Skip to main content
Purinergic Signalling logoLink to Purinergic Signalling
. 2023 Feb 13;19(3):551–564. doi: 10.1007/s11302-023-09924-3

In vivo phenotypic validation of adenosine receptor-dependent activity of non-adenosine drugs

Cuiying Xiao 1,#, Oksana Gavrilova 2,#, Naili Liu 2, Sarah A Lewicki 3, Marc L Reitman 1, Kenneth A Jacobson 3,
PMCID: PMC10539256  PMID: 36781825

Abstract

Some non-adenosinergic drugs are reported to also act through adenosine receptors (ARs). We used mouse hypothermia, which can be induced by agonism at any of the four ARs, as an in vivo screen for adenosinergic effects. An AR contribution was identified when a drug caused hypothermia in wild type mice that was diminished in mice lacking all four ARs (quadruple knockout, QKO). Alternatively, an adenosinergic effect was identified if a drug potentiated adenosine-induced hypothermia. Four drugs (dipyridamole, nimodipine, cilostazol, cyclosporin A) increased the hypothermia caused by adenosine. Dipyridamole and nimodipine probably achieved this by inhibition of adenosine clearance via ENT1. Two drugs (cannabidiol, canrenoate) did not cause hypothermia in wild type mice. Four other drugs (nifedipine, ranolazine, ketamine, ethanol) caused hypothermia, but the hypothermia was unchanged in QKO mice indicating non-adenosinergic mechanisms. Zinc chloride caused hypothermia and hypoactivity; the hypoactivity was blunted in the QKO mice. Interestingly, the antidepressant amitriptyline caused hypothermia in wild type mice that was amplified in the QKO mice. Thus, we have identified adenosine-related effects for some drugs, while other candidates do not affect adenosine signaling by this in vivo assay. The adenosine-modulating drugs could be considered for repurposing based on predicted effects on AR activation.

Supplementary Information

The online version contains supplementary material available at 10.1007/s11302-023-09924-3.

Keywords: Purinergic receptors, Nucleosides, Adenosine receptor, Prodrug, Pain, Steatohepatitis, Drug repurposing, Equilibrative nucleoside transport, Hypothermia, Knockout mice

Introduction

Adenosine acts as an autocrine or paracrine signal by activating four G protein-coupled receptors (adenosine receptors, ARs) that have been the focus of extensive medicinal chemical and drug development efforts [17]. Generally, extracellular adenosine elicits protective actions to restore the stability of an organism in response to challenges or stresses. The ARs are distributed widely throughout the body and mediate local, often tissue-specific, effects [1, 2]. There is a rich history and experience in the development of selective synthetic AR agonists [2, 3, 5, 7].

In addition to drugs developed for their agonism or antagonism at the four ARs, some drugs designed for other actions coincidentally act as AR antagonists [8]. One example of off-target binding of an approved drug is the antimalarial mefloquine, which is an A2A receptor antagonist [9]. In fact, most ligand chemotypes found fortuitously or by computational approaches to bind to ARs do so as antagonists [1012]. Other drugs with coincidental AR antagonism include experimental Alzheimer’s drug etazolate, dopamine agonist (3,4-dihydroxy-phenylamino)-2-imidazoline (DPI) [10, 13], anxiogenic β-carbolines (e.g. methyl 6,7-dimethoxy-4-ethyl-β-carboline-3-carboxylate) [14], flavonoid derivatives hispidol and galangin [15], and 1,4-dihydropyridines such as nicardipine [16]. Drugs listed in databases as binding to the A2AAR include tamoxifen, imiquimod, and sildenafil [17] (Table S1).

Other drugs enhance AR signaling indirectly, by increasing adenosine availability to the ARs. This can occur by inhibiting adenosine’s cellular uptake, metabolism, and/or degradation, raising extracellular adenosine levels [1820]. For example, diverse compounds inhibit nucleoside uptake through the equilibrative transporters (ENT1–3) or concentrative (CNTs) transporters of the SLC29 family. The antithrombotic P2Y12 receptor antagonist ticagrelor and ethanol and cannabidiol are all reported to raise adenosine levels by inhibiting its transport [2124]. In a screen of 1625 diverse molecules, more than half bound to ENT1 with a Ki value < 10 µM [25], suggesting that additional drugs may share this property. Other compounds inhibit intracellular adenosine kinase, thereby reducing cellular uptake of adenosine via equilibrative transporters. The antimetabolite methotrexate increases intracellular adenosine levels (and thus indirectly extracellular levels) by increasing levels of 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), and this is proposed to contribute to methotrexate’s therapeutic benefit in rheumatoid arthritis [26]. For still other compounds, the mechanism of action (MoA) leading to activation of one or more AR subtypes is unknown [8].

Pharmacological modulation of ARs can be evaluated in vivo using mouse models in which specific components of the signaling pathways are genetically deleted [27, 28]. Mice with one or more of the ARs genetically knocked out (KO mice), either globally or in a tissue-specific manner, are important tools for exploring the interaction of drugs with pathways [2731]. They are particularly useful for the ARs, since adenosine often acts locally so the relevant primary site of systemic adenosinergic drug action can be difficult to determine. A mouse line in which all four ARs have been globally deleted is useful for investigating adenosine physiology [27]. At baseline these mice resemble wild type mice by most criteria examined, including body temperature regulation (diurnal variation, response to stress, and torpor), suggesting that the ARs are more important in allostatic rather than homeostatic functions.

Adenosine can cause hypothermia (and hypoactivity) in mice by individual activation of each of the four AR [29, 30, 32], and the quadruple AR knockout mice (QKOs) no longer respond to adenosine administration [27]. Here we evaluated compounds that have previously been reported to have adenosinergic actions, using mouse hypothermia as a sensitive, standardized assay, with a goal of identifying drugs that might be repurposed for altering adenosinergic signaling.

Methods

Chemicals and mice

Chemicals were of reagent grade and obtained from Sigma-Aldrich (St. Louis, MO), unless noted. All compounds were administered intraperitoneally (i.p., 10 ml/g body weight). Cannabidiol (CBD, stored at –80 °C) was dissolved freshly in 1:1:18 dimethyl sulfoxide (DMSO):Tween 80:saline prior to the injection (10 mg/kg). Cilostazol (10 mg/kg), dipyridamole (10 and 30 mg/kg), nimodipine (10, 20, and 30 mg/kg), and nifedipine (10 and 20 mg/kg, Tocris), sildefanil citrate (1, 3, 10, and 30 mg/kg), cyclosporine (30 mg/kg, Tocris) were dissolved in 15:15:70 DMSO: Kolliphor EL: saline. Canrenoate (0.3, 1 and 3 mg/kg), ZnCl2 (1, 3, 10 and 30 mg/kg), ethanol (1, 2, and 3 g/kg), and ketamine (Zetamine, VetOne, 1, 3, 10, and 30 mg/kg) were dissolved in saline, ranolazine (25 and 50 mg/kg) in PBS, and amitriptyline (20 mg/kg) in 10% DMSO. The animal protocol for the in vivo studies was approved by the NIDDK Animal Care and Use Committee. All experiments were performed on male mice. QKO mice on a mixed genetic background were generated as reported and compared to male wild type C57BL/6 J mice (Jackson Laboratories, Bar Harbor, ME) as controls [27]. Mice were kept at ~ 21–22 °C in a 12:12-h light–dark cycle, and chow (NIH-07, Envigo Inc., Madison, WI) and water were provided ad libitum.

Body temperature

Surgical operations to implant G2 E-mitters intraperitoneally were performed on the mice at least seven days prior to experimentation. Core body temperature (Tb) and locomotor activity were measured continuously by telemetry (ER4000 energizer/receivers, and VitalView software, Starr Life Sciences, Oakmont, PA) with data collection intervals of 1 min. The Tb response was followed for up to 24 h after drug injection. The indicated drug was injected 20–25 min before adenosine (100 mg/kg, i.p.). Two standard analysis intervals were used to calculate mean Tb. Using 0–60 min after injection (timing from the second injection when two injections were done) includes the increase in Tb and physical activity due to handling and is a sensitive measure, able to detect transient or small hypothermic effects. The mean Tb and time below 34 °C measured 0–300 min after injection better discriminate larger, longer duration effects.

Statistics

All data are expressed as the mean ± SEM. Data were tested for statistical significance.

by two-tailed, unpaired Student’s t test, or two-way ANOVA followed by post hoc Holm-Sidak multiple comparison tests as appropriate. A P value of less than 0.05 was considered significant.

Results

To assess in vivo action of putative adenosinergic drugs, we used mouse hypothermia. Single doses of test compounds were administered i.p., and the effect on Tb and locomotor activity were monitored by telemetry in C57BL/6 J (WT) mice. An AR contribution was identified when a drug caused hypothermia in WT mice that was diminished in mice lacking all four AR receptors (QKO mice). In addition, we also tested the ability of drugs to potentiate adenosine-induced hypothermia in WT mice. The properties of the tested compounds and the proposed interaction with adenosine system are shown in Table 1; the summary of the results is in Table 2.

Table 1.

Compounds examined for effects on adenosinergic signaling

Compound Use or indicationa Main mechanism Proposed Interaction with adenosine system Human ENT1, Ki or KD (nM) Reference for adenosine interaction
Dipyridamole Vasodilator PDE3 inhibitor, ENT1 inhibitor ENT1 inhibition 2.6 [21]
Nimodipine Subarachnoid hemorrhage Ca+2 channel blocker ENT1 inhibition 52 [33]
Cilostazol Anti-claudication PDE3 inhibitor NDb 10,000 [25]
Cyclosporine A Immuno-suppressant Calcineurin inhibitor Inhibition of adenosine uptake ND *
Cannabidiol Antiepileptic unclear ENT1 inhibition 200 [23, 3638]
Canrenoate Diuretic Mineralocorticoid antagonist ND ND [40]
Nifedipine Anti-hypertensive Ca+2 channel blocker ND 13,700 [43]
Ranolazine Anti-angina Sodium current inhibition Increased cardiac adenosine ND [44]
Ketamine Antidepressant NMDA receptor antagonist Stimulation of presynaptic A1AR ND [46]
Ethanol Psychoactive drug Ion channel modulator Multiple 200,000,000 [4749]
Zinc chloride Zinc deficiency Unclear ND ND [50]
Amitriptyline Antidepressant Serotonin/norepi-nephrine reuptake inhibitor ND ND [51]
Sildenafil Erectile dysfunction, vasodilator PDE5 inhibitor ND ND **, ***

aAll except ethanol are approved by the United States FDA

bND, not determined

*Guieu et al. [34]

**Lee et al. [43]

***Table S1

Table 2.

Summary of drugs screened for adenosinergic effects using mouse hypothermia. aAll drugs given i.p. b100 mg/kg adenosine, i.p. cIndicates significant drug × adenosine interaction by two-way ANOVA. dIndicates additive drug and adenosine effects without significant interaction by two-way ANOVA. eND, not determined. fQKO has more hypothermia. Color key: Green indicates significant reduction. Orange indicates the effect in QKO is different from wild type mice. Blue indicates that drug + adenosine has a different effect than either alone. Gray indicates that results demonstrate the drug has an adenosinergic effect

graphic file with name 11302_2023_9924_Tab2_HTML.jpg

Drugs inhibiting adenosine transport

Inhibition of adenosine transport (Table 1) increases extracellular adenosine concentrations [2]. We reported previously that the benchmark ENT1 inhibitor 6-S-[(4-nitrophenyl)methyl]-6-thioinosine (NBMPR, 1 mg/kg i.p.) induced a slight hypothermia in mice and profoundly increased the hypothermic effect of a subsequent dose of adenosine (100 mg/kg i.p.) [27].

The vasodilator dipyridamole inhibits both PDE3 and ENT1 [21]. Dipyridamole (10 mg/kg i.p.) itself produced no hypothermia but increased the hypothermia caused by subsequent adenosine (100 mg/kg i.p.), with a significant adenosine × dipyridamole interaction (Fig. 1, Table S1). The likely explanation for dipyridamole’s adenosinergic effects is inhibition of ENT1. However, a higher dipyridamole dose (30 mg/kg i.p.) increased activity (Fig. 1c, f) with a nonsignificant rise of Tb (Fig. 1a, b, d). This dose has not been tested in QKO mice or studied further.

Fig. 1.

Fig. 1

Effects of dipyridamole. a Vehicle or dipyridamole (10 or 30 mg/kg) treatment of WT mice. b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 7–8/group. g Treatment of QKO and control mice with dipyridamole (10 mg/kg). h Mean Tb at 0–60 min, i mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 4–6/group. m Effect of dipyridamole (10 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. n Mean Tb at 0–60 min, o mean activity at 0–60 min, p mean Tb at 0–300 min, q time below 34 °C, and r mean activity at 0–300 min; n = 5–6/group. Statistical analyses are in Table S2

The Ca2+ channel blocker nimodipine is coincidentally an ENT1 inhibitor [33]. Nimodipine (10, 20 mg/kg i.p.) caused hypothermia and hypoactivity that were diminished in QKO mice (Fig. 2a–l). Nimodipine treatment also augmented adenosine-induced hypothermia (Fig. 2mr). Thus, nimodipine has both adenosinergic (such as via ENT1 inhibition) and non-adenosinergic (not lost in the QKO mice) actions.

Fig. 2.

Fig. 2

Effects of nimodipine. a Vehicle or nimodipine (10 or 20 mg/kg) treatment of WT mice. b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 7–11/group. g Treatment of QKO and control mice with nimodipine (20 mg/kg). h Mean Tb at 0–60 min, (i) mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 5–6/group. m Effect of nimodipine (10 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. n Mean Tb at 0–60 min, o mean activity at 0–60 min, p mean Tb at 0–300 min, q time below 34 °C, and r mean activity at 0–300 min; n = 5–6/group. Statistical analyses are in Table S2

The quinolinone PDE3 inhibitor cilostazol [25] caused hypoactivity and non-significant hypothermia and augmented adenosine-induced hypothermia but is a poor ENT1 inhibitor (Fig. 3). Taken together, these results suggest that at the doses tested, dipyridamole and nimodipine have some adenosinergic effects, likely by ENT1 inhibition, while cilostazol has a different mode of action.

Fig. 3.

Fig. 3

Effects of cilostazol (10 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. a Time course, b mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 10–11/group. Statistical analyses are in Table S2

The immunosuppressive drug cyclosporine A, a calcineurin inhibitor, was reported to inhibit adenosine uptake by red blood cells [34] and T-lymphocytes [35]. Cyclosporine A (30 mg/kg, i.p.) caused a slight hypothermia and hypoactivity that were not clearly reduced in QKO mice (Fig. 4a–f). However, cyclosporine treatment significantly increased adenosine-induced hypothermia (Fig. 4g–l). These data suggest that cyclosporine A has both adenosinergic and non-adenosinergic action.

Fig. 4.

Fig. 4

Effects of cyclosporine A. a Treatment of QKO and control mice with cyclosporine A (30 mg/kg). b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 7–14/group. g Effect of cyclosporine A (30 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. h Mean Tb at 0–60 min, i mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 10–12/group. Statistical analyses are in Table S2

Drugs without hypothermic effects

The anti-epileptic drug cannabidiol, a putative ENT1 inhibitor [23, 3639], had no effect on body temperature and did not augment adenosine-induced hypothermia at 10 mg/kg, i.p. (Fig. S1).

The diuretic canrenoate has cardioprotective effects which are absent in mice lacking either CD73 or the A2BAR, suggesting dependence on extracellular adenosine [40]. However, canrenoate (0.3, 1, and 3 mg/kg, i.p.) did not reduce Tb in WT mice (Fig. S2). At 3 mg/kg, canrenoate tended to increase Tb and, therefore, was not investigated further.

The PDE5 inhibitor sildenafil may modulate antinociception via multiple AR subtypes [41]. However, sildenafil (1, 3, 10, and 30 mg/kg, i.p.) had no effect Tb in WT mice (Fig. S5). Therefore, alone it does not appear to have AR agonist activity, but we have not evaluated its possible indirect action.

Drugs with non-adenosinergic hypothermic effects

Nifedipine is a Ca2+ channel blocker used as an antihypertensive agent with a lower affinity at ENT1 but reported to have adenosinergic actions [42, 43]. Nifedipine (10 mg/kg) caused hypothermia without significant reduction of activity in both WT and QKO mice, suggesting non-adenosinergic action (Fig. 5a–l). However, nifedipine treatment potentiated adenosine-induced hypothermia in additive manner (Fig. 4m–r).

Fig. 5.

Fig. 5

Effects of nifedipine. a Vehicle or nifedipine (10 or 20 mg/kg) treatment of WT mice. b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 6–18/group. g Treatment of QKO and control mice with nifedipine (10 mg/kg). h Mean Tb at 0–60 min, i mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 10–18/group. m Effect of nifedipine (10 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. n Mean Tb at 0–60 min, o mean activity at 0–60 min, p mean Tb at 0–300 min, q time below 34 °C, and r mean activity at 0–300 min; n = 8–11/group. Statistical analyses are in Table S2

The anti-angina drug ranolazine may exert its beneficial effects by increasing myocardial adenosine levels [44]. Ranolazine (50 mg/kg, i.p.) itself reduced Tb and activity in both WT and QKO mice and had no effect on adenosine-induced hypothermia (Fig. 6).

Fig. 6.

Fig. 6

Effects of ranolazine. a Vehicle or ranolazine (12.5, 25, or 50 mg/kg) treatment of WT mice. b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 6–16/group. g Treatment of QKO and control mice with ranolazine (50 mg/kg). h Mean Tb at 0–60 min, i mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 8–10/group. m Effect of ranolazine (50 mg/kg) pretreatment on adenosine (100 mg/kg) induced hypothermia. n Mean Tb at 0–60 min, o mean activity at 0–60 min, p mean Tb at 0–300 min, q time below 34 °C, and r mean activity at 0–300 min; n = 5–6/group. Statistical analyses are in Table S2

The antidepressant ketamine was reported to boost adenosinergic signaling [45, 46]. Ketamine was tested at four doses (1, 3, 10, and 30 mg/kg, i.p.). Only the highest dose caused significant reduction of Tb with no changes in activity (Fig. S4a–f). However, the hypothermic effect was the same in WT and QKO (Fig. S4g–l), suggesting a non-adenosinergic MoA for the ketamine-induced hypothermia.

Ethanol may modulate adenosine signaling by multiple mechanisms [4749]. Ethanol (2 or 3 g/kg) reduced Tb similarly in both WT and QKO mice and did not potentiate adenosine-induced hypothermia, suggesting non-adenosinergic action (Fig. S5).

Zinc chloride elicits an antidepressant-like effect in the mouse model of forced swimming, with some of the effect attributed to enhanced adenosine signaling [50]. Zinc chloride induced a robust hypothermic effect at 10 mg/kg, but not at lower doses; however, the effect was similar in WT and QKO mice (Fig. 7). Of note, at 30 mg/kg, the dose used by Lobato et al. [40], zinc chloride caused death in four out of four WT mice; the difference in the results could be due to the different genetic background, C57BL/6 vs Swiss.

Fig. 7.

Fig. 7

Effects of zinc chloride. a Vehicle or zinc chloride (1, 3, 10, or 30 mg/kg) treatment of WT mice. b Mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 4–9/group. g Treatment of QKO and control mice with zinc chloride (10 mg/kg). h Mean Tb at 0–60 min, i mean activity at 0–60 min, j mean Tb at 0–300 min, k time below 34 °C, and l mean activity at 0–300 min; n = 10–12/group. Statistical analyses are in Table S2

The antidepressant amitriptyline was also reported to modulate adenosine signaling [51]. Amitriptyline (20 mg/kg) reduced both Tb and activity in WT mice, but this reduction was slightly increased in the QKO mice, consistent with non-adenosinergic MoA (Fig. 8).

Fig. 8.

Fig. 8

Effects of amitriptyline (20 mg/kg) treatment of QKO and control mice. a Time course, b mean Tb at 0–60 min, c mean activity at 0–60 min, d mean Tb at 0–300 min, e time below 34 °C, and f mean activity at 0–300 min; n = 8/group. Statistical analyses are in Table S2

Taken together, these data show that the hypothermic effects of nifedipine, ranolazine, ketamine, ethanol, zinc chloride, and amitriptyline are independent of AR signaling.

Discussion

Following up on indirect evidence and prior hypotheses, we have directly tested pharmacologically important substances for adenosinergic effects using mouse hypothermia as an in vivo assay. The thirteen drugs examined can mainly be divided into three groups, (1) evidence for adenosinergic effect, (2) hypothermia via non-adenosinergic mechanisms, and (3) no effect at all in this assay.

A major strength of using hypothermia as an in vivo screen for adenosinergic actions is that it detects agonism at any of the four ARs. The detailed mechanisms are not characterized in all cases. For example, A3AR agonists activate peripheral mast cells in mice, causing degranulation and histamine release, increased vascular permeability, and hypotension [52]. The hypothermia is caused by the histamine acting via H1 receptors [53]. Classically, central A1AR were identified as mediators of adenosine hypothermia [5456]. Additional studies suggested that activation of A1AR on neurons both within and outside the blood–brain barrier can cause hypothermia [57]. Within the brain, activation of A1AR-expressing neurons in the dorsomedial hypothalamus, but not the preoptic area, causes hypothermia [57]. Agonism at peripheral A2AAR causes hypothermia, possibly via vasodilation and hypotension [32, 58]. Finally, agonism of central A2BAR also causes hypothermia, with the mechanistic details not yet determined [32]. It is conceivable that that some of the activities observed here for drugs that permeate the BBB occur at the CNS level.

Since hypothermia is caused by at least five different AR/site combinations, demonstration of hypothermia and its lack in the QKO mice is a first step in characterizing the adenosinergic actions of a drug. Further experiments are needed to determine if the drug is a direct agonist or antagonist at one or more of the AR, or if it modulates a different target, changing adenosine levels either at particular anatomic sites or throughout the body. Non-receptor targets could include adenosine transporters [18], adenosine deaminase [59], and adenosine kinase inhibitors [27, 60]. Hypothermia can be a sensitive test for some drugs, but might miss potent adenosinergic actions, such as central adenosinergic effects for a drug that does not pass the blood–brain barrier. Even for compounds that do reach the relevant ARs, hypothermia may occur at a higher dose than required for other actions via that AR. It is important to remember that the hypothermia screen depends on the affinity of the tested drug for mouse AR and that a drug’s affinity can vary widely among species [61]. There may also be species differences in the biology, with one example being mast cell expression (or not) of A3AR [62].

An adenosinergic effect of a drug can also be identified by its ability to potentiate adenosine-induced hypothermia, an important approach for screening compounds blocking adenosine transport. While the inhibition of transport alone might not produce significant hypothermia, co-administration of adenosine with the test substance increases the sensitivity of the assay. We previously demonstrated that the ENT1 inhibitor NBMPR by itself produced a hint of hypothermia, but it greatly augmented adenosine-induce hypothermia [27]. Here we show that four drugs reported to inhibit adenosine transport (dipyridamole [21], nimodipine [33], cilostazol [63], and cyclosporine A [34, 35]) also increased the hypothermia caused by adenosine; these results are consistent with inhibition of adenosine transport. In vivo studies indicate that dipyridamole alone, as an ENT2 inhibitor, can increase A2BAR activation, e.g. in colitis and lung injury mouse models [64, 65]. Dipyridamole and nimodipine probably increased adenosine levels through inhibition of adenosine clearance via ENT1. In contrast to dipyridamole which itself did not reduce Tb, nimodipine alone caused hypothermia in WT mice that was diminished in QKO mice. This indicates that nimodipine has both adenosinergic (such as via ENT1 inhibition) and non-adenosinergic actions.

CBD is reported to be a sub-micromolar ENT1 inhibitor [23, 3638], but CBD did not enhance adenosine-induced hypothermia. Thus, CBD does not appear to be sufficiently efficacious as an ENT1 inhibitor under the in vivo conditions tested. Similarly, no adenosinergic effects were detected for canrenoate or sildenafil.

Cyclosporine A has been shown to increase plasma adenosine levels in kidney transplant recipients and inhibit adenosine uptake in red blood cells [34]. In T lymphocytes, it had dual action and inhibited both adenosine transport and adenosine kinase activity [35]. The enhancement of adenosine-induced hypothermia by cyclosporine A is consistent with inhibition of adenosine transport; however, it is unknown if this effect is mediated by ENT1.

The PDE3 inhibitor cilostazol is an anti-claudication drug that has been reported to inhibit adenosine transport in vitro and in vivo [63]. That MoA is thought to contribute to its cardioprotective and anti-ischemic neuroprotective effects [66]. Cilostazol did not induce hypothermia itself, but enhanced adenosine-induced hypothermia in wild type mice. Since cilostazol is a poor human ENT1 inhibitor (ref. 27 in Table 1), it might act at different targets. Its potency at mouse ENT1 is not reported. Taken together, these results suggest that at the doses tested, dipyridamole and nimodipine have some adenosinergic effects, likely by ENT1 inhibition, while cilostazol may have a different mode of action. Nevertheless, the relationship of cilostazol to potential human adenosinergic signaling was strengthened by a report that in acute coronary syndrome patients it raised plasma adenosine levels [67].

Ethanol has been hypothesized to act in the brain by reducing adenosine uptake [68]. ENT1−/− mice were less sensitive to acute effects of ethanol and showed an increase in alcohol consumption. While the hypothermic effects of ethanol were partially blunted in the A2AAR KO mouse [69], we have not detected any adenosinergic effects in the hypothermia assay. Unexpectedly, both ethanol and cannabidiol, that weakly block ENT1, did not cause adenosine-induced hypothermia. This likely reflects an insufficient degree of ENT1 inhibition under these conditions.

Nifedipine is a calcium channel blocker used as an antihypertensive. It caused hypothermia by itself and enhanced adenosine-induced hypothermia. However, nifedipine hypothermia was not altered in the QKO mice. Nifedipine may potentiate adenosine hypothermia via its hypotensive actions. Our results do not support the proposed adenosinergic effects of nifedipine [42].

The antidepressant, amitriptyline, also has antinociceptive actions reported to depend on A3AR, because this effect was attenuated by a co-administered A3AR antagonist MRS1191 [51]. Amitriptyline itself induced hypothermia, but there was no reduction (actually a slight enhancement) in the QKO mice. Therefore, in vivo activation of A3AR by amitriptyline was not detectable and whether loss of ARs enhances amitriptyline-induced hypothermia requires further investigation.

There are many drugs and chemicals that can cause hypothermia, potentially for therapeutic application [70]. Here we have identified an adenosinergic mechanism for some compounds and found that others caused hypothermia via non-adenosinergic mechanisms because their effect remained in QKO mice. We have not investigated the mechanisms of these non-adenosinergic drugs.

Adenosine and adenosine receptor signaling has been implicated in many biological processes [17]. Extracellular adenosine can be elevated during disease conditions by generation from nucleotides or by transcriptional control of the hypoxia-inducible factor 1α (HIF1A) pathway during hypoxia [71]. Numerous exogenous AR agonists and antagonists have beneficial therapeutic effects in animal models, and many have been tested in clinical trials. However, currently, short-acting, parenteral agonists, adenosine and regadenoson, are the only AR agonists approved for human use [5]. Regadenoson is also being examined for treatment of sickle cell disease, glioblastoma (opening the blood brain barrier) and other conditions (ClinicalTrials.gov Identifier: NCT03971734, accessed November 15, 2022) [72, 73]. Thus, identification of approved drugs that elevate adenosine in vivo could lead to expanded indications for these compounds. Dipyridamole has been repurposed as a potential treatment of Covid19 (ClinicalTrials.gov Identifiers: NCT04391179, NCT04424901, accessed November 15, 2022) based on the anti-inflammatory effects of adenosine elevation [74]. Other conditions in which adenosine could have a beneficial effect include pain, inflammation, steatohepatitis, and seizures. Identifying and characterizing adenosinergic actions is a promising approach for repurposing approved drugs.

Supplementary Information

Below is the link to the electronic supplementary material.

Acknowledgements

We thank the Intramural Research Program of the National Institutes of Health National Institute of Diabetes and Digestive and Kidney Diseases for support (ZIADK075063; ZIADK031117).

Abbreviations

AR

Adenosine receptor

CBD

Cannabidiol

ENT

Equilibrative nucleoside transporter

GPCR

G protein-coupled receptor

MoA

Mechanism of action

NBMPR

6-S-[(4-Nitrophenyl)methyl]-6-thioinosine

NMDA

N-methyl-D-aspartate

PDE

Phosphodiesterase

SLC

Solute carrier

Tb

Core body temperature

QKO

Quadruple knockout of adenosine receptors

WT

Wild-type

Cuiying Xiao

received her MD degree from Southwest Medical University, and PhD in genetics from Sichuan University/West China Medical Center before starting her postdoctoral fellowship in NIDDK, NIH in early 2004. She has been a staff scientist in NIDDK since late 2011 and has conducted research using mouse models to understand metabolic rate regulation, body temperature regulation, and drug treatments for obesity. Her research goal is to make advances in the treatment of obesity. Inline graphic

Author contribution

K.A.J., O.G., and M.L.R. conceptualized the experiments, analyzed data, and wrote the main manuscript text; C.X., O.G., N.L., and S.A.L. performed experiments, analyzed data, and prepared figures and tables. All authors reviewed the manuscript.

Funding

This work was supported by the Intramural Research Program of the National Institutes of Health National Institute of Diabetes and Digestive and Kidney Diseases for support (ZIADK075063 to M.L.R.; ZIADK031117 to K.A.J.).

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Declarations

Ethical approval

All animal procedures were conducted with approval of the NIDDK Institutional Animal Care and Use Committee (IUCAC), protocol number K016-DEOB-23. Standards of the Association for Assessment and Accreditation of Laboratory Animal Care, International (AAALAC) were followed, and work was performed in an AAALAC-accredited facility. Human studies—not applicable.

Conflict of interest

Cuiying Xiao declares that he/she has no conflict of interest. Oksana Gavrilova declares that he/she has no conflict of interest. Naili Liu declares that he/she has no conflict of interest. Sarah A. Lewicki declares that he/she has no conflict of interest. Marc L. Reitman declares that he/she has no conflict of interest. Kenneth A. Jacobson declares that he/she has no conflict of interest.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Cuiying Xiao and Oksana Gavrilova contributed equally to this work.

References

  • 1.Chen JF, Eltzschig HK, Fredholm BB. Adenosine receptors as drug targets–what are the challenges? Nat Rev Drug Discov. 2013;12(4):265–286. doi: 10.1038/nrd3955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K. Pharmacology of adenosine receptors: the state of the art. Physiol Rev. 2018;98(3):1591–1625. doi: 10.1152/physrev.00049.2017. [DOI] [PubMed] [Google Scholar]
  • 3.Congreve M, Brown GA, Borodovsky A, Lamb ML. Targeting adenosine A2A receptor antagonism for treatment of cancer. Exp Opin Drug Discov. 2018;13(11):997–1003. doi: 10.1080/17460441.2018.1534825. [DOI] [PubMed] [Google Scholar]
  • 4.Gao ZG, Jacobson KA. A2B adenosine receptor and cancer. Int J Mol Sci. 2019;20:5139. doi: 10.3390/ijms20205139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jacobson KA, Tosh DK, Jain S, Gao ZG. Historical and current adenosine receptor agonists in preclinical and clinical development. Frontiers Cell Neurosci. 2019;13:124. doi: 10.3389/fncel.2019.00124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Cronstein B, Sitkovsky M. Adenosine and adenosine receptors in the pathogenesis and treatment of rheumatic diseases. Nat Rev Rheumatol. 2017;13:41–51. doi: 10.1038/nrrheum.2016.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Antonioli L, Lucarini E, Lambertucci C, Fornai M, Pellegrini C, Benvenuti L, et al. The anti-Inflammatory and pain-relieving effects of AR170, an adenosine A3 receptor agonist, in a rat model of colitis. Cells. 2020;9(6):1509. doi: 10.3390/cells9061509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Jacobson KA, Reitman ML. Adenosine-related mechanisms in non-adenosine receptor drugs. Cells. 2020;9:956. doi: 10.3390/cells9040956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Gillespie RJ, Adams DR, Bebbington D, Benwell K, Cliffe IA, Dawson CE, Dourish CT, Fletcher A, Gaur S, Giles PR, Jordan AM, Knight AR, Knutsen LJS, Lawrence A, Lerpiniere J, Misra A, Porter RHP, Pratt RM, Shepherd R, Upton R, Ward SE, Weiss SM, Williamson DS. Antagonists of the human adenosine A2A receptor. Part 1: Discovery and synthesis of thieno[3,2-d]pyrimidine-4-methanone derivatives. Bioorg Med Chem Lett. 2008;18:2916–2919. doi: 10.1016/j.bmcl.2008.03.075. [DOI] [PubMed] [Google Scholar]
  • 10.Daly JW, Hong O, Padgett WL, Shamim MT, Jacobson KA, Ukena D. Non-xanthine heterocycles: activity as antagonists of A1- and A2-adenosine receptors. Biochem Pharmacol. 1988;37:655–664. doi: 10.1016/0006-2952(88)90139-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Katritch V, Jaakola VP, Lane JR, Lin J, IJzerman AP, Yeager M, Kufareva I, Stevens RC, Abagyan R. Structure-based discovery of novel chemotypes for adenosine A2A receptor antagonists. J Med Chem. 2010;53:1799–1809. doi: 10.1021/jm901647p. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rodríguez D, Chakraborty S, Warnick E, Crane S, Gao ZG, O’Connor RO, Jacobson KA, Carlsson J. Structure-based screening of uncharted chemical space for atypical adenosine receptor agonists. ACS Chem Biol. 2016;11:2763–2772. doi: 10.1021/acschembio.6b00357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Daly JW, Hong O, Padgett WL, Shamim MT, Jacobson KA, Ukena D. Non-xanthine heterocycles: activity as antagonists of A1- and A2-adenosine receptors. Biochem Pharmacol. 1988;37(4):655–664. doi: 10.1016/0006-2952(88)90139-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Phillis JW, O'Regan MH. The role of adenosine in the central actions of the benzodiazepines. Prog Neuropsychopharmacol Biol Psychiatry. 1988;12(4):389–404. doi: 10.1016/0278-5846(88)90100-5. [DOI] [PubMed] [Google Scholar]
  • 15.Jacobson KA, Moro S, Manthey JA, West PL, Ji XD. Interactions of flavones and other phytochemicals with adenosine receptors. Adv Exp Med Biol. 2002;505:163–171. doi: 10.1007/978-1-4757-5235-9_15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.van Rhee AM, Jiang JL, Melman N, Olah ME, Stiles GL, Jacobson KA. Interaction of 1,4-dihydropyridine and pyridine derivatives with adenosine receptors: selectivity for A3 receptors. J Med Chem. 1996;39(15):2980–2989. doi: 10.1021/jm9600205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Schon MP, Schon M, Klotz KN. The small antitumoral immune response modifier imiquimod interacts with adenosine receptor signaling in a TLR7- and TLR8-independent fashion. J Invest Dermatol. 2006;126(6):1338–1347. doi: 10.1038/sj.jid.5700286. [DOI] [PubMed] [Google Scholar]
  • 18.Boswell-Casteel RC, Hays FA. Equilibrative nucleoside transporters—a review. Nucleosides, Nucleotides Nucleic Acids. 2017;36:7–30. doi: 10.1080/15257770.2016.1210805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Vlachodimou A, Konstantinopoulou K, IJzerman AP, Heitman LH, Affinity, binding kinetics and functional characterization of draflazine analogues for human equilibrative nucleoside transporter 1 (SLC29A1) Biochem Pharmacol. 2020;172:113747. doi: 10.1016/j.bcp.2019.113747. [DOI] [PubMed] [Google Scholar]
  • 20.Boison D. Adenosine kinase: exploitation for therapeutic gain. Pharmacol Rev. 2013;65(3):906–943. doi: 10.1124/pr.112.006361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Armstrong C, Summers L, Ewart S, Nylander JE, van Sidaway JJ, Giezen. Characterization of the adenosine pharmacology of ticagrelor reveals therapeutically relevant inhibition of equilibrative nucleoside transporter 1. J Cardiovasc Pharmacol Ther. 2014;19:209–219. doi: 10.1177/1074248413511693. [DOI] [PubMed] [Google Scholar]
  • 22.Cattaneo M, Schulz R, Nylander S. Adenosine-mediated effects of ticagrelor: Evidence and potential clinical relevance. J Am Coll Cardiol. 2014;63(23):2503–2509. doi: 10.1016/j.jacc.2014.03.031. [DOI] [PubMed] [Google Scholar]
  • 23.Carrier EJ, Auchampach JA, Hillard CJ. Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression. Proc Natl Acad Sci USA. 2006;103:7895–7900. doi: 10.1073/pnas.0511232103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Nagy LE. Ethanol metabolism and inhibition of nucleoside uptake lead to increased extracellular adenosine in hepatocytes. Am J Physiol. 1992;262(5 Pt 1):C1175–C1180. doi: 10.1152/ajpcell.1992.262.5.C1175. [DOI] [PubMed] [Google Scholar]
  • 25.Ribeiro LR, Storer RI. A semi-quantitative translational pharmacology analysis to understand the relationship between in vitro ENT1 inhibition and the clinical incidence of dyspnoea and bronchospasm. Toxicol Appl Pharmacol. 2017;317:41–50. doi: 10.1016/j.taap.2016.12.021. [DOI] [PubMed] [Google Scholar]
  • 26.Tian H, Cronstein BN. Understanding the mechanisms of action of methotrexate implications for the treatment of rheumatoid arthritis. Bull NYU Hosp Joint Dis. 2007;65:168–173. [PubMed] [Google Scholar]
  • 27.Xiao C, Liu N, Jacobson KA, Gavrilova O, Reitman ML. Physiology and effects of nucleosides in mice lacking all four adenosine receptors. PLoS Biol. 2019;17(3):e3000161. doi: 10.1371/journal.pbio.3000161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lopes CR, Lourenço VS, Tomé ÂR, Cunha RA, Canas PM. Use of knockout mice to explore CNS effects of adenosine. Biochem Pharmacol. 2021;187:114367. doi: 10.1016/j.bcp.2020.114367. [DOI] [PubMed] [Google Scholar]
  • 29.Carlin JL, Tosh DK, Xiao C, Piñol RA, Chen Z, Salvemini D, Gavrilova O, Jacobson KA, Reitman ML. Peripheral adenosine A3 receptor activation causes regulated hypothermia in mice that is dependent on central histamine H1 receptors. J Pharmacol Exp Therap. 2016;356:474–482. doi: 10.1124/jpet.115.229872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Carlin JL, Jain S, Gizewski E, Wan TC, Tosh DK, Xiao C, Auchampach JA, Jacobson KA, Gavrilova O, Reitman ML. Hypothermia in mouse is caused by adenosine A1 and A3 receptor agonists and AMP via three distinct mechanisms. Neuropharmacology. 2017;114:101–113. doi: 10.1016/j.neuropharm.2016.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kochanek PM, Vagni VA, Janesko KL, Washington CB, Crumrine PK, Garman RH, et al. Adenosine A1 receptor knockout mice develop lethal status epilepticus after experimental traumatic brain injury. J Cereb Blood Flow Metab. 2006;26(4):565–575. doi: 10.1038/sj.jcbfm.9600218. [DOI] [PubMed] [Google Scholar]
  • 32.Carlin JL, Jain S, Duroux R, Suresh RR, Xiao C, Auchampach JA, et al. (2018) Activation of adenosine A2A or A2B receptors causes hypothermia in mice. Neuropharmacology. 2018;139:268–278. doi: 10.1016/j.neuropharm.2018.02.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Striessnig J, Zernig G, Glossmann H. Glossmann Human red-blood-cell Ca2+-antagonist binding sites. Evidence for an unusual receptor coupled to the nucleoside transporter. Eur J Biochem. 1985;150:67–77. doi: 10.1111/j.1432-1033.1985.tb08989.x. [DOI] [PubMed] [Google Scholar]
  • 34.Guieu R, Dussol B, Devaux C, Sampol J, Brunet P, Rochat H, et al. Interactions between cyclosporine A and adenosine in kidney transplant recipients. Kidney Int. 1998;53(1):200–204. doi: 10.1046/j.1523-1755.1998.00727.x. [DOI] [PubMed] [Google Scholar]
  • 35.Spychala J, Mitchell BS. Cyclosporin A and FK506 decrease adenosine kinase activity and adenosine uptake in T-lymphocytes. J Lab Clin Med. 2002;140(2):84–91. doi: 10.1016/S0022-2143(02)00022-7. [DOI] [PubMed] [Google Scholar]
  • 36.Liou GI, Auchampach JA, Hillard CJ, Zhu G, Yousufzai B, Mian S, Khan S, Khalifa Y. Mediation of cannabidiol anti-inflammation in the retina by equilibrative nucleoside transporter and A2A adenosine receptor. Investig Ophthalmol Vis Sci. 2008;49:5526–5531. doi: 10.1167/iovs.08-2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Castillo A, Tolón MR, Fernández-Ruiz J, Romero J, Martinez-Orgado J. The neuroprotective effect of cannabidiol in an in vitro model of newborn hypoxic–ischemic brain damage in mice is mediated by CB2 and adenosine receptors. Neurobiol Dis. 2010;37:434–440. doi: 10.1016/j.nbd.2009.10.023. [DOI] [PubMed] [Google Scholar]
  • 38.Mijangos-Moreno S, Poot-Aké A, Arankowsky-Sandoval G, Murillo-Rodríguez E. Intrahypothalamic injection of cannabidiol increases the extracellular levels of adenosine in nucleus accumbens in rats. Neurosci Res. 2014;84:60–63. doi: 10.1016/j.neures.2014.04.006. [DOI] [PubMed] [Google Scholar]
  • 39.Vitale RM, Iannotti FA, Amodeo P (2021) The (poly)pharmacology of cannabidiol in neurological and neuropsychiatric disorders: molecular mechanisms and targets. Int J Mol Sci 22(9):4876. 10.3390/ijms22094876 [DOI] [PMC free article] [PubMed]
  • 40.Schmidt K, Tissier R, Ghaleh B, Drogies T, Felix SB, Krieg T. Cardioprotective effects of mineralocorticoid receptor antagonists at reperfusion. Eur Heart J. 2010;31:1655–1662. doi: 10.1093/eurheartj/ehp555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Lee HG, Kim WM, Choi JI, Yoon MH. Roles of adenosine receptor subtypes on the antinociceptive effect of sildenafil in rat spinal cord. Neurosci Lett. 2010;480(3):182–185. doi: 10.1016/j.neulet.2010.06.025. [DOI] [PubMed] [Google Scholar]
  • 42.Swanson TH, Green CL. Nifedipine: more than a calcium channel blocker. Gen Pharmacol. 1986;17(3):255–260. doi: 10.1016/0306-3623(86)90038-8. [DOI] [PubMed] [Google Scholar]
  • 43.Li RW, Tse CM, Man RY, Vanhoutte PM, Leung GP. Inhibition of human equilibrative nucleoside transporters by dihydropyridine-type calcium channel antagonists. Eur J Pharmacol. 2007;568(1–3):75–82. doi: 10.1016/j.ejphar.2007.04.033. [DOI] [PubMed] [Google Scholar]
  • 44.Le DE, Davis CM, Wei K, Zhao Y, Cao Z, Nugent M, Scott KLL, Liu L, Nagarajan S, Alkayed NJ, et al. Ranolazine may exert its beneficial effects by increasing myocardial adenosine levels. Am J Physiol Heart Circ Physiol. 2020;318:H189–H202. doi: 10.1152/ajpheart.00217.2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lazarevic V, Yang Y, Flais I, et al. Ketamine decreases neuronally released glutamate via retrograde stimulation of presynaptic adenosine A1 receptors. Mol Psychiatry. 2021;26:7425–7435. doi: 10.1038/s41380-021-01246-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Mazar J, Rogachev B, Shaked G, Ziv NY, Czeiger D, Chaimovitz C, Zlotnik M, Mukmenev I, Byk G, Douvdevani A. Involvement of adenosine in the antiinflammatory action of ketamine. Anesthesiology. 2005;102:1174–1181. doi: 10.1097/00000542-200506000-00017. [DOI] [PubMed] [Google Scholar]
  • 47.Ramadan A, Naydenova Z, Stevanovic K, Rose JB, Coe IR. The adenosine transporter, ENT1, in cardiomyocytes is sensitive to inhibition by ethanol in a kinase-dependent manner: implications for ethanol-dependent cardioprotection and nucleoside analog drug cytotoxicity. Purinergic Signal. 2014;10(2):305–312. doi: 10.1007/s11302-013-9391-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Naassila M, Ledent C, Daoust M. Low ethanol sensitivity and increased ethanol consumption in mice lacking adenosine A2A receptors. J Neurosci. 2002;22:10487–10493. doi: 10.1523/JNEUROSCI.22-23-10487.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Nam HW, Bruner RC, Choi DS. Adenosine signaling in striatal circuits and alcohol use disorders. Mol Cells. 2013;36:195–202. doi: 10.1007/s10059-013-0192-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lobato KR, Binfaré RW, Budni J, Rosa AO, Santos ARS, Rodrigues ALS. Involvement of the adenosine A1 and A2A receptors in the antidepressant-like effect of zinc in the forced swimming test. Prog Neuro-Psychopharmacol Biol Psych. 2010;32:994–999. doi: 10.1016/j.pnpbp.2008.01.012. [DOI] [PubMed] [Google Scholar]
  • 51.Kim Y, Kwon SY, Jung HS, Park YJ, Kim YS, In JH, Choi JW, Kim JA, Joo JD. Amitriptyline inhibits the MAPK/ERK and CREB pathways and proinflammatory cytokines through A3AR activation in rat neuropathic pain models. Korean J Anesthesiol. 2019;72:60–67. doi: 10.4097/kja.d.18.00022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Salvatore CA, Tilley SL, Latour AM, Fletcher DS, Koller BH, Jacobson MA. Disruption of the A3 adenosine receptor gene in mice and its effect on stimulated inflammatory cells. J Biol Chem. 2000;275(6):4429–4434. doi: 10.1074/jbc.275.6.4429. [DOI] [PubMed] [Google Scholar]
  • 53.Carlin JL, Tosh DK, Xiao C, Pinol RA, Chen Z, Salvemini D, et al. Peripheral adenosine A3 receptor activation causes regulated hypothermia in mice that is dependent on central histamine H1 receptors. J Pharmacol Exp Ther. 2016;356(2):474–482. doi: 10.1124/jpet.115.229872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Anderson R, Sheehan MJ, Strong P. Characterization of the adenosine receptors mediating hypothermia in the conscious mouse. Br J Pharmacol. 1994;113(4):1386–1390. doi: 10.1111/j.1476-5381.1994.tb17151.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Shintani M, Tamura Y, Monden M, Shiomi H. Characterization of N6-cyclohexyladenosine-induced hypothermia in Syrian hamsters. J Pharmacol Sci. 2005;97(3):451–454. doi: 10.1254/jphs.SC0040178. [DOI] [PubMed] [Google Scholar]
  • 56.Tupone D, Madden CJ, Morrison SF. Central activation of the A1 adenosine receptor (A1AR) induces a hypothermic, torpor-like state in the rat. J Neurosci. 2013;33(36):14512–14525. doi: 10.1523/JNEUROSCI.1980-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Province HS, Xiao C, Mogul AS, Sahoo A, Jacobson KA, Pinol RA, et al. Activation of neuronal adenosine A1 receptors causes hypothermia through central and peripheral mechanisms. PLoS ONE. 2020;15(12):e0243986. doi: 10.1371/journal.pone.0243986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ledent C, Vaugeois JM, Schiffmann SN, Pedrazzini T, El Yacoubi M, Vanderhaeghen JJ, et al. Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature. 1997;388(6643):674–678. doi: 10.1038/41771. [DOI] [PubMed] [Google Scholar]
  • 59.Bhagavatham SKS, Khanchandani P, Kannan V, et al. Adenosine deaminase modulates metabolic remodeling and orchestrates joint destruction in rheumatoid arthritis. Sci Rep. 2021;11:15129. doi: 10.1038/s41598-021-94607-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Jarvis MF, Mikusa J, Chu KL, Wismer CT, Honore P, Kowaluk EA, et al. Comparison of the ability of adenosine kinase inhibitors and adenosine receptor agonists to attenuate thermal hyperalgesia and reduce motor performance in rats. Pharmacol Biochem Behav. 2002;73(3):573–581. doi: 10.1016/S0091-3057(02)00840-7. [DOI] [PubMed] [Google Scholar]
  • 61.Alnouri MW, Jepards S, Casari A, Schiedel AC, Hinz S, Muller CE. Selectivity is species-dependent: Characterization of standard agonists and antagonists at human, rat, and mouse adenosine receptors. Purinergic Signal. 2015;11(3):389–407. doi: 10.1007/s11302-015-9460-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Rudich N, Ravid K, Sagi-Eisenberg R. Mast cell adenosine receptors function: a focus on the A3 adenosine receptor and inflammation. Front Immunol. 2012;3:134. doi: 10.3389/fimmu.2012.00134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Bai Y, Muqier Murakami H, Iwasa M, Sumi S, Yamada Y, Minatoguchi S. Cilostazol protects the heart against ischaemia reperfusion injury in a rabbit model of myocardial infarction: focus on adenosine, nitric oxide and mitochondrial ATP-sensitive potassium channels. Clin Exp Pharmacol Physiol. 2011;38(10):658–665. doi: 10.1111/j.1440-1681.2011.05550.x. [DOI] [PubMed] [Google Scholar]
  • 64.Aherne CM, Collins CB, Rapp CR, Olli KE, Perrenoud L, Jedlicka P, Bowser JL, Mills TW, Karmouty-Quintana H, Blackburn MR, Eltzschig HK. Coordination of ENT2-dependent adenosine transport and signaling dampens mucosal inflammation. JCI Insight. 2018;3(20):e121521. doi: 10.1172/jci.insight.121521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Eckle T, Hughes K, Ehrentraut H, Brodsky KS, Rosenberger P, Choi DS, Ravid K, Weng T, Xia Y, Blackburn MR, Eltzschig HK. Crosstalk between the equilibrative nucleoside transporter ENT2 and alveolar Adora2b adenosine receptors dampens acute lung injury. FASEB J. 2013;27(8):3078–3089. doi: 10.1096/fj.13-228551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kanlop N, Chattipakorn S, Chattipakorn N. Effects of cilostazol in the heart. J Cardiovasc Med. 2011;12:88–95. doi: 10.2459/JCM.0b013e3283439746. [DOI] [PubMed] [Google Scholar]
  • 67.Xue Y, Wang Z, Wu H, Li X, Chen J, Lv Q. Cilostazol increases adenosine plasma concentration in patients with acute coronary syndrome. J Clin Pharm Ther. 2021;46:328–332. doi: 10.1111/jcpt.13284. [DOI] [PubMed] [Google Scholar]
  • 68.Choi DS, Cascini MG, Mailliard W, Young H, Paredes P, McMahon T, Messing RO. The type 1 equilibrative nucleoside transporter regulates ethanol intoxication and preference. Nat Neurosci. 2004;7:855–861. doi: 10.1038/nn1288. [DOI] [PubMed] [Google Scholar]
  • 69.El Yacoubi M, Ledent C, Parmentier M, Costentin J, Vaugeois JM. Caffeine reduces hypnotic effects of alcohol through adenosine A2A receptor blockade. Neuropharmacology. 2003;45(7):977–985. doi: 10.1016/s0028-3908(03)00254-5. [DOI] [PubMed] [Google Scholar]
  • 70.Liu K, Khan H, Geng X, Zhang J, Ding Y. Pharmacological hypothermia: a potential for future stroke therapy? Neurolog Res. 2016;38(6):478–490. doi: 10.1080/01616412.2016.1187826. [DOI] [PubMed] [Google Scholar]
  • 71.Poth JM, Brodsky K, Ehrentraut H, Grenz A, Eltzschig HK. Transcriptional control of adenosine signaling by hypoxia-inducible transcription factors during ischemic or inflammatory disease. J Mol Med (Berl) 2013;91(2):183–193. doi: 10.1007/s00109-012-0988-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nathan DG, Field J, Lin G, Neuberg D, Majerus E, Onyekwere O, Keefer J, Okam M, Ross A, Linden J. Sickle cell disease (SCD), iNKT cells, and regadenoson infusion. Trans Am Clin Climatol Assoc. 2012;123:312–317. [PMC free article] [PubMed] [Google Scholar]
  • 73.Jackson S, George RT, Lodge MA, Piotrowski A, Wahl RL, Gujar SK, Grossman SA. The effect of regadenoson on the integrity of the human blood-brain barrier, a pilot study. J Neurooncol. 2017;132(3):513–519. doi: 10.1007/s11060-017-2404-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kanthi Y, Knight JS, Zuo Y. Pinsky DJ (2020) New (re)purpose for an old drug: purinergic modulation may extinguish the COVID-19 thromboinflammatory firestorm. JCI Insight. 2020;5:e140971. doi: 10.1172/jci.insight.140971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Goldman N, et al. Adenosine A1 receptors mediate local anti-nociceptive effects of acupuncture. Nature Neurosci. 2010;13:883–888. doi: 10.1038/nn.2562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Shi Y, Dai Q, Ji B, Huang L, Zhuang X, Mo Y, Wang J. Electroacupuncture pretreatment prevents cognitive impairment induced by cerebral ischemia–reperfusion via adenosine A1 receptors in rats. Front Aging Neurosci. 2021;13:680706. doi: 10.3389/fnagi.2021.680706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Coppi E, Cherchi F, Lucarini E, Ghelardini C, Pedata F, Jacobson KA, Di Cesare ML, Pugliese AM, Salvemini D. Uncovering the mechanisms of A3 adenosine receptor-mediated pain control. Int J Mol Sci. 2021;22:7952. doi: 10.3390/ijms22157952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Nair AB, Jacob S. A simple practice guide for dose conversion between animals and human. J Basic Clin Pharm. 2016;7(2):27–31. doi: 10.4103/0976-0105.177703. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data Availability Statement

The data that support the findings of this study are available from the corresponding author upon reasonable request.


Articles from Purinergic Signalling are provided here courtesy of Springer

RESOURCES