Abstract
Importance of the field
Ischemia-reperfusion (IR) injury is a common clinical problem after transplantation as well as myocardial infarction and stroke. IR initiates an inflammatory response leading to rapid tissue damage. Adenosine, produced in response to IR, is generally considered as a protective signaling molecule and elicits its physiological responses through four distinct adenosine receptors. The short half-life, lack of specificity, and rapid metabolism limits the use of adenosine as a therapeutic agent. Thus intense research efforts have focused on the synthesis and implementation of specific adenosine receptor agonists and antagonists as potential therapeutic agents for a variety of inflammatory conditions including IR injury.
Areas covered by this review
This review summarizes current knowledge on IR injury with a focus on lung, heart, and kidney, and examines studies that have advanced our understanding of the role of adenosine receptors and the therapeutic potential of adenosine receptor agonists and antagonists for the prevention of IR injury.
What the reader will gain
The reader will gain insight into the role of adenosine receptor signaling in IR injury.
Take home message
No clinical therapies are currently available that specifically target IR injury; however, targeting of specific adenosine receptors may offer therapeutic strategies in this regard.
Keywords: inflammation, innate immunity, therapeutic targets, transplantation, preconditioning
Introduction
Ischemia is defined as a lack of blood supply to an organ or tissue, resulting in oxygen deprivation of cells. Without restoration of oxygenated blood, ischemic tissues soon undergo necrosis and die. Thus, restoration of blood flow, or reperfusion, is the definitive treatment for ischemia. Although ischemia itself is a serious condition, reperfusion of ischemic tissue paradoxically imparts further tissue injury, especially after prolonged ischemia. This injury, defined as reperfusion injury, has widespread clinical relevance and is encountered in a variety of surgical settings (e.g. transplantation, cardiopulmonary bypass and aneurysm repair) as well as non-surgical settings (e.g. myocardial infarction, stroke, hemorrhage, trauma and shock). The sum total of tissue injury imposed by ischemia and reperfusion is defined as ischemia-reperfusion (IR) injury. IR injury therefore involves a complex cascade of events including oxidative stress, inflammation, and interactions between many cell types. In the transplant setting, IR injury is unavoidable, and, in addition to short term effects on organ function, can have long-term effects on allograft survival [1]. If severe enough, IR injury can also lead to systemic inflammatory effects, multiorgan dysfunction and death [2].
Currently, no therapeutic agents are clinically available specifically for the prevention or treatment of IR injury. The single established strategy to limit IR injury is early reperfusion of the ischemic tissue. Despite a multitude of successful experimental interventions to reduce IR injury, very few have been successfully translated into clinical practice. Various pharmacological and non-pharmacological therapies may help reduce IR injury. For example, non-pharmacological, surgical strategies which may limit IR injury in patients include ischemic preconditioning, postconditioning, and remote preconditioning [3, 4]. Improvements in hypothermic preservation solutions have been instrumental in reducing early graft failure. Several pharmacological preconditioning agents have been investigated such as nicorandil (K-ATP channel opener), adenosine, sodium-hydrogen exchange inhibitors, and statins. Adenosine is known to play an intimate role in preconditioning and postconditioning [5–7]. Other pharmacological strategies which have shown little to no success in clinical trials include antioxidants (e.g. allopurinol), anti-inflammatory drugs (e.g. methylprednisolone), and vasodilators (e.g. prostaglandins, dopamine) [3, 8]. However, one example of a potentially promising agent has been the use of cyclosporine A to inhibit mitochondrial permeability transition pore (mPTP) opening [9].
In a variety of experimental settings and animal models, including models of IR, adenosine has been described as a retaliatory metabolite which serves as a protective agent with anti-inflammatory properties [10]. Adenosine is a purine nucleoside produced intracellularly and extracellularly during high stress conditions such as inflammation and IR [11]. Signaling through four known receptors, adenosine can have multiple effects depending on which receptors are activated and on which cells these receptors reside. Significant progress has been made in our understanding of the multifaceted role of adenosine signaling in tissue IR and inflammation, and new approaches and strategies are now being uncovered as a means to target adenosine receptor signaling as a therapeutic avenue to curtail tissue injury and inflammation. This review summarizes current knowledge on IR injury with a focus on the lung, heart, and kidney; and highlights studies that have advanced our understanding of the role of adenosine receptors and the therapeutic potential of adenosine receptor agonists and antagonists in the prevention of IR injury.
1.1 Ischemia-reperfusion injury
Within moments of reperfusion of ischemic tissue, reactive oxygen species (ROS) are generated (e.g. superoxide, peroxynitrite, hydrogen peroxide, and hydroxyl radical) [12, 13]. ROS generation contributes to a rapid inflammatory response via direct oxidative injury to cells and stimulation of proinflammatory mediators such as cytokines, chemokines and cell adhesion molecules. This inflammatory cascade is a key mediator of IR injury, and considerable studies have now established that an innate immune response is a significant component of this cascade. Reperfusion also entails complement activation and the release of cleavage fragments known as anaphylatoxins which are potent inflammatory mediators [14]. Microvascular dysfunction characterizes the “no-reflow” phenomenon, which refers to the failure of blood to reperfuse a previously ischemic tissue at the microvascular level even after blood flow has been restored at the macrovascular level. Mechanisms contributing to the no-reflow phenomenon include platelet-leukocyte aggregation, leukocyte-endothelial adhesion, reduced vasorelaxation capacity, and accumulation of interstitial fluid [15].
Circulating and resident leukocytes as well as tissue resident cells contribute to the immune response to IR injury. Orchestration of immune cell responses after IR has been made possible with the application of multi-color flow cytometry and the use of genetically modified mice, before which quantitative analysis relied primarily on immunohistochemical labeling of small samples of tissue. For example, a comprehensive leukocyte profile following kidney IR has been described using multi-color flow cytometry [16]. Tissue resident dendritic cells, neutrophils, macrophages, lymphocytes and platelets have been shown to contribute to IR injury [17]. Neutrophils, which are the major leukocyte population to rapidly infiltrate tissues following IR, are attracted by chemokines including MCP-1 (CCL2), KC (CXCL1), RANTES (CCL5), and IL-17. Infiltrating neutrophils can impose significant tissue injury through ROS generation and further release of cytokines and proteases. Damaged vascular endothelium provides an anchoring site for neutrophils via upregulation of adhesion molecules such as P-selectin, L-selectin and intercellular adhesion molecule (ICAM) [18–20]. Monocytes, macrophages and dendritic cells are heterogeneous and contribute to early and late phases of IR injury [21, 22]. Recent studies have also documented that rapid activation and infiltration of T cells also mediate IR injury [23]. Depletion of neutrophils [24], macrophages [25], or T cells [26] have been shown to attenuate IR injury. Thus IR-induced inflammation entails a potent innate immune response involving the activation and infiltration of multiple leukocyte populations including neutrophils, macrophages, dendritic cells and T cells.
In addition to leukocytes, resident, tissue (non-leukocyte) cells are also involved in the pathogenesis of IR injury. For example, cardiac myocytes release IL-6 to contribute to myocardial dysfunction after IR [27], and alveolar epithelial cells contribute to pulmonary IR injury via release of chemokines such as KC and MIP-2 [28]. Thus, a variety of key cellular and molecular targets exist for which therapeutic interventions could be designed to attenuate the onset of inflammation initiated by IR.
1.2. Ischemic conditioning
Different organs have the ability to be preconditioned by a nonlethal period of ischemia, rendering the target organ refractory to further ischemia-induced dysfunction (ischemic preconditioning) such as the small intestine [29], liver [30], brain [31], lung [32], and kidney [33]. Mechanisms and pathways that are also involved in IR injury (described above in section 1.1), which are attenuated by activation of adenosine receptors, are important targets for ischemic preconditioning and postconditioning [34]. For example, many studies are now showing that preconditioning and postconditioning may involve similar signaling pathways upon reperfusion of the ischemic heart such as the reperfusion injury salvage kinase (RISK) pathway, redox signaling, and mPTP [35–37]. Adenosine is largely thought to serve as a mediator of ischemic preconditioning [38]. Similar to preconditioning, ischemic postconditioning was first described in 2003 and involves the interruption of reperfusion by intermittent, brief episodes of ischemia before extended reperfusion in order to provide protection [39]. Various studies suggest that the protection mediated by postconditioning is also mediated in part by adenosine receptor signaling [40]. Finally, another concept, called remote conditioning, is defined as brief ischemia in an organ or tissue which provides a protective effect in a distant or remote organ which undergoes the primary ischemic insult. This brief ischemia can occur either before or after the primary ischemic event to be termed remote pre- or post-conditioning, and there is evidence that adenosine receptor signaling is also involved in remote conditioning [40–42]. Thus pre- and postconditioning as well as remote conditioning all appear to invoke similar protective pathways which may involve adenosine receptor signaling. It is important to note here that there is a distinct difference between the potential role of endogenous adenosine and the “preconditioning-like” protection afforded by the administration of adenosine either before or after ischemia.
1.3 Adenosine and adenosine receptors
Adenosine is a nucleoside locally released in response to cellular stress such as injury, IR, and inflammation [10]. Extracellular adenosine largely accumulates from the breakdown of adenine nucleotides that are liberated from damaged cells. These nucleotides are rapidly converted to adenosine by a family of nucleotidases including CD39 and CD73 [11]. In addition, two cytosolic enzymes important in the removal of adenosine from tissues, adenosine deaminase and adenosine kinase, will rapidly metabolize adenosine when extracellular adenosine traverses the cell membrane via the equilabrative nucleoside transporters (ENT) according to the concentration gradient of adenosine. Deficiency of adenosine deaminase results in adenosine levels which can increase dramatically in some tissues [43]. This mechanism is thought to contribute importantly to the pathophysiology underlying one form of severe combined immunodeficiency: adenosine deaminase (ADA) deficiency.
Adenosine exerts its effects through binding to any of four G protein-coupled adenosine receptors designated as A1R, A2AR, A2BR and A3R [44]. These receptors reside on a wide variety of cell types to control, either by inhibiting or stimulating, adenylyl cyclase and subsequently modulating intracellular levels of cyclic AMP (cAMP). A1R and A3R signaling is linked to inhibitory G proteins to down-regulate cAMP whereas A2AR and A2BR signaling is linked to stimulatory G proteins to up-regulate cAMP with subsequent transcriptional activation of cAMP-regulated genes (Figure 1). In addition to these classical pathways, adenosine receptor signaling can also activate other pathways involving phospholipase C, mitogen activated protein kinase (MAPK), phosphatidylinositol 3-kinase (PI3K), or Akt pathways to influence gene expression [45] (summarized in Figure 1). The wide variety of physiological effects of adenosine, which may include not only protective, but also deleterious effects, depend upon many factors including: 1) differences in receptor affinities for adenosine, 2) extracellular concentration of adenosine, 3) expression of receptors among various cell types, 4) differences in receptor densities on cell membranes, and 5) duration of adenosine exposure.
Figure 1. Adenosine receptor signaling pathways.
A2AR and A2BR activation increase cyclic AMP (cAMP) levels with subsequent activation of the PKA → CREB pathway to increase transcription of cAMP-regulated genes, while A1R and A3R activation decrease cAMP levels. In addition to mediating gene expression via cAMP, adenosine receptor activation can also influence gene expression via the PI3K → Akt/PBK or PLC → MAPK pathways, among others not shown here. CREB, cAMP response element binding protein; MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase; PLC, phospholipase C; PK, protein kinase.
Adenosine has the highest affinity for A1R and A2AR, an intermediate affinity for A3R, and the lowest affinity for A2BR [46, 47]. Adenosine concentrations under basal conditions are sufficient to activate A1R, A2AR and A3R. On the other hand, A2BR requires higher adenosine concentrations to be significantly activated, concentrations typically attained after tissue injury. The expression of adenosine receptors are under dynamic regulation and can be up- or down-regulated depending on the cell type and nature of the physiological stress. For example, A2AR expression can increase under inflammatory conditions [48], and the A2BR expression can increase during hypoxic and ischemic conditions [49].
Research into the role of adenosine receptor signaling in organ injury and disease has undergone rapid and significant progress due to the development of several critical tools. First, potent and selective synthetic agonists and antagonists for each adenosine receptor have been developed to enable researchers to specifically test the role of each receptor in almost any model [50]. Second, gene knockout (−/−) mice with targeted deletions of each adenosine receptor have now been generated and have been important in exploring the physiological and pathophysiological roles that these receptors play. The utilization of these agonists/antagonists and knockout mice in various studies of IR injury are discussed below.
2. Adenosine receptors in lung IR injury
All four adenosine receptors are expressed in lungs of mice [51] and humans [52]. Adenosine is well recognized as exhibiting protective effects in various models of IR injury [53], and a study by Hsu et al. was one of the first to demonstrate this for the lung [54]. Most studies into lung IR injury have focused on the A2AR or A3R, where activation of either is protective. The roles of A1R and A2BR remain poorly understood.
2.1 A1R in lung IR injury
Studies on the role of A1R in lung IR injury have been conflicting as the A1R has been shown to have both pro- and anti-inflammatory effects [55]. Activation of A1R in a variety of cell types can produce proinflammatory effects. For example, in neutrophils, A1R activation induces chemotaxis, adherence to endothelium, and generation of ROS [56]. Although A1R activation has been shown to be protective after IR in the heart [57] and kidney [58], an early study by Neely and Keith concluded that A1R antagonists (XAC and DPCPX) block IR injury in the lung [59]. More recent studies, however, have suggested that the A1R may be protective. Using a blood-perfused rabbit lung model, it was recently shown that activation of A1R (via CCPA) during reperfusion attenuates lung IR injury [60]. Yildiz et al. evaluated the role of adenosine in ischemic preconditioning using an in vivo rat lung IR model [61]. Here, preconditioning in the form of adenosine infusion or a brief ischemic exposures prior to sustained ischemia prevented injury, and, importantly, these protective effects were abolished by DPCPX. This suggests that preconditioning entails adenosine signaling via the A1R. Furthermore, studies with A1R−/− mice suggest that A1R activation is protective in a model of sepsis [62], and it has recently been shown that the A1R on hematopoietic cells mediates (reduces) transendothelial and transepithelial migration in a model of LPS-induced lung injury [63]. In contrast, blockade of A1R was found to prevent injury in a feline model of LPS-induced lung injury [64]. Thus the role of A1R in lung IR injury and other inflammatory conditions remains controversial, which suggests that the A1R has a more complex role than previously realized. This complexity likely arises from a variety of differences among these studies including differences in organ and tissue models, A1R signaling pathways among cell/tissue types, and A1R expression among cell types within an organ or between species.
2.2 A2AR in lung IR injury
A rich history of studies provides overwhelming evidence that A2AR is the major adenosine receptor mediating anti-inflammatory properties of adenosine [55]. An early study by Khimenko et al. was the first to demonstrate that selective activation of A2AR is protective in the setting of lung IR [65]. Here, treatment of isolated rat lungs with an A2AR agonist, CGS-21680, reversed endothelial damage after IR. There have been a series of more recent supportive studies as well. Sharma et al. demonstrated that ATL-313, an A2AR agonist [66], prevented lung IR injury in wild-type mice and that ATL-313 had no protective effect in A2AR−/− mice [67]. In rabbit lungs, it was also shown that ATL-313 has an additive protective effect when administered both prior to ischemia as well as during reperfusion, an effect that was abolished by the A2AR antagonist, ZM241385 [68]. There is now evidence that the protective effect of ATL-313 in lung IR injury is largely derived through the activation of A2ARs on CD4+ T lymphocytes and neutrophils [69]. In a more mechanistic study, Rivo et al. concluded that A2AR-mediated protection is associated with decreased apoptosis and involves ERK1/2 activation and modulation of Bcl-2 and Bax expression [70].
In an effort to move beyond small animal models toward more clinically relevant models, the protective effects of A2AR activation was recently tested in a porcine lung transplantation model in which the lungs were ischemic for six hours prior to transplant [71]. In this study, recipient pigs were administered ATL146e, an A2AR agonist, intravenously for three hours after lung transplantation. This study demonstrated that ATL146e attenuated lung injury and inflammation and preserved pulmonary function after transplantation. Collectively, these studies, and others, suggest that specific A2AR activation may be a promising therapeutic target for the prevention of IR injury and primary graft function in lung transplant patients.
2.3 A2BR in lung IR injury
The A2BR is expressed on a wide variety of cells including most inflammatory cells and, in the lung, are highly expressed on alveolar epithelial cells [72]. Expression of A2BR is induced under stresses such as injury, oxidative stress, or by TNF-α [73]. As with other adenosine receptors, both anti- and pro-inflammatory activities have been attributed to A2BR activation. However, the fact that the A2BR has relatively low affinity for adenosine suggests that it may serve a more prominent role in pathological conditions in which high levels of adenosine are achieved.
The role of A2BR in lung IR injury has not been well studied. A recent study by Anvari et al. showed that lung IR injury is attenuated in A2BR−/− mice, suggesting that A2BR is proinflammatory in this setting [74]. Also in this study, use of chimeric mice demonstrated that the proinflammatory effects of A2BR were attributed to A2BR signaling on resident pulmonary cells and not bone-marrow-derived cells. A proinflammatory role of A2BR has been reflected in several other studies as well [75–77]. On the other hand, some studies suggest an anti-inflammatory role for A2BR in different settings of acute lung injury. For example, Schingnitz et al. demonstrated a protective role of A2BR signaling in endotoxin-mediated lung injury, and use of bone marrow chimeric mice demonstrated that this is largely mediated via A2BR signaling in pulmonary cells, suggesting that A2BR agonists may be potentially useful for the treatment of endotoxin-induced lung injury [78]. These seemingly contradictory studies of A2BR may be due its various signaling partners in different tissues and whether the signal in the system used originates from bone marrow cells or from tissues. Continued efforts to better define the role of A2BR in lung IR injury will be critical to the development of adenosine receptor agonist/antagonist-based therapeutics.
2.4 A3R in lung IR injury
The A3R is widely expressed with the highest levels found in the lung and liver. The role of A3R in inflammatory conditions, including IR injury, is ambiguous. Although there is evidence for both protective and detrimental effects, most studies suggest that A3R activation is protective in lung IR. Using a feline model, Rivo et al. found that IB-MECA, an A3R agonist, administered before ischemia or during reperfusion, attenuated injury and apoptosis [79]. These protective effects of IB-MECA were blocked by MRS-1191, an A3R antagonist. This same group has also provided evidence that the protective effects of A3R activation may be mediated in part through upregulation of phosphorylated ERK [80] as well as inhibition of p38 MAPK [81]. In addition to IR injury, activation of A3R was shown to serve an anti-inflammatory function in a bleomycin-induced model of pulmonary inflammation and fibrosis [82]. In LPS-induced pulmonary inflammation, a recent study by Wagner et al. demonstrated that administration of the A3R agonist, CI-IB-MECA, attenuated microvascular permeability and neutrophil migration in lungs after LPS exposure, suggesting that pharmacologic modulation of A3R signaling may be a promising strategy for lung inflammation [83]. This study also found that CI-IB-MECA required A3Rs on both hematopoietic and nonhematopoietic cells to reduce transmigration in vivo. Thus, overall, it appears that the most promising therapeutic strategies to prevent lung IR injury may involve the use of A2AR and/or A3R agonists in transplant recipients. It will likely be important for these agonists to be very specific so as to not engage potentially proinflammatory adenosine receptors such as A1R or A2BR.
3. Adenosine receptors in kidney IR injury
Extracellular adenosine has a critical role in mediating physiological properties in the kidney [84–86]. Adenosine constricts afferent arterioles, thereby reducing filtered sodium loading and controlling metabolic work of the kidney, vasodilation, and NaCl transport in the distal nephron. These vascular and epithelial effects control metabolic function in the kidney. In addition, adenosine has an important effect on the regulation of inflammatory processes in the kidney. Early studies using a nonselective adenosine receptor antagonist, theophylline, attenuated the reduction in renal blood flow and glomerular filtration rateobserved after IR [87, 88]. In contrast, intravenous infusion of adenosine protected kidneys from IR injury. Kidney IR in mice deficient in CD39 or CD73, enzymes necessary for adenosine production, resulted in enhanced injury, highlighting the protective effects of adenosine and a critical role for adenosine-generating enzymes [89, 90]. These reports in which adenosine receptor antagonists and agonists reduced injury are likely explained by the nonselective effect of these compounds, and the use of more selective pharmacological reagents in recent studies have helped to shed more light onto the mechanism of action.
3.1 A1R in kidney IR injury
In rodents, A1R activation protected kidneys from IR injury [91, 92]. In these studies, animals treated with an A1R agonist before renal ischemia showed reduced IR injury. Further support of the role of A1Rs in mediating protection comes from studies in which A1R−/− mice showed increased kidney IR injury [58]. Thus activation of A1Rs appears to serve a protective role against renal IR injury. This protective effect could be due to: 1) over-expression of A1Rs in proximal tubule cells leading to reduced renal proximal tubule cell necrosis and apoptosis via upregulation of heat shock protein 27 [93, 94] and 2) enhanced A1R-mediated vasoconstriction with subsequent induction of hypoxia-inducible factors (HIFs) and HIF-dependent, protective genes [93]. In other models of acute kidney injury (AKI); however, A1R antagonism leads to renal protection. In cisplatin-induced AKI, tonapofylline, an A1R antagonist, reduced injury [95]. In radiocontrast-induced AKI, A1R−/− mice or wild-type mice treated with a selective A1R antagonist, DPCPX, were protected from kidney injury [96]. In both of these models, vasoconstriction contributed to injury, and A1R blockade was thought to reduce vasoconstriction.
3.2 A2AR in kidney IR injury
The use of A2AR-specific agonists and antagonists has revealed in numerous studies that A2AR signaling is protective in renal IR injury (reviewed in [97]). Since the initial observation that A2AR activation ameliorates IR injury in rat kidneys [98], significant advances have been made in characterizing the mechanism and target of action for A2AR. ATL146e, an A2AR agonist, markedly reduced the damaging effects of IR when infusion was initiated prior to, or at the time of ischemia [99]. Dose-dependent studies in mice indicated that infusion of 1–10 ng/kg/min of ATL146e was the most effective dose [99] and that this protective dose was not due to changes in systemic blood pressure [98]. When mice were subjected to kidney IR and treated with rolipram, a PDE IV inhibitor, or ATL146e during IR, delivery of either drug was associated with reduced renal injury in a dose-dependent manner, and maximal (additive) protection was observed when both compounds were administered together [99]. It was then demonstrated that the protective mechanism of ATL146e was, at least in part, mediated through neutrophils, where ATL146e significantly reduced neutrophil accumulation in renal tissues after IR [99]. Using high resolution microscopy combined with flow cytometry, it was also demonstrated that A2AR agonists reduced transmigration of neutrophils from the intravascular space to the interstitium as well as decreased vascular permeability [100]. Additionally, the enhanced expression of adhesion molecules P-selectin and ICAM-1 following IR was reduced by A2AR agonists [20]. These observations are consistent with the idea that A2AR agonists limit IR injury due to an inhibitory effect on neutrophil adhesion.
Given the ubiquitous expression of A2ARs, the precise target of A2AR agonist action in mediating tissue protection in vivo has only recently been described. Using bone marrow chimeric mice, it was determined that the protective effect of A2AR agonists following IR was due to A2AR signaling on bone marrow-derived cells [101]. Subsequent studies have now demonstrated that activation of A2ARs on CD4+ T cells, and not macrophages, mediates tissue protection [97].
3.3 A2BR and A3R in kidney IR injury
Few studies have examined the role of A2BR or A3R in kidney IR injury or AKI. In one recent study, renal protection by ischemic preconditioning was abolished in A2BR−/− mice and in wild-type mice administered PSB1115, an A2BR antagonist [102]. In contrast, protection by ischemic preconditioning was observed in A3R−/− mice [102]. Using chimeric mice, this study also demonstrated a critical role of renal A2BR (as opposed to A2BR on hematopoietic cells) in mediating the protective effects of ischemic preconditioning.
Lee et al. demonstrated that an A3R agonist worsened kidney injury while an antagonist improved renal function after IR in mice [103]. In accordance, A3R−/− mice displayed significant functionaland morphological renal protection from IR injury [103]. A study by Lee et al. also demonstrated that A3R activation prior to renal IR worsens injury whereas A3R antagonism protects renal function [92]. On the other hand, it has been shown that A3R activation confers significant protection from renal injury in murine septic peritonitis [104]. Thus further studies will be required to understand the roles of A2BR and A3R in kidney IR injury.
4. Adenosine receptors in cardiac IR injury
Gene expression of all four adenosine receptors have been detected in the rat heart, including cultured cardiomyocytes [105] and cardiac fibroblasts [106]. These results are consistent with the pharmacologic studies undertaken in many species over many years, which for the most part support a functionally significant role for each adenosine receptor in the heart [107]. Adenosine itself has long been known to exert cardioprotective effects in various models of IR injury [53, 108], effects that are traditionally understood in terms of adenosine triggering a preconditioned state through A1R stimulation [109]. However, cardioprotective roles for the A2AR [110] and A3R [111] were demonstrated not long thereafter. More recent studies have even suggested a cardioprotective role for the A2BR [112]. Thus the cardioprotective effects of A1R signaling are better understood and characterized than A2AR and A3R signaling, and until recently, little was known about the cardioprotective potential of the A2BR.
4.1 A1R in cardiac IR injury
The phenomenon of ischemic preconditioning was first discovered in the heart [113], and the role of adenosine in mediating the effects of preconditioning via A1R activation was discovered soon after [6]. It is now well established that activation of not only the A1R, but also other G-protein coupled receptors, can induce preconditioning. Indeed, opioids and bradykinin can stimulate signal transduction pathways which ultimately protect the mitochondria by inhibiting the opening of the mitochondrial permeability transition pore (mPTP), thereby protecting mitochondria from lysis due to calcium overload and salvaging cardiomyocytes from cell death. The downstream signal transduction mechanisms and end effectors mediating ischemic preconditioning have been the subject of intensive study for many years (reviewed in [114–116]). In addition to the many elegant pharmacologic studies conducted in a wide variety of animal models, the central importance of the A1R in preconditioning has also been demonstrated in transgenic mouse lines that overexpress A1R via a cardiac-specific promoter [57, 117]. Nevertheless, it is important to note that classic ischemic preconditioning (and preconditioning mimetics such as adenosine, bradykinin and opioids) need to be applied in advance of the index event and are thought to protect cardiomyocytes against the effects of prolonged ischemia and/or calcium overload upon reperfusion. Thus while preconditioning induces a very effective and reproducible form of cardioprotection, the clinical utility is limited to applications in which the ischemic event can be predicted (such as cardiac transplantation). An alternative or supplemental strategy for cardioprotection is to limit myocellular death by minimizing the added damage imposed by calcium overload and inflammation during reperfusion. These approaches may be used in a wider range of clinical applications since they may be effective even when treatment is delayed until the time of reperfusion.
4.2 A2AR in cardiac IR injury
Soon after the discovery of preconditioning, it was reported that the intracoronary administration of adenosine during reperfusion could also substantially reduce infarct size [118]. In contrast to the direct effects of preconditioning on cardiomyocytes, the cardioprotective effects of adenosine during reperfusion were primarily attributed to decreased neutrophil infiltration in the ischemic zone and preservation of endothelial structure in the subendocardium. Interestingly, the same group later reported that the salutary effect of adenosine on infarct size was lost if treatment was delayed by only three hours post-reperfusion [119]. Subsequent work using more selective adenosine receptor agonists established that this form of cardioprotection was mediated primarily by the A2AR [120, 121]. The ability of selective A2AR agonists to reduce infarct size when administered at, shortly before, or during reperfusion has since been confirmed in studies of a wide variety of animal models including pigs [122], dogs [123, 124], rabbits [125], rats [126] and mice. Of note, the cardioprotection afforded by selective A2AR agonists is abrogated in A2AR−/− mice [127], as well as lymphocyte-deficient Rag-1−/− mice reconstituted with CD4+ T cells from A2AR−/− mice, thus implicating CD4+ T cells in the pathophysiology of IR injury [128].
The timing of administration for A2AR agonists during reperfusion makes them far more suitable than A1R agonists for clinical applications such as direct percutaneous intervention for acute myocardial infarct. As such, the timing of reperfusion therapy shares much in common with the more recently discovered phenomenon of postconditioning [39]. Similar to preconditioning, postconditioning involves a series of coronary occlusions and reperfusions, but the series of ischemic episodes is imposed at the end (rather than the beginning) of the coronary occlusion. It remains to be definitively established whether postconditioning functions primarily by reducing ischemic injury to cardiomyocytes or by reducing the additional injury imposed by reperfusion. However, the efficacy of postconditioning in reducing neutrophil infiltration and preserving endothelial function is well established [129], and the salutary effect of postconditioning on infarct size is retained in A1R−/− mice whereas it is greatly attenuated in A2AR−/− mice [130]. Thus, it would appear that preconditioning and A1R agonists protect cardiomyocytes against ischemic injury, while postconditioning and A2AR agonists function primarily by inhibiting inflammation and thus attenuating reperfusion injury to the heart.
4.3 A2BR in cardiac IR injury
Similar to the A2AR and its agonists, the A2BR has also been implicated in ischemic postconditioning [112], and its agonists have been shown to reduce infarct size when administered during the first hour of reperfusion in rabbits [131]. Differences in animal models and experimental preparations (Langendorff vs. open-chest) led to healthy debate regarding the relative importance of the two receptors in reducing infarct size when their agonists were administered during reperfusion. However, a recent study found that both A2AR and A2BR activation were required to achieve maximal protection against IR injury in rats [132], and this observation was supported by an independent laboratory using an open-chest murine model of IR injury [133]. The mechanisms underlying the additive effect of combined A2AR and A2BR stimulation have yet to be fully defined, but the signal transduction pathways implicated in A2BR-mediated cardioprotection involve cardiomyocytes, whereas the inflammatory system has been more strongly implicated in A2AR-mediated cardioprotection, suggesting that the two-pronged strategy targets both ischemic and reperfusion injury.
4.4 A3R in cardiac IR injury
The A2AR and A2BR are primarily coupled through the Gs G-protein coupled receptors, whereas the A1R and A3R are Gi-coupled [53]. Thus one might expect A3R activation to share aspects with A1R activation, which exerts only modest cardioprotection and then principally during ischemia [134]. However, unlike selective A1R agonists [135], selective A3R agonists have been shown to trigger cardioprotection not only when applied prior to ischemia, but also when administered at reperfusion [136]. When studied in isolated rat cardiomyocytes subjected to simulated ischemia/reoxygenation injury, A3R activation exerted anti-apoptotic and anti-necrotic effects when administered at reoxygenation [137]. Another proposed mechanism by which A3R reduces infarct size is by attenuating neutrophil activation and infiltration. For example, Jordan et al. have shown that A3R activation at reperfusion attenuates neutrophil-mediated reperfusion injury [111]. Thus, therapeutic strategies targeting A3R activation may also be promising for the reduction of cardiac reperfusion injury. It remains to be seen whether the combination of A2AR, A2BR and A3R stimulation during reperfusion offers any additional cardioprotection over the combination of A2AR and A2BR in pre-clinical models of myocardial infarction. Even so, the relative tissue abundance (and perhaps even the primary function) of the four adenosine receptors may vary from species to species, and more work will be needed to identify the most potent combination of adenosine receptor agonists (or mixed agonists) for reducing infarct size in humans.
5. Adenosine receptors in liver IR injury
All four adenosine receptors are expressed in hepatocytes, where they stimulate the rates of glycogenolysis, gluconeogenesis, and ureagenesis [138]. One of the first reports of the protective effects of adenosine in the liver after IR was a study by Satoru et al. which showed that augmentation of endogenous adenosine by inhibition of nucleoside transport attenuated liver IR injury in dogs [139].
5.1 A1R in liver IR injury
Most studies, but not all, suggest that A1R activation is protective in the setting of liver IR. The protective effects of ozone oxidative preconditioning have been attributed to A1R activation [140], and, in another study, A1R activation was found to induce hepatic delayed preconditioning while A1R antagonist blocked preconditioning [141]. Although A1R−/− mice and wild-type mice treated with an A1R antagonist developed worse liver IR injury, the A1R agonist, CCPA, failed to protect against injury [142], suggesting that the endogenous A1R is protective. On the other hand, administration of A1R antagonists have been shown to elicit beneficial effects against hepatic IR injury in a canine model [143] and a pig model of non-heart beating donor liver transplantation [144]. Thus, as with other organs, the role of A1R in liver IR injury remains unclear.
5.2 A2AR in liver IR injury
As with the lung, liver, and heart, ample evidence exists which demonstrates that A2AR activation confers protective and anti-inflammatory effects against liver IR injury. An early study by Harada et al. demonstrated that selective A2AR agonists, YT-146 and CGS-21680C, reduce liver IR injury in rats by inhibiting leukocyte activation [145]. Several more recent studies have now determined that the protective effects of A2AR against liver IR injury are due to the inhibition of NKT cell activation and chemokine production [146–148].
5.3 A2BR and A3R in liver IR injury
The roles for A2BR and A3R in liver IR injury remain obscure. To date, no studies have examined the A2BR and very few have evaluated the A3R. Guinzberg et al. [149] have provided evidence for a protective role for A3R in the liver by evaluating the role of A3R in the responses of hepatocytes to inosine, a purine nucleoside formed by adenosine deamination which has been shown to bind to A3R to promote its effects [150]. Guinzberg et al. also showed that, in perfused rat liver as well as isolated hepatocytes, hypoxia-reoxygenation stimulated the release of inosine, adenosine, and glucose; effects which were attenuated by an A3R antagonist [149]. They concluded that after ischemia, inosine contributes to the maintenance of homeostasis by releasing glucose from the liver through A3R activation. Further studies will be required to better define the roles for A2BR and A3R in liver IR injury.
6. Adenosine receptors in IR injury of other tissues
Literature on the role of adenosine receptors in IR injury of tissues other than lung, kidney, heart and liver is much less extensive. An A2AR agonist was shown to improve islet cell transplant outcome by attenuating innate immune responses [151]. Most studies of ischemic brain injury conclude that A2AR antagonists and adenosine appear to be neuroprotective, depending on the nature of the brain injury and associated pathology [152, 153]. The mechanisms by which ischemic brain injury is modulated by A2AR are poorly understood. Several studies have demonstrated that A2AR agonists are protective in the setting of spinal cord IR [154, 155]. In addition, Miyazaki et al. demonstrated the involvement of A1R activation in the neuroprotective effects of adenosine in spinal cord ventral horn [156]. In models of intestinal IR injury, agonists for both A1R [157] and A2BR [158] have been shown to provide protection.
7. Conclusion
The most extensively studied adenosine receptors in the setting of IR injury have been the A1R and A2AR. Nearly all studies indicate that A2AR activation results in potent, protective, anti-inflammatory effects. This is likely the primary mechanism responsible for adenosine-mediated protection, at least in acute inflammatory conditions. However, controversy remains regarding the role of other adenosine receptors, where both pro- and anti-inflammatory effects have been observed. Potent and selective adenosine receptor agonists and antagonists have been, and will remain, important tools to help define these roles. While the anti-inflammatory properties of A2AR activation are well characterized in multiple tissue systems, it appears that the roles of other adenosine receptors in IR injury may be more complex and tissue-dependent, which is likely due to multifaceted patterns of expression and signaling pathways for these receptors. However, there is now building and significant evidence which support protective effects of A1R and A3R in IR injury. Further studies will be necessary to decipher the roles for these receptors in various settings of IR injury.
8. Expert Opinion
A general conceptual framework for understanding IR injury is presented in Figure 2, which employs data derived from a murine model of myocardial infarction as an example [159]. Figure 2 illustrates the concept that the experimentally-defined relationship between infarct size and duration of ischemia (the sigmoidal curve) is actually the sum total of two separate components: ischemic injury (gray region) and reperfusion injury (white area between the sigmoidal curve and the gray region). In this scheme, the gray area representing ischemic injury is hypothetical and could differ in shape and/or size. Nevertheless, ischemic injury clearly dominates in the setting of extended or long-term occlusion and certainly disappears if the period of occlusion is extremely brief. A major component of reperfusion injury is inflammatory injury, although other mechanisms are also known to contribute to reperfusion injury. Figure 2 illustrates the point that the efficacy of agents that focus on reperfusion injury, such as A2AR agonists, is predicted to wane with increasing durations of ischemia. Conversely, therapies that minimize both ischemic injury and reperfusion injury will be needed to achieve maximal tissue protection over the widest range of ischemic events. It should be noted that the slope of the line defining ischemic injury almost certainly varies depending on the tissue undergoing ischemia. Thus heart and brain would likely exhibit steeper slopes than other tissues (e.g., liver, lung and kidney) where metabolic demand is far lower and reperfusion injury may become the predominant mode of injury.
Figure 2. Conceptual framework for separating reperfusion injury from ischemic injury.

As shown, the final infarct size indicated by the empirically-determined sigmoidal line represents the sum total of the ischemic damage to the tissue and the additional injury imposed by reperfusion, a major component of which is inflammation. The white area directly underneath the sigmoidal curve represents reperfusion injury, while the shape and size of the gray area is hypothetical due to difficulties in determining it experimentally. Adopted with permission from Xu et al. [159] and used with permission from the American Physiological Society.
Currently, no therapeutic strategies are available that specifically target IR injury. Perhaps strategies aimed toward augmenting adenosine-mediated protective effects could provide such a therapy. The development and use of selective adenosine receptor agonists and antagonists have been instrumental in understanding the role of adenosine receptors in IR injury. More importantly, these agents, and newer generation agents with greater selectivity and potency, will be key to the development of future adenosine receptor-targeted therapies to prevent IR injury. In light of current research, the most promising therapeutic strategies will likely involve the administration of selective A1R, A2AR, or A3R agonists to patients immediately upon organ transplantation, or as quickly as possible after episodes of IR such as myocardial infarct or trauma to minimize injury imposed by inflammation.
As pointed out in the introduction, despite many experimental studies which suggest a beneficial effect of anti-inflammatory agents in preventing IR injury, most anti-inflammatory therapies, at least in the heart [8] and kidney [160], which have been tested in the clinical setting have failed. Possible reasons for these negative results are nicely discussed in a recent review by Dirksen et al. [8] and may involve differences in the methodology and biology between experimental and clinical studies. This could reflect an incomplete understanding of the intricacies of IR injury. Indeed, IR injury is a very complex process, involving more than just inflammation, and it is quite possible that a single intervention cannot be expected to completely prevent IR injury. However, experimental anti-inflammatory studies are progressing, and novel anti-inflammatory strategies continue to be explored such as neutralization of proinflammatory cytokines, prevention of cytokine secretion, administration of anti-inflammatory cytokines, and cell-based therapies [161].
The application of A2AR agonists toward treatments for inflammation has been discussed in detail by Lappas and colleagues [162]. One advantage of using an A2AR agonist is that minimal side effects have been reported in animals. For example, there are no adverse side effects of ATL146e treatment, due to its short half life (minutes), with the exception of transient hypotension following administration at high doses. However, at much lower doses, which effectively block inflammation in IR studies (1–50 ng/kg/min), there are no hemodynamic effects [71, 99]. Thus A2AR agonists show promise in future clinical studies directed towards the treatment and prevention of IR injury.
The role of adenosine receptors other than A2AR in IR injury and whether agonists or antagonists for these receptors can be used to attenuate injury remains unclear and will likely vary depending on the organ/species affected. Overall, studies suggest a protective role for A1R and A3R agonists in IR injury, although A3R antagonists appear to attenuate kidney injury. The role of A2BR in modulating inflammatory responses to injury is confounding, complex, and likely tissue-specific. Animal studies of the A2BR have been hindered by the limited availability of well-characterized, selective agonists. In addition, A2BR−/− mice have only recently been generated [163]. Further investigations will be required to better understand the diverse A2BR-mediated signaling pathways in various cell types after IR.
An advantage to utilizing adenosine receptor agonists to attenuate IR injury is that an acute treatment would likely be sufficient and efficacious; possibly in the range of hours rather than days. This therapeutic window is based upon many studies in both animals and humans that have documented acute inflammation that begins immediately upon reperfusion that is initiated by rapid activation and infiltration of leukocytes. Blocking this inflammatory cascade appears to be sufficient to reduce acute graft dysfunction after transplant or cardiac dysfunction after infarct. Because IR injury to single organs leads to systemic inflammation and multiorgan dysfunction, compounds such as adenosine receptor agonists may not only benefit the ischemic organ but may also attenuate dysfunction of distal organs.
A continued push to test the efficacy of adenosine receptor agonists in humans to ameliorate IR injury may be a worthwhile pursuit. One caveat to keep in mind for this strategy is that different adenosine receptors can result in different effects, and in fact, activation of some receptors, especially under high adenosine levels, may even be detrimental under certain conditions. Thus, the specificity of adenosine receptor agonists will be paramount so as to not engage other, potentially detrimental receptors. In conclusion, the development of small molecule therapies to prevent IR injury is sorely needed, and may now be getting nearer to reality due to the many pre-clinical studies that continue to advance our understanding of the actions of adenosine receptor agonists in various models. Such a therapy will not only help the patient acutely after IR, but will also have a significant impact on organ survival in the long-term.
Article highlights.
Ischemia-reperfusion injury involves a complex cascade of events including oxidative stress, inflammation, and interactions between many cell types.
Adenosine is a retaliatory, protective metabolite locally released in response to cellular stress such as ischemia-reperfusion and inflammation.
Adenosine exerts its effects through binding to any of four G protein-coupled adenosine receptors designated as A1R, A2AR, A2BR and A3R.
Adenosine receptor signaling has a wide variety of physiological effects, often protective, depending on receptor type activation, cell/tissue type, and physiologic conditions.
In the lung, A1R and A2AR have been shown to be protective, A2BR may be detrimental, and the role of A1R remains controversial.
In the kidney, A1R and A2AR have been shown to be protective, whereas the roles of A2BR and A3R are less understood.
In the heart, A1R is cardioprotective via preconditioning effects, A2AR, and A3R are also protective, and there is building evidence that A2BR may be protective.
In the liver, A1R and A2AR have been shown to be protective, whereas the roles of A2BR and A3R remain obscure.
Less extensive studies suggest that adenosine receptor signaling, especially A2AR, is protective in other tissues such as islet cell transplant, brain, spinal cord, and intestine.
Adenosine receptor agonists may be a promising therapeutic strategy against ischemia-reperfusion injury, and continued push to test the efficacy of adenosine receptor agonists in humans to treat ischemia-reperfusion injury is likely a worthwhile pursuit.
Acknowledgments
We thank Ashish Sharma, Zequan Yang, Y.J. Day, Li Li, Liping Huang, and Hong Ye (University of Virginia) who have provided much of the scientific data for this review. We also thank Drs. Joel Linden (La Jolla Institute for Allergy & Immunology) and Diane Rosin (University of Virginia) for helpful discussions.
Abbreviations
- −/−
gene knockout
- ADA
adenosine deaminase
- AKI
acute kidney injury
- cAMP
cyclic AMP
- ENT
equilabrative nucleoside transporter
- GPCR
G-protein coupled receptor
- HIF
hypoxia-inducible factor
- ICAM
intercellular adhesion molecule
- IR
ischemia reperfusion
- MAPK
mitogen activated protein kinase
- MODS
multiple organ dysfunction syndrome
- mPTP
mitochondrial permeability transition pore
- ROS
reactive oxygen species
- SIRS
systemic inflammatory response syndrome
Footnotes
Declaration of interest
V Laubach is sponsored by NIH/NHLBI grants R01HL077301 and R01HL092953 and receives research support from the Roche Organ Transplant Research Foundation, Meggen, Switzerland. B French is sponsored by NIH/NHLBI grant R01HL092305, AHA grant 09GRNT2261123 and receives research support from the AstraZeneca/UVA Strategic Alliance.
M Okusa is sponsored by NIH/NIDDK grants RO1DK56223 and RO1DK62324 and received research support from Genzyme (Genzyme Renal Innovations Program) and PGx Health (previously, Adenosine Therapeutics, LLC).
Bibliography
- 1.Fiser SM, Tribble CG, Long SM, et al. Ischemia-reperfusion injury after lung transplantation increases risk of late bronchiolitis obliterans syndrome. Ann Thorac Surg. 2002;73:1041–7. doi: 10.1016/s0003-4975(01)03606-2. [DOI] [PubMed] [Google Scholar]
- 2.Neary P, Redmond HP. Ischaemia-reperfusion injury and the systemic inflammatory response syndrome. In: Grace PA, Mathie RT, editors. Ischaemia-Reperfusion Injury. Oxford: Blackwell Science; 1999. pp. 123–36. [Google Scholar]
- 3.Bahde R, Spiegel HU. Hepatic ischaemia-reperfusion injury from bench to bedside. Br J Surg. 2010;97:1461–75. doi: 10.1002/bjs.7176. [DOI] [PubMed] [Google Scholar]
- 4.Kharbanda RK. Cardiac conditioning: a review of evolving strategies to reduce ischaemia-reperfusion injury. Heart. 2010;96:1179–86. doi: 10.1136/hrt.2009.179101. [DOI] [PubMed] [Google Scholar]
- 5.Kin H, Zatta AJ, Lofye MT, et al. Postconditioning reduces infarct size via adenosine receptor activation by endogenous adenosine. Cardiovasc Res. 2005;67:124–33. doi: 10.1016/j.cardiores.2005.02.015. [DOI] [PubMed] [Google Scholar]
- 6.Liu GS, Thornton J, Van Winkle DM, Stanley AW, Olsson RA, Downey JM. Protection against infarction afforded by preconditioning is mediated by A1 adenosine receptors in rabbit heart. Circulation. 1991;84:350–6. doi: 10.1161/01.cir.84.1.350. [DOI] [PubMed] [Google Scholar]
- 7.Yang XM, 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]
- 8.Dirksen MT, Laarman GJ, Simoons ML, Duncker DJ. Reperfusion injury in humans: a review of clinical trials on reperfusion injury inhibitory strategies. Cardiovasc Res. 2007;74:343–55. doi: 10.1016/j.cardiores.2007.01.014. [DOI] [PubMed] [Google Scholar]
- 9.Piot C, Croisille P, Staat P, et al. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med. 2008;359:473–81. doi: 10.1056/NEJMoa071142. [DOI] [PubMed] [Google Scholar]
- 10.Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 2007;14:1315–23. doi: 10.1038/sj.cdd.4402132. [DOI] [PubMed] [Google Scholar]
- 11.Zimmermann H. Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedebergs Arch Pharmacol. 2000;362:299–309. doi: 10.1007/s002100000309. [DOI] [PubMed] [Google Scholar]
- 12.Becker LB. New concepts in reactive oxygen species and cardiovascular reperfusion physiology. Cardiovasc Res. 2004;61:461–70. doi: 10.1016/j.cardiores.2003.10.025. [DOI] [PubMed] [Google Scholar]
- 13.Li C, Jackson RM. Reactive species mechanisms of cellular hypoxia-reoxygenation injury. Am J Physiol Cell Physiol. 2002;282:C227–41. doi: 10.1152/ajpcell.00112.2001. [DOI] [PubMed] [Google Scholar]
- 14.Collard CD, Lekowski R, Jordan JE, Agah A, Stahl GL. Complement activation following oxidative stress. Mol Immunol. 1999;36:941–8. doi: 10.1016/s0161-5890(99)00116-9. [DOI] [PubMed] [Google Scholar]
- 15.Rezkalla SH, Kloner RA. No-reflow phenomenon. Circulation. 2002;105:656–62. doi: 10.1161/hc0502.102867. [DOI] [PubMed] [Google Scholar]
- 16.Li L, Huang L, Sung SS, et al. The chemokine receptors CCR2 and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury. Kidney Int. 2008;74:1526–37. doi: 10.1038/ki.2008.500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kaczorowski DJ, Tsung A, Billiar TR. Innate immune mechanisms in ischemia/reperfusion. Front Biosci (Elite Ed) 2009;1:91–8. doi: 10.2741/E10. [DOI] [PubMed] [Google Scholar]
- 18.Farhood A, McGuire GM, Manning AM, Miyasaka M, Smith CW, Jaeschke H. Intercellular adhesion molecule 1 (ICAM-1) expression and its role in neutrophil-induced ischemia-reperfusion injury in rat liver. J Leukoc Biol. 1995;57:368–74. [PubMed] [Google Scholar]
- 19.Lefer AM, Weyrich AS, Buerke M. Role of selectins, a new family of adhesion molecules, in ischaemia-reperfusion injury. Cardiovasc Res. 1994;28:289–94. doi: 10.1093/cvr/28.3.289. [DOI] [PubMed] [Google Scholar]
- 20.Okusa MD, Linden J, Huang L, JMR, Macdonald TL, Huynh LP. A2a-Adenosine receptor mediated inhibition of renal injury and neutrophil adhesion. Am J Physiol. 2000;279:F809–18. doi: 10.1152/ajprenal.2000.279.5.F809. [DOI] [PubMed] [Google Scholar]
- 21.Li L, Okusa MD. Macrophages, dendritic cells, and kidney ischemia-reperfusion injury. Semin Nephrol. 2010;30:268–77. doi: 10.1016/j.semnephrol.2010.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ysebaert DK, De Greef KE, Vercauteren SR, et al. Identification and kinetics of leukocytes after severe ischaemia/reperfusion renal injury. Nephrol Dial Transplant. 2000;15:1562–74. doi: 10.1093/ndt/15.10.1562. [DOI] [PubMed] [Google Scholar]
- 23.Linfert D, Chowdhry T, Rabb H. Lymphocytes and ischemia-reperfusion injury. Transplant Rev (Orlando) 2009;23:1–10. doi: 10.1016/j.trre.2008.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Breda MA, Drinkwater DC, Laks H, et al. Prevention of reperfusion injury in the neonatal heart with leukocyte-depleted blood. J Thorac Cardiovasc Surg. 1989;97:654–65. [PubMed] [Google Scholar]
- 25.Zhao M, Fernandez LG, Doctor A, et al. Alveolar macrophage activation is a key initiation signal for acute lung ischemia-reperfusion injury. Am J Physiol Lung Cell Mol Physiol. 2006;291:L1018–26. doi: 10.1152/ajplung.00086.2006. [DOI] [PubMed] [Google Scholar]
- 26.Yokota N, Daniels F, Crosson J, Rabb H. Protective effect of T cell depletion in murine renal ischemia-reperfusion injury. Transplantation. 2002;74:759–63. doi: 10.1097/00007890-200209270-00005. [DOI] [PubMed] [Google Scholar]
- 27.Yamauchi-Takihara K, Ihara Y, Ogata A, Yoshizaki K, Azuma J, Kishimoto T. Hypoxic stress induces cardiac myocyte-derived interleukin-6. Circulation. 1995;91:1520–4. doi: 10.1161/01.cir.91.5.1520. [DOI] [PubMed] [Google Scholar]
- 28.Sharma AK, Fernandez LG, Awad AS, Kron IL, Laubach VE. Proinflammatory response of alveolar epithelial cells is enhanced by alveolar macrophage-produced TNF-alpha during pulmonary ischemia-reperfusion injury. Am J Physiol Lung Cell Mol Physiol. 2007;293:L105–13. doi: 10.1152/ajplung.00470.2006. [DOI] [PubMed] [Google Scholar]
- 29.Osborne DL, Aw TY, Cepinskas G, Kvietys PR. Development of ischemia/reperfusion tolerance in the rat small intestine. An epithelium-independent event. J Clin Invest. 1994;94:1910–8. doi: 10.1172/JCI117541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Peralta C, Hotter G, Closa D, Gelpi E, Bulbena O, Rosello-Catafau J. Protective effect of preconditioning on the injury associated to hepatic ischemia-reperfusion in the rat: role of nitric oxide and adenosine. Hepatology. 1997;25:934–7. doi: 10.1002/hep.510250424. [DOI] [PubMed] [Google Scholar]
- 31.Heurteaux C, Lauritzen I, Widmann C, Lazdunski M. Essential role of adenosine, adenosine A1 receptors, and ATP-sensitive K+ channels in cerebral ischemic preconditioning. Proc Natl Acad Sci U S A. 1995;92:4666–70. doi: 10.1073/pnas.92.10.4666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Du ZY, Hicks M, Winlaw D, Spratt P, Macdonald P. Ischemic preconditioning enhances donor lung preservation in the rat. J Heart Lung Transplant. 1996;15:1258–67. [PubMed] [Google Scholar]
- 33.Islam CF, Mathie RT, Dinneen MD, Kiely EA, Peters AM, Grace PA. Ischaemia-reperfusion injury in the rat kidney: the effect of preconditioning. Br J Urol. 1997;79:842–7. doi: 10.1046/j.1464-410x.1997.00209.x. [DOI] [PubMed] [Google Scholar]
- 34.Hausenloy DJ, Yellon DM. Preconditioning and postconditioning: united at reperfusion. Pharmacol Ther. 2007;116:173–91. doi: 10.1016/j.pharmthera.2007.06.005. [DOI] [PubMed] [Google Scholar]
- 35.Hausenloy DJ, Yellon DM. Reperfusion injury salvage kinase signalling: taking a RISK for cardioprotection. Heart Fail Rev. 2007;12:217–34. doi: 10.1007/s10741-007-9026-1. [DOI] [PubMed] [Google Scholar]
- 36.Hausenloy DJ, Yellon DM. Time to take myocardial reperfusion injury seriously. N Engl J Med. 2008;359:518–20. doi: 10.1056/NEJMe0803746. [DOI] [PubMed] [Google Scholar]
- 37.Morin D, Assaly R, Paradis S, Berdeaux A. Inhibition of mitochondrial membrane permeability as a putative pharmacological target for cardioprotection. Curr Med Chem. 2009;16:4382–98. doi: 10.2174/092986709789712871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Downey JM, Liu GS, Thornton JD. Adenosine and the anti-infarct effects of preconditioning. Cardiovasc Res. 1993;27:3–8. doi: 10.1093/cvr/27.1.3. [DOI] [PubMed] [Google Scholar]
- 39.Zhao ZQ, Corvera JS, Halkos ME, et al. Inhibition of myocardial injury by ischemic postconditioning during reperfusion: comparison with ischemic preconditioning. Am J Physiol-Heart Circul Physiol. 2003;285:H579–88. doi: 10.1152/ajpheart.01064.2002. [DOI] [PubMed] [Google Scholar]
- 40.Kaur S, Jaggi AS, Singh N. Molecular aspects of ischaemic postconditioning. Fundam Clin Pharmacol. 2009;23:521–36. doi: 10.1111/j.1472-8206.2009.00733.x. [DOI] [PubMed] [Google Scholar]
- 41.Kerendi F, Kin H, Halkos ME, et al. Remote postconditioning. Brief renal ischemia and reperfusion applied before coronary artery reperfusion reduces myocardial infarct size via endogenous activation of adenosine receptors. Basic Res Cardiol. 2005;100:404–12. doi: 10.1007/s00395-005-0539-2. [DOI] [PubMed] [Google Scholar]
- 42.Tapuria N, Kumar Y, Habib MM, Abu Amara M, Seifalian AM, Davidson BR. Remote ischemic preconditioning: a novel protective method from ischemia reperfusion injury--a review. J Surg Res. 2008;150:304–30. doi: 10.1016/j.jss.2007.12.747. [DOI] [PubMed] [Google Scholar]
- 43.Hershfield MS. New insights into adenosine-receptor-mediated immunosuppression and the role of adenosine in causing the immunodeficiency associated with adenosine deaminase deficiency. Eur J Immunol. 2005;35:25–30. doi: 10.1002/eji.200425738. [DOI] [PubMed] [Google Scholar]
- 44.Fredholm BB, Jzerman AP, Jacobson KA, Klotz KN, Linden J. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001;53:527–52. [PMC free article] [PubMed] [Google Scholar]
- 45.Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008;7:759–70. doi: 10.1038/nrd2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fredholm BB, Irenius E, Kull B, Schulte G. Comparison of the potency of adenosine as an agonist at human adenosine receptors expressed in Chinese hamster ovary cells. Biochem Pharmacol. 2001;61:443–8. doi: 10.1016/s0006-2952(00)00570-0. [DOI] [PubMed] [Google Scholar]
- 47.Londos C, Wolff J, Cooper DMF. Adenosine receptors and adenylate cyclase interactions. In: Berne RM, Rall TW, Rubio R, editors. Regulatory Function of Adenosine. Boston: Martinus Nijhoff; 1982. pp. 17–32. [Google Scholar]
- 48.Thiel M, Caldwell CC, Sitkovsky MV. The critical role of adenosine A2A receptors in downregulation of inflammation and immunity in the pathogenesis of infectious diseases. Microbes Infect. 2003;5:515–26. doi: 10.1016/s1286-4579(03)00068-6. [DOI] [PubMed] [Google Scholar]
- 49.Eltzschig HK, Ibla JC, Furuta GT, et al. Coordinated adenine nucleotide phosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role of ectonucleotidases and adenosine A2B receptors. J Exp Med. 2003;198:783–96. doi: 10.1084/jem.20030891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat Rev Drug Discov. 2006;5:247–64. doi: 10.1038/nrd1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fan M, Qin W, Mustafa SJ. Characterization of adenosine receptor(s) involved in adenosine-induced bronchoconstriction in an allergic mouse model. Am J Physiol Lung Cell Mol Physiol. 2003;284:L1012–9. doi: 10.1152/ajplung.00353.2002. [DOI] [PubMed] [Google Scholar]
- 52.Salvatore CA, Jacobson MA, Taylor HE, Linden J, Johnson RG. Molecular cloning and characterization of the human A3 adenosine receptor. Proc Natl Acad Sci U S A. 1993;90:10365–9. doi: 10.1073/pnas.90.21.10365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Linden J. Molecular approach to adenosine receptors: receptor-mediated mechanisms of tissue protection. Annu Rev Pharmacol Toxicol. 2001;41:775–87. doi: 10.1146/annurev.pharmtox.41.1.775. [DOI] [PubMed] [Google Scholar]
- 54.Hsu K, Wang D, Wu SY, Shen CY, Chen HI. Ischemia-reperfusion lung injury attenuated by ATP-MgCl2 in rats. J Appl Physiol. 1994;76:545–52. doi: 10.1152/jappl.1994.76.2.545. [DOI] [PubMed] [Google Scholar]
- 55.Blackburn MR, Vance CO, Morschl E, Wilson CN. Adenosine receptors and inflammation. Handb Exp Pharmacol. 2009:215–69. doi: 10.1007/978-3-540-89615-9_8. [DOI] [PubMed] [Google Scholar]
- 56.Cronstein BN, Daguma L, Nichols D, Hutchison AJ, Williams M. The adenosine/neutrophil paradox resolved: human neutrophils possess both A1 and A2 receptors that promote chemotaxis and inhibit O2 generation, respectively. J Clin Invest. 1990;85:1150–7. doi: 10.1172/JCI114547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Yang Z, Cerniway RJ, Byford AM, Berr SS, French BA, Matherne GP. Cardiac overexpression of A1-adenosine receptor protects intact mice against myocardial infarction. Am J Physiol Heart Circ Physiol. 2002;282:H949–55. doi: 10.1152/ajpheart.00741.2001. [DOI] [PubMed] [Google Scholar]
- 58.Lee HT, Xu H, Nasr SH, Schnermann J, Emala CW. A1 adenosine receptor knockout mice exhibit increased renal injury following ischemia and reperfusion. Am J Physiol Renal Physiol. 2004;286:F298–306. doi: 10.1152/ajprenal.00185.2003. [DOI] [PubMed] [Google Scholar]
- 59.Neely CF, Keith IM. A1 adenosine receptor antagonists block ischemia-reperfusion injury of the lung. Am J Physiol. 1995;268:L1036–46. doi: 10.1152/ajplung.1995.268.6.L1036. [DOI] [PubMed] [Google Scholar]
- 60.Gazoni LM, Walters DM, Unger EB, Linden J, Kron IL, Laubach VE. Activation of A1, A2A, or A3 adenosine receptors attenuates lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2010;140:440–6. doi: 10.1016/j.jtcvs.2010.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Yildiz G, Demiryurek AT, Gumusel B, Lippton H. Ischemic preconditioning modulates ischemia-reperfusion injury in the rat lung: role of adenosine receptors. Eur J Pharmacol. 2007;556:144–50. doi: 10.1016/j.ejphar.2006.11.008. [DOI] [PubMed] [Google Scholar]
- 62.Gallos G, Ruyle TD, Emala CW, Lee HT. A1 adenosine receptor knockout mice exhibit increased mortality, renal dysfunction, and hepatic injury in murine septic peritonitis. Am J Physiol Renal Physiol. 2005;289:F369–76. doi: 10.1152/ajprenal.00470.2004. [DOI] [PubMed] [Google Scholar]
- 63.Ngamsri KC, Wagner R, Vollmer I, Stark S, Reutershan J. Adenosine receptor A1 regulates polymorphonuclear cell trafficking and microvascular permeability in lipopolysaccharide-induced lung injury. J Immunol. 2010;185:4374–84. doi: 10.4049/jimmunol.1000433. [DOI] [PubMed] [Google Scholar]
- 64.Neely CF, Jin J, Keith IM. A1-adenosine receptor antagonists block endotoxin-induced lung injury. Am J Physiol. 1997;272:L353–61. doi: 10.1152/ajplung.1997.272.2.L353. [DOI] [PubMed] [Google Scholar]
- 65.Khimenko PL, Moore TM, Hill LW, et al. Adenosine A2 receptors reverse ischemia-reperfusion lung injury independent of beta-receptors. J Appl Physiol. 1995;78:990–6. doi: 10.1152/jappl.1995.78.3.990. [DOI] [PubMed] [Google Scholar]
- 66.Rieger JM, Brown ML, Sullivan GW, Linden J, Macdonald TL. Design, synthesis, and evaluation of novel A2A adenosine receptor agonists. J Med Chem. 2001;44:531–9. doi: 10.1021/jm0003642. [DOI] [PubMed] [Google Scholar]
- 67.Sharma AK, Linden J, Kron IL, Laubach VE. Protection from pulmonary ischemia-reperfusion injury by adenosine A2A receptor activation. Respir Res. 2009;10:58. doi: 10.1186/1465-9921-10-58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gazoni LM, Laubach VE, Mulloy DP, et al. Additive protection against lung ischemia-reperfusion injury by adenosine A2A receptor activation before procurement and during reperfusion. J Thorac Cardiovasc Surg. 2008;135:156–65. doi: 10.1016/j.jtcvs.2007.08.041. [DOI] [PubMed] [Google Scholar]
- 69.Sharma AK, Laubach VE, Ramos SI, et al. Adenosine A2A receptor activation on CD4+ T lymphocytes and neutrophils attenuates lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2010;139:474–82. doi: 10.1016/j.jtcvs.2009.08.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Rivo J, Zeira E, Galun E, Einav S, Linden J, Matot I. Attenuation of reperfusion lung injury and apoptosis by A2A adenosine receptor activation is associated with modulation of Bcl-2 and Bax expression and activation of extracellular signal-regulated kinases. Shock. 2007;27:266–73. doi: 10.1097/01.shk.0000235137.13152.44. [DOI] [PubMed] [Google Scholar]
- 71.Reece TB, Ellman PI, Maxey TS, et al. Adenosine A2A receptor activation reduces inflammation and preserves pulmonary function in an in vivo model of lung transplantation. J Thorac Cardiovasc Surg. 2005;129:1137–43. doi: 10.1016/j.jtcvs.2004.11.042. [DOI] [PubMed] [Google Scholar]
- 72.Cagnina RE, Ramos SI, Marshall MA, Wang G, Frazier CR, Linden J. Adenosine A2B receptors are highly expressed on murine type II alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol. 2009;297:L467–74. doi: 10.1152/ajplung.90553.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.St Hilaire C, Koupenova M, Carroll SH, Smith BD, Ravid K. TNF-alpha upregulates the A2B adenosine receptor gene: The role of NAD(P)H oxidase 4. Biochem Biophys Res Commun. 2008;375:292–6. doi: 10.1016/j.bbrc.2008.07.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Anvari F, Sharma AK, Fernandez LG, et al. Tissue-derived proinflammatory effect of adenosine A2B receptor in lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2010;140:871–7. doi: 10.1016/j.jtcvs.2010.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Kolachala VL, Vijay-Kumar M, Dalmasso G, et al. A2B adenosine receptor gene deletion attenuates murine colitis. Gastroenterology. 2008;135:861–70. doi: 10.1053/j.gastro.2008.05.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhong H, Belardinelli L, Maa T, Zeng D. Synergy between A2B adenosine receptors and hypoxia in activating human lung fibroblasts. Am J Respir Cell Mol Biol. 2005;32:2–8. doi: 10.1165/rcmb.2004-0103OC. [DOI] [PubMed] [Google Scholar]
- 77.Zhong H, Wu Y, Belardinelli L, Zeng D. A2B adenosine receptors induce IL-19 from bronchial epithelial cells, resulting in TNF-alpha increase. Am J Respir Cell Mol Biol. 2006;35:587–92. doi: 10.1165/rcmb.2005-0476OC. [DOI] [PubMed] [Google Scholar]
- 78.Schingnitz U, Hartmann K, Macmanus CF, et al. Signaling through the A2B adenosine receptor dampens endotoxin-induced acute lung injury. J Immunol. 2010;184:5271–9. doi: 10.4049/jimmunol.0903035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rivo J, Zeira E, Galun E, Matot I. Activation of A3 adenosine receptor provides lung protection against ischemia-reperfusion injury associated with reduction in apoptosis. Am J Transplant. 2004;4:1941–8. doi: 10.1111/j.1600-6143.2004.00620.x. [DOI] [PubMed] [Google Scholar]
- 80.Matot I, Weiniger CF, Zeira E, Galun E, Joshi BV, Jacobson KA. A3 adenosine receptors and mitogen-activated protein kinases in lung injury following in vivo reperfusion. Crit Care. 2006;10:R65. doi: 10.1186/cc4893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Matot I, Einav S, Weiniger CF, et al. Lung injury after in vivo reperfusion: outcome at 27 hours after reperfusion. Anesthesiology. 2008;109:269–78. doi: 10.1097/ALN.0b013e31817f5b90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Morschl E, Molina JG, Volmer JB, et al. A3 adenosine receptor signaling influences pulmonary inflammation and fibrosis. Am J Respir Cell Mol Biol. 2008;39:697–705. doi: 10.1165/rcmb.2007-0419OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Wagner R, Ngamsri KC, Stark S, Vollmer I, Reutershan J. Adenosine receptor A3 is a critical mediator in LPS-induced pulmonary inflammation. Am J Physiol Lung Cell Mol Physiol. 2010;299:L502–12. doi: 10.1152/ajplung.00083.2010. [DOI] [PubMed] [Google Scholar]
- 84.Spielman WS, Arend LJ. Adenosine receptors and signaling in the kidney. Hypertension. 1991;17:117–30. doi: 10.1161/01.hyp.17.2.117. [DOI] [PubMed] [Google Scholar]
- 85.Vallon V, Muhlbauer B, Osswald H. Adenosine and kidney function. Physiol Rev. 2006;86:901–40. doi: 10.1152/physrev.00031.2005. [DOI] [PubMed] [Google Scholar]
- 86.Vallon V, Osswald H. Adenosine receptors and the kidney. Handb Exp Pharmacol. 2009:443–70. doi: 10.1007/978-3-540-89615-9_15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Gouyon J-B, Guignard J-P. Theophylline prevents the hypoxemia-induced renal hemodynamic changes in rabbits. Kidney Int. 1988;33:1078–83. doi: 10.1038/ki.1988.114. [DOI] [PubMed] [Google Scholar]
- 88.Lin J-J, Churchill PC, Bidani AK. Effect of theophylline on the initiation phase of postischemic acute renal failure. JLabClinMed. 1986;108:150–4. [PubMed] [Google Scholar]
- 89.Grenz A, Zhang H, Eckle T, et al. Protective role of ecto-5′-nucleotidase (CD73) in renal ischemia. J Am Soc Nephrol. 2007;18:833–45. doi: 10.1681/ASN.2006101141. [DOI] [PubMed] [Google Scholar]
- 90.Grenz A, Zhang H, Hermes M, et al. Contribution of E-NTPDase1 (CD39) to renal protection from ischemia-reperfusion injury. FASEB J. 2007;21:2863–73. doi: 10.1096/fj.06-7947com. [DOI] [PubMed] [Google Scholar]
- 91.Joo JD, Kim M, Horst P, et al. Acute and delayed renal protection against renal ischemia and reperfusion injury with A1 adenosine receptors. Am J Physiol Renal Physiol. 2007;293:F1847–57. doi: 10.1152/ajprenal.00336.2007. [DOI] [PubMed] [Google Scholar]
- 92.Lee HT, Emala CW. Protective effects of renal ischemic preconditioning and adenosine pretreatment: role of A(1) and A(3) receptors. Am J Physiol Renal Physiol. 2000;278:F380–7. doi: 10.1152/ajprenal.2000.278.3.F380. [DOI] [PubMed] [Google Scholar]
- 93.Kim M, Chen SW, Park SW, D’Agati VD, Yang J, Lee HT. Kidney-specific reconstitution of the A1 adenosine receptor in A1 adenosine receptor knockout mice reduces renal ischemia-reperfusion injury. Kidney Int. 2009;75:809–23. doi: 10.1038/ki.2008.699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Lee HT, Kim M, Jan M, Penn RB, Emala CW. Renal tubule necrosis and apoptosis modulation by A1 adenosine receptor expression. Kidney Int. 2007;71:1249–61. doi: 10.1038/sj.ki.5002227. [DOI] [PubMed] [Google Scholar]
- 95.Gill A, Wortham K, Costa D, Davis W, Ticho B, Whalley E. Protective effect of tonapofylline (BG9928), an adenosine A1 receptor antagonist, against cisplatin-induced acute kidney injury in rats. Am J Nephrol. 2009;30:521–6. doi: 10.1159/000248762. [DOI] [PubMed] [Google Scholar]
- 96.Lee HT, Jan M, Bae SC, et al. A1 adenosine receptor knockout mice are protected against acute radiocontrast nephropathy in vivo. Am J Physiol Renal Physiol. 2006;290:F1367–75. doi: 10.1152/ajprenal.00347.2005. [DOI] [PubMed] [Google Scholar]
- 97.Li L, Okusa MD. Blocking the Immune respone in ischemic acute kidney injury: the role of adenosine 2A agonists. Nature Clinical Practice Nephrology. 2006;2:432–44. doi: 10.1038/ncpneph0238. [DOI] [PubMed] [Google Scholar]
- 98.Okusa MD, Linden J, Macdonald T, Huang L. Selective A2A-adenosine receptor activation during reperfusion reduces ischemia-reperfusion injury in rat kidney. Am J Physiol. 1999;277:F404–12. doi: 10.1152/ajprenal.1999.277.3.F404. [DOI] [PubMed] [Google Scholar]
- 99.Okusa MD, Linden J, Huang L, Rosin DL, Smith DF, Sullivan G. Enhanced protection from renal ischemia-reperfusion injury with A2A-adenosine receptor activation and PDE 4 inhibition. Kidney Int. 2001;59:2114–25. doi: 10.1046/j.1523-1755.2001.00726.x. [DOI] [PubMed] [Google Scholar]
- 100.Awad AS, Rouse M, Huang L, et al. Compartmentalization of neutrophils in the kidney and lung following acute ischemic kidney injury. Kidney Int. 2009:689–98. doi: 10.1038/ki.2008.648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Day Y-J, Huang L, McDuffie MJ, et al. Renal protection from ischemia mediated by A2A adenosine receptors on bone marrow-derived cells. J Clin Invest. 2003;112:883–91. doi: 10.1172/JCI15483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Grenz A, Osswald H, Eckle T, et al. The reno-vascular A2B adenosine receptor protects the kidney from ischemia. PLoS Med. 2008;5:e137. doi: 10.1371/journal.pmed.0050137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Lee HT, Ota-Setlik A, Xu H, D’Agati VD, Jacobson MA, Emala CW. A3 adenosine receptor knockout mice are protected against ischemia- and myoglobinuria-induced renal failure. Am J Physiol Renal Physiol. 2003;284:F267–73. doi: 10.1152/ajprenal.00271.2002. [DOI] [PubMed] [Google Scholar]
- 104.Lee HT, Kim M, Joo JD, Gallos G, Chen JF, Emala CW. A3 adenosine receptor activation decreases mortality and renal and hepatic injury in murine septic peritonitis. Am J Physiol Regul Integr Comp Physiol. 2006;291:R959–69. doi: 10.1152/ajpregu.00034.2006. [DOI] [PubMed] [Google Scholar]
- 105.Grden M, Podgorska M, Szutowicz A, Pawelczyk T. Altered expression of adenosine receptors in heart of diabetic rat. J Physiol Pharmacol. 2005;56:587–97. [PubMed] [Google Scholar]
- 106.Epperson SA, Brunton LL, Ramirez-Sanchez I, Villarreal F. Adenosine receptors and second messenger signaling pathways in rat cardiac fibroblasts. Am J Physiol Cell Physiol. 2009;296:C1171–7. doi: 10.1152/ajpcell.00290.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Auchampach JA, Bolli R. Adenosine receptor subtypes in the heart: therapeutic opportunities and challenges. Am J Physiol Heart Circ Physiol. 1999;276:H1113–6. doi: 10.1152/ajpheart.1999.276.3.H1113. [DOI] [PubMed] [Google Scholar]
- 108.Toombs CF, McGee S, Johnston WE, Vinten-Johansen J. Myocardial protective effects of adenosine. Infarct size reduction with pretreatment and continued receptor stimulation during ischemia. Circulation. 1992;86:986–94. doi: 10.1161/01.cir.86.3.986. [DOI] [PubMed] [Google Scholar]
- 109.Thornton JD, Liu GS, Olsson RA, Downey JM. Intravenous pretreatment with A1-selective adenosine analogues protects the heart against infarction. Circulation. 1992;85:659–65. doi: 10.1161/01.cir.85.2.659. [DOI] [PubMed] [Google Scholar]
- 110.Zhao ZQ, Sato H, Williams MW, Fernandez AZ, Vinten-Johansen J. Adenosine A2-receptor activation inhibits neutrophil-mediated injury to coronary endothelium. Am J Physiol Heart Circ Physiol. 1996;271:H1456–64. doi: 10.1152/ajpheart.1996.271.4.H1456. [DOI] [PubMed] [Google Scholar]
- 111.Jordan JE, Thourani VH, Auchampach JA, Robinson JA, Wang N-P, Vinten-Johansen J. A3 adenosine receptor activation attenuates neutrophil function and neutrophil-mediated reperfusion injury. Am J Physiol Heart Circ Physiol. 1999;277:H1895–905. doi: 10.1152/ajpheart.1999.277.5.H1895. [DOI] [PubMed] [Google Scholar]
- 112.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. Cardiovascular Research. 2006;70:308–14. doi: 10.1016/j.cardiores.2006.02.014. [DOI] [PubMed] [Google Scholar]
- 113.Murry CE, Jennings RB, Reimer KA. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–36. doi: 10.1161/01.cir.74.5.1124. [DOI] [PubMed] [Google Scholar]
- 114.Downey J, Davis A, Cohen M. Signaling pathways in ischemic preconditioning. Heart Failure Reviews. 2007;12:181–8. doi: 10.1007/s10741-007-9025-2. [DOI] [PubMed] [Google Scholar]
- 115.Gross ER, Gross GJ. Ischemic preconditioning and myocardial infarction: An update and perspective. Drug Discovery Today: Disease Mechanisms. 2007;4:165–74. doi: 10.1016/j.ddmec.2007.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Yellon DM, Downey JM. Preconditioning the Myocardium: From Cellular Physiology to Clinical Cardiology. Physiol Rev. 2003;83:1113–51. doi: 10.1152/physrev.00009.2003. [DOI] [PubMed] [Google Scholar]
- 117.Matherne GP, Linden J, Byford AM, Gauthier NS, Headrick JP. Transgenic A1 adenosine receptor overexpression increases myocardial resistance to ischemia. Proc Natl Acad Sci U S A. 1997;94:6541–6. doi: 10.1073/pnas.94.12.6541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Olafsson B, Forman MB, Puett DW, et al. Reduction of reperfusion injury in the canine preparation by intracoronary adenosine: importance of the endothelium and the no- reflow phenomenon. Circulation. 1987;76:1135–45. doi: 10.1161/01.cir.76.5.1135. [DOI] [PubMed] [Google Scholar]
- 119.Babbitt DG, Virmani R, Vildibill HD, Jr, Norton ED, Forman MB. Intracoronary adenosine administration during reperfusion following 3 hours of ischemia: Effects on infarct size, ventricular function, and regional myocardial blood flow. Am Heart J. 1990;120:808–18. doi: 10.1016/0002-8703(90)90196-5. [DOI] [PubMed] [Google Scholar]
- 120.Jordan JE, Zhao ZQ, Sato H, Taft S, Vinten-Johansen J. Adenosine A2 receptor activation attenuates reperfusion injury by inhibiting neutrophil accumulation, superoxide generation and coronary endothelial adherence. J Pharmacol Exp Ther. 1997;280:301–9. [PubMed] [Google Scholar]
- 121.Nakamura M, Zhao Z-Q, Clark KL, Velez DV, Guyton RA, Vinten-Johansen J. A novel adenosine analog, AMP579, inhibits neutrophil activation, adherence and neutrophil-mediated injury to coronary vascular endothelium. Eur J Pharmacol. 2000;397:197–205. doi: 10.1016/s0014-2999(00)00234-x. [DOI] [PubMed] [Google Scholar]
- 122.Lasley RD, Jahania MS, Mentzer RM., Jr Beneficial effects of adenosine A2a agonist CGS-21680 in infarcted and stunned porcine myocardium. Am J Physiol Heart Circ Physiol. 2001;280:H1660–6. doi: 10.1152/ajpheart.2001.280.4.H1660. [DOI] [PubMed] [Google Scholar]
- 123.Glover DK, Riou LM, Ruiz M, et al. Reduction of infarct size and postischemic inflammation from ATL-146e, a highly selective adenosine A2A receptor agonist, in reperfused canine myocardium. Am J Physiol Heart Circ Physiol. 2005;288:H1851–8. doi: 10.1152/ajpheart.00362.2004. [DOI] [PubMed] [Google Scholar]
- 124.Schlack W, Schafer M, Uebing A, Schafer S, Borchard U, Thamer V. Adenosine-A2-receptor activation at reperfusion reduces infarct size and improves myocardial wall function in dog heart. J Cardiovasc Pharmacol. 1993;22:89–96. doi: 10.1097/00005344-199307000-00015. [DOI] [PubMed] [Google Scholar]
- 125.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;123:332–8. doi: 10.1016/0002-8703(92)90643-a. [DOI] [PubMed] [Google Scholar]
- 126.Dai W, Hale SL, Nayak R, Kloner RA. ATL 313, a selective A(2A) adenosine receptor agonist, reduces myocardial infarct size in a rat ischemia/reperfusion model. Open Cardiovasc Med J. 2009;3:166–72. doi: 10.2174/1874192400903010166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Yang Z, Day Y-J, Toufektsian M-C, et al. Infarct-sparing effect of A2A-adenosine receptor activation is due primarily to its action on lymphocytes. Circulation. 2005;111:2190–7. doi: 10.1161/01.CIR.0000163586.62253.A5. [DOI] [PubMed] [Google Scholar]
- 128.Yang Z, Day Y-J, Toufektsian M-C, et al. Myocardial infarct-sparing effect of adenosine A2A receptor activation is due to its action on CD4+ T lymphocytes. Circulation. 2006;114:2056–64. doi: 10.1161/CIRCULATIONAHA.106.649244. [DOI] [PubMed] [Google Scholar]
- 129.Granfeldt A, Lefer DJ, Vinten-Johansen J. Protective ischaemia in patients: preconditioning and postconditioning. Cardiovascular Research. 2009;83:234–46. doi: 10.1093/cvr/cvp129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Morrison RR, Tan XL, Ledent C, Mustafa SJ, Hofmann PA. Targeted deletion of A2A adenosine receptors attenuates the protective effects of myocardial postconditioning. Am J Physiol Heart Circ Physiol. 2007;293:H2523–9. doi: 10.1152/ajpheart.00612.2007. [DOI] [PubMed] [Google Scholar]
- 131.Kuno A, Solenkova NV, Solodushko V, 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:H1288–95. doi: 10.1152/ajpheart.00209.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Xi J, McIntosh R, Shen X, et al. Adenosine A2A and A2B receptors work in concert to induce a strong protection against reperfusion injury in rat hearts. Journal of Molecular & Cellular Cardiology. 2009;47:684–90. doi: 10.1016/j.yjmcc.2009.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.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:H1262–4. doi: 10.1152/ajpheart.00181.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhao ZQ, Nakanishi K, McGee DS, Tan P, Vinten-Johansen J. A1 receptor mediated myocardial infarct size reduction by endogenous adenosine is exerted primarily during ischaemia. Cardiovascular Research. 1994;28:270–9. doi: 10.1093/cvr/28.2.270. [DOI] [PubMed] [Google Scholar]
- 135.Baxter GF, Hale SL, Miki T, et al. Adenosine A1 agonist at reperfusion trial (AART): results of a three-center, blinded, randomized, controlled experimental infarct study. Cardiovasc Drugs Ther. 2000;14:607–14. doi: 10.1023/a:1007850527878. [DOI] [PubMed] [Google Scholar]
- 136.Auchampach JA, Ge Z-D, Wan TC, Moore J, Gross GJ. A3 adenosine receptor agonist IB-MECA reduces myocardial ischemia-reperfusion injury in dogs. Am J Physiol Heart Circ Physiol. 2003;285:H607–13. doi: 10.1152/ajpheart.01001.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Maddock HL, Mocanu MM, Yellon DM. Adenosine A(3) receptor activation protects the myocardium from reperfusion/reoxygenation injury. Am J Physiol Heart Circ Physiol. 2002;283:H1307–13. doi: 10.1152/ajpheart.00851.2001. [DOI] [PubMed] [Google Scholar]
- 138.Ralevic V, Burnstock G. Receptors for purines and pyrimidines. Pharmacol Rev. 1998;50:413–92. [PubMed] [Google Scholar]
- 139.Todo S, Zhu Y, Zhang S, et al. Attenuation of ischemic liver injury by augmentation of endogenous adenosine. Transplantation. 1997;63:217–23. doi: 10.1097/00007890-199701270-00007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Leon Fernandez OS, Ajamieh HH, Berlanga J, et al. Ozone oxidative preconditioning is mediated by A1 adenosine receptors in a rat model of liver ischemia/ reperfusion. Transpl Int. 2008;21:39–48. doi: 10.1111/j.1432-2277.2007.00568.x. [DOI] [PubMed] [Google Scholar]
- 141.Ajamieh HH, Candelario-Jalil E, Fernandez OS, Gerbes AL. Ischaemic and pharmacological preconditionings protect liver via adenosine and redox status following hepatic ischaemia/reperfusion in rats. Clin Sci (Lond) 2008;115:69–77. doi: 10.1042/CS20070415. [DOI] [PubMed] [Google Scholar]
- 142.Kim J, Kim M, Song JH, Lee HT. Endogenous A1 adenosine receptors protect against hepatic ischemia reperfusion injury in mice. Liver Transpl. 2008;14:845–54. doi: 10.1002/lt.21432. [DOI] [PubMed] [Google Scholar]
- 143.Magata S, Taniguchi M, Suzuki T, et al. The effect of antagonism of adenosine A1 receptor against ischemia and reperfusion injury of the liver. J Surg Res. 2007;139:7–14. doi: 10.1016/j.jss.2006.09.021. [DOI] [PubMed] [Google Scholar]
- 144.Net M, Valero R, Almenara R, et al. The effect of normothermic recirculation is mediated by ischemic preconditioning in NHBD liver transplantation. Am J Transplant. 2005;5:2385–92. doi: 10.1111/j.1600-6143.2005.01052.x. [DOI] [PubMed] [Google Scholar]
- 145.Harada N, Okajima K, Murakami K, et al. Adenosine and selective A(2A) receptor agonists reduce ischemia/reperfusion injury of rat liver mainly by inhibiting leukocyte activation. J Pharmacol Exp Ther. 2000;294:1034–42. [PubMed] [Google Scholar]
- 146.Cao Z, Yuan Y, Jeyabalan G, et al. Preactivation of NKT cells with alpha-GalCer protects against hepatic ischemia-reperfusion injury in mouse by a mechanism involving IL-13 and adenosine A2A receptor. Am J Physiol Gastrointest Liver Physiol. 2009;297:G249–58. doi: 10.1152/ajpgi.00041.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Day YJ, Marshall MA, Huang L, McDuffie MJ, Okusa MD, Linden J. Protection from ischemic liver injury by activation of A2A adenosine receptors during reperfusion: inhibition of chemokine induction. Am J Physiol Gastrointest Liver Physiol. 2004;286:G285–93. doi: 10.1152/ajpgi.00348.2003. [DOI] [PubMed] [Google Scholar]
- 148.Lappas CM, Day YJ, Marshall MA, Engelhard VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J Exp Med. 2006;203:2639–48. doi: 10.1084/jem.20061097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Guinzberg R, Cortes D, Diaz-Cruz A, Riveros-Rosas H, Villalobos-Molina R, Pina E. Inosine released after hypoxia activates hepatic glucose liberation through A3 adenosine receptors. Am J Physiol Endocrinol Metab. 2006;290:E940–51. doi: 10.1152/ajpendo.00173.2005. [DOI] [PubMed] [Google Scholar]
- 150.Jin X, Shepherd RK, Duling BR, Linden J. Inosine binds to A3 adenosine receptors and stimulates mast cell degranulation. J Clin Invest. 1997;100:2849–57. doi: 10.1172/JCI119833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Chhabra P, Wang K, Zeng Q, et al. Adenosine A(2A) agonist administration improves islet transplant outcome: Evidence for the role of innate immunity in islet graft rejection. Cell Transplant. 2010;19:597–612. doi: 10.3727/096368910X491806. [DOI] [PubMed] [Google Scholar]
- 152.Chen JF, Pedata F. Modulation of ischemic brain injury and neuroinflammation by adenosine A2A receptors. Curr Pharm Des. 2008;14:1490–9. doi: 10.2174/138161208784480126. [DOI] [PubMed] [Google Scholar]
- 153.Phillis JW. The effects of selective A1 and A2a adenosine receptor antagonists on cerebral ischemic injury in the gerbil. Brain Res. 1995;705:79–84. doi: 10.1016/0006-8993(95)01153-6. [DOI] [PubMed] [Google Scholar]
- 154.Cassada DC, Gangemi JJ, Rieger JM, et al. Systemic adenosine A2A agonist ameliorates ischemic reperfusion injury in the rabbit spinal cord. Ann Thorac Surg. 2001;72:1245–50. doi: 10.1016/s0003-4975(01)03057-0. [DOI] [PubMed] [Google Scholar]
- 155.Li Y, Oskouian RJ, Day YJ, et al. Mouse spinal cord compression injury is reduced by either activation of the adenosine A2A receptor on bone marrow-derived cells or deletion of the A2A receptor on non-bone marrow-derived cells. Neuroscience. 2006;141:2029–39. doi: 10.1016/j.neuroscience.2006.05.014. [DOI] [PubMed] [Google Scholar]
- 156.Miyazaki N, Nakatsuka T, Takeda D, Nohda K, Inoue K, Yoshida M. Adenosine modulates excitatory synaptic transmission and suppresses neuronal death induced by ischaemia in rat spinal motoneurones. Pflugers Arch. 2008;457:441–51. doi: 10.1007/s00424-008-0542-1. [DOI] [PubMed] [Google Scholar]
- 157.Ozacmak VH, Sayan H. Pretreatment with adenosine and adenosine A1 receptor agonist protects against intestinal ischemia-reperfusion injury in rat. World J Gastroenterol. 2007;13:538–47. doi: 10.3748/wjg.v13.i4.538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hart ML, Jacobi B, Schittenhelm J, Henn M, Eltzschig HK. Cutting Edge: A2B Adenosine receptor signaling provides potent protection during intestinal ischemia/reperfusion injury. J Immunol. 2009;182:3965–8. doi: 10.4049/jimmunol.0802193. [DOI] [PubMed] [Google Scholar]
- 159.Xu Y, Huo Y, Toufektsian MC, et al. Activated platelets contribute importantly to myocardial reperfusion injury. Am J Physiol Heart Circ Physiol. 2006;290:H692–9. doi: 10.1152/ajpheart.00634.2005. [DOI] [PubMed] [Google Scholar]
- 160.Bajwa A, Kinsey GR, Okusa MD. Immune mechanisms and novel pharmacological therapies of acute kidney injury. Curr Drug Targets. 2009;10:1196–204. doi: 10.2174/138945009789753174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Steffens S, Montecucco F, Mach F. The inflammatory response as a target to reduce myocardial ischaemia and reperfusion injury. Thromb Haemost. 2009;102:240–7. doi: 10.1160/TH08-12-0837. [DOI] [PubMed] [Google Scholar]
- 162.Lappas CM, Sullivan GW, Linden J. Adenosine A2A agonists in development for the treatment of inflammation. Expert Opin Investig Drugs. 2005;14:797–806. doi: 10.1517/13543784.14.7.797. [DOI] [PubMed] [Google Scholar]
- 163.Yang D, Zhang Y, Nguyen HG, et al. The A2B adenosine receptor protects against inflammation and excessive vascular adhesion. J Clin Invest. 2006;116:1913–23. doi: 10.1172/JCI27933. [DOI] [PMC free article] [PubMed] [Google Scholar]

