Abstract
Adenosine increases coronary flow mainly through the activation of A2A and A2B adenosine receptors (ARs). However, the mechanisms for the regulation of coronary flow are not fully understood. We previously demonstrated that adenosine-induced increase in coronary flow is in part through NADPH oxidase (Nox) activation, which is independent of activation of either A1 or A3ARs. In this study, we hypothesize that adenosine-mediated increase in coronary flow through Nox activation depends on A2A but not A2BARs. Functional studies were conducted using isolated Langendorff-perfused mouse hearts. Hydrogen peroxide (H2O2) production was measured in isolated coronary arteries from WT, A2AAR knockout (KO), and A2BAR KO mice using dichlorofluorescein immunofluorescence. Adenosine-induced concentration-dependent increase in coronary flow was attenuated by the specific Nox2 inhibitor gp91 ds-tat or reactive oxygen species (ROS) scavenger EUK134 in both WT and A2B but not A2AAR KO isolated hearts. Similarly, the A2AAR selective agonist CGS-21680-induced increase in coronary flow was significantly blunted by Nox2 inhibition in both WT and A2BAR KO, while the A2BAR selective agonist BAY 60-6583-induced increase in coronary flow was not affected by Nox2 inhibition in WT. In intact isolated coronary arteries, adenosine-induced (10 μM) increase in H2O2 formation in both WT and A2BAR KO mice was attenuated by Nox2 inhibition, whereas adenosine failed to increase H2O2 production in A2AAR KO mice. In conclusion, adenosine-induced increase in coronary flow is partially mediated by Nox2-derived H2O2, which critically depends upon the presence of A2AAR.
Keywords: Adenosine, A2A receptor knockout, NADPH oxidase, Hydrogen peroxide, Coronary flow
Introduction
Coronary flow is tightly regulated to maintain a consistently high level of myocardial oxygen extraction over a wide range of myocardial demands [1–3]. This tight regulation is dependent on numerous vasoconstrictor and vasodilator influences, exerted by the autonomic nervous system, endothelium, and myocardium [3–6]. Adenosine, a well-known locally released metabolite, has been postulated as one of the important agents responsible for coronary vascular tone regulation in various conditions [3, 5, 7–9], among which adenosine has also been thought to regulate resting coronary flow [10, 11]. However, the role of adenosine in regulating resting coronary flow remains controversial [3]. This discrepancy may be due to different animal models, differences in species, and/or different agonists and antagonists used in these studies. It is well established that the coronary effects of adenosine are mediated through the activation of its four subtypes of receptors, namely A1, A2A, A2B, and A3 receptors (ARs) [9, 12, 13]. The role of these receptors has been studied in various species [12]. With an overall coronary vasodilation, A2AAR predominantly and A2BAR minimally contribute to dilating coronary vasculature [14–16], whereas A1AR and A3AR counteract the effects of A2A/A2BARs resulting in a diminished coronary vasodilation [15, 17, 18]. At post-receptor levels, several effector pathways of adenosine-mediated coronary flow have been reported, such as activation of nitric oxide (NO) pathway [19, 20], cyclic adenosine 5′-monophosphate (cAMP)-dependent pathway [21], activation of potassium channels [9, 21], and the involvement of H2O2 [19]. However, the downstream effectors linked to the activation of ARs in this process are not completely understood.
H2O2, a molecule within the reactive oxygen species (ROS) family, has been proposed to serve as a pivotal vasodilator in coronary flow regulation in human, canine, porcine, and murine coronary vasculature [19, 22–26]. NADPH oxidases (Nox) are the major source of ROS in the vasculature that plays both physiological and pathophysiological roles in the control of vascular tone [27–29]. The Nox family consists of seven members, Nox1-5, Doux1, and Doux2, among which Nox2 (gp91 Phox) forms the major source of H2O2 in cells stimulated with growth factors or cytokines under normal circumstances [30, 31]. Indeed, in the coronary vasculature, Nox2 has been proposed to be a functionally relevant source of H2O2 that mediates agonist-induced vasodilation [32]. Several recent studies have shown the interaction between adenosine and ROS via the regulation of Nox activity. For instance, inhibition or deletion of Nox2 leads to the attenuation of adenosine vasoconstrictor responses in aortas [33] and renal arterioles [34] and adenosine vasodilator responses in cerebral arteries [35]. However, little is known about the involvement of Nox2-derived ROS in adenosine-mediated flow response in the coronary circulation. Although activation of A1 and A3AR has been shown to have vascular effects through ROS [33, 36], our recent studies have already excluded the involvement of A1 and A3ARs in ROS-mediated regulation of coronary flow [37]. Therefore, with the focus on the adenosine signaling, the first aim of the present study was to further investigate whether A2A and/or A2BAR are involved in Nox2-derived ROS-mediated coronary vasodilation. Given the previous findings that adenosine A2AAR coupled to H2O2 contributes to coronary reactive hyperemia [38] and Nox2 acts as a functional source for H2O2 resulting in coronary vasodilation [32], the second aim of the present study was to examine whether Nox2-derived ROS production contributing to adenosine-mediated coronary flow is through H2O2 and which adenosine receptors are required in this process. We performed exogenous infusion of adenosine in Langendorff-perfused isolated hearts as well as immunohistochemistry in isolated coronary arteries from wild type (WT), A2AAR knockout (KO), and A2BAR KO mice for these studies.
Methods
Materials
EUK134 was purchased from Cayman Chemical (Ann Arbor, MI, USA), gp91 ds-tat was purchased from Anaspec (Fremont, CA, USA), and BAY 60-6583 was obtained as a gift from Bayer AG (Leverkusen, Germany). All other chemicals (adenosine, CGS-21680 and 2′,7′-dichloro-fluorescein diacetate (DCFH-DA)) were purchased from Sigma-Aldrich (St. Louis, MO, USA). Stock solutions of adenosine, BAY 60-6583, CGS-21680, EUK134, and DCFH-DA were made in dimethyl sulfoxide (DMSO), whereas gp91 ds-tat was dissolved in distilled water.
Animals
All experimental protocols were approved by the Institutional Animal Care and Use Committee at West Virginia University School of Medicine. We followed guidelines set forth by National Institutes of Health regarding the care and use of laboratory animals. WT mice (C57BL/6 background) were purchased from The Jackson Laboratory (Bar Harbor, ME, USA). A2A and A2BAR KO mice on an inbred C57BL/6 background were generously provided by Dr. Catherine Ledent (Universite Libre de Bruxelles, Belgium) and Dr. Stephen Tilley (University of North Carolina, Chapel Hill, NC, USA), respectively. Mice were kept in cages with 12:12-h light-dark cycles and maintained on a standard laboratory diet with access to water ad libitum. The absence of A2A and A2BARs at mRNA level in A2AAR KO and A2BAR KO mice has been confirmed by our previous studies using PCR in isolated aortas, mesenteric arteries, and coronary arteries [16, 20, 39–41]. With difficulties in collecting large amount of tissues for Western blot, one of our recent studies has confirmed the absence of A2AAR protein level in A2AAR KO mice in isolated coronary arteries using immunohistochemistry [40]. Our functional data also confirmed the lack of CGS-21680 (a selective A2AAR agonist)-induced coronary flow response in isolated hearts of A2AAR KO mice and the lack of BAY 60-6583 (a selective A2B AR agonist)-induced coronary flow response in isolated hearts of A2BAR KO mice [16, 20].
Langendorff-perfused mouse heart preparation
Mice (14–18 weeks) of either sex were anesthetized with pentobarbital sodium (50 mg kg−1, i.p.). Mice were weighed before hearts were rapidly removed into heparinized (5 U ml−1) ice-cold Krebs-Henseleit buffer containing (in mM) 119 NaCl, 11 glucose, 22 NaHCO3, 4.7 KCl, 1.2 KH2PO4, 1.2 MgSO4, 2.5 CaCl2, 2 pyruvate, and 0.5 EDTA. After removal of the surrounding tissue, the aorta was rapidly cannulated with a 20-gauge, blunt-ended needle; then, the heart was continuously perfused with 37 °C buffer bubbled with 95 % O2/5 % CO2 at a constant perfusion pressure of 80 mmHg [20, 40]. Subsequently, through an opening in the left atrium, a fluid-filled balloon made of plastic wrap was inserted into the left ventricle across the mitral valve. The balloon was connected to a pressure transducer for continuous measurement of left ventricular developed pressure (LVDP) and heart rate. The heart was then immersed in a water-jacketed perfusate bath maintained at 37 °C and beats spontaneously. Coronary flow was continuously measured with an ultrasonic flow probe (Transonic Systems, Ithaca, NY, USA) placed in the aortic perfusion line. A PowerLab Chart data acquisition system (AD Instruments, Colorado Springs, CO, USA) was used for data acquisition. Hearts were allowed to equilibrate for 30 to 45 min before starting experimental protocols. Hearts with persistent arrhythmias were excluded from the study. As a positive control, a 15-s flow occlusion resulted in a twofold increase in coronary flow over baseline [19, 38], indicating that buffer-perfused hearts have minimum hypoxia and coronary vessels have the capacity to further dilate.
Langendorff experimental protocols
After equilibrium, baseline coronary flow, heart rate (HR), and LVDP were measured. Adenosine concentration response curves (10−8–10−5 M) were acquired in perfused hearts from WT, A2AAR KO, and A2BAR KO mice. Each concentration of adenosine was infused for 5 min followed by a minimum of 5 min of perfusion for drug washout [16, 37]. In separate experiments, the specific Nox2 inhibitor gp91 ds-tat (1 μM) [42] or the superoxide dismutase and catalase-mimicking drug EUK134 (50 μM) was perfused for 20 min before acquiring adenosine concentration response curves (10−8–10−5 M) in perfused hearts from those mice [37].
In another two experimental groups, the selective A2AAR agonist CGS-21680 concentration response curves (10−10–10−6 M) were acquired in perfused hearts from WT, A2AAR KO, and A2BAR KO mice, while the selective A2BAR agonist BAY 60-658 concentration response curves (10−10–10−6 M) were acquired in perfused hearts only from WT mice. In separate experiments, the specific Nox2 inhibitor gp91 ds-tat (1 μM) was perfused for 20 min before acquiring CGS-21680 and BAY 60-658 concentration response curves (10−10–10−6 M), respectively [16, 37]. All compounds were infused at a rate of 1/100 ml min−1 of the coronary flow through an injection port directly proximal to the aortic cannula using a microinjection pump (Harvard Apparatus, Holliston, MA, USA) [16, 20, 37].
Fluorescence detection of H2O2 in mouse coronary arteries
Left coronary arteries (with diameter of 50–120 μm) were isolated from WT, A2AAR KO, and A2BAR KO mice. Then, coronary arteries were incubated in DCFH-DA (10 μM) prepared in DMEM (ATCC, Manassas, VA, USA) for 30 min at 37 °C followed by 10-min wash [40]. Vessels were then pinned on a layer of silicone gel lying on a plastic culture dish and incubated with DMEM buffer maintained at 37 °C. Baseline control images were obtained by optical xyz sectioning with a vertical depth of 1 μm using a Zeiss water immersion objective (W N-AchroplanX40/0.75 numerical aperture) on a confocal microscope (LSM 510, Zeiss, Heidelberg, Germany). Once the baseline control images were acquired, adenosine (10 μM) was added into the culture dish and images were taken at 5 min (in some vessels at both 5 and 10 min) after adenosine incubation. We previously showed that a maximal fluorescence intensity of DCFH-DA was reached after 15 min of adenosine incubation [40], and there was no difference in fluorescence intensity between 5 and 10 min in the present study (data not shown). The data at 5 min after adenosine incubation was used for analysis. In separate sets of experiments, the arteries were treated with the Nox2 inhibitor gp91 ds-tat (1 μM) for 20 min and baseline control images were obtained before the addition of adenosine. After 5 min of adenosine incubation, additional images were taken. Hydrogen peroxide (200 μM) served as the positive control at the end of experiments [37, 40]. Only the bottom half of vessels (more proximal to the scanner) were scanned by xyz sectioning, in order to avoid the quenching as much as possible. ImageJ software was used for fluorescence image analysis. Stacks of regions of interest (ROIs) were selected on the basis of the outline of the vessel indicated by the fluorescence.
Statistical analysis
Langendorff baseline data for WT, A2AAR KO, and A2BAR KO groups were compared using one-way ANOVA followed by post hoc analysis using Bonferroni’s test. The effects of drug treatment on concentration response curves of adenosine, CGS-21680, and BAY 60-6583 were analyzed using two-way ANOVA followed by post hoc analysis using Bonferroni’s test. Since the absolute coronary flow changes proportionally with heart mass, the coronary flow was presented as ml min−1 g−1 wet heart weight [16, 37, 43]. With regard to imaging, the mean fluorescence intensity of each ROI (including those on both endothelial cells and smooth muscle cells) was calculated by subtraction of the background signal and changes in fluorescence intensity were presented as a ratio normalized to control [40]. Image data were analyzed using paired t test. All the data are presented as mean ± SEM; n represents the number of animals. Statistical significance was accepted when P < 0.05.
Results
Baseline function in isolated WT, A2AAR KO, and A2BAR KO mouse hearts
Table 1 summarizes the baseline functional parameters for heart rate, LVDP, and coronary flow in WT, A2AAR KO, and A2BAR KO mice after 30 min of equilibration of isolated hearts. Average heart weight to body weight ratio was significantly different between A2AAR KO and A2BAR KO mice, which is due to a heavier body weight in A2BAR KO mice as compared to A2AAR KO mice (27 ± 0.7 g in A2BAR KO mice vs. 24 ± 0.5 g in A2AAR KO mice), while the heart weight between the groups was comparable. However, no significant differences were observed in heart rate, LVDP, and baseline coronary flow from those mice (P > 0.05, by one-way ANOVA) (Table 1).
Table 1.
Baseline data for WT, A2AAR KO, and A2BAR KO mouse hearts
| WT (n = 21) | A2AAR KO (n = 15) | A2BAR KO (n = 17) | |
|---|---|---|---|
| Age, week | 16 ± 0.3 | 15 ± 0.3 | 16 ± 0.4 |
| No. of mice | 21 | 15 | 17 |
| BW, g | 25 ± 0.8 | 24 ± 0.5 | 27 ± 0.7 |
| HW, mg | 96.2 ± 2.6 | 99.1 ± 2.6 | 98.4 ± 2.4 |
| HW/BW, % | 0.39 ± 0.01 | 0.42 ± 0.01 | 0.37 ± 0.02* |
| CF, ml min−1 g−1 | 18 ± 0.6 | 18 ± 0.9 | 20 ± 0.4 |
| HR, beats min−1 | 437 ± 10 | 391 ± 8 | 446 ± 10 |
| LVDP, mmHg | 90 ± 4 | 90 ± 5 | 80 ± 4 |
All parameters were collected after 30 min of equilibration in a Langendorff preparation. Values are means ± SEM
WT wild type, KO knockout mice, BW body weight, HW heart weight, CF coronary flow, HR heart rate, LVDP left ventricular developed pressure
*P < 0.05 compared to A2AAR KO
Effects of Nox2 inhibition and A2AAR/A2BAR deletion on adenosine-induced increase in coronary flow
Adenosine produced a concentration-dependent increase in coronary flow in WT mice (Emax: 38.13 ± 0.78 ml min−1 g−1, data obtained from both Figs. 1a and 2a), which was significantly attenuated by Nox2 inhibition with gp91 ds-tat (Fig. 1a). In agreement with our previous studies [37], these findings indicate that adenosine-induced increase in coronary flow in WT mice is mediated in part through Nox2 activation.
Fig. 1.
Effects of NADPH oxidase (Nox2) inhibition on adenosine-induced increase in coronary flow. Shown are the effects of gp91 ds-tat (1 μM) on adenosine (Ado) concentration response curves (10−8 to 10−5 M) in isolated hearts of wild type (WT) (a, n = 6), A2A receptor (AR) knockout (KO) (b, n = 6), and A2BAR KO mice (c, n = 6). Values are mean ± SEM. *Significant difference vs. corresponding control points using two-way ANOVA followed by post hoc analysis using Bonferroni’s test (P < 0.05)
Fig. 2.
Effects of ROS scavenging on adenosine-induced increase in coronary flow. Shown are the effects of EUK134 (50 μM) on adenosine (Ado) concentration response curves (10−8 to 10−5 M) in isolated hearts of WT (a, n = 5), A2AAR KO (b, n = 4), and A2BAR KO mice (c, n = 6). Values are mean ± SEM. *Significant difference vs. corresponding control points using two-way ANOVA followed by post hoc analysis using Bonferroni’s test (P < 0.05)
Notably, inhibition of Nox2 by gp91 ds-tat in A2AAR KO mice had no effect on adenosine-induced increase in coronary flow (Fig. 1b) but significantly attenuated the increased coronary flow in A2BAR KO mice (Fig. 1c). All together, these findings indicate that adenosine-induced increase in coronary flow is partially mediated via activation of Nox2 and likely depends on A2A but not A2BARs.
Effects of ROS scavenging and A2A/A2BAR deletion on adenosine-induced increase in coronary flow
In accordance with previous studies from our laboratory [37], ROS scavenging with EUK134 markedly attenuated the adenosine-induced increase in coronary flow in WT mice (Fig. 2a). Similar to the effects of Nox2 inhibition, ROS scavenging with EUK134 had no effect on adenosine-induced increase in coronary flow in A2AAR KO mice (Fig. 2b) but significantly attenuated the increased coronary flow in A2BAR KO mice (Fig. 2c). All together, these findings indicate that ROS production is involved in adenosine-induced increase in coronary flow, which depends on A2A but not A2BAR activation.
Effects of Nox2 inhibition and A2AAR/A2BAR deletion on A2AAR/A2BAR agonist-mediated increase in coronary flow
To further investigate the role of A2AAR in this process, the selective A2AAR agonist CGS-21680 concentration response curves were acquired. CGS-21680 produced a concentration-dependent increase in coronary flow in WT, which was significantly blunted by Nox2 inhibition with gp91 ds-tat (Fig. 3a). A similar finding was also observed in A2BAR KO mice in which CGS-21680-induced increase in coronary flow was significantly attenuated by the Nox2 inhibitor gp91 ds-tat (Fig. 3c). As expected, CGS-21680 failed to increase coronary flow in A2AAR KO mice (Fig. 3b). To further exclude the involvement of A2BAR, the selective A2BAR agonist BAY 60-6583 concentration response curves were acquired in WT mice. BAY 60-6583 produced a concentration-dependent increase in coronary flow, which was not affected by the Nox2 inhibitor gp91 ds-tat (Fig. 4). Taken together, these findings indicate that adenosine-induced increase in coronary flow is partially mediated by Nox2-derived ROS, which requires A2AARs.
Fig. 3.
Effects of Nox2 inhibition on adenosine A2AAR agonist-induced increase in coronary flow. Shown are the effects of gp91 ds-tat (1 μM) on CGS-21680 (CGS, the A2AAR selective agonist) concentration response curves (10−10 to 10−6 M) in isolated hearts of WT (a, n = 6), A2AAR KO (b, n = 5), and A2BAR KO mice (c, n = 5). Values are mean ± SEM. *Significant difference vs. corresponding control points using two-way ANOVA followed by post hoc analysis using Bonferroni’s test (P < 0.05)
Fig. 4.
Effects of Nox2 inhibition on adenosine A2BAR agonist-induced increase in coronary flow. Shown are the effects of gp91 ds-tat (1 μM) on BAY 60-6583 (BAY, the A2BAR selective agonist) concentration response curves (10−10 to 10−6 M) in isolated hearts of WT mice (n = 4). Values are mean ± SEM
Effects of Nox2 inhibition and A2AAR/A2BAR deletion on H2O2 production by adenosine in isolated coronary arteries
To assess the potential involvement of vasodilator ROS (H2O2) in adenosine-induced increase in coronary flow, DCFH-DA immunofluorescence was performed on intact isolated coronary arteries. As shown in Fig. 5, adenosine (10 μM) increased H2O2 generation in both WT and A2BAR KO mice (Fig. 5a), as evidenced by increases in fluorescence intensity ratio in adenosine-treated tissues (1.36 ± 0.04 and 1.24 ± 0.02 times corresponding baselines, respectively, P < 0.05) as compared to corresponding baseline (Fig. 5b). H2O2 production was observed in both endothelial cells (ECs, arrow head) as well as smooth muscle cells (SMCs, arrow) (Fig. 1a). Of note, adenosine-increased H2O2 production was inhibited by the Nox2 inhibitor gp91 ds-tat in both WT and A2BAR KO mice to baseline levels (Fig. 5a, b). More importantly, adenosine failed to increase H2O2 production from A2AAR KO mice either in the presence or absence of Nox2 inhibition (Fig. 5a, b). Hydrogen peroxide (200 μM) added at the end of experiments further increased fluorescence (Fig. 5a), indicating that the dye did not reach the saturation point [40]. Taken together, these data further suggest that Nox2 is an important source for H2O2 production, which depends on the activation of A2AARs by adenosine.
Fig. 5.

Effects of Nox2 inhibition on adenosine-induced H2O2 production in isolated coronary arteries. a Representative confocal fluorescence images showing changes in DCFH-DA (dichlorofluorescin) fluorescence intensity before (baseline) and after 5 min of adenosine (Ado) (10 μM) or 10 min of H2O2 (200 μM) stimulation in the presence and absence of gp91 ds-tat (1 μM) from WT, A2AAR KO, and A2BAR KO mice. Scale bar: 50 μm. b Adenosine-induced increase in H2O2 in isolated coronary arteries of WT (n = 8), A2AAR KO (n = 5), and A2BAR KO mice (n = 5). Values are mean ± SEM. *P < 0.05 vs. corresponding control (by paired t test)
Discussion
This study examined the hypothesis that A2A and/or A2BAR-mediated coronary vasodilation involves Nox2-derived ROS. With the focus on the adenosine signaling pathway, we measured changes in coronary flow in isolated hearts from WT, A2AAR, and A2B AR KO mice in response to Nox2 inhibition (gp91 ds-tat) or ROS scavenging (EUK134) during exogenous adenosine infusion. In addition, the downstream effector (H2O2) of ROS in this process was also assessed in isolated coronary arteries from WT, A2AAR, and A2B AR KO mice. The main findings of the present study are as follows: (1) either Nox2 inhibition with gp91 ds-tat or ROS scavenging with EUK134 attenuated the adenosine-induced increase in coronary flow in WT and A2BAR KO mice but not in A2AAR KO mice; (2) Nox2 inhibition blunted the A2AAR agonist-increased coronary flow in WT and A2BAR KO mice, whereas the A2BAR agonist responses were unaltered by Nox2 inhibition in WT mice; (3) furthermore, adenosine-mediated increase in H2O2 formation in intact isolated coronary arteries of WT and A2BAR KO mice was inhibited by Nox2 inhibition, whereas the H2O2 formation was not affected by adenosine either with or without Nox2 inhibition in A2AAR KO mice. The implications of these findings are discussed below.
Adenosine-induced increase in coronary flow is mediated in part through Nox2-derived ROS
Adenosine is known to regulate coronary vascular tone in various species. We [20, 44] and others [10, 11, 45] previously demonstrated that adenosine also plays a physiological role in regulating coronary resting flow in human [11], swine [10], dogs [45], guinea pigs [46], and mice [20, 44], while some other groups failed to observe an endogenous role of adenosine [3, 47, 48], which leaves this scientific topic controversial. This discrepancy may be due to different animal models, differences in species, and/or different agonists and antagonists applied in these studies [19]. However, the application of exogenous adenosine to increase coronary flow for the investigation of adenosine receptor-mediated signaling pathways has been well established [37, 44]. Thus, in agreement with previous studies from our laboratory [37, 44, 49] and others [14], adenosine produced a large increase in coronary flow in isolated mouse hearts, supporting the overall effect of vasodilation by adenosine in the coronary circulation [14, 15, 37, 50, 51].
Several recent studies showed the interaction between adenosine and ROS via the activation of Nox, which has been reported to be the major source of ROS in the vasculature and involved in vascular tone regulation [27, 28]. Among Nox isoforms, Nox2 has been extensively studied in various vascular beds. Nox2 inhibition or deletion attenuated the adenosine-mediated vasoconstriction in aortas [33] and renal arterioles [34]. In contrast, Nox2 inhibition or ROS scavenging blunted the adenosine-induced vasodilation in cerebral arteries [35] and coronary circulation [37], both of which suggest that adenosine is capable of generating ROS in these vasculatures. Consistent with our previous findings in the coronary circulation but in contrast to those in other vascular beds [33, 34, 37], Nox2 inhibition or ROS scavenging significantly attenuated the adenosine-mediated increase in coronary flow, suggesting that adenosine-mediated coronary vasodilation is in part attributed to Nox2-derived ROS. This heterogeneity might be due to vasoconstrictor ROS vs. vasodilator ROS [29, 31] generated from different vascular beds (coronary vs. renal artery) [34, 37]. Furthermore, in the coronary vasculature, the varying effects of ROS may depend on which source ROS are generated from (NADPH vs. uncoupled NO synthase) [31, 52, 53] or the pathophysiological conditions [32, 52, 53], e.g., diabetes, and the right ventricular hypertrophy [54–56]. In accordance with our previous studies [37], the effect of ROS scavenging with EUK134 was comparable with the effect of Nox2 inhibition with gp91 ds-tat in WT isolated hearts (P = 0.24), suggesting that the majority of ROS generated by adenosine is likely from Nox2, which leads to increases in coronary flow. Although EUK134 is capable of scavenging both vasoconstrictor and vasodilator ROS [57], the observation that EUK134 significantly attenuated the adenosine-mediated increase in coronary flow in one of our previous studies [37] as well as in the present study suggest that vasodilator ROS are likely generated by adenosine.
In the healthy coronary vasculature, several recent studies addressed a crucial role of NADPH oxidase in coronary vasodilation in response to various agonists. Indeed, downregulation of Nox activity by cytosolic p47phox knockdown or inhibition of Nox2 attenuated the coronary vasodilation to vascular endothelial growth factor (VEGF) [52] and bradykinin [32], respectively. In addition, Nox2 overexpression enhanced the coronary vasodilation in response to both VEGF and acetylcholine [53], further confirming that the vasodilator ROS is derived from Nox in this process. More importantly, Larsen and colleagues proposed that Nox2 is a functionally relevant source of H2O2 that mediates agonist-induced coronary vasodilation [32]. In accordance with this concept, we found in the present study as well as one of our previous studies [40] that adenosine-increased H2O2 formation in intact isolated coronary arteries was attenuated by not only Nox2 inhibition but also catalase [40]. The production of H2O2 induced by adenosine might stem from the intermediate ROS product namely, superoxide anion [27, 28, 31], as we previously reported that adenosine is also capable of producing superoxide [37]. Thus, it is likely that H2O2 acts as a downstream effector of superoxide contributing to adenosine-induced increase in coronary flow. Regarding the source of H2O2, we believe that H2O2 produced from EC and SMC of coronary arteries may play a major role in adenosine-mediated coronary flow. This idea is based on our previous [40] as well as present findings that the increased H2O2 was observed in both EC (arrow head) and SMC (arrow) (Fig. 5a). In addition, functional A2AAR is reported to be expressed on both EC and SMC [9], which is in agreement with our previous data showing positive A2AAR staining in both cell types of isolated mouse coronary arteries [40]. Therefore, it is reasonable to speculate that adenosine may act through A2AAR in both cell types producing H2O2, and activation of A2AAR-H2O2 pathway may result in different downstream vasodilator mechanisms, as activation of A2AAR in different cell types of coronary artery stimulates either cAMP-PKA or cGMP-PKG signaling [9, 12, 58, 59]. On the other hand, the cross talk of these vasodilator mechanisms exists. For instance, there is a possibility that endothelium-derived H2O2 may elicit further H2O2 production in SMC through activation of Nox [60], independent of A2AAR activation on SMC to activate PKG pathway, as H2O2 has been shown to be able to regulate both PKA and PKG pathways [61]. Taken together, these findings support Nox2 as a relevant source for H2O2 in the coronary vasculature [32] and suggest that the involvement of Nox2-derived ROS in adenosine-mediated coronary vasodilation is through H2O2 production.
Involvement of Nox2-derived ROS in adenosine-induced increase in coronary flow requires the presence of A2AARs
We [9, 16] and others [14, 15] have demonstrated that adenosine-mediated coronary vasodilation is predominantly attributed to A2AAR as compared to vasodilator A2BAR. In contrast, adenosine A1 and A3ARs counteract the adenosine-mediated coronary vasodilation [15, 17, 18]. Despite the findings that all four adenosine receptor subtypes are expressed and functional in the coronary vasculature [13], their roles in regulating coronary vascular tone through generation of ROS remain unclear. Given the fact that Nox has been suggested to be the major source of ROS in coronary vasculature [32, 52], it is not clear whether this leads to the generation of ROS by adenosine. We recently found that activation of A3AR enhances Nox2-derived ROS contributing to vascular contraction in mouse aorta [33], suggesting a role of A3AR in relation to ROS production. In contrast, in the coronary circulation, we have already excluded the involvement of A1 and A3ARs in increased flow that is coupled to the activation of Nox2 [37].
In the present study, we further investigated the role of adenosine A2A and A2BARs in this process. We found that adenosine A2A but not A2BARs are required for Nox2-mediated coronary vasodilation, as evidenced by the attenuation of adenosine-increased coronary flow by Nox2 inhibition (gp91 ds-tat) or ROS scavenging (EUK134) in both WT and A2BAR KO mice but not in A2AAR KO mice. Furthermore, the selective A2AAR stimulation-induced increase in coronary flow was significantly attenuated by Nox2 inhibition in both WT and A2BAR KO mice, whereas Nox2 inhibition failed to blunt the increased coronary flow upon the selective A2BAR stimulation in WT mice. Taken together, these findings suggest that A2AAR could be a potential mediator of adenosine-induced ROS generation contributing to increases in coronary flow. Moreover, in the isolated coronary arteries, adenosine-increased H2O2 formation was inhibited by Nox2 blockade in both WT and A2BAR KO mice but not in A2AAR KO mice, further suggesting that the involvement of Nox2-derived ROS in adenosine-induced increase in coronary flow requires the presence of A2AAR. In contrast to coronary circulation, both A2A and A2BARs have been observed to generate ROS accounting for the vasodilation in cerebral circulation [35]. This discrepancy may be due to the different distribution, expression, and the downstream effectors of A2A and A2BARs between these vascular beds [12, 13].
A2AAR-mediated coronary vasodilation through Nox2-derived ROS observed in the present study may bolster the functional influences of A2AAR by integrating the vascular actions of the generated ROS to the previously known A2AAR-mediated releases of other vasodilators (such as NO [20] and prostacyclin [62]). Furthermore, pertinent to this effect, it may also be possible that adenosine-generated ROS is involved in mediating reactive or functional hyperemia, despite the role of adenosine in functional hyperemia remaining controversial [19]. Indeed, the involvement of adenosine in either reactive or functional hyperemia has been shown to be mediated by endogenously released vasodilators, including NO, prostacyclin, and endothelium-derived hyperpolarizing factor [38, 40, 63, 64]. The influence and interaction of these vasoactive factors in generation of ROS by A2AAR in the coronary circulation remain unclear. Although we previously reported that NO and H2O2 function in a parallel manner contributing to the reactive hyperemia [40], future studies focusing on this aspect will provide greater insights into the interaction of these vasoactive factors.
Conclusions and implications
The present study shows that adenosine induces generation of ROS (likely H2O2) through the activation of Nox2 resulting in coronary vasodilation. This whole process requires the presence of A2AAR. These findings further support NADPH oxidase as a functionally relevant source of ROS for regulating coronary vascular tone. As both adenosine and ROS are generated and released under pathophysiological conditions [9, 65] and generated ROS mainly exerts a detrimental influence on the coronary vasculature [54–56], future studies addressing the role of A2AAR in ROS-mediated coronary vascular tone regulation in pathophysiological conditions will bring us greater insights into mechanisms underlying ischemic heart disease.
Acknowledgments
The authors would like to thank Dr. Xueping Zhou for assistance with the immunofluorescence staining studies. This study was supported by NIH grants of HL027339, HL09444, HL071802, and U54GM104942.
References
- 1.Feigl EO. Coronary physiology. Physiol Rev. 1983;63(1):1–205. doi: 10.1152/physrev.1983.63.1.1. [DOI] [PubMed] [Google Scholar]
- 2.Duncker DJ, Bache RJ, Merkus D. Regulation of coronary resistance vessel tone in response to exercise. J Mol Cell Cardiol. 2012;52(4):802–813. doi: 10.1016/j.yjmcc.2011.10.007. [DOI] [PubMed] [Google Scholar]
- 3.Duncker DJ, Bache RJ. Regulation of coronary blood flow during exercise. Physiol Rev. 2008;88(3):1009–1086. doi: 10.1152/physrev.00045.2006. [DOI] [PubMed] [Google Scholar]
- 4.Durand MJ, Gutterman DD. Diversity in mechanisms of endothelium-dependent vasodilation in health and disease. Microcirculation. 2013;20(3):239–247. doi: 10.1111/micc.12040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Deussen A, Ohanyan V, Jannasch A, Yin L, Chilian W. Mechanisms of metabolic coronary flow regulation. J Mol Cell Cardiol. 2012;52(4):794–801. doi: 10.1016/j.yjmcc.2011.10.001. [DOI] [PubMed] [Google Scholar]
- 6.Zhou Z, de Beer VJ, Bender SB, Jan Danser AH, Merkus D, Laughlin MH, Duncker DJ. Phosphodiesterase-5 activity exerts a coronary vasoconstrictor influence in awake swine that is mediated in part via an increase in endothelin production. Am J Physiol Heart Circ Physiol. 2014;306(6):H918–927. doi: 10.1152/ajpheart.00331.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Berne RM. Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. Am J Physiol. 1963;204:317–322. doi: 10.1152/ajplegacy.1963.204.2.317. [DOI] [PubMed] [Google Scholar]
- 8.Tune JD, Gorman MW. Feigl EO (2004) Matching coronary blood flow to myocardial oxygen consumption. J Appl Physiol. 1985;97(1):404–415. doi: 10.1152/japplphysiol.01345.2003. [DOI] [PubMed] [Google Scholar]
- 9.Mustafa SJ, Morrison RR, Teng B, Pelleg A. Adenosine receptors and the heart: role in regulation of coronary blood flow and cardiac electrophysiology. Handb Exp Pharmacol. 2009;193:161–188. doi: 10.1007/978-3-540-89615-9_6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Duncker DJ, Stubenitsky R, Verdouw PD. Role of adenosine in the regulation of coronary blood flow in swine at rest and during treadmill exercise. Am J Physiol. 1998;275(5 Pt 2):H1663–1672. doi: 10.1152/ajpheart.1998.275.5.H1663. [DOI] [PubMed] [Google Scholar]
- 11.Edlund A, Conradsson T, Sollevi A. A role for adenosine in coronary vasoregulation in man. Effects of theophylline and enprofylline. Clin Physiol. 1995;15(6):623–636. doi: 10.1111/j.1475-097X.1995.tb00549.x. [DOI] [PubMed] [Google Scholar]
- 12.Burnstock G, Ralevic V. Purinergic signaling and blood vessels in health and disease. Pharmacol Rev. 2014;66(1):102–192. doi: 10.1124/pr.113.008029. [DOI] [PubMed] [Google Scholar]
- 13.Headrick JP, Ashton KJ, Rose'meyer RB, Peart JN. Cardiovascular adenosine receptors: expression, actions and interactions. Pharmacol Ther. 2013;140(1):92–111. doi: 10.1016/j.pharmthera.2013.06.002. [DOI] [PubMed] [Google Scholar]
- 14.Flood A, Headrick JP. Functional characterization of coronary vascular adenosine receptors in the mouse. Br J Pharmacol. 2001;133(7):1063–1072. doi: 10.1038/sj.bjp.0704170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sato A, Terata K, Miura H, Toyama K, Loberiza FR, Jr, Hatoum OA, Saito T, Sakuma I, Gutterman DD. Mechanism of vasodilation to adenosine in coronary arterioles from patients with heart disease. Am J Physiol Heart Circ Physiol. 2005;288(4):H1633–1640. doi: 10.1152/ajpheart.00575.2004. [DOI] [PubMed] [Google Scholar]
- 16.Sanjani MS, Teng B, Krahn T, Tilley S, Ledent C, Mustafa SJ (2011) Contributions of A2A and A2B adenosine receptors in coronary flow responses in relation to the KATP channel using A2B and A2A/2B double-knockout mice. Am J Physiol Heart Circ Physiol 301(6):H2322–2333 [DOI] [PMC free article] [PubMed]
- 17.Talukder MA, Morrison RR, Jacobson MA, Jacobson KA, Ledent C, Mustafa SJ (2002) Targeted deletion of adenosine A(3) receptors augments adenosine-induced coronary flow in isolated mouse heart. Am J Physiol Heart Circ Physiol 282(6):H2183–2189 [DOI] [PMC free article] [PubMed]
- 18.Tawfik HE, Teng B, Morrison RR, Schnermann J, Mustafa SJ (2006) Role of A1 adenosine receptor in the regulation of coronary flow. Am J Physiol Heart Circ Physiol 291(1):H467–472 [DOI] [PubMed]
- 19.Zhou X, Teng B, Tilley S, Ledent C, Mustafa SJ (2014) Metabolic hyperemia requires ATP-sensitive K+ channels and H2O2 but not adenosine in isolated mouse hearts. Am J Physiol Heart Circ Physiol 307(7):H1046–1055 [DOI] [PMC free article] [PubMed]
- 20.Teng B, Ledent C, Mustafa SJ (2008) Up-regulation of A2B adenosine receptor in A2A adenosine receptor knockout mouse coronary artery. J Mol Cell Cardiol 44(5):905–914 [DOI] [PMC free article] [PubMed]
- 21.Fredholm BB, Arslan G, Halldner L, Kull B, Schulte G, Wasserman W. Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch Pharmacol. 2000;362(4-5):364–374. doi: 10.1007/s002100000313. [DOI] [PubMed] [Google Scholar]
- 22.Liu Y, Bubolz AH, Mendoza S, Zhang DX, Gutterman DD (2011) H2O2 is the transferrable factor mediating flow-induced dilation in human coronary arterioles. Circ Res 108(5):566–573 [DOI] [PMC free article] [PubMed]
- 23.Rogers PA, Dick GM, Knudson JD, Focardi M, Bratz IN, Swafford AN Jr, Saitoh S, Tune JD, Chilian WM (2006) H2O2-induced redox-sensitive coronary vasodilation is mediated by 4-aminopyridine-sensitive K+ channels. Am J Physiol Heart Circ Physiol 291(5):H2473–2482 [DOI] [PubMed]
- 24.Yada T, Shimokawa H, Hiramatsu O, Kajita T, Shigeto F, Goto M, Ogasawara Y, Kajiya F. Hydrogen peroxide, an endogenous endothelium-derived hyperpolarizing factor, plays an important role in coronary autoregulation in vivo. Circulation. 2003;107(7):1040–1045. doi: 10.1161/01.CIR.0000050145.25589.65. [DOI] [PubMed] [Google Scholar]
- 25.Owen MK, Witzmann FA, McKenney ML, Lai X, Berwick ZC, Moberly SP, Alloosh M, Sturek M, Tune JD. Perivascular adipose tissue potentiates contraction of coronary vascular smooth muscle: influence of obesity. Circulation. 2013;128(1):9–18. doi: 10.1161/CIRCULATIONAHA.112.001238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yada T, Shimokawa H, Hiramatsu O, Shinozaki Y, Mori H, Goto M, Ogasawara Y, Kajiya F. Important role of endogenous hydrogen peroxide in pacing-induced metabolic coronary vasodilation in dogs in vivo. J Am Coll Cardiol. 2007;50(13):1272–1278. doi: 10.1016/j.jacc.2007.05.039. [DOI] [PubMed] [Google Scholar]
- 27.Takac I, Schroder K, Brandes RP. The Nox family of NADPH oxidases: friend or foe of the vascular system? Curr Hypertens Rep. 2012;14(1):70–78. doi: 10.1007/s11906-011-0238-3. [DOI] [PubMed] [Google Scholar]
- 28.Amanso AM, Griendling KK. Differential roles of NADPH oxidases in vascular physiology and pathophysiology. Front Biosci (Schol Ed) 2012;4:1044–1064. doi: 10.2741/S317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Frazziano G, Champion HC, Pagano PJ. NADPH oxidase-derived ROS and the regulation of pulmonary vessel tone. Am J Physiol Heart Circ Physiol. 2012;302(11):H2166–2177. doi: 10.1152/ajpheart.00780.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Schroder K. Isoform specific functions of Nox protein-derived reactive oxygen species in the vasculature. Curr Opin Pharmacol. 2010;10(2):122–126. doi: 10.1016/j.coph.2010.01.002. [DOI] [PubMed] [Google Scholar]
- 31.Taverne YJ, Bogers AJ, Duncker DJ, Merkus D. Reactive oxygen species and the cardiovascular system. Oxid Med Cell Longev. 2013;2013:862423. doi: 10.1155/2013/862423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Larsen BT, Bubolz AH, Mendoza SA, Pritchard KA, Jr, Gutterman DD. Bradykinin-induced dilation of human coronary arterioles requires NADPH oxidase-derived reactive oxygen species. Arterioscler Thromb Vasc Biol. 2009;29(5):739–745. doi: 10.1161/ATVBAHA.108.169367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.El-Awady MS, Ansari HR, Fil D, Tilley SL, Mustafa SJ (2011) NADPH oxidase pathway is involved in aortic contraction induced by A3 adenosine receptor in mice. J Pharmacol Exp Ther 338(2):711–717 [DOI] [PMC free article] [PubMed]
- 34.Carlstrom M, Lai EY, Ma Z, Patzak A, Brown RD, Persson AE. Role of NOX2 in the regulation of afferent arteriole responsiveness. Am J Physiol Regul Integr Comp Physiol. 2009;296(1):R72–79. doi: 10.1152/ajpregu.90718.2008. [DOI] [PubMed] [Google Scholar]
- 35.Gebremedhin D, Weinberger B, Lourim D, Harder DR. Adenosine can mediate its actions through generation of reactive oxygen species. J Cereb Blood Flow Metab. 2010;30(10):1777–1790. doi: 10.1038/jcbfm.2010.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Narayan P, Mentzer RM Jr, Lasley RD (2001) Adenosine A1 receptor activation reduces reactive oxygen species and attenuates stunning in ventricular myocytes. J Mol Cell Cardiol 33(1):121–129 [DOI] [PubMed]
- 37.El-Awady MS, Rajamani U, Teng B, Tilley SL, Mustafa SJ. Evidence for the involvement of NADPH oxidase in adenosine receptors-mediated control of coronary flow using A and A knockout mice. Physiol Rep. 2013;1(3) doi: 10.1002/phy2.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sharifi-Sanjani M, Zhou X, Asano S, Tilley S, Ledent C, Teng B, Dick GM, Mustafa SJ (2013) Interactions between A(2A) adenosine receptors, hydrogen peroxide, and KATP channels in coronary reactive hyperemia. Am J Physiol Heart Circ Physiol 304(10):H1294–1301 [DOI] [PMC free article] [PubMed]
- 39.Teng B, Fil D, Tilley SL, Ledent C, Krahn T, Mustafa SJ. Functional and RNA expression profile of adenosine receptor subtypes in mouse mesenteric arteries. J Cardiovasc Pharmacol. 2013;61(1):70–76. doi: 10.1097/FJC.0b013e318278575e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhou X, Teng B, Tilley S, Mustafa SJ (2013) A1 adenosine receptor negatively modulates coronary reactive hyperemia via counteracting A2A-mediated H2O2 production and KATP opening in isolated mouse hearts. Am J Physiol Heart Circ Physiol 305(11):H1668–1679 [DOI] [PMC free article] [PubMed]
- 41.Ponnoth DS, Sanjani MS, Ledent C, Roush K, Krahn T, Mustafa SJ (2009) Absence of adenosine-mediated aortic relaxation in A(2A) adenosine receptor knockout mice. Am J Physiol Heart Circ Physiol 297(5):H1655–1660 [DOI] [PMC free article] [PubMed]
- 42.Rey FE, Cifuentes ME, Kiarash A, Quinn MT, Pagano PJ (2001) Novel competitive inhibitor of NAD(P)H oxidase assembly attenuates vascular O(2)(-) and systolic blood pressure in mice. Circ Res 89(5):408–414 [DOI] [PubMed]
- 43.van Nierop BJ, Coolen BF, Bax NA, Dijk WJ, van Deel ED, Duncker DJ, Nicolay K, Strijkers GJ. Myocardial perfusion MRI shows impaired perfusion of the mouse hypertrophic left ventricle. Int J Cardiovasc Imaging. 2014;30(3):619–628. doi: 10.1007/s10554-014-0369-0. [DOI] [PubMed] [Google Scholar]
- 44.Talukder MA, Morrison RR, Ledent C, Mustafa SJ (2003) Endogenous adenosine increases coronary flow by activation of both A2A and A2B receptors in mice. J Cardiovasc Pharmacol 41(4):562–570 [DOI] [PubMed]
- 45.Tune JD, Richmond KN, Gorman MW, Olsson RA, Feigl EO. Adenosine is not responsible for local metabolic control of coronary blood flow in dogs during exercise. Am J Physiol Heart Circ Physiol. 2000;278(1):H74–84. doi: 10.1152/ajpheart.2000.278.1.H74. [DOI] [PubMed] [Google Scholar]
- 46.Headrick JP, Ely SW, Matherne GP, Berne RM. Myocardial adenosine, flow, and metabolism during adenosine antagonism and adrenergic stimulation. Am J Physiol. 1993;264(1 Pt 2):H61–70. doi: 10.1152/ajpheart.1993.264.1.H61. [DOI] [PubMed] [Google Scholar]
- 47.Dole WP, Yamada N, Bishop VS, Olsson RA. Role of adenosine in coronary blood flow regulation after reductions in perfusion pressure. Circ Res. 1985;56(4):517–524. doi: 10.1161/01.RES.56.4.517. [DOI] [PubMed] [Google Scholar]
- 48.Saito D, Steinhart CR, Nixon DG, Olsson RA. Intracoronary adenosine deaminase reduces canine myocardial reactive hyperemia. Circ Res. 1981;49(6):1262–1267. doi: 10.1161/01.RES.49.6.1262. [DOI] [PubMed] [Google Scholar]
- 49.Morrison RR, Talukder MA, Ledent C, Mustafa SJ (2002) Cardiac effects of adenosine in A(2A) receptor knockout hearts: uncovering A(2B) receptors. Am J Physiol Heart Circ Physiol 282(2):H437–444 [DOI] [PubMed]
- 50.Berwick ZC, Payne GA, Lynch B, Dick GM, Sturek M, Tune JD (2010) Contribution of adenosine A(2A) and A(2B) receptors to ischemic coronary dilation: role of K(V) and K(ATP) channels. Microcirculation 17(8):600–607 [DOI] [PMC free article] [PubMed]
- 51.Zhou Z, Merkus D, Cheng C, Duckers HJ, Jan Danser AH, Duncker DJ. Uridine adenosine tetraphosphate is a novel vasodilator in the coronary microcirculation which acts through purinergic P1 but not P2 receptors. Pharmacol Res. 2013;67(1):10–17. doi: 10.1016/j.phrs.2012.09.011. [DOI] [PubMed] [Google Scholar]
- 52.Feng J, Damrauer SM, Lee M, Sellke FW, Ferran C, Abid MR. Endothelium-dependent coronary vasodilatation requires NADPH oxidase-derived reactive oxygen species. Arterioscler Thromb Vasc Biol. 2010;30(9):1703–1710. doi: 10.1161/ATVBAHA.110.209726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Shafique E, Choy WC, Liu Y, Feng J, Cordeiro B, Lyra A, Arafah M, Yassin-Kassab A, Zanetti AV, Clements RT, Bianchi C, Benjamin LE, Sellke FW, Abid MR. Oxidative stress improves coronary endothelial function through activation of the pro-survival kinase AMPK. Aging (Albany NY) 2013;5(7):515–530. doi: 10.18632/aging.100569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lu T, Chai Q, Yu L, d'Uscio LV, Katusic ZS, He T, Lee HC. Reactive oxygen species signaling facilitates FOXO-3a/FBXO-dependent vascular BK channel beta1 subunit degradation in diabetic mice. Diabetes. 2012;61(7):1860–1868. doi: 10.2337/db11-1658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lu X, Dang CQ, Guo X, Molloi S, Wassall CD, Kemple MD, Kassab GS. Elevated oxidative stress and endothelial dysfunction in right coronary artery of right ventricular hypertrophy. J Appl Physiol (1985) 2011;110(6):1674–1681. doi: 10.1152/japplphysiol.00744.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kassan M, Choi SK, Galan M, Lee YH, Trebak M, Matrougui K. Enhanced p22phox expression impairs vascular function through p38 and ERK1/2 MAP kinase-dependent mechanisms in type 2 diabetic mice. Am J Physiol Heart Circ Physiol. 2014;306(7):H972–980. doi: 10.1152/ajpheart.00872.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lawler JM, Kim JH, Kwak HB, Barnes WS. Redox modulation of diaphragm contractility: Interaction between DHPR and RyR channels. Free Radic Biol Med. 2010;49(12):1969–1977. doi: 10.1016/j.freeradbiomed.2010.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Rekik M, Mustafa JS (2003) Modulation of A2A adenosine receptors and associated Galphas proteins by ZM 241385 treatment of porcine coronary artery. J Cardiovasc Pharmacol 42(6):736–744 [DOI] [PubMed]
- 59.Olanrewaju HA, Mustafa SJ (2000) Adenosine A(2A) and A(2B) receptors mediated nitric oxide production in coronary artery endothelial cells. Gen Pharmacol 35(3):171–177 [DOI] [PubMed]
- 60.Cai H. Hydrogen peroxide regulation of endothelial function: origins, mechanisms, and consequences. Cardiovasc Res. 2005;68(1):26–36. doi: 10.1016/j.cardiores.2005.06.021. [DOI] [PubMed] [Google Scholar]
- 61.Burgoyne JR, Eaton P (2013) Detecting disulfide-bound complexes and the oxidative regulation of cyclic nucleotide-dependent protein kinases by H2O2. Methods Enzymol 528:111–128 [DOI] [PubMed]
- 62.Maddock HL, Broadley KJ, Bril A, Khandoudi N. Effects of adenosine receptor agonists on guinea-pig isolated working hearts and the role of endothelium and NO. J Pharm Pharmacol. 2002;54(6):859–867. doi: 10.1211/0022357021779041. [DOI] [PubMed] [Google Scholar]
- 63.Otomo J, Nozaki N, Tomoike H. Roles of nitric oxide and adenosine in the regulation of coronary conductance in the basal state and during reactive hyperemia. Jpn Circ J. 1997;61(5):441–449. doi: 10.1253/jcj.61.441. [DOI] [PubMed] [Google Scholar]
- 64.Duffy SJ, Castle SF, Harper RW, Meredith IT. Contribution of vasodilator prostanoids and nitric oxide to resting flow, metabolic vasodilation, and flow-mediated dilation in human coronary circulation. Circulation. 1999;100(19):1951–1957. doi: 10.1161/01.CIR.100.19.1951. [DOI] [PubMed] [Google Scholar]
- 65.Cai H, Griendling KK, Harrison DG. The vascular NAD(P)H oxidases as therapeutic targets in cardiovascular diseases. Trends Pharmacol Sci. 2003;24(9):471–478. doi: 10.1016/S0165-6147(03)00233-5. [DOI] [PubMed] [Google Scholar]




