pan K-T, Teng C-H, Wang D-L, Wang AHJ, Khoo K-H, Meng T-C. Cysteine S-nitrosylation protects protein-tyrosine phosphatase 1B against oxidation-induced permanent inactivation. J Biol Chem. 2008;283:35265–35272. doi: 10.1074/jbc.M805287200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Haendeler J, Hoffmann J, Tischler V, Berk B, Zeiher A, Dimmeler S. Redox regulatory and anti-apoptotic functions of thioredoxin depend on S-nitrosylation at cysteine 69. Nat Cell Biol. 2002;4:743–749. doi: 10.1038/ncb851. [DOI] [PubMed] [Google Scholar]
- 145.Haendeler J, Hoffmann J, Zeiher A, Dimmeler S. Antioxidant effects of statins via S-nitrosylation and activation of thioredoxin in endothelial cells: a novel vasculoprotective function of statins. Circulation. 2004;110:856–861. doi: 10.1161/01.CIR.0000138743.09012.93. [DOI] [PubMed] [Google Scholar]
- 146.Hashemy SI, Holmgren A. Regulation of the catalytic activity and structure of human thioredoxin 1 via oxidation and S-nitrosylation of cysteine residues. J Biol Chem. 2008;283:21890–21898. doi: 10.1074/jbc.M801047200. [DOI] [PubMed] [Google Scholar]
- 147.Hashemy SI, Johansson C, Berndt C, Lillig CH, Holmgren A. Oxidation and S-nitrosylation of cysteines in human cytosolic and mitochondrial glutaredoxins: effects on structure and activity. J Biol Chem. 2007;282:14428–14436. doi: 10.1074/jbc.M700927200. [DOI] [PubMed] [Google Scholar]
- 148.Fang J, Nakamura T, Cho D-H, Gu Z, Lipton SA. S-nitrosylation of peroxiredoxin 2 promotes oxidative stress-induced neuronal cell death in Parkinson's disease. Proc Natl Acad Sci USA. 2007;104:18742–18747. doi: 10.1073/pnas.0705904104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Saraiva RM, Minhas KM, Zheng M, Pitz E, Treuer A, Gonzalez D, Schuleri KH, Vandegaer KM, Barouch LA, Hare JM. Reduced neuronal nitric oxide synthase expression contributes to cardiac oxidative stress and nitroso-redox imbalance in ob/ob mice. Nitric Oxide. 2007;16:331–338. doi: 10.1016/j.niox.2006.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Hara M, Agrawal N, Kim S, Cascio M, Fujimuro M, Ozeki Y, Takahashi M, Cheah J, Tankou S, Hester L, Ferris C, Hayward S, Snyder S, Sawa A. S-nitrosylated GAPDH initiates apoptotic cell death by nuclear translocation following Siah1 binding. Nat Cell Biol. 2005;7:665–674. doi: 10.1038/ncb1268. [DOI] [PubMed] [Google Scholar]
- 151.Sen N, Hara M, Shafique Ahmad A, Cascio M, Kamiya A, Ehmsen J, Aggrawal N, Hester L, Dore S, Snyder S, Sawa A. GOSPEL: a neuroprotective protein that binds to GAPDH upon S-nitrosylation. Neuron. 2009;63:81–91. doi: 10.1016/j.neuron.2009.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Foster D, Van Eyk J, Marbán E, O'Rourke B. Redox signaling and protein phosphorylation in mitochondria: progress and prospects. J Bioenerg Biomembr. 2009;41:159–168. doi: 10.1007/s10863-009-9217-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Nadtochiy SM, Burwell LS, Brookes PS. Cardioprotection and mitochondrial S-nitrosation: effects of S-nitroso-2-mercaptopropionyl glycine (SNO-MPG) in cardiac ischemia-reperfusion injury. J Mol Cell Cardiol. 2007;42:812–825. doi: 10.1016/j.yjmcc.2007.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Asanuma H, Nakai K, Sanada S, Minamino T, Takashima S, Ogita H, Fujita M, Hirata A, Wakeno M, Takahama H, Kim J, Asakura M, Sakuma I, Kitabatake A, Hori M, Komamura K, Kitakaze M. S-nitrosylated and pegylated hemoglobin, a newly developed artificial oxygen carrier, exerts cardioprotection against ischemic hearts. J Mol Cell Cardiol. 2007;42:924–930. doi: 10.1016/j.yjmcc.2006.12.001. [DOI] [PubMed] [Google Scholar]
- 155.Hogg N, Broniowska KA, Novalija J, Kettenhofen NJ, Novalija E. Role of S-nitrosothiol transport in the cardioprotective effects of S-nitrosocysteine in rat hearts. Free Radic Biol Med. 2007;43:1086–1094. doi: 10.1016/j.freeradbiomed.2007.06.016. [DOI] [PubMed] [Google Scholar]