Skip to main content
Purinergic Signalling logoLink to Purinergic Signalling
. 2014 Oct 29;10(4):529–564. doi: 10.1007/s11302-014-9427-2

Purinergic signalling and immune cells

Geoffrey Burnstock 1,2,, Jean-Marie Boeynaems 3
PMCID: PMC4272370  PMID: 25352330

Abstract

This review article provides a historical perspective on the role of purinergic signalling in the regulation of various subsets of immune cells from early discoveries to current understanding. It is now recognised that adenosine 5′-triphosphate (ATP) and other nucleotides are released from cells following stress or injury. They can act on virtually all subsets of immune cells through a spectrum of P2X ligand-gated ion channels and G protein-coupled P2Y receptors. Furthermore, ATP is rapidly degraded into adenosine by ectonucleotidases such as CD39 and CD73, and adenosine exerts additional regulatory effects through its own receptors. The resulting effect ranges from stimulation to tolerance depending on the amount and time courses of nucleotides released, and the balance between ATP and adenosine. This review identifies the various receptors involved in the different subsets of immune cells and their effects on the function of these cells.

Keywords: ATP , UTP, Lymphocytes, Neutrophils, Mast cells, Microglia, Macrophages, Purinoceptors

Synopsis

Introduction

Purinergic signalling in the main subsets of immune cells

Polymorphonuclear leukocytes

Neutrophils

Eosinophils

Basophils

Mast cells

Section summary

Monocytes, macrophages and microglia

Monocytes

Macrophages

Microglia

Section summary

Dendritic cells

P1 receptors

P2 receptors

Section summary

Lymphocytes

T and B lymphocytes

Natural killer (NK and NKT) cells

Section summary

Concluding remarks

Introduction

Although nucleotides, such as adenosine 5′-triphosphate (ATP) and uridine 5′-triphosphate (UTP), are mainly intracellular, they are released into the extracellular fluids by various mechanisms. One of them is cell damage and death: both necrotic and apoptotic cells release ATP and other nucleotides that thus constitute “danger signals” or damage associated molecular pattern [13]. In the absence of cell death, they are also released in response to various types of stress: mechanical stimulation (stretch, shear stress) [4], hypoxia or pathogen invasion [5, 6]. Specific mechanisms of release include: exocytosis of secretory granules, vesicular transport [7, 8] and membrane channels, such as ATP-binding cassette transporters, pannexins [911] and connexins [12]. In particular, nucleotides are released by exocytosis during platelet aggregation and synaptic transmission. For many years, cells of the immune system were not considered to be innervated, but there is increasing recognition that nerves can influence the immune system and the field of neuroimmunology is growing rapidly [1315].

Once in the extracellular fluids, nucleotides are rapidly degraded by a variety of ectonucleotidases [16], such as the ENTPDases, like CD39 that degrades ATP into adenosine 5′-diphosphate (ADP) and ADP into adenosine monophosphate (AMP) and CD73/5′-nucleotidase that converts AMP into adenosine. Receptors for extracellular nucleotides and their degradation products such as adenosine have been progressively characterized. Subdivision of purinergic receptors between P1 (adenosine) and P2 (ATP, ADP) was proposed by Burnstock in 1978 [17]. A further subdivision of P2 receptors between P2Y and P2X was made in 1985 [18]. It is now well established that signalling by extracellular nucleotides is mediated by these two families of receptors, the molecular structure of which has been characterized: P2Y receptors are metabotropic G protein-coupled while P2X receptors are oligomeric ion channels.

Numerous reviews on various aspects of purinergic signalling in the immune system are available (Table 1). In the history and development of knowledge about purinergic signalling, early workers focussed on adenosine, while those concerned with ATP rarely referred to adenosine. This is obviously an inadequate approach since the effects of ATP and adenosine, its breakdown product that is rapidly produced by ectonucleotidases, are intimately related. In this review, purinergic signalling in immune cells will be covered in a comprehensive and historical way, following the increase in knowledge from the early discoveries to current understanding. The review will consider the major subsets of immune cells and, for each of them, address the mechanisms of nucleotides release and adenosine generation, as well as the repertoire of functional P1 and P2 receptors that they express.

Table 1.

Reviews on the role of purinergic signalling in the immune system

General reviews on purinergic signalling in the immune system [474, 537543]
Immune regulation by extracellular nucleotides [544551]
Immune regulation by adenosine [552563]
Ectonucleotidases and immune responses [426, 564566]
Purinergic signalling in neutrophils [567569]
Purinergic signalling in eosinophils [570]
Purinergic signalling in mast cells [571, 572]
Purinergic signalling in monocytes [573]
Purinergic signalling in macrophages [574]
Purinergic signalling in microglia [344, 575, 576]
Purinergic signalling in dendritic cells [577, 578]
Purinergic signalling in lymphocytes [579586]
P2X7 receptors and immune cells [383, 503, 587589]
P2X7 receptors, macrophage function and bacteria [590594]

Purinergic signalling in the main subsets of immune cells

Polymorphonuclear leukocytes

Neutrophils

P1 receptors

Ectoenzymes that hydrolyse ATP have been observed on guinea pig polymorphonuclear leukocytes [1921]. In particular, both CD39 and CD73 ectonucleotidases are present on neutrophil membranes [22]. Furthermore, neutrophils express mRNA for A1, A2A, A2B and A3 receptors [23], but the mRNA for A2A and A3 receptors are the most abundant [24]. Adenosine was shown to be a physiological modulator inhibiting the generation of superoxide (O2 ) anion by neutrophils via A2 receptors [2529]. Not surprisingly, dipyridamole, which prevents the uptake of adenosine, thereby increasing extracellular levels, inhibits O2 generation by neutrophils [30]. Adenosine also inhibited the degranulation induced by the chemotactic peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP) [28], phagocytosis [31] and the bactericidal function of neutrophils [32]. It was proposed that the inhibitory actions of adenosine on neutrophils were due to calcium entry blockade [3335]. Adenosine inhibited neutrophil respiratory bursts in association with an increase in cyclic AMP (cAMP) and reduction in [Ca2+]i [36, 37]. Occupancy of A2A receptors by adenosine inhibits fMLP-induced neutrophil activation via cAMP and protein kinase A regulated events [38]. Caffeine intake results in increase in cAMP accumulation and decrease in O2 anion production in human neutrophils, mediated by A2A receptors [39]. Activation of A2A receptors also inhibited the expression and release of various cytokines and chemokines after lipopolysaccharide (LPS) stimulation of neutrophils [40]. But other studies showed that both the A2B and the A3 receptors can also play a role in these inhibitory actions. Tumour necrosis factor-α (TNF-α) production by neutrophils following renal ischemia-reperfusion was increased in A2B-deficient mice [41]. Activation of A2B receptors also inhibited fMLP-induced O2 production [42]. The A3 receptor is also involved in the inhibition of O2 production [43] and of degranulation [44]. Adenosine downregulated ligand-stimulated leukotriene B4 biosynthesis in neutrophil suspensions [45], but it potentiated neutrophil cyclooxygenase-2 via A2A receptors [46].

In contrast, there are discrepant reports concerning the action of adenosine on neutrophil chemotaxis. It was claimed in 1982 that adenosine had no effect on the chemotaxis of neutrophils, although it did enhance the inhibition of chemotaxis by 3-deaza-(±)aristeromycin [47]. However, it was reported later that adenosine promotes neutrophil chemotaxis [48], perhaps via A1 receptors [49]. It was shown recently that the recruitment of neutrophils and other leukocytes in the lung during influenza infection is reduced in A1-deficient mice [50]. In contrast, LPS-induced recruitment of neutrophils in the lung was increased in A2A-deficient mice and experiments with chimeric mice revealed that this involves a direct inhibitory effect of the A2A receptor in myeloid cells [51]. Similar results were obtained in A2B-deficient mice [52]. Interestingly, in A2A-deficient mice, neutrophils were increased in the alveolar space [51], whereas they were increased in the interstitium of A2B-deficient mice [52]. Chen et al. [24] showed that adenosine stimulates neutrophil migration and amplifies the action of chemotactic signals through A3 receptors that are recruited to the leading edge (see Fig. 1). In A3-deficient mice, the recruitment of neutrophils was reduced in the lung during sepsis [53] and in the colon after induction of colitis by dextran sulphate [54]. Interestingly neutrophil chemotaxis requires excitatory signals at the front and inhibitory signals at the back of cells. This inhibitory signal at the back might be mediated by adenosine acting on A2A receptors [55] (Fig. 1).

Fig. 1.

Fig. 1

Proposed model of neutrophil chemotaxis. As previously reported, stimulation of chemoattractant receptors induces local release of ATP through PANX1 channels at the site that first encounters the chemoattractant. Autocrine feedback via P2Y2 receptors amplifies the chemotactic signal and triggers cell polarization, whereby cells assume an elongated shape, and PANX1, CD39 (NTPDase1) and A3 adenosine receptors accumulate at the leading edge. In the current study, we found that A2A receptors are translocated from the leading edge toward the back of polarized neutrophils and that inhibitory signaling via A2A receptor-dependent cAMP accumulation inhibits excitatory chemotactic signalling by blocking FPR-dependent ERK and p38 MAPK activation globally with the exception of the leading edge. ALP alkaline phosphatase, ADO adenosine, PIP3 phosphatidylinositol (3,4,5)-triphosphate. (Reproduced from [55], with permission from the American Society for Biochemistry and Molecular Biology)

There is evidence that adenosine can modulate the interaction of neutrophils with pathogens. A3 receptors aggregate in highly polarised immunomodulatory microdomains of human neutrophil membranes. They promote the formation of filipodia-like projections (cytonemes) that can extend up to 100 μm to tether pathogens. Exposure to bacteria or an A3 agonist stimulates the formation of these projections and bacterial phagocytosis, whereas an A3 antagonist inhibits cytoneme formation [56].

Neutrophil adherence to endothelium was enhanced via A1 receptors and inhibited via A2 receptors [57, 58]. It is now believed that adenosine generated from ATP by CD39 and CD73 on the vascular surface functions as an anti-adhesive signal for neutrophil binding to microvascular endothelia through activation of neutrophil adenosine A2A and A2B receptors [59]. Activation of A2A receptors also inhibits expression of α4/β1 integrin on human neutrophils [60]. Human neutrophils activated by fMLP increased the number of cell surface β2 integrins on endothelial cells and induced the shedding of L-selectin. These effects were inhibited by adenosine, most likely via the A2A receptor [61]. A2 receptor activation inhibited neutrophil injury to coronary endothelium [62]. Adenosine also acts on endothelial receptors, thereby promoting vascular barrier function, providing a mechanism to dampen vascular leak syndrome during neutrophil–endothelial interactions [63] and regulating neutrophil chemotaxis [64]. Exposure of human endothelial cells to hypoxia/re-oxygenation caused increased neutrophil adhesion, an effect prevented by adenosine [65]. Adenosine also reduced the stimulatory effect of neutrophils on tissue factor-dependent coagulant activity of endothelial cells as a result of the inhibition of neutrophil adhesion to endothelial cells mediated by A2 receptors [66].

Adenosine might also play a role in the regulation of neutrophil number. Synergistic effects of granulocyte colony-stimulating factor and dipyridamole increased neutrophil production in mice [67]. Both effects were inhibited by adenosine deaminase (ADA). Theophylline has an immunomodulatory action on neutrophil apoptosis via A2A receptor antagonism [68].

The expression of adenosine receptors on neutrophils can be modulated in pathological conditions and following various interventions. A2A receptors on freshly isolated human neutrophils are upregulated after stimulation by LPS or TNF-α, and this may represent a feedback mechanism to control inflammation [69]. A2B receptor activity in neutrophils is reduced in patients with systemic sclerosis [70]. A 4.6-fold decrease in adenosine-mediated inhibition of neutrophils from patients with septic shock was reported [71]. Hypertonic saline upregulates A3 receptor expression on activated neutrophils and increases acute lung injury after sepsis [72]. Alterations in the functional expression of both A2A and A3 receptors in human neutrophils treated with pulsing electromagnetic fields have been reported [73, 74].

P2 receptors

ATP induces an increase in [Ca2+]i in human [75] and mouse [76] neutrophils. ATP and UTP, acting via P2U (i.e. P2Y2 and/or P2Y4) receptors, coupled to the inositol 1,4,5-trisphosphate pathway and increased [Ca2+]i [37]. This was associated with a priming of neutrophils for enhanced O2 generation when stimulated by other agonists [37, 77, 78]. The release of Ca2+ from thapsigargin-sensitive intracellular stores is essential for this nucleotide-induced priming in human neutrophils [79], indicating mediation via P2Y receptors. Enhanced O2 responses of rat neutrophils stimulated by formyl chemotactic peptide were evoked by ATP and ADP, whereas adenosine and AMP were inhibitory [8082]. ATP and UTP also stimulated granule secretion from human neutrophils [83, 84] and potentiated the secretion induced by chemotactic peptides [78]. They also induced neutrophil aggregation [78, 85].

Human neutrophils release ATP from the leading edge of the cell surface to amplify chemotaxic signals and direct cell orientation by feedback via P2Y2 receptors (Fig. 1) [24, 55, 86]. The importance of this mechanism in pathology is demonstrated by studies showing that the infiltration of neutrophils in the smoke-injured lung [87] and in the liver damaged by toxic agents [88] is decreased in P2Y2 knockout (−/−) mice. Chen et al. [24] also showed that neutrophil ectonucleotidases hydrolyze ATP to adenosine, which, via A3 receptors, also promoted cell migration (Fig. 1). In agreement with this concept, both P2Y2 and A3 receptors control the recruitment of neutrophils to the lungs in a mouse model of sepsis [53]. Neutrophil chemotaxis requires excitatory signals at the front and inhibitory signals at the back of cells that regulate cell migration. P2Y2 receptors, as well as A3 receptors, were shown to contribute to excitatory signals at the front, while adenosine acting on A2A receptors contributed to the inhibitory signal at the back [55] (Fig. 1).

The P2Y14 receptor was shown to be functionally expressed on human neutrophils [89], and uridine-diphosphate (UDP)-sugars promoted Rho-mediated signalling and chemotaxis in human neutrophils [90], which was blocked by a P2Y14 antagonist [91].

Neutrophil apoptosis induced by ATP was inhibited by P2Y11 receptor activation, and it was suggested that targeting of P2Y11 receptors could retain the immune functions of neutrophils and reduce the injurious effects of increased neutrophil longevity during inflammation [92]. A later paper showed that P2Y11 receptors mediate ATP-enhanced chemotactic responses of rat neutrophils [93].

RT-PCR and Northern blot analysis revealed the presence of P2X7 receptors on neutrophils and 2′ (3′)-O-(4-benzoylbenzoyl) ATP (BzATP), a potent P2X7 receptor agonist, stimulated production of O2 [23, 94]. A role of P2X7 in protection against neutrophil apoptosis has been reported [95, 96]. Neutrophil accumulation in the skin during croton oil-induced irritant contact dermatitis was reduced in P2X7-deficient mice [97]. However, it was claimed more recently that human neutrophils do not express P2X7 receptors [98]. In an RT-PCR study of human neutrophils, mRNA for P2X1 was strongly expressed, while mRNA for P2X4 and P2X5 was weakly expressed and P2X7 mRNA was not detected [99]. P2X1 receptors mediate neutrophil chemotaxis via Rho kinase activation [100]. A study using P2X1 receptor knockout mice led to the conclusion that P2X1 receptors play a protective role in endotoxaemia by negatively regulating systemic neutrophil activation, thereby limiting the oxidative response, coagulation, and organ damage [101].

Eosinophils

P1 receptors

A3 receptors were identified on human eosinophils and their activation led to increased [Ca2+]i [102]. However, the role of adenosine and A3 receptor signalling on this cell type remains controversial with both pro- and anti-inflammatory activities of adenosine being reported. Adenosine was shown to potentiate the production of O2 by guinea pig pulmonary eosinophils [103]. However, an inhibition of degranulation and O2 anion release from human eosinophils was observed later and shown to be mediated by A3 receptors [104]. In human eosinophils, adenosine inhibits chemotaxis via the A3 receptor [105, 106], whereas a stimulatory effect has been observed in eosinophils of ADA-deficient mice [107].

P2 receptors

Nucleotides were shown to stimulate human eosinophils, and it was suggested that since ATP is released from autonomic nerves and activated platelets, it could modulate the migration and other activities of eosinophils in vivo [76]. Thrombin-stimulated platelets secrete ATP, a chemotactic factor that attracts eosinophils [108]. ATP was shown to be a potent activator of eosinophils, suggesting a role for ATP in the pathogenesis of eosinophilic inflammation as an activator of pro-inflammatory effector functions [109]. Expression of P2Y1, P2Y2, P2Y4, P2Y6, P2Y11 and P2X1, P2X4, P2X5 and P2X7 receptor mRNA has been observed in human eosinophils (see Fig. 2) [99, 110]. It was also shown in this paper that purinoceptors mediate increase in [Ca2+]i and the production of reactive oxygen intermediates. The functional characterization of P2Y and P2X receptors on human eosinophils was undertaken, and it was shown that UTP and ATP had a greater stimulatory effect on the production of reactive oxygen metabolites, actin polymerization and chemotaxis than the selective P2X receptor agonists α,β-methylene ATP and BzATP, suggesting a predominant role of P2Y receptors [111]. However, P2Y and P2X agonists had similar effects regarding intracellular calcium transients and the adhesion molecule CD11b. In a study of human eosinophils, ATP was shown to trigger secretion of eosinophil cationic protein, probably via P2Y2 receptors, while ATP induced interleukin (IL)-8, probably via P2Y6, P2X1 and P2X7 receptors [112]. Autocrine release of ATP and P2 receptors, presumably P2Y2, were shown to play a pivotal role in human eosinophil degranulation and production of pro-inflammatory cytokines in response to the endogenous danger signal, crystalline uric acid [113]. Human eosinophils respond also to ADP via P2Y12 receptors to elicit eosinophil secretion of peroxidase [114]. The use of knockout mice has allowed us to demonstrate the crucial role of P2Y2 receptors in the accumulation of eosinophils in the lungs during allergic inflammation. This involves both a direct chemotactic effect of ATP mediated by the eosinophil P2Y2 receptor [115] and an indirect effect on endothelial cells, where ATP via P2Y2 stimulates the expression of VCAM-1 that mediates eosinophil adherence and infiltration, and its soluble form that is chemotactic for eosinophils [116].

Fig. 2.

Fig. 2

P2 receptors expressed by human eosinophils. a P2Y receptors. b P2X receptors. (Reproduced from [110], with permission from Elsevier)

Basophils

P1 receptors

It was reported that human basophils have a receptor for adenosine that mediates inhibition of immunoglobulin (Ig)E-mediated histamine release [117, 118]. In later papers, it was shown that the inhibitory effect of adenosine is mediated by an A2 receptor and cAMP increase [119121].

P2 receptors

Activation of permeabilised rat basophilic leukaemia cells (RBL-2H3) by adenosine-5′-O-(3-thio)triphosphate led to secretion of allergic and inflammatory mediators [122]. In a recent paper, it was shown that degranulation and histamine release from human basophils, associated with type 1 allergy, was evoked by UTP and particularly UDP, suggesting mediation by P2Y2 and/or P2Y4 and P2Y6 receptors [123].

Mast cells

P1 receptors

Potentiation of A23187 calcium ionophore-induced mast cell release of histamine by adenosine was initially reported [124]. Anti-IgE-induced release of histamine from mast cells was also enhanced by adenosine [125130], as was β-hexosaminidase release from bone marrow-derived mast cells [131]. Histamine release from human adenoidal mast cells induced by concanavalin A or acetylcholine was also enhanced by adenosine [132]. Although other mechanisms have been proposed [133, 134], the potentiation of histamine release by adenosine appears to be mediated by A3 receptors, since it was mimicked by selective A3 agonists [135, 136] and abolished in A3-deficient mice [137]. Furthermore, it was shown that the increase of cutaneous vascular permeability and extravasation of plasma proteins in response to adenosine was abolished in mast cell-deficient mice as well as in A3-deficient mice [138]. Similarly, adenosine-induced bronchoconstriction was attenuated in mast cell-deficient and A3-deficient mice [139].

The response of human lung mast cells to adenosine was biphasic: low concentrations of adenosine potentiated release of histamine, while high concentrations elicited inhibition [140]. Adenosine also inhibited IgE-dependent degranulation of human skin mast cells via A2A receptors [141]. Both A2A and A2B receptors were identified on mouse bone marrow-derived mast cells [142]. Using knockout mice, it was demonstrated that the inhibition of mast cell degranulation by adenosine in mediated by the A2B receptor, while the combined action of A2B and A2A receptors is responsible for the inhibition of cytokine production [143]. However, the role of adenosine receptors in mast cell regulation is more complex, since the stimulatory effect of adenosine on the release of angiogenic factors such as vascular endothelial growth factor (VEGF) was shown to involve a cooperation between A2B and A3 receptors [144, 145]. Furthermore, in umbilical cord blood-derived mast cells, IL-4 increased the potentiating effect of adenosine on degranulation via an upregulation of A2B receptors, whereas these receptors were shown previously to be inhibitory in murine mast cells [146].

P2 receptors

ATP was reported early to evoke calcium-dependent histamine release with degranulation of rat mast cells [147152]. It was suggested that the source of ATP may be innervating nerve fibres [153, 154], as discussed below. A correlation was shown between the ATP levels in rat peritoneal mast cells and histamine released by the anaphylactic reaction and compound 48/80 [155, 156]. ATP was shown to induce cytokine expression and apoptosis via P2X7 receptors on murine mast cells [157], supporting the earlier recognition of ATP-induced pore formation in rat peritoneal mast cells [158]. Interestingly, colitis was improved in mast cell-deficient mice as well as in those mice reconstituted with P2X7 −/− mast cells, showing the role of mast cell activation by ATP via the P2X7 receptor in intestinal inflammation [159]. ATP-induced cytokine and chemokine expression could also be mediated by P2X1 and P2X3 receptors on murine mast cells [160]. Functional expression of P2X1, P2X4 and P2X7 receptors in human lung mast cells was presented [161]. Mast cells are a major source of protein arginine deiminase, and it was shown that ATP induced protein arginine deiminase 2-dependent citrullination in mast cells via P2X7 receptors [162].

G protein-coupled P2Y receptors were also shown to mediate mast cell activation [163, 164]. UDP-glucose acting via P2Y14 receptors was shown to be a mediator of mast cell degranulation and considered as a potential therapeutic target for allergic conditions [165]. In a recent paper, all P2Y receptor subtypes were shown to be expressed in variable levels by human LAD2 mast cells [166]. Although P2Y4 and P2Y11 receptors were highly expressed, they did not appear to play a major role in degranulation, whereas P2Y14 receptors did.

Autonomic nerves as well as sensory-motor nerves innervate immune cells and release ATP as a cotransmitter in close vicinity of immune cells [167]. Indeed in accordance with the definition of the autonomic neuroeffector junction, close contact of nerve varicosities with effector cells in effect constitutes innervation, albeit of a transient nature [168, 169]. Mast cells were the first claimed to be innervated [170]. Antidromic stimulation of sensory nerves increased degranulation of mast cells in the skin, and this effect was mimicked by ATP [171]. Close opposition of nerve varicosities containing small and large vesicles and mast cells in the mucosa of intestine was shown with electron microscopy [154, 172] and also in cerebral blood vessels [173] (Fig. 3). Synovial mast cell activity that contributes to inflammation in joints was shown to be influenced by both unmyelinated afferent and sympathetic efferent nerves [174]. Sympathetic and trigeminal sensory nerve fibres influence rat dural mast cells and have been shown to play a role in the pathophysiology of vascular headache [175]. Functional interactions between sensory nerves and mast cells of the dura mater have been described in both normal and in inflammatory conditions [176]. Vagus nerve stimulation modulates histamine content in mast cells in the rat jejunal mucosa [177]. From a study of co-cultures of nerves and mast cells, it was concluded that ATP released from activated mast cells was an important mediator to activate nerves [178, 179]. While substance P released from nerves activated mast cells, ATP released from mast cells in response to anti-IgE antibody activated superior cervical ganglia neurons. Few investigations have been carried out about the influence of nerves on non-mast cell immune cells, but evidence has been presented that nerve fibres form close relationships with other immune cells, such as eosinophils [180], macrophages [181], and T and B lymphocytes [182184].

Fig. 3.

Fig. 3

a Close apposition between rat mast cell protease 1 immunoreactive and calcitonin gene-related peptide immunoreactive nerve fibres observed by confocal microscopy. b Ultrathin section of rabbit middle cerebral artery showing granular cells (G) separated by a distance of less than 200 nm. V varicosities; arrowheads basement membranes. Magnification, ×29374. (a Reproduced from [176] and b from [173], with permission from Elsevier)

Section summary

Adenosine and ATP have opposite effects on O2 generation and other functions of neutrophils: adenosine has an inhibitory effect, mediated mainly by A2A and A2B receptors, while ATP has a potentiating effect. On the other hand, ATP and adenosine cooperate to amplify the migration of neutrophils induced by chemotactic signals: this involves a stimulatory effect mediated by P2Y2 and A3 receptors expressed at the front of the neutrophils and an inhibitory effect of A2A receptors expressed at the back of the cells.

ATP via P2Y2 receptors also plays an important role in the migration of eosinophils and their accumulation in the lungs during allergic inflammation. Adenosine exerts a dual effect on mast cell degranulation: stimulation through A3 receptors and inhibition via A2A and A2B receptors.

Monocytes, macrophages and microglia

Monocytes

P1 receptors

Adenosine was initially reported to inhibit the production of the second complement component (C2) of human monocytes [185], and this effect was later shown to be mediated by A2 receptors [186]. Subsequently, it was also shown that A1 receptors were expressed on cultured human monocytes and rheumatoid synovial fluid mononuclear phagocytes [187]. Enhancement of Fcγ receptor-mediated phagocytosis was induced via A1 receptors, while A2 receptors mediated reduction of Fcγ phagocytosis in cultured monocytes. TNF-α production in human monocytes was inhibited by P1 receptor agonists [188]. Both A2A and A2B receptors were shown to be involved in the inhibition of TNF-α production [189]. Activation of A2A receptors also inhibited IL-12 and stimulated IL-10 production by human monocytes [190, 191]. These actions may contribute to suppression of Th1 responses. However, the effect of P1 receptor agonists on cytokine release from human mononuclear cells was shown to depend on the specific Toll-like receptor (TLR) subtype used for stimulation: the A2A agonist CGS21680 inhibited TLR4-mediated TNF-α release, but potentiated TLR3- and TLR5-mediated IL-6 release [192]. Activation of A2A receptors also inhibited LPS-induced IL-18 production, expression of adhesion molecules and production of TNF-α, in human monocytes [193, 194]. Activation of A1 receptors promoted multinucleated giant cell formation by human monocytes [195]. Adenosine analogues were shown to produce apoptosis of human mononuclear cells via A2A and A3 receptors [196].

P2 receptors

ATP and ADP were initially shown to increase [Ca2+]i in monocytes and to regulate the activity of adhesion receptors CD11b/CD18 [197]. ATP and ADP activated human promonocytic U-937 cells apparently via different P2 receptor subtypes [198]. mRNA for P2X7 and P2Y2 receptors was shown to be expressed by human THP-1 monocytic cells and monocytes, and the presence of these receptors was supported by pharmacological data [199201]. P2X7 receptor expression in THP-1 monocytes was positively modulated by pro-inflammatory stimuli and negatively modulated by cAMP, a classic anti-inflammatory second messenger [202]. P2X7 receptors mediated ATP-induced IL-1β release from human and canine monocytes [203205], an effect requiring priming by LPS [206]. This mechanism plays a major role in the physiological control of IL-1β secretion by monocytes. Indeed microbial components acting on different pathogen-sensing receptors, as well as the danger signals uric acid and C3a, induced the activation of human monocytes and their secretion of IL-1β and IL-18 through a process involving, as an initial event, the release of ATP [207209]. This was followed by the autocrine stimulation of P2X7 receptors and inflammasome activation [210] (Fig. 4). Indeed, IL-1β secretion was inhibited by apyrase as well as by P2X7 antagonists. Additional evidence in favour of the involvement of P2X7 was the observation that the P2X7 receptor polymorphism Glu496Ala, which is associated with a loss of function, impaired ATP-induced IL-1β release from human monocytes [211].

Fig. 4.

Fig. 4

Hypothetical sequence of events leading to P2X7 receptor and pannexin 1 (panx-1)-mediated inflammasome activation. Pathogen-associated molecular patterns (PAMPs) bind to Toll-like receptors (TLRs) and drive interleukin (IL)-1β gene expression and accumulation of the pro-cytokine. Extracellular ATP binds to the P2X7 receptor and triggers K+ efflux and panx-1 activation. The functional significance of K+ efflux is unknown, although it might facilitate or even precipitate inflammasome activation. Likewise, the mechanism of panx-1 activation by the P2X7 receptor is unknown. Panx-1 in turn activates the inflammasome. Data suggest that the ion-carrying activity of panx-1 is unnecessary for inflammasome activation. The activated inflammasome then cleaves pro-IL-1β. Thus, stimulation of the inflammasome by extracellular ATP can be split into two steps: (a) recruitment and activation of panx-1 by the P2X7 receptor and (b) activation of the inflammasome by panx-1. Colour coding: white PAMP, red TLR, green NALP3 inflammasome, orange protein–protein interaction domains, further subdivided into orange square, ASC apoptosis-associated speck-like protein containing a caspase-recruitment domain and orange octagon, pyrin domain; yellow FIIND domain, light blue caspase domain (Casp-1), dark blue biologically active IL-1β and IL-1β propiece, violet P2X7 receptor, light green panx-1. (Reproduced from [210], with permission from Elsevier)

ATP was initially described as a chemoattractant for monocytes [212, 213]. More recently apoptotic thymocytes were found to release nucleotides leading to the recruitment of monocytes [3]. This release is mediated by pannexin 1 channels, as demonstrated by the use of pharmacological inhibitors and small interfering RNA (siRNA), and involves the activation of pannexin 1 by caspases [9]. Monocyte recruitment by apoptotic cells supernatants, demonstrated inter alia in the murine air-pouch model, was decreased in P2Y2 −/− mice, leading to impaired clearance of the apoptotic cells. These data clearly identify ATP as a find-me signal acting through the P2Y2 receptor that recruits monocytes in order to clear apoptotic cells.

Other effects of extracellular nucleotides on monocytes include increased surface expression of Mac-1 integrin [214], secretion of IL-8 that might involve P2Y2 and P2Y6 receptors [215, 216], inhibition of soluble HLA-G secretion [217], secretion of VEGF [218] and modulation of phagocytosis [219]. These last 3 effects involve P2X7 receptors. In human monocytes, ATP was reported to increase cAMP via the P2Y11 receptor, and thereby to inhibit proinflammatory cytokines production and to increase the release of IL-10 [213].

Macrophages

P1 receptors

Chemotaxis and lysosomal secretion were shown to be inhibited by adenosine and analogues in the mouse macrophage cell line RAW 264 or murine peritoneal macrophages [220, 221]. Adenosine was reported to inhibit TNF-α expression, induced by LPS in the mouse macrophage cell lines J774.1 [222] and RAW264.7 [223], whereas it potentiated nitric oxide synthase (NOS) expression induced by LPS in RAW 264.7 mouse macrophages [224, 225]. Interferon (IFN)-γ upregulated A2B receptor expression in macrophages [226], while TNF-α or LPS induced A2A expression via nuclear factor-κB, as part of a feedback mechanism for macrophage deactivation [227, 228]. TNF-α release from macrophages was inhibited by adenosine via A2A and A2B receptors [229232] and IL-10 production was augmented by adenosine acting through A2B [233] or A2A [234, 235] receptors. Interestingly, it was shown that pro-inflammatory macrophages (M1 cells that release TNF-α) have a low expression of ecto-nucleotidases and rate of ATP hydrolysis as compared to anti-inflammatory macrophages (M2 cells that release IL-10) [236]. A2A receptors also upregulated the expression of peroxisome proliferator-activated receptors [237] and hypoxia-inducible factor 1 [238]; this could contribute to the anti-inflammatory and tissue-protecting action of adenosine. A2A receptors mediated upregulation of vascular endothelial growth factor expression in murine [239] and human [240] macrophages. On the other hand, activation of A3 receptors stimulates matrix metalloproteinase-9 secretion by macrophages [241], and glucocorticoids promote survival of macrophages through stimulation of A3 receptors [242].

P2 receptors

Early reports showed that ATP permeabilised the plasma membrane to fluorescent dyes [243, 244], promoted cation fluxes [245247], increased [Ca2+]i, induced a respiratory burst and O2 generation [248, 249], inhibited phagocytosis [250] and induced cytotoxicity [251] and cell lysis [252] in a variety of macrophage populations. ATP was also shown to stimulate phosphoinositides hydrolysis and eicosanoid synthesis in mouse peritoneal macrophages [253]. Oxidized ATP (oxATP) was shown to irreversibly inhibit the permeabilization of the plasma membrane, but not the fast mobilization of Ca2+ induced by ATP in macrophages, supporting the expression of P2X7, then called P2Z, receptors in the J774 macrophage cell line [254]. P2X7 receptors were also shown to be expressed by BAC1.2F5 mouse macrophages, mediating both pore-forming and phospholipase (PL)-D activity [255], and in human monocyte-derived macrophages [256, 257].

Later studies demonstrated the involvement of the P2X7 receptor in several responses of macrophages to danger, in particular the proinflammatory response mediated by IL-1β secretion, bacterial killing and the associated macrophage death. ATP was shown to promote the maturation and release of IL-1β from macrophages [258, 259], via P2X7 receptors [260, 261]. ATP-induced secretion of IL-1β was abolished in macrophages from P2X7-deficient mice and involved inflammasome assembly and caspase-1 activation [262264]. Activation of the inflammasome and release of IL-1β in macrophages dying through autophagy [265] or stimulated by serum amyloid A [266] involved the release of ATP and the activation of P2X7. P2X7 −/− mice showed increased survival after lung adenoviral infection, resulting from a decreased production of IL-1β by macrophages [264]. These mice were also protected against smoke-induced lung inflammation and emphysema, as a result of decreased activation of lung macrophages [267].

P2X7-mediated ATP-induced killing of mycobacteria by human macrophages was initially reported in 1997 [268]. This seminal observation was later confirmed in numerous studies. Mycobacterial killing involved phagosome–lysosome fusion [269] that was induced by the rise of Ca2+ and the activation of PLD resulting from P2X7 activation [270]. It was decreased in macrophages from P2X7 −/− mice [271]. Infection by mycobacteria upregulated the expression of P2X7 and its activation by ATP not only enhanced intracellular bacterial killing but also induced the apoptosis of macrophages [272] or autophagy [273]. This dual response was missing in macrophages from P2X7 −/− mice [271]. ATP-induced bacterial killing was abrogated in macrophages from individuals homozygous for a loss of function P2X7 polymorphism [274] and reduced by 50 % in heterozygous subjects [275]. Additional polymorphisms leading to similar consequences were described later [276]. Furthermore, the pattern of gene expression in response to ATP was different in patients with tuberculosis and controls, suggesting that a defective function of P2X7 might lead to the development of tuberculosis [277]. Infection by parasites, such as Leishmania amazonensis [278, 279] and Toxoplasma gondii [280, 281], also increased the expression of P2X7 that mediated a dual response of parasite killing and macrophage apoptosis.

The P2X7 receptor is also involved in various additional responses of macrophages. ATP released by LPS increased NOS expression and NO production in RAW 264.7 macrophages via P2X7 receptors [282288]. The P2X7 receptor was also associated with the generation of reactive oxygen species (ROS) [289291] and leukotriene B4 [279, 292]. Activation of P2X7 receptors on macrophages induces the activation and release of tissue factor and thus favours thrombosis [293, 294]. Phagocytosis of nonopsonised beads and heat-killed bacteria was increased by P2X7 over-expression, showing that it can behave as a scavenger receptor, but this effect was inhibited by ATP [219, 295]. Loss of function polymorphisms of P2X7 and P2X4 receptors were associated with reduced phagocytosis and were overrepresented in patients with macular degeneration [296]. P2X7 receptors play a role in the generation of macrophage-derived giant cells, a hallmark of chronic inflammation [297]. Spontaneous cell fusion was indeed described in macrophage cultures expressing high levels of the P2X7 receptors [298]. Furthermore, the formation of multinucleated giant cells was inhibited by P2X7 antagonists and in macrophages from P2X7-deficient mice [299, 300].

Despite the dominant role of P2X7 in macrophages, evidence has accumulated to support the role of additional receptors. Multiple P2X and P2Y receptor subtypes were identified in mouse J774, spleen and peritoneal macrophages [301]. In an extensive study, mRNA for P2X1, P2X4, P2X5, P2X7, P2Y2, P2Y4, P2Y6, P2Y11, P2Y13 and P2Y14 receptors were all expressed by human alveolar macrophages [302]. It was suggested that other P2X receptor subtypes, in addition to P2X7 receptors, were involved in the ATP-mediated current in human macrophages [303]. In particular it was shown that a small slowly-desensitising ATP-induced current was abolished in P2X4 −/− mice [304]. This P2X4 response might contribute to the P2X7-induced cell death that was reduced by siRNA against P2X4 [305, 306]. It has been reported that HIV binding to macrophages stimulates the release of ATP and that P2X1 is necessary for the entry of HIV in macrophages [307]. P2Y receptors are also expressed and functional. Low concentrations of ATP were shown to activate PLC and IL-6 transcription [308]. Studies of P2Y2 and P2Y4 receptor knockout mice led to the conclusion that P2Y2 receptors are the dominant P2Y receptor subtype in mouse peritoneal macrophages [309]. Nucleotides, released by apoptotic cells, through pannexin 1 [310], act as ‘find-me’ signals to promote P2Y2-dependent recruitment of phagocytic macrophages (as well as monocytes and dendritic cells (DCs)) and this recruitment is reduced in P2Y2-deficient mice [3]. The chemoattractant effect of C5a on macrophages was amplified by the release of ATP and the autocrine stimulation of P2Y2 and also P2Y12 receptors [311]. P2Y2 receptors also mediate potentiation of prostaglandin E2 release involved in the induction of NOS [312, 313] and stimulate the production of monocyte chemoattractant protein-1 (MCP-1)/chemokine (C-C motif) ligand 2 (CCL2) [314]. Furthermore LPS potentiated nucleotide-induced inflammatory gene expression via upregulation of P2Y2 receptors [315]. P2Y6 receptor expression also increased following macrophage activation [309]. Indeed the amount of IL-6 and macrophage inflammatory protein-2 released in response to LPS was significantly enhanced in the presence of UDP, and this effect was lost in the macrophages of P2Y6 knockout mice [316]. Activation of P2Y6 receptors increased the clearance of Escherichia coli and improved survival to peritonitis through the release of MCP-1 and enhancement of macrophage chemotaxis [317]. The P2Y11 receptor was also reported to be functional in macrophages [318]. These authors observed that ATP released from LPS-activated macrophages by vesicular exocytosis activated the P2Y11 receptor, leading to a M1 polarisation characterized by an increased production of IL-12 [318].

Microglia

P1 receptors

The first evidence of a role of adenosine and its receptors in microglia was derived from the observation of effects of propentophylline, a neuroprotective xanthine derivative that increases the extracellular concentration of adenosine by inhibiting its transport into cells [319]. Propentofylline was shown to inhibit the production of ROS by microglial cells [320, 321], their uptake of amyloid precursor protein [322, 323] and their proliferation and release of TNF-α [324]. Further studies showed that microglia express all subtypes of adenosine receptors. Enhanced activation of microglia associated with worsened demyelination and axonal damage was observed in A1 receptor knockout mice subjected to experimental allergic encephalomyelitis [325]. ATP-triggered migration of microglia was inhibited in A1 −/− as well as CD39−/− mice [326]. The A3 receptor is also involved in microglial process extension and migration [327]. On the other hand, ATP acted as a repellent for LPS-treated microglia and induced process retraction; these actions were associated with the upregulation of A2A receptors [328]. A2A receptor knockout mice also displayed enhanced microglial activation in a model of experimental autoimmune encephalomyelitis (EAE) [329].

P2 receptors

It was initially reported that ATP, but not ADP, induced an inward current in microglia [330], associated with an increase in cytosolic Ca2+ [331]. Further pharmacological studies suggested that these responses were mediated by P2Y receptors [332, 333]. It was later shown that the ATP effect on Ca2+ influx was mimicked by BzATP and inhibited by oxATP, supporting the role of the P2z or P2X7 receptor [334]. This receptor was shown to mediate the secretion of IL-1β induced by ATP or by LPS via the release of ATP [335], and to induce microglia cell death [336] as well as microglia-mediated injury of neurons [337]. The P2X7 receptor was also shown to be involved in microglial activation by amyloid β [338]. After nerve injury, the P2X4 receptor was upregulated in the spinal cord and selectively expressed in microglia [339]. The tactile allodynia induced by nerve injury was suppressed by antisense oligodeoxynucleotides silencing P2X4 receptors. Knockdown of the P2X4 receptor by siRNA inhibited migration of microglia [340].

Following brain injury, microglia extrude processes and migrate toward sites of tissue damage. Polarisation, process extension and chemotaxis did not occur in P2Y12-deficient mice, while baseline motility was normal [341]. Furthermore, in living P2Y12-deficient mice, branch extension toward sites of cortical damage was decreased. Microglial activation leads to the downregulation of P2Y12 receptors and the upregulation of P2Y6 receptors [342, 343]. Activation of P2Y6 receptors by UDP stimulates phagocytosis and the uptake of microspheres. In vivo an upregulation of P2Y6 was observed following administration of kainic that damages neurons, leading to microglia activation. Taken together these findings show that ADP, acting through P2Y12, is a find-me signal for microglia, whereas UDP, acting on P2Y6, behaves as an eat-me signal [344] (Fig. 5).

Fig. 5.

Fig. 5

Independence of P2Y12 receptor-mediated migration and P2Y6 receptor-mediated phagocytosis in microglia. a Release/leakage of adenine nucleotides/nucleosides and uridine nucleotides from injured neurons. When neurons or cells are injured or dead, high concentrations of ATP (∼mM) and UTP at a concentration of less than 10 % are leaked. Compared with ATP/ADP/adenosine, UTP/UDP should be transient and localized signals. b Changes in P2Y12 and P2Y6 receptors in microglia according to their activation stages. Insert shows pharmacological characterization of P2Y6 receptor. UDP is a selective agonist to the P2Y6 receptor, and thus, it does not stimulate P2Y12, P2X4, A1, or A2A receptors. Similarly, the P2Y6 receptor is a very selective receptor for UDP, and therefore, is not activated by ATP, ADP, or adenosine (Ado). Resting microglia express no or only faint P2Y6 receptors; whereas, they express P2Y12 receptors adequately. When microglia are activated, they increase P2Y6 receptors; whereas, they decrease P2Y12 receptors. Only when activated microglia meet UDP at the injured sites do they sense UDP as an eat-me signal. c Microglial migration and phagocytosis are controlled by distinct P2 receptors. When microglia sense ATP/ADP by P2Y12 receptors, they extrude their processes, followed by migration toward the injured sites. These microglial motilities are not affected by UDP/P2Y6 receptors. When activated, microglia upregulate P2Y6 receptors, and if they sense the eat-me signal UDP, they start to phagocytose the dead cells or debris. The phagocytic responses are not affected by the activation of P2Y12 receptors nor by other P2 or P1 receptors. (Reproduced from [344], with permission from Springer)

Section summary

ATP released from apoptotic cells constitutes a find-me signal that attracts monocytes/macrophages, an action mediated by the P2Y2 receptor. It stimulates bacterial killing and macrophage apoptosis thereby contributing to decrease the bacterial and parasite burden: this action is mediated by the P2X7 receptor. ATP also exerts a proinflammatory effect through the secretion of IL1-β, which is mediated by the P2X7 receptor and NLRP3 inflammasome.

In contrast, adenosine exerts an inhibitory effect on monocytes/macrophages mediated by A2A and A2B receptors.

Multiple P1 and P2 receptors have been shown to play a role in microglia. The P2X7 receptor is involved in IL-1β secretion and cell death. P2Y12, P2X4, A1 and A3 receptors stimulate process extension and migration, whereas the A2A receptor is inhibitory. On the other hand the P2Y6 receptor is upregulated in activated microglia and triggers microglial phagocytosis.

Dendritic cells

P1 receptors

CD39 and CD73 ectonucleotidases [345] as well as A1, A2A and A3 but not A2B receptors [346] are expressed by human monocyte-derived DCs. In immature DCs, adenosine induced calcium transients but no increase in cAMP. This resulted in actin polymerization, chemotaxis [346] and increased expression of co-stimulatory molecules [347]. Maturation of DCs by LPS resulted in downregulation of A1 and A3 receptor mRNA, whereas A2A receptors were still expressed [346]. In these mature DCs, adenosine increased cAMP and inhibited IL-12 and TNF-α production, whereas it enhanced IL-10 secretion [346, 347]. These results show that adenosine can act as a chemotaxin for immature human DCs and induce their semi-maturation, characterized by a reduced capacity to induce a Th1 polarisation of CD4+ T lymphocytes [347]. Adenosine via cAMP also decreased the capacity of human DCs to prime CD8+ T cells [348].

In murine monocyte-derived DCs, adenosine also impaired maturation and inhibited the production of IL-12, leading to tolerance: this effect was mediated by the A2B receptor instead of the A2A receptor active in human cells [349351]. IL-27 is a cytokine produced by DCs that suppresses Th1 and Th17 responses and limits inflammation in several experimental models. The suppressive action of IL-27 was mediated at least in part by the induction of CD39 in DCs and the resulting accumulation of adenosine [352]. However, the observation that adenosine could also promote the development of murine Th17 cells, via the A2B receptor- mediated production of IL-6, added an additional complexity [353]. The A2B receptor was upregulated in EAE and A2B knockout mice developed less severe EAE than wild-type mice [354]. In human plasmacytoid DCs, adenosine plays a dual role by initially recruiting immature cells to sites of inflammation, an effect mediated by A1 receptors, and by subsequently inhibiting the production of IL-6 and IFN-α, via the A2A receptor [355].

The physiological importance of the inhibitory effect of adenosine on DCs is supported by observations on the role of ADA. Indeed the high ADA activity of DCs might help to maintain them in an active state [356]. ADA has been shown to be upregulated in DCs from non-obese diabetic (NOD) mice leading to their spontaneous activation and autoimmune T cell activation [357]. Paradoxically DCs from CD39−/− mice exhibited impaired antigen-presenting capacity and ability to induce a Th2 response [358, 359]. This resulted in decreased allergic contact hypersensitivity [358] and allergic airway inflammation [359]. This was explained not by a defect in adenosine formation but by an increased accumulation of ATP leading to the desensitization of P2Y receptors (see below).

Finally it must be mentioned that inosine has been reported to induce DCs chemotaxis independently from adenosine receptors [360]. On the other hand AMP was shown to mimic the inhibitory effects of adenosine on DCs, and these effects were maintained in CD73-deficient mice and could not be explained by adenosine contamination of AMP [361]. The mechanisms of these effects remain unknown.

P2 receptors

Human DCs express mRNA for almost all known P2 receptors [345, 362364] and extracellular nucleotides exert multiple effects on them ranging from chemotaxis to control of cytokine release and induction of cell death. P2Y but not P2X agonists are potent chemotactic stimuli for immature but not mature DCs [364]. Chemotaxis was associated with a rise in intracellular Ca2+ and actin polymerization and involved the activation of Gi. Allergen challenge was shown to cause acute accumulation of ATP in the airways of asthmatic subjects and mice with experimentally induced asthma that resulted in the recruitment of DCs [1]. That recruitment was mediated by the P2Y2 receptor. Indeed, in vitro the ATP-induced migration of P2Y2-deficient DCs was strongly decreased as compared to DCs from wild-type mice [115]. The attraction of DCs to the lungs in a model of allergic inflammation induced by ovalbumin was also decreased in P2Y2 −/− mice [115]. Decreased attraction of DCs to the airways might also explain the higher mortality of P2Y2 −/− mice with lung infection by pneumonia virus of mice, as a consequence of lowered immune response and viral clearance [365]. Interestingly, the formation of ATP gradients at a site of inflammation can also inhibit transiently the migration of human DCs, via the P2Y11 receptor, and thereby prolong the time of encounter with antigens [366]. Conversely, antagonism of the P2Y11 receptor might improve the migration of antigen-loaded DCs to the lymph nodes. In addition to these direct effects on migration, ATP modulated the expression of chemokine receptors, with an induction of CXCR4 and a reduction of CCR5 [367], and inhibited the release of CCL2 and CCL3 chemokines [368].

Nucleotides were also shown to modulate the maturation of DCs. Schnurr et al. [369] initially reported that ATP stimulates the expression of CD83 and the secretion of IL-12 by human monocyte-derived DCs. This action was shown to be mediated by the P2Y11 receptor and a rise in cAMP [370]. However, la Sala et al. [371] confirmed that ATP stimulates the maturation of DCs but observed an inhibitory effect on the release of IL-12 stimulated by LPS, leading to an impaired ability to initiate Th1 responses. These apparent discrepancies were resolved by the demonstration that ATP, via P2Y11, increased IL-12p40 but inhibited the production of IL-12p70 [372]. Furthermore ATP synergized with LPS and sCD40L to stimulate IL-10 production. This led to the conclusion that ATP, via the P2Y11 receptor, induces a semi-maturation of DCs, characterized by an increased expression of co-stimulatory molecules and a decreased production of bioactive IL-12, leading to increased Th2 responses or tolerance. Additional studies showed that ATP via the P2Y11 receptor produced an impressive upregulation of the expression of thrombospondin-1 and indoleamine 2,3-dioxygenase that could play a major role in tolerance [373]. A systematic study of the effect of ATP on gene expression in DCs revealed a P2Y11-mediated stimulatory effect on the expression of VEGF-A, that has immunosuppressive effects in addition to its angiogenic action [374], and amphiregulin, that can exert an angiogenic and tumorigenic action [375].

Other P2Y receptors were found to be expressed on monocyte-derived DCs. ATP can modulate the function of DCs directly via a cAMP increase mediated by P2Y11 receptors and indirectly via its degradation into ADP, which acts on P2Y1 receptors; these distinct mechanisms combine to inhibit inflammatory cytokine production, particularly IL-12, but have a differential effect on IL-10 [376]. P2Y12 receptors are also expressed by murine DCs and their activation increased antigen endocytosis with subsequent enhancement of T cell activation [377]. UDP, but not UTP, stimulated the release of CXC-chemokine 8 from mature human DCs, via P2Y6 receptors [378]. UTP and UDP also acted on murine DCs to mobilize intracellular Ca2+ and to induce cytokine production [379]. Human immature monocyte-derived DCs express P2Y14 receptors that mediate an increase in [Ca2+]i in response to agonists [380]. In plasmacytoid DCs, UTP, UDP and UDP-glucose were shown to inhibit IFN-α production [381].

Like in macrophages, ATP induced in DCs the NLRP3/ASC inflammasome signalling complexes that drive proteolytic maturation and secretion of the proinflammatory cytokines IL-1β and IL-18 [382, 383]. This action was mediated by the P2X7 receptor, which is functionally expressed on DCs [384]. P2X7 receptors were shown to be present in microvesicles shed from DCs together with IL-1β and caspase-1 and caspase-3 [385]. P2X7-deficient DCs fail to release IL-1β in response to LPS and ATP [386]. This might explain the resistance to allergic contact dermatitis observed in P2X7-deficient mice [386]. Additional P2X7-mediated effects of ATP on DCs include shedding of CD23 [387], release of tissue factor-bearing microparticles [388] and apoptosis [389, 390].

In the intestine ATP released from commensal bacteria induced the differentiation of Th17 CD4+ cells via the activation of lamina propria CD11c+ antigen-presenting cells, apparently via a P2X receptor [391]. The number of Th17 cells was increased in mice deficient in ENTPDase7, which is preferentially expressed on epithelial cells of the small intestine [392].

Section summary

ATP can exert multiple actions on DCs, mediated by distinct receptors: chemotaxis mediated mainly by the P2Y2 receptor; semi-maturation, characterized by increased expression of co-stimulatory molecules and inhibition by IL-12, which is mediated by the P2Y11 receptor and associated with a Th2 response or tolerance; induction of NLRP3/ASC inflammasome signalling complexes, mediated by the P2X7 receptor, that leads to secretion of IL-1β and a proinflammatory effect; and enhanced antigen endocytosis mediated by the P2Y12 receptor.

Adenosine acting on the A2A or A2B receptor exerts complex effects on DCs: as ATP it impairs Th1 polarisation and favours Th2 and/or tolerance, but it can also favour Th17 cell development.

Lymphocytes

T and B lymphocytes

P1 receptors

Adenosine was reported to cause an increase in cAMP in lymphocytes as well as in thymocytes [393396] and to have powerful inhibitory effects on lymphocyte proliferation [397] and the immune response in humans, particularly those who have inherited deficiency of ADA [398]. The destruction of tumour cells by mouse lymphocytes was shown to be inhibited by adenosine, and this effect was potentiated by an inhibitor of ADA [399]. It was suggested that this effect of adenosine may contribute to the lack of immune response associated with ADA deficiency.

ATPase, ADPase, 5′-nucleotidase and ADA have been shown to be present on human lymphocytes [400403]. Although it was claimed that adenosine release results from the intracellular degradation of ATP to adenosine, later studies showed that extracellular adenosine is generated following the release of ATP and its extracellular breakdown [404, 405]. Human B lymphocytes showed high degrading activity, while T lymphocytes were reported to be unable to degrade extracellular nucleotides [404]. However, ecto-ATPase activity was reported on cytolytic T lymphocytes [406], and E-NTPDase activity was upregulated within 15 min of T cell stimulation [407]. Furthermore, a subset of T regulatory (Treg) cells expresses CD39 and CD73 ectonucleotidases (see below). However it was suggested that CD39 is not the exclusive switch of the immune system to trigger immunosuppression, and that an alternative adenosine-generating axis is operating [408]. This axis involves the enzymes CD38 (a nicotinamide adenine dinucleotide (NAD+) nucleosidase) and CD303a (an ecto nucleotide pyrophosphatase).

A2A receptors were shown to be expressed on T lymphocytes [409411]. A2B receptors were also shown to be expressed on human T lymphocytes, and it was suggested that they play a role in lymphocyte deactivation by adenosine [412]. In another study, it was suggested that A2A receptors vary in their expression on T cell functional subsets and may regulate cytokine production in activated T lymphocytes [413]. There was lower expression of A2A receptors on B cells. A3 receptor mRNA and protein were shown to be expressed in both resting and activated human lymphocytes and under activating conditions they are upregulated [414]. Stimulation of A1 and A3 receptors were reported to block the inhibitory action mediated by A2A receptors [415]. Exposure to adenosine prior to antigenic stimulation also induced a desensitization of cAMP accumulation leading to a stronger response to antigenic stimulation [416].

Conclusive evidence for the major role of A2A receptor in the regulation of T lymphocytes came out of the study of A2A-deficient mice. cAMP accumulation in response to adenosine was decreased in T cells from A2A +/− mice and almost abolished in those of A2A −/− mice [417]. In CD4+ T cells, a selective A2A agonist had a major inhibitory effect on the T cell receptor (TCR)-mediated production of IFN-γ and this effect was decreased by 50 % in cells of A2A +/− mice and completely abolished in those from A2A −/− mice [418]. A2A receptor activation inhibited T cell proliferation and IL-2 production whether the cells were expanded under Th1 or Th2-skewing conditions, and again this inhibition was abolished in A2A-deficient mice [419]. Furthermore, TCR stimulation caused a rapid increase in A2A mRNA, both in Th1 and Th2 cells [418, 419].

Adenosine via A2A receptors exerts other effects on T cells. The apoptotic effect of adenosine on resting T cells was inhibited in A2A −/− mice [417, 420]. On the other hand, adenosine via A2A receptor inhibited activation-induced cell death of already activated T cells [421]. Furthermore, the A2A receptor contributes to the maintenance of a normal number of naive T cells by inhibiting TCR-induced activation [422]. Adenosine also inhibits T cell mobility [423], migration to lymph nodes [424] and adhesion to the endothelium [425].

The importance of the A2A-mediated inhibitory effect of adenosine on T cells was underscored by the discovery that CD39 is selectively expressed on Treg cells (see Fig. 6) [426] that are essential for maintaining peripheral tolerance [427, 428]. In human T cells, CD39 is expressed primarily by immunosuppressive Treg cells that express the Foxp3 transcription factor, and its activity is enhanced by TCR ligation [429]. CD73 is also expressed on CD4+/CD25+/Foxp3 Treg cells [430432]. However, subsets of Treg cells expressing CD39, but not CD73, have been identified [433]. Inhibition of ADA activity further enhanced Treg-mediated immunosuppression [432].

Fig. 6.

Fig. 6

The CD39/CD73 pathway modulates regulatory T cell (Treg) activity. The activation of T cell receptor (TCR), expressed on Tregs, induces CD39 activity. This increment of ATP-metabolizing activity is critical for the immunosuppressive activity of Tregs because it facilitates the pericellular generation of adenosine, a substantial component of the immunosuppressive and anti-inflammatory functions of Tregs. The inhibitory action of Treg-derived adenosine can be ascribed to the activation of A2A receptors expressed on T effector cells, which undergo reduced immune activity. In addition, adenosine generation triggers a self-reinforcing loop of Treg functions because the stimulation of A2A receptors expressed on these cells elicits their expansion and increases their immunoregulatory activity. (Reproduced from [426], with permission from Elsevier)

Several studies have shown the impact of adenosine-mediated inhibition of T cells in various models of disease. Defective adenosine-induced cAMP accumulation and immunosuppression were reported in T lymphocytes of patients with systemic lupus erythematosus [434, 435]. A2A receptor activation during reperfusion after ischemia protected the myocardium from infarction and this effect was dependent on an inhibition of T cell accumulation [436, 437]. A2A receptor agonists attenuated allograft rejection and alloantigen recognition by an action on T lymphocytes [438], suppressed the development of graft-versus-host disease [439, 440] and attenuated experimental autoimmune myasthenia gravis [441]. CD39 and CD73 expressed on Treg cells led to a local accumulation of adenosine that protected against Helicobacter induced gastritis [442]. Treg cells suppressed contact hypersensitivity reactions by a CD39 and adenosine-dependent mechanism [425]. In other models, the action of adenosine proved to be deleterious. A2A-deficient mice were protected from the lethal effect of sepsis, due to preserved lymphocyte function and decreased immunosuppressive IL-10 [443]. CD39 and CD73 expressed on ovarian cancer cells generate adenosine that exerts an immunosuppressive effect, which was relieved by siRNAs against CD39 and CD73 and by an A2A antagonist [444]. In HIV infection, Treg inhibitory effects were relieved by CD39 downregulation and reproduced by an A2A agonist [445, 446]. Furthermore, a polymorphism of the CD39 gene was identified, that is associated with downregulation of CD39 and slower progression to AIDS [446].

Few studies have been performed on B lymphocytes. Accumulation of cAMP produced by adenosine in B cells stimulated by Staphyloccocus aureus suppressed IgM production [447]. On the other hand B cells coexpress CD39 and CD73 and adenosine inhibited B cell proliferation and cytokine expression [448]. Activated B cells also inhibited T cell proliferation and cytokine production [448].

P2 receptors

Early reports showed that ATP protected rat lymphocytes against the loss of intracellular enzymes into the medium [449, 450] and that receptors for ATP were present on lymphocytes [451]. The action of ATP on lymphocytes is complex: ATP was reported to stimulate DNA synthesis in a subpopulations of T cells [452, 453], but ATP was also shown to be highly toxic to human lymphocytes and to thymocytes, causing permeabilization of the plasma membrane and cell death [454, 455]. It was later shown that ATP increased cytosolic Ca2+ in mouse thymocytes [456458] and stimulated the PLC pathway in human B lymphocytes [459]. On the other hand, an ATP4− receptor-operated sodium channel was identified on human lymphocytes [460, 461]. ATP-gated channels were also identified in human lymphoblasts [462].

Important advances were made in 1994 and the following years with the identification on human lymphocytes of P2Z, now called P2X7, receptors antagonised by oxATP [463] and by the isoquinoline derivative KN-62 [464]. P2X7 receptors were also identified specifically in human B lymphocytes [465, 466] and murine T lymphocytes [467]. P2X7 receptors were implicated in the mitogenic stimulation of human T lymphocytes purified from peripheral blood [468]. ATP and the selective P2X7 agonist BzATP caused plasma membrane depolarisation and a Ca2+ influx in T lymphocytes. ATP or BzATP alone had no effect on lymphocyte proliferation but potentiated the action of mitogens such as anti-CD3 [468]. Transfection of lymphoid cells lacking P2X7 receptors with P2X7 cDNA increased their proliferation [469]. Later studies showed that TCR stimulation triggers the release of ATP through pannexin-1 hemichannels [470] and vesicular exocytosis [471], and upregulates P2X7 expression [472]. siRNA silencing of P2X7 inhibited T cell activation, which was also lower in C57BL/6 mice that express a poorly functional P2X7 receptor, as compared to BALB/c mice that express fully functional P2X7 receptors [472]. Shockwaves increased T cell proliferation through ATP release and P2X7 activation [473]. Thus ATP released through pannexin 1 channels enhances T call activation in an autocrine manner (Fig. 7; [474]). But it is also involved in a paracrine communication that leads to calcium waves in neighbouring lymphocytes and a reduction of T cell motility in lymph nodes that would favour T cell scanning of antigen-loaded DCs [475]. However, P2X7 receptors also induced the shedding of L-selectin (CD62L) from T cells, which accompanies T cell activation and allows T cells to move away from lymph nodes and enter the circulation [476482].

Fig. 7.

Fig. 7

Purinergic signalling in T cell activation. Antigen recognition by T cells involves the formation of an immune synapse between a T cell and an antigen-presenting cell (APC). The immune synapse contains a large number of signalling molecules that are required for T cell activation, including T cell receptors (TCRs), MHC molecules, co-stimulatory receptors and the purinergic signalling receptors P2X1, P2X4 and P2X7. In response to TCR and CD28 stimulation, pannexin 1, P2X1 receptors and P2X4 receptors translocate to the immune synapse. ATP released through pannexin 1 promotes autocrine signalling via the P2X receptors. Confinement of ATP in the immune synapse results in a powerful autocrine feedback mechanism that facilitates the signal amplification required for antigen recognition. P2 receptors expressed and ATP released by APCs may also have important roles in regulating the antigen recognition process. NFAT nuclear factor of activated T cells. (Reproduced from [474], with permission from Springer)

ATP induced the lysis of CD4+ thymocytes and peripheral CD4+ T cells [483] and the apoptosis of murine thymocytes [484, 485]. T lymphocyte subsets express different levels of P2X7 and high levels are associated with ATP-induced cell death [486]. P2X7 receptor-mediated cell death was also shown to differ between different stages of murine T cell maturation [487]. Interestingly mouse Treg cells express a higher level of P2X7 and their activation by ATP leads to their depletion [488, 489]. P2X7 −/− mice have increased levels of Treg cells [490]. The P2X7 receptor was also involved in T cell death induced by NAD+ through the ADP-ribosylating ectoenzyme, ART2. Indeed ART2-catalyzed ADP-ribosylation activates P2X7 receptors [491493]. In particular Treg cells express ART2 and can be depleted by intravenous injection of NAD+ [494]. However, ATP (1 mM) enhanced the proliferation and immunosuppressive ability of human Treg cells, whereas it induced apoptosis of CD4+ T cells [495]. The dual action of the P2X7 receptor on growth versus death clearly depends on the concentration of ATP, with stimulatory effects at 250 nM and inhibition at 1 mM [495]. This could be related to the existence of two states of activation of the P2X7 receptor: cation-selective channel or large conductance non-selective pore [496].

Numerous studies have shown the importance of the lymphocyte P2X7 receptor in various models of inflammatory diseases. In some of these models, inhibition or deficiency of P2X7 was associated with decreased immune reactions. Mycobacterium tuberculosis infected P2X7 −/− mice had an increased microbial burden in the lung and pulmonary infiltrates contained a higher number of Treg cells [497]. oxATP was shown to inhibit T cell-mediated autoimmunity in models of autoimmune type 1 diabetes and encephalitis in mice [498]. CD38 knockout NOD mice develop accelerated type 1 diabetes. This was corrected by coablation of P2X7 [499]. oxATP delayed islet allograft rejection [500] and increased cardiac transplant survival in mice [501]; these effects were associated with decreased T cell activation. However in other models P2X7 deficiency was associated with increased immune reactions, illustrating the dual role of P2X7 receptors emphasized previously. Following oral infection with Listeria monocytogenes, P2X7-deficient mice showed enhanced CD8 responses in the intestinal mucosa, which can be explained by the proapoptotic effect of P2X7 on intestinal CD8 cells [502]. Graft versus host disease was enhanced in P2X7 −/− mice, and this is associated with T cell expansion and reduced Treg cells [503]. EAE was also exacerbated in P2X7 −/− mice as a result of decreased apoptosis of T lymphocytes [504] and increased T cell cytokine production [505].

P2X receptors other than P2X7 have been shown to play a role in T cell control. RT-PCR studies had shown that P2X1, P2X2 and P2X6 were expressed by murine thymocytes in addition to P2X7 [506]. ATP released through pannexin hemichannels following TCR stimulation amplified T cell activation not only through P2X7 receptors [472] but also via P2X1 and P2X4 receptors, as demonstrated by the use of siRNA [474, 507]. Hypertonic saline is known to increase T cell function [508]: it acts through the release of ATP and the activation of P2X1, P2X4 and P2X7 receptors, as shown also by gene silencing [509]. Both P2X7 and P2X4 are also involved in the activation of unconventional βγ T cells [510, 511].

Although P2X receptors and particularly P2X7 play a major role in lymphocytes, there is some evidence for the role of P2Y receptors as well. Upregulation of P2Y2 receptor mRNA expression was described as an immediate early gene response in activated thymocytes [512] and P2Y2 receptors were shown to be involved in ATP-induced T cell migration [495]. The P2Y6 receptor was shown to be expressed in activated T cells infiltrating in inflammatory bowel disease [513]. Antagonists of the P2Y6 receptor blocked murine T cell activation [514], but these results must be interpreted with caution since T cells of P2Y6-deficient mice exhibited an increased activity in a model of allergic pulmonary inflammation, suggesting that the P2Y6 receptor plays an inhibitory rather than a stimulatory role [515]. P2Y14 receptors were shown to be functionally expressed by mouse spleen-derived T lymphocytes [516]. Adenine nucleotides inhibited CD4+ T cell activation via an increase in cAMP induced by an unidentified P2Y receptor [517].

Natural killer (NK and NKT) cells

P1 receptors

NK cell activity was shown to be inhibited by adenosine and A2 receptor agonists that increase cAMP [518]. Later studies demonstrated the involvement of A2A receptors [519]. Adenosine via the A2A receptor inhibited IFN-γ production by NKT cells, a subset of T cells with natural killer activity [520], but increased their production of IL-4 and IL-10 [521]. Mice were protected against liver reperfusion injury and concanavalin A (ConA)-induced hepatitis by adenosine acting on the A2A receptor on NKT cells, and this protection was abolished in A2A −/− mice [520, 522]. Sickle cell disease results in disseminated microvascular ischemia and reperfusion injury that leads to the activation of NKT cells and the upregulation of A2A receptors [523525]. Activation of A2A receptors in NY1DD mice with sickle cell disease reduced pulmonary inflammation and injury [523]. In a phase I study, the A2A agonist regadenoson was administered to patients with sickle cell disease and was shown to inhibit the activation of NKT cells [524]. A2A −/− mice were protected against tumor metastasis, and this protection was associated with increased NK cell maturation and cytotoxic function [526]. On the other hand, an A3 receptor agonist was shown to potentiate NK cell cytotoxic activity [527] and IFN-γ production [528].

P2 receptors

Inhibition of human and mouse NK cell reactivity via nucleotide receptors was reported [529532]. It was later shown that ATP inhibits cell killing by NK cells via the P2Y11 receptor and an increase in cAMP [533]. On the other hand, NKT cells express the P2X7 receptor, the activation of which can lead to either apoptosis or cell activation [534536]. In vitro NAD induced rapid apoptosis of NKT cells that was mediated by the P2X7 receptor, but its injection in Con A-treated mice enhanced cytokine production by NK cells and liver injury, that was decreased in P2X7 knockout mice [534]. In CD39-deficient mice, apoptosis of NKT cells was increased leading to protection against ConA-induced liver injury [535] or hyperoxic lung injury [536].

Section summary

The release of ATP through pannexin hemichannels or vesicular exocytosis amplifies in an autocrine way the TCR-mediated activation of T lymphocytes. This amplification is mediated by the P2X7 receptor, and also by P2X1 and P2X4 receptors. But activation of P2X7 can also induce T cell death. The resulting effect (activation or death) depends on the particular subset of T cells and on the concentration of ATP.

Adenosine exerts inhibitory effects on T lymphocytes, which are mediated by the A2A receptor. Treg cells over-express the ectonucleotidases CD39 and CD73 that sequentially convert ATP into AMP and adenosine, and their immunosuppressive action is partially mediated by adenosine.

Concluding remarks

Extracellular nucleotides and adenosine exert a variety of effects on distinct subsets of immune cells via a wide spectrum of receptor subtypes (Table 2). These actions can be both stimulatory and inhibitory, and the balance between the two critically depends on the amount and time course of nucleotide release. This is consistent with the role of ATP and its degradation product adenosine as danger signals that stimulate the immune response following injury but moderate this response when it becomes excessive and deleterious.

Table 2.

Expression profiles and functional responses of the purinergic receptor subtypes in different immune cells

Inflammatory cell type Functional response to purines P2 receptor subtype (expression profile and/or involvement in functional response)
Neutrophils Undefined roles P2Y1, P2Y4, P2Y11, P2Y14 and P2X7
Calcium mobilization P2Y2
Actin polymerization P2Y2
Primary granule release P2Y2
Chemotaxis P2Y2, P2Y6 and P2X1
Reduced cAMP accumulation P2Y14
Delay in constitutive neutrophil apoptosis P2Y11
Macrophages Undefined roles P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, P2Y14 and P2X1–P2X6
Intracellular calcium increase P2Y1, P2Y2, P2Y4, P2Y11, P2X4 and P2X7
IL-1β/IL-18 maturation/release via the NLRP3 inflammasome, caspase-1 and cytosolic K+ depletion P2X7
Release of cathepins, PGE2, MMP-9 phosphatidilserine (caspase independent) P2X7
Promoting chemotaxis/phagocytosis P2Y2, P2Y12, P2X1 and P2X3
Regulation of autophagy P2X4 and P2X7
Multinucleated giant cells formation P2X7
Dendritic cells Undefined roles P2X1, P2X4, P2X5, P2X7, P2Y1, P2Y4, P2Y6 and P2Y11
Regulation in cytokine release P2Y11
DC maturation P2Y11, P2Y12 and P2Y14
Apoptosis P2X7
DC migration P2Y2 and P2Y11
Lymphocytes
 B and T cells Undefined roles P2X2, P2X3, P2X5, P2X6 and all P2Y
T cell activation (p38 MAPK activation and IL-2 gene transcription ) P2X1, P2X4 and P2X7
T cell activation (CD62L shedding) P2X7
cAMP accumulation P2Y14
Inhibition of immunosuppressive potential of Tregs P2X7
 Natural killer cells Regulation of NK cytoxicity and chemotaxis P2Y11
 Eosinophils Undefined roles P2Y1, P2Y4, P2Y6, P2Y11, P2Y14, P2X1, P2X4 and P2X7
Chemotaxis P2Y2
Release of chemokines and cytokines P2Y2, P2X1, P2X7 and P2Y6
 Mast cells Undefined roles P2X1, P2X4, P2X6, P2X7, P2Y1, P2Y2, P2Y11, P2Y12 and P2Y13
Degranulation P2Y13 and P2Y14

Reproduced from [550], with permission from Springer

Neutrophils and eosinophils

ATP released from neutrophils amplifies their attraction by chemotactic signals via the P2Y2 receptor and after its degradation to adenosine via the A3 receptor, one example of cooperation between P1 and P2 receptors. The P2Y2 receptor is also involved in the recruitment of eosinophils in the lung during allergic inflammation. On the other hand ATP and adenosine have opposite effects on O2 production and other functions of neutrophils: potentiation by ATP and inhibition by adenosine.

Monocytes/macrophages and microglia

ATP released from apoptotic cells constitutes a find-me signal that attracts monocytes and macrophages, an action mediated by the P2Y2 receptor. Via the P2X7 receptor, ATP stimulates NLRP3 inflammasome activation and IL-1β secretion by macrophages, their killing of bacteria and their apoptosis.

ADP acting on the P2Y12 receptor induces the polarisation and migration of microglia. UDP acting on the P2Y6 receptor stimulates their phagocytic activity. ADP and UDP have, thus, a complementary action of find-me and eat-me signals, respectively, involving a cooperation between two distinct P2Y receptor subtypes. The P2X4, A1 and A3 receptors have also been shown to play a role in microglia migration, whereas the A2A receptor is inhibitory.

Dendritic cells

ATP can exert an immunostimulatory effect on DCs via P2X7 receptor activation. But it can also activate the P2Y11 receptor leading to a semi-maturation state characterized by the upregulation of co-stimulatory molecules and the inhibition of IL-12 production, which impairs the Th1 response and favours tolerance or a Th2 response. The balance between these opposite effects depends on the amount of ATP released and the time course of this release.

Other specific functions of DCs can be activated by nucleotides via distinct P2Y receptor subtypes: chemotaxis by the P2Y2 receptor and antigen endocytosis by the P2Y12 receptor. Adenosine acting on the A2A (human) or A2B (mouse) receptors exerts complex effects on DCs: as ATP it impairs Th1 polarisation and favours Th2 and/or tolerance, but it also favours Th17 cell development.

Lymphocytes

The release of ATP through pannexin hemichannels or vesicular exocytosis amplifies in an autocrine way the TCR-mediated activation of T lymphocytes. This amplification is mediated by the P2X1, P2X4 and P2X7 receptors. On the other hand, Treg cells over-express the ectonucleotidases CD39 and CD73 that sequentially convert ATP into AMP and adenosine, which binds to A2A receptors on effector T cells and suppresses their function.

Neuroimmunology

Contrary to earlier beliefs, the evidence that immune cells are innervated, albeit by nerve varicosities that form occasional close appositions, is convincing. This will be important for future studies of neuroimmunology that might reveal additional roles of ATP and purinergic signalling.

Acknowledgments

We are greatly indebted to the superb supporting work in the preparation of this manuscript by Dr. Gill Knight.

Contributor Information

Geoffrey Burnstock, Email: g.burnstock@ucl.ac.uk.

Jean-Marie Boeynaems, Email: jmboeyna@ulb.ac.be.

References

  • 1.Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, Muskens F, Hoogsteden HC, Luttmann W, Ferrari D, Di Virgilio F, Virchow JC, Jr, Lambrecht BN. Extracellular ATP triggers and maintains asthmatic airway inflammation by activating dendritic cells. Nat Med. 2007;13:913–919. doi: 10.1038/nm1617. [DOI] [PubMed] [Google Scholar]
  • 2.Di Virgilio F, Boeynaems JM, Robson SC. Extracellular nucleotides as negative modulators of immunity. Curr Opin Pharmacol. 2009;9:507–513. doi: 10.1016/j.coph.2009.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, Park D, Woodson RI, Ostankovich M, Sharma P, Lysiak JJ, Harden TK, Leitinger N, Ravichandran KS. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461:282–286. doi: 10.1038/nature08296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Homolya L, Steinberg TH, Boucher RC. Cell to cell communication in response to mechanical stress via bilateral release of ATP and UTP in polarized epithelia. J Cell Biol. 2000;150:1349–1360. doi: 10.1083/jcb.150.6.1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.McNamara N, Khong A, McKemy D, Caterina M, Boyer J, Julius D, Basbaum C. ATP transduces signals from ASGM1, a glycolipid that functions as a bacterial receptor. Proc Natl Acad Sci U S A. 2001;98:9086–9091. doi: 10.1073/pnas.161290898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Seror C, Melki MT, Subra F, Raza SQ, Bras M, Saidi H, Nardacci R, Voisin L, Paoletti A, Law F, Martins I, Amendola A, Abdul-Sater AA, Ciccosanti F, Delelis O, Niedergang F, Thierry S, Said-Sadier N, Lamaze C, Metivier D, Estaquier J, Fimia GM, Falasca L, Casetti R, Modjtahedi N, Kanellopoulos J, Mouscadet JF, Ojcius DM, Piacentini M, Gougeon ML, Kroemer G, Perfettini JL. Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. J Exp Med. 2011;208:1823–1834. doi: 10.1084/jem.20101805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lazarowski ER, Shea DA, Boucher RC, Harden TK. Release of cellular UDP-glucose as a potential extracellular signaling molecule. Mol Pharmacol. 2003;63:1190–1197. doi: 10.1124/mol.63.5.1190. [DOI] [PubMed] [Google Scholar]
  • 8.Zhong X, Malhotra R, Guidotti G. ATP uptake in the Golgi and extracellular release require Mcd4 protein and the vacuolar H+-ATPase. J Biol Chem. 2003;278:33436–33444. doi: 10.1074/jbc.M305785200. [DOI] [PubMed] [Google Scholar]
  • 9.Chekeni FB, Elliott MR, Sandilos JK, Walk SF, Kinchen JM, Lazarowski ER, Armstrong AJ, Penuela S, Laird DW, Salvesen GS, Isakson BE, Bayliss DA, Ravichandran KS. Pannexin 1 channels mediate 'find-me' signal release and membrane permeability during apoptosis. Nature. 2010;467:863–867. doi: 10.1038/nature09413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Forsyth AM, Wan J, Owrutsky PD, Abkarian M, Stone HA. Multiscale approach to link red blood cell dynamics, shear viscosity, and ATP release. Proc Natl Acad Sci U S A. 2011;108:10986–10991. doi: 10.1073/pnas.1101315108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Kim JE, Kang TC. The P2X7 receptor-pannexin-1 complex decreases muscarinic acetylcholine receptor-mediated seizure susceptibility in mice. J Clin Invest. 2011;121:2037–2047. doi: 10.1172/JCI44818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Anselmi F, Hernandez VH, Crispino G, Seydel A, Ortolano S, Roper SD, Kessaris N, Richardson W, Rickheit G, Filippov MA, Monyer H, Mammano F. ATP release through connexin hemichannels and gap junction transfer of second messengers propagate Ca2+ signals across the inner ear. Proc Natl Acad Sci U S A. 2008;105:18770–18775. doi: 10.1073/pnas.0800793105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Elenkov IJ, Wilder RL, Chrousos GP, Vizi ES. The sympathetic nerve - an integrative interface between two supersystems: the brain and the immune system. Pharmacol Rev. 2000;52:595–638. [PubMed] [Google Scholar]
  • 14.Serafeim A, Gordon J. The immune system gets nervous. Curr Opin Pharmacol. 2001;1:398–403. doi: 10.1016/s1471-4892(01)00069-8. [DOI] [PubMed] [Google Scholar]
  • 15.Bienenstock J, Goetzl EJ, Blennerhassett MG. Autonomic Neuroimmunology. London: Taylor & Francis; 2003. pp. 139–170. [Google Scholar]
  • 16.Deaglio S, Robson SC. Ectonucleotidases as regulators of purinergic signaling in thrombosis, inflammation, and immunity. Adv Pharmacol. 2011;61:301–332. doi: 10.1016/B978-0-12-385526-8.00010-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Burnstock G. A basis for distinguishing two types of purinergic receptor. In: Straub RW, Bolis L, editors. Cell membrane receptors for drugs and hormones: a multidisciplinary approach. New York: Raven Press; 1978. pp. 107–118. [Google Scholar]
  • 18.Burnstock G, Kennedy C. Is there a basis for distinguishing two types of P2-purinoceptor? Gen Pharmacol. 1985;16:433–440. doi: 10.1016/0306-3623(85)90001-1. [DOI] [PubMed] [Google Scholar]
  • 19.DePierre JW, Karnovsky ML. Ecto-enzymes of the guinea pig polymorphonuclear leukocyte. I. Evidence for an ecto-adenosine monophosphatase, adenosine triphosphatase, and -p-nitrophenyl phosphates. J Biol Chem. 1974;249:7111–7120. [PubMed] [Google Scholar]
  • 20.Newby AC. Role of adenosine deaminase, ecto-(5'-nucleotidase) and ecto-(non-specific phosphatase) in cyanide-induced adenosine monophosphate catabolism in rat polymorphonuclear leucocytes. Biochem J. 1980;186:907–918. doi: 10.1042/bj1860907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Smith GP, Peters TJ. Subcellular localization and properties of pyridoxal phosphate phosphatases of human polymorphonuclear leukocytes and their relationship to acid and alkaline phosphatase. Biochim Biophys Acta. 1981;661:287–294. doi: 10.1016/0005-2744(81)90017-6. [DOI] [PubMed] [Google Scholar]
  • 22.Eltzschig HK, Weissmuller T, Mager A, Eckle T. Nucleotide metabolism and cell-cell interactions. Methods Mol Biol. 2006;341:73–87. doi: 10.1385/1-59745-113-4:73. [DOI] [PubMed] [Google Scholar]
  • 23.Chen Y, Shukla A, Namiki S, Insel PA, Junger WG. A putative osmoreceptor system that controls neutrophil function through the release of ATP, its conversion to adenosine, and activation of A2 adenosine and P2 receptors. J Leukoc Biol. 2004;76:245–253. doi: 10.1189/jlb.0204066. [DOI] [PubMed] [Google Scholar]
  • 24.Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A, Nizet V, Insel PA, Junger WG. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314:1792–1795. doi: 10.1126/science.1132559. [DOI] [PubMed] [Google Scholar]
  • 25.Cronstein BN, Kramer SB, Weissmann G, Hirschhorn R. Adenosine: a physiological modulator of superoxide anion generation by human neutrophils. J Exp Med. 1983;158:1160–1177. doi: 10.1084/jem.158.4.1160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cronstein BN, Rosenstein ED, Kramer SB, Weissmann G, Hirschhorn R. Adenosine; a physiologic modulator of superoxide anion generation by human neutrophils. Adenosine acts via an A2 receptor on human neutrophils. J Immunol. 1985;135:1366–1371. [PubMed] [Google Scholar]
  • 27.Cronstein BN, Kubersky SM, Weissmann G, Hirschhorn R. Engagement of adenosine receptors inhibits hydrogen peroxide (H2O2−) release by activated human neutrophils. Clin Immunol Immunopathol. 1987;42:76–85. doi: 10.1016/0090-1229(87)90174-7. [DOI] [PubMed] [Google Scholar]
  • 28.Schrier DJ, Imre KM. The effects of adenosine agonists on human neutrophil function. J Immunol. 1986;137:3284–3289. [PubMed] [Google Scholar]
  • 29.Sun WC, Moore JN, Hurley DJ, Vandenplas ML, Murray TF. Effects of stimulation of adenosine A2A receptors on lipopolysaccharide-induced production of reactive oxygen species by equine neutrophils. Am J Vet Res. 2007;68:649–656. doi: 10.2460/ajvr.68.6.649. [DOI] [PubMed] [Google Scholar]
  • 30.Colli S, Tremoli E. Multiple effects of dipyridamole on neutrophils and mononuclear leukocytes: adenosine-dependent and adenosine-independent mechanisms. J Lab Clin Med. 1991;118:136–145. [PubMed] [Google Scholar]
  • 31.Mircevová L, Viktora L, Hermanová E. Inhibition of phagocytosis of polymorphonuclear leucocytes by adenosine and HoCl3 in vitro. Med Biol. 1984;62:326–330. [PubMed] [Google Scholar]
  • 32.Hardart GE, Sullivan GW, Carper HT, Mandell GL. Adenosine and 2-phenylaminoadenosine (CV-1808) inhibit human neutrophil bactericidal function. Infect Immun. 1991;59:885–889. doi: 10.1128/iai.59.3.885-889.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Laghi Pasini F, Capecchi PL, Ceccatelli L, Orrico A, Pasqui AL, Di Perri T. Effect of extracellular Ca++ on in vitro adenosine-dependent inhibition of neutrophil function. Int J Immunotherapy. 1986;11:225–232. [Google Scholar]
  • 34.Laghi Pasini F, Capecchi PL, Pasqui AL, Ceccatelli L, Mazza S, Gistri A, Di Perri T. Adenosine system and cell calcium translocation: interference of calcium channel blockers. Exp Gerontol. 1990;25:383–391. doi: 10.1016/0531-5565(90)90076-e. [DOI] [PubMed] [Google Scholar]
  • 35.Tsuruta S, Ito S, Mikawa H. Adenosine inhibits divalent cation influx across human neutrophil plasma membrane via surface adenosine A2 receptors. Cell Signal. 1992;4:543–551. doi: 10.1016/0898-6568(92)90023-2. [DOI] [PubMed] [Google Scholar]
  • 36.Nielson CP, Vestal RE. Effects of adenosine on polymorphonuclear leucocyte function, cyclic 3': 5'-adenosine monophosphate, and intracellular calcium. Br J Pharmacol. 1989;97:882–888. doi: 10.1111/j.1476-5381.1989.tb12028.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhang Y, Palmblad J, Fredholm BB. Biphasic effect of ATP on neutrophil functions mediated by P2U and adenosine A2A receptors. Biochem Pharmacol. 1996;51:957–965. doi: 10.1016/0006-2952(95)02403-4. [DOI] [PubMed] [Google Scholar]
  • 38.Thibault N, Burelout C, Harbour D, Borgeat P, Naccache PH, Bourgoin SG. Occupancy of adenosine A2a receptors promotes fMLP-induced cyclic AMP accumulation in human neutrophils: impact on phospholipase D activity and recruitment of small GTPases to membranes. J Leukoc Biol. 2002;71:367–377. [PubMed] [Google Scholar]
  • 39.Varani K, Portaluppi F, Gessi S, Merighi S, Vincenzi F, Cattabriga E, Dalpiaz A, Bortolotti F, Belardinelli L, Borea PA. Caffeine intake induces an alteration in human neutrophil A2A adenosine receptors. Cell Mol Life Sci. 2005;62:2350–2358. doi: 10.1007/s00018-005-5312-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.McColl SR, St-Onge M, Dussault AA, Laflamme C, Bouchard L, Boulanger J, Pouliot M. Immunomodulatory impact of the A2A adenosine receptor on the profile of chemokines produced by neutrophils. FASEB J. 2006;20:187–189. doi: 10.1096/fj.05-4804fje. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Grenz A, Kim JH, Bauerle JD, Tak E, Eltzschig HK, Clambey ET. Adora2b adenosine receptor signaling protects during acute kidney injury via inhibition of neutrophil-dependent TNF-α release. J Immunol. 2012;189:4566–4573. doi: 10.4049/jimmunol.1201651. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 42.van der Hoeven D, Wan TC, Gizewski ET, Kreckler LM, Maas JE, Van Orman J, Ravid K, Auchampach JA. A role for the low-affinity A2B adenosine receptor in regulating superoxide generation by murine neutrophils. J Pharmacol Exp Ther. 2011;338:1004–1012. doi: 10.1124/jpet.111.181792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.van der Hoeven D, Wan TC, Auchampach JA. Activation of the A3 adenosine receptor suppresses superoxide production and chemotaxis of mouse bone marrow neutrophils. Mol Pharmacol. 2008;74:685–696. doi: 10.1124/mol.108.048066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bouma MG, Jeunhomme TM, Boyle DL, Dentener MA, Voitenok NN, van den Wildenberg FA, Buurman WA. Adenosine inhibits neutrophil degranulation in activated human whole blood: involvement of adenosine A2 and A3 receptors. J Immunol. 1997;158:5400–5408. [PubMed] [Google Scholar]
  • 45.Krump E, Picard S, Mancini J, Borgeat P. Suppression of leukotriene B4 biosynthesis by endogenous adenosine in ligand-activated human neutrophils. J Exp Med. 1997;186:1401–1406. doi: 10.1084/jem.186.8.1401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cadieux JS, Leclerc P, St-Onge M, Dussault AA, Laflamme C, Picard S, Ledent C, Borgeat P, Pouliot M. Potentiation of neutrophil cyclooxygenase-2 by adenosine: an early anti-inflammatory signal. J Cell Sci. 2005;118:1437–1447. doi: 10.1242/jcs.01737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Garcia-Castro I, Mato JM, Vasanthakumar G, Wiesmann WP, Schiffmann E, Chiang PK. Paradoxical effects of adenosine on neutrophil chemotaxis. J Biol Chem. 1983;258:4345–4349. [PubMed] [Google Scholar]
  • 48.Rose FR, Hirschhorn R, Weissmann G, Cronstein BN. Adenosine promotes neutrophil chemotaxis. J Exp Med. 1988;167:1186–1194. doi: 10.1084/jem.167.3.1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Salmon JE, Cronstein BN. Fcγ Receptor-mediated functions in neutrophils are modulated by adenosine receptor occupancy. A1 receptors are stimulatory and A2 receptors are inhibitory. J Immunol. 1990;145:2235–2240. [PubMed] [Google Scholar]
  • 50.Aeffner F, Woods PS, Davis IC. Activation of A 1-adenosine receptors promotes leukocyte recruitment to the lung and attenuates acute lung injury in mice infected with influenza A/WSN/33 (H1N1) virus. J Virol. 2014;88:10214–10227. doi: 10.1128/JVI.01068-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Reutershan J, Cagnina RE, Chang D, Linden J, Ley K. Therapeutic anti-inflammatory effects of myeloid cell adenosine receptor A2a stimulation in lipopolysaccharide-induced lung injury. J Immunol. 2007;179:1254–1263. doi: 10.4049/jimmunol.179.2.1254. [DOI] [PubMed] [Google Scholar]
  • 52.Konrad FM, Witte E, Vollmer I, Stark S, Reutershan J. Adenosine receptor A2b on hematopoietic cells mediates LPS-induced migration of PMNs into the lung interstitium. Am J Physiol Lung Cell Mol Physiol. 2012;303:L425–L438. doi: 10.1152/ajplung.00387.2011. [DOI] [PubMed] [Google Scholar]
  • 53.Inoue Y, Chen Y, Hirsh MI, Yip L, Junger WG. A3 and P2Y2 receptors control the recruitment of neutrophils to the lungs in a mouse model of sepsis. Shock. 2008;30:173–177. doi: 10.1097/shk.0b013e318160dad4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Butler M, Sanmugalingam D, Burton VJ, Wilson T, Pearson R, Watson RP, Smith P, Parkinson SJ. Impairment of adenosine A3 receptor activity disrupts neutrophil migratory capacity and impacts innate immune function in vivo. Eur J Immunol. 2012;42:3358–3368. doi: 10.1002/eji.201242655. [DOI] [PubMed] [Google Scholar]
  • 55.Bao Y, Chen Y, Ledderose C, Li L, Junger WG. Pannexin 1 channels link chemoattractant receptor signalling to local excitation and global inhibition responses at the front and back of polarized neutrophils. J Biol Chem. 2013;288:22650–22657. doi: 10.1074/jbc.M113.476283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Corriden R, Self T, Akong-Moore K, Nizet V, Kellam B, Briddon SJ, Hill SJ. Adenosine-A3 receptors in neutrophil microdomains promote the formation of bacteria-tethering cytonemes. EMBO Rep. 2013;14:726–732. doi: 10.1038/embor.2013.89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cronstein BN, Levin RI, Philips M, Hirschhorn R, Abramson SB, Weissmann G. Neutrophil adherence to endothelium is enhanced via adenosine A1 receptors and inhibited via adenosine A2 receptors. J Immunol. 1992;148:2201–2206. [PubMed] [Google Scholar]
  • 58.Asako H, Wolf RE, Granger DN. Leukocyte adherence in rat mesenteric venules: effects of adenosine and methotrexate. Gastroenterology. 1993;104:31–37. doi: 10.1016/0016-5085(93)90832-w. [DOI] [PubMed] [Google Scholar]
  • 59.Eltzschig HK, Thompson LF, Karhausen J, Cotta RJ, Ibla JC, Robson SC, Colgan SP. Endogenous adenosine produced during hypoxia attenuates neutrophil accumulation: coordination by extracellular nucleotide metabolism. Blood. 2004;104:3986–3992. doi: 10.1182/blood-2004-06-2066. [DOI] [PubMed] [Google Scholar]
  • 60.Sullivan GW, Lee DD, Ross WG, DiVietro JA, Lappas CM, Lawrence MB, Linden J. Activation of A2A adenosine receptors inhibits expression of α4/β1 integrin (very late antigen-4) on stimulated human neutrophils. J Leukoc Biol. 2004;75:127–134. doi: 10.1189/jlb.0603300. [DOI] [PubMed] [Google Scholar]
  • 61.Thiel M, Chambers JD, Chouker A, Fischer S, Zourelidis C, Bardenheuer HJ, Arfors KE, Peter K. Effect of adenosine on the expression of β2 integrins and L-selectin of human polymorphonuclear leukocytes in vitro. J Leukoc Biol. 1996;59:671–682. doi: 10.1002/jlb.59.5.671. [DOI] [PubMed] [Google Scholar]
  • 62.Zhao ZQ, Sato H, Williams MW, Fernandez AZ, Vinten-Johansen J. Adenosine A2-receptor activation inhibits neutrophil-mediated injury to coronary endothelium. Am J Physiol. 1996;271:H1456–H1464. doi: 10.1152/ajpheart.1996.271.4.H1456. [DOI] [PubMed] [Google Scholar]
  • 63.Eltzschig HK, MacManus CF, Colgan SP. Neutrophils as sources of extracellular nucleotides: functional consequences at the vascular interface. Trends Cardiovasc Med. 2008;18:103–107. doi: 10.1016/j.tcm.2008.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Corriden R, Chen Y, Inoue Y, Beldi G, Robson SC, Insel PA, Junger WG. Ecto-nucleoside triphosphate diphosphohydrolase 1 (E-NTPDase1/CD39) regulates neutrophil chemotaxis by hydrolyzing released ATP to adenosine. J Biol Chem. 2008;283:28480–28486. doi: 10.1074/jbc.M800039200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kilian JG, Nakhla S, Sieveking DP, Celermajer DS. Adenosine prevents neutrophil adhesion to human endothelial cells after hypoxia/reoxygenation. Int J Cardiol. 2005;105:322–326. doi: 10.1016/j.ijcard.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 66.Watanabe T, Tokuyama S, Yasuda M, Sasaki T, Yamamoto T. Involvement of adenosine A2 receptors in the changes of tissue factor-dependent coagulant activity induced by polymorphonuclear leukocytes in endothelial cells. Jpn J Pharmacol. 2002;88:407–413. doi: 10.1254/jjp.88.407. [DOI] [PubMed] [Google Scholar]
  • 67.Pospísil M, Hofer M, Znojil V, Vácha J, Netiková J, Holá J. Synergistic effect of granulocyte colony-stimulating factor and drugs elevating extracellular adenosine on neutrophil production in mice. Blood. 1995;86:3692–3697. [PubMed] [Google Scholar]
  • 68.Yasui K, Agematsu K, Shinozaki K, Hokibara S, Nagumo H, Nakazawa T, Komiyama A. Theophylline induces neutrophil apoptosis through adenosine A2A receptor antagonism. J Leukoc Biol. 2000;67:529–535. doi: 10.1002/jlb.67.4.529. [DOI] [PubMed] [Google Scholar]
  • 69.Fortin A, Harbour D, Fernandes M, Borgeat P, Bourgoin S. Differential expression of adenosine receptors in human neutrophils: up-regulation by specific Th1 cytokines and lipopolysaccharide. J Leukoc Biol. 2006;79:574–585. doi: 10.1189/jlb.0505249. [DOI] [PubMed] [Google Scholar]
  • 70.Bazzichi L, Trincavelli L, Rossi A, De Feo F, Lucacchini A, Bombardieri S, Martini C. A2B adenosine receptor activity is reduced in neutrophils from patients with systemic sclerosis. Arthritis Res Ther. 2005;7:R189–R195. doi: 10.1186/ar1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Kaufmann I, Hoelzl A, Schliephake F, Hummel T, Chouker A, Lysenko L, Peter K, Thiel M. Effects of adenosine on functions of polymorphonuclear leukocytes from patients with septic shock. Shock. 2007;27:25–31. doi: 10.1097/01.shk.0000238066.00074.90. [DOI] [PubMed] [Google Scholar]
  • 72.Inoue Y, Chen Y, Pauzenberger R, Hirsh MI, Junger WG. Hypertonic saline up-regulates A3 adenosine receptor expression of activated neutrophils and increases acute lung injury after sepsis. Crit Care Med. 2008;36:2569–2575. doi: 10.1097/CCM.0b013e3181841a91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Varani K, Gessi S, Merighi S, Iannotta V, Cattabriga E, Spisani S, Cadossi R, Borea PA. Effect of low frequency electromagnetic fields on A2A adenosine receptors in human neutrophils. Br J Pharmacol. 2002;136:57–66. doi: 10.1038/sj.bjp.0704695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Varani K, Gessi S, Merighi S, Iannotta V, Cattabriga E, Pancaldi C, Cadossi R, Borea PA. Alteration of A3 adenosine receptors in human neutrophils and low frequency electromagnetic fields. Biochem Pharmacol. 2003;66:1897–1906. doi: 10.1016/s0006-2952(03)00454-4. [DOI] [PubMed] [Google Scholar]
  • 75.Kuroki M, Takeshige K, Minakami S. ATP-induced calcium mobilization in human neutrophils. Biochim Biophys Acta. 1989;1012:103–106. doi: 10.1016/0167-4889(89)90017-7. [DOI] [PubMed] [Google Scholar]
  • 76.Saito H, Ebisawa M, Reason DC, Ohno K, Kurihara K, Sakaguchi N, Ohgimi A, Saito E, Akasawa A, Akimoto K. Extracellular ATP stimulates interleukin-dependent cultured mast cells and eosinophils through calcium mobilization. Int Arch Allergy Appl Immunol. 1991;94:68–70. doi: 10.1159/000235327. [DOI] [PubMed] [Google Scholar]
  • 77.Kuhns DB, Wright DG, Nath J, Kaplan SS, Basford RE. ATP induces transient elevations of [Ca2+] i in human neutrophils and primes these cells for enhanced O2− generation. Lab Invest. 1988;58:448–453. [PubMed] [Google Scholar]
  • 78.Seifert R, Wenzel K, Eckstein F, Schultz G. Purine and pyrimidine nucleotides potentiate activation of NADPH oxidase and degranulation by chemotactic peptides and induce aggregation of human neutrophils via G proteins. Eur J Biochem. 1989;181:277–285. doi: 10.1111/j.1432-1033.1989.tb14722.x. [DOI] [PubMed] [Google Scholar]
  • 79.Tuluc F, Bredetean O, Brailoiu E, Meshki J, Garcia A, Dun NJ, Kunapuli SP. The priming effect of extracellular UTP on human neutrophils: Role of calcium released from thapsigargin-sensitive intracellular stores. Purinergic Signal. 2005;1:359–368. doi: 10.1007/s11302-005-0039-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ward PA, Cunningham TW, McCulloch KK, Phan SH, Powell J, Johnson KJ. Platelet enhancement of O2− responses in stimulated human neutrophils. Identification of platelet factor as adenine nucleotide. Lab Invest. 1988;58:37–47. [PubMed] [Google Scholar]
  • 81.Ward PA, Cunningham TW, McCulloch KK, Johnson KJ. Regulatory effects of adenosine and adenine nucleotides on oxygen radical responses of neutrophils. Lab Invest. 1988;58:438–447. [PubMed] [Google Scholar]
  • 82.Axtell RA, Sandborg RR, Smolen JE, Ward PA, Boxer LA. Exposure of human neutrophils to exogenous nucleotides causes elevation in intracellular calcium, transmembrane calcium fluxes, and an alteration of a cytosolic factor resulting in enhanced superoxide production in response to FMLP and arachidonic acid. Blood. 1990;75:1324–1332. [PubMed] [Google Scholar]
  • 83.Cockcroft S, Stutchfield J. The receptors for ATP and fMetLeuPhe are independently coupled to phospholipases C and A2 via G-protein(s). Relationship between phospholipase C and A2 activation and exocytosis in HL60 cells and human neutrophils. Biochem J. 1989;263:715–723. doi: 10.1042/bj2630715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Cockcroft S, Stutchfield J. ATP stimulates secretion in human neutrophils and HL60 cells via a pertussis toxin-sensitive guanine nucleotide-binding protein coupled to phospholipase C. FEBS Lett. 1989;245:25–29. doi: 10.1016/0014-5793(89)80184-x. [DOI] [PubMed] [Google Scholar]
  • 85.Ford-Hutchinson AW. Aggregation of rat neutrophils by nucleotide triphosphates. Br J Pharmacol. 1982;76:367–371. doi: 10.1111/j.1476-5381.1982.tb09229.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.O'Grady SM. Purinergic signaling and immune cell chemotaxis. Focus on "the UDP-sugar-sensing P2Y14 receptor promotes Rho-mediated signaling and chemotaxis in human neutrophils". Am J Physiol Cell Physiol. 2012;303:C486–C487. doi: 10.1152/ajpcell.00184.2012. [DOI] [PubMed] [Google Scholar]
  • 87.Cicko S, Lucattelli M, Muller T, Lommatzsch M, De Cunto G, Cardini S, Sundas W, Grimm M, Zeiser R, Durk T, Zissel G, Boeynaems JM, Sorichter S, Ferrari D, Di Virgilio F, Virchow JC, Lungarella G, Idzko M. Purinergic receptor inhibition prevents the development of smoke-induced lung injury and emphysema. J Immunol. 2010;185:688–697. doi: 10.4049/jimmunol.0904042. [DOI] [PubMed] [Google Scholar]
  • 88.Ayata CK, Ganal SC, Hockenjos B, Willim K, Vieira RP, Grimm M, Robaye B, Boeynaems JM, Di Virgilio F, Pellegatti P, Diefenbach A, Idzko M, Hasselblatt P. Purinergic P2Y2 receptors promote neutrophil infiltration and hepatocyte death in mice with acute liver injury. Gastroenterology. 2012;143:1620–1629. doi: 10.1053/j.gastro.2012.08.049. [DOI] [PubMed] [Google Scholar]
  • 89.Scrivens M, Dickenson JM. Functional expression of the P2Y14 receptor in human neutrophils. Eur J Pharmacol. 2006;543:166–173. doi: 10.1016/j.ejphar.2006.05.037. [DOI] [PubMed] [Google Scholar]
  • 90.Sesma JI, Kreda SM, Steinckwich-Besancon N, Dang H, García-Mata R, Harden TK, Lazarowski ER. The UDP-sugar-sensing P2Y14 receptor promotes Rho-mediated signaling and chemotaxis in human neutrophils. Am J Physiol Cell Physiol. 2012;303:C490–C498. doi: 10.1152/ajpcell.00138.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Barrett MO, Sesma JI, Ball CB, Jayasekara PS, Jacobson KA, Lazarowski ER, Harden TK. A selective high-affinity antagonist of the P2Y14 receptor inhibits UDP-glucose-stimulated chemotaxis of human neutrophils. Mol Pharmacol. 2013;84:41–49. doi: 10.1124/mol.113.085654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Vaughan KR, Stokes L, Prince LR, Marriott HM, Meis S, Kassack MU, Bingle CD, Sabroe I, Surprenant A, Whyte MK. Inhibition of neutrophil apoptosis by ATP is mediated by the P2Y11 receptor. J Immunol. 2007;179:8544–8553. doi: 10.4049/jimmunol.179.12.8544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Alkayed F, Kashimata M, Koyama N, Hayashi T, Tamura Y, Azuma Y. P2Y11 purinoceptor mediates the ATP-enhanced chemotactic response of rat neutrophils. J Pharmacol Sci. 2012;120:288–295. doi: 10.1254/jphs.12173fp. [DOI] [PubMed] [Google Scholar]
  • 94.Suh BC, Kim JS, Namgung U, Ha H, Kim KT. P2X7 nucleotide receptor mediation of membrane pore formation and superoxide generation in human promyelocytes and neutrophils. J Immunol. 2001;166:6754–6763. doi: 10.4049/jimmunol.166.11.6754. [DOI] [PubMed] [Google Scholar]
  • 95.Nagaoka I, Tamura H, Hirata M. An antimicrobial cathelicidin peptide, human CAP18/LL-37, suppresses neutrophil apoptosis via the activation of formyl-peptide receptor-like 1 and P2X7. J Immunol. 2006;176:3044–3052. doi: 10.4049/jimmunol.176.5.3044. [DOI] [PubMed] [Google Scholar]
  • 96.Christenson K, Björkman L, Tängemo C, Bylund J. Serum amyloid A inhibits apoptosis of human neutrophils via a P2X7-sensitive pathway independent of formyl peptide receptor-like 1. J Leukoc Biol. 2008;83:139–148. doi: 10.1189/jlb.0507276. [DOI] [PubMed] [Google Scholar]
  • 97.da Silva GL, Sperotto ND, Borges TJ, Bonorino C, Takyia CM, Coutinho-Silva R, Campos MM, Zanin RF, Morrone FB. P2X7 receptor is required for neutrophil accumulation in a mouse model of irritant contact dermatitis. Exp Dermatol. 2013;22:184–188. doi: 10.1111/exd.12094. [DOI] [PubMed] [Google Scholar]
  • 98.Martel-Gallegos G, Rosales-Saavedra MT, Reyes JP, Casas-Pruneda G, Toro-Castillo C, Pérez-Cornejo P, Arreola J. Human neutrophils do not express purinergic P2X7 receptors. Purinergic Signal. 2010;6:297–306. doi: 10.1007/s11302-010-9178-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mohanty JG, Raible DG, McDermott LJ, Pelleg A, Schulman ES. Effects of purine and pyrimidine nucleotides on intracellular Ca 2+ in human eosinophils: activation of purinergic P2Y receptors. J Allergy Clin Immunol. 2001;107:849–855. doi: 10.1067/mai.2001.114658. [DOI] [PubMed] [Google Scholar]
  • 100.Lecut C, Frederix K, Johnson DM, Deroanne C, Thiry M, Faccinetto C, Marée R, Evans RJ, Volders PG, Bours V, Oury C. P2X1 ion channels promote neutrophil chemotaxis through Rho kinase activation. J Immunol. 2009;183:2801–2809. doi: 10.4049/jimmunol.0804007. [DOI] [PubMed] [Google Scholar]
  • 101.Lecut C, Faccinetto C, Delierneux C, van Oerle R, Spronk HM, Evans RJ, El BJ, Bours V, Oury C. ATP-gated P2X1 ion channels protect against endotoxemia by dampening neutrophil activation. J Thromb Haemost. 2012;10:453–465. doi: 10.1111/j.1538-7836.2011.04606.x. [DOI] [PubMed] [Google Scholar]
  • 102.Kohno Y, Ji X, Mawhorter SD, Koshiba M, Jacobson KA. Activation of A3 adenosine receptors on human eosinophils elevates intracellular calcium. Blood. 1996;88:3569–3574. [PMC free article] [PubMed] [Google Scholar]
  • 103.Walker BA. Effects of adenosine on guinea pig pulmonary eosinophils. Inflammation. 1996;20:11–21. doi: 10.1007/BF01487741. [DOI] [PubMed] [Google Scholar]
  • 104.Ezeamuzie CI, Philips E. Adenosine A3 receptors on human eosinophils mediate inhibition of degranulation and superoxide anion release. Br J Pharmacol. 1999;127:188–194. doi: 10.1038/sj.bjp.0702476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Knight D, Zheng X, Rocchini C, Jacobson M, Bai T, Walker B. Adenosine A3 receptor stimulation inhibits migration of human eosinophils. J Leukoc Biol. 1997;62:465–468. doi: 10.1002/jlb.62.4.465. [DOI] [PubMed] [Google Scholar]
  • 106.Walker BA, Jacobson MA, Knight DA, Salvatore CA, Weir T, Zhou D, Bai TR. Adenosine A3 receptor expression and function in eosinophils. Am J Respir Cell Mol Biol. 1997;16:531–537. doi: 10.1165/ajrcmb.16.5.9160835. [DOI] [PubMed] [Google Scholar]
  • 107.Young HW, Molina JG, Dimina D, Zhong H, Jacobson M, Chan LN, Chan TS, Lee JJ, Blackburn MR. A3 adenosine receptor signaling contributes to airway inflammation and mucus production in adenosine deaminase-deficient mice. J Immunol. 2004;173:1380–1389. doi: 10.4049/jimmunol.173.2.1380. [DOI] [PubMed] [Google Scholar]
  • 108.Burgers JA, Schweizer RC, Koenderman L, Bruijnzeel PL, Akkerman JW. Human platelets secrete chemotactic activity for eosinophils. Blood. 1993;81:49–55. [PubMed] [Google Scholar]
  • 109.Dichmann S, Idzko M, Zimpfer U, Hofmann C, Ferrari D, Luttmann W, Virchow C, Jr, Di Virgilio F, Norgauer J. Adenosine triphosphate-induced oxygen radical production and CD11b up-regulation: Ca++ mobilization and actin reorganization in human eosinophils. Blood. 2000;95:973–978. [PubMed] [Google Scholar]
  • 110.Ferrari D, Idzko M, Dichmann S, Purlis D, Virchow C, Norgauer J, Chiozzi P, Di Virgilio F, Luttmann W. P2 purinergic receptors of human eosinophils: characterization and coupling to oxygen radical production. FEBS Lett. 2000;486:217–224. doi: 10.1016/s0014-5793(00)02306-1. [DOI] [PubMed] [Google Scholar]
  • 111.Idzko M, Dichmann S, Panther E, Ferrari D, Herouy Y, Virchow C, Jr, Luttmann W, Di Virgilio F, Norgauer J. Functional characterization of P2Y and P2X receptors in human eosinophils. J Cell Physiol. 2001;188:329–336. doi: 10.1002/jcp.1129. [DOI] [PubMed] [Google Scholar]
  • 112.Idzko M, Panther E, Bremer HC, Sorichter S, Luttmann W, Virchow CJ, Jr, Di Virgilio F, Herouy Y, Norgauer J, Ferrari D. Stimulation of P2 purinergic receptors induces the release of eosinophil cationic protein and interleukin-8 from human eosinophils. Br J Pharmacol. 2003;138:1244–1250. doi: 10.1038/sj.bjp.0705145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kobayashi T, Kouzaki H, Kita H. Human eosinophils recognize endogenous danger signal crystalline uric acid and produce proinflammatory cytokines mediated by autocrine ATP. J Immunol. 2010;184:6350–6358. doi: 10.4049/jimmunol.0902673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Muniz VS, Thompson GA, Barbosa-Pereira C, Patrasso-Salgado B, Neves JS. Expression and functional roles of the purinergic receptor P2Y12 in human eosinophils. Inflamm Res. 2011;60:S145–S146. [Google Scholar]
  • 115.Müller T, Robaye B, Vieira RP, Ferrari D, Grimm M, Jakob T, Martin SF, Di Virgilio F, Boeynaems JM, Virchow JC, Idzko M. The purinergic receptor P2Y2 receptor mediates chemotaxis of dendritic cells and eosinophils in allergic lung inflammation. Allergy. 2010;65:1545–1553. doi: 10.1111/j.1398-9995.2010.02426.x. [DOI] [PubMed] [Google Scholar]
  • 116.Vanderstocken G, Bondue B, Horckmans M, Di PL, Robaye B, Boeynaems JM, Communi D. P2Y2 receptor regulates VCAM-1 membrane and soluble forms and eosinophil accumulation during lung inflammation. J Immunol. 2010;185:3702–3707. doi: 10.4049/jimmunol.0903908. [DOI] [PubMed] [Google Scholar]
  • 117.Marone G, Findlay SR, Lichtenstein LM. Adenosine receptor on human basophils: modulation of histamine release. J Immunol. 1979;123:1473–1477. [PubMed] [Google Scholar]
  • 118.Marone G, Triggiani M, Kagey-Sobotka A, Lichtenstein LM, Condorelli M. Adenosine receptors on human basophils and lung mast cells. Adv Exp Med Biol. 1986;195 Pt B:35–42. doi: 10.1007/978-1-4684-1248-2_6. [DOI] [PubMed] [Google Scholar]
  • 119.Church MK, Holgate ST, Hughes PJ. Adenosine inhibits and potentiates IgE-dependent histamine release from human basophils by an A2-receptor mediated mechanism. Br J Pharmacol. 1983;80:719–726. doi: 10.1111/j.1476-5381.1983.tb10063.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Marone G, Vigorita S, Antonelli C, Torella G, Genovese A, Condorelli M. Evidence for an adenosine A2/Ra receptor on human basophils. Life Sci. 1985;36:339–345. doi: 10.1016/0024-3205(85)90119-5. [DOI] [PubMed] [Google Scholar]
  • 121.Hughes PJ, Church MK. Inhibition of immunological and nonimmunological histamine release from human basophils by adenosine analogues that act at P-sites. Biochem Pharmacol. 1986;35:1809–1816. doi: 10.1016/0006-2952(86)90297-2. [DOI] [PubMed] [Google Scholar]
  • 122.Ludowyke RI, Scurr LL. Calcium-independent secretion by ATP gamma S from a permeabilized rat basophilic leukaemia cell line (RBL-2H3) Cell Signal. 1994;6:223–231. doi: 10.1016/0898-6568(94)90080-9. [DOI] [PubMed] [Google Scholar]
  • 123.Nakano M, Kudo F, Nakamura T, Kasai K, Ito K, Takami H, Ito K. Uracil nucleotides enhance the degranulation of human basophils induced by anti-IgE antibody via a purinergic receptor (P6016) J Immunol. 2013;190:59. [Google Scholar]
  • 124.Marquardt DL, Parker CW, Sullivan TJ. Potentiation of mast cell mediator release by adenosine. J Immunol. 1978;120:871–878. [PubMed] [Google Scholar]
  • 125.Burt DS, Stanworth DR. The effect of ribose and purine modified adenosine analogues on the secretion of histamine from rat mast cells induced by ionophore A23187. Biochem Pharmacol. 1983;32:2729–2732. doi: 10.1016/0006-2952(83)90083-7. [DOI] [PubMed] [Google Scholar]
  • 126.Hughes PJ, Holgate ST, Church MK. Adenosine inhibits and potentiates IgE-dependent histamine release from human lung mast cells by an A2-purinoceptor mediated mechanism. Biochem Pharmacol. 1984;33:3847–3852. doi: 10.1016/0006-2952(84)90050-9. [DOI] [PubMed] [Google Scholar]
  • 127.Church MK, Hughes PJ. Adenosine potentiates immunological histamine release from rat mast cells by a novel cyclic AMP-independent cell-surface action. Br J Pharmacol. 1985;85:3–5. doi: 10.1111/j.1476-5381.1985.tb08823.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Church MK, Hughes PJ, Vardey CJ. Studies on the receptor mediating cyclic AMP-independent enhancement by adenosine of IgE-dependent mediator release from rat mast cells. Br J Pharmacol. 1986;87:233–242. doi: 10.1111/j.1476-5381.1986.tb10176.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Leoutsakos A, Pearce FL. The effect of adenosine and its analogues on cyclic AMP changes and histamine secretion from rat peritoneal mast cells stimulated by various ligands. Biochem Pharmacol. 1986;35:1373–1379. doi: 10.1016/0006-2952(86)90284-4. [DOI] [PubMed] [Google Scholar]
  • 130.Campos BG, Ferreira RR, Gomes JC. The potentiation of the histamine release induced by adenosine in mast cells from guinea pig lung and heart: sharp dependence on the time of preincubation. Pharmacol Res. 2000;41:291–297. doi: 10.1006/phrs.1999.0589. [DOI] [PubMed] [Google Scholar]
  • 131.Marquardt DL, Walker LL, Wasserman SI. Adenosine receptors on mouse bone marrow-derived mast cells: functional significance and regulation by aminophylline. J Immunol. 1984;133:932–937. [PubMed] [Google Scholar]
  • 132.Delmich K, Eichelberg D, Schmutzler W. The effects of adenosine and of some adenosine analogues on the concanavalin A- or acetylcholine-induced histamine release from human adenoidal mast cells. Agents Actions. 1985;16:141–143. doi: 10.1007/BF01983122. [DOI] [PubMed] [Google Scholar]
  • 133.Lohse MJ, Maurer K, Gensheimer HP, Schwabe U. Dual actions of adenosine on rat peritoneal mast cells. Naunyn Schmiedebergs Arch Pharmacol. 1987;335:555–560. doi: 10.1007/BF00169124. [DOI] [PubMed] [Google Scholar]
  • 134.Auchampach JA, Jin X, Wan TC, Caughey GH, Linden J. Canine mast cell adenosine receptors: cloning and expression of the A3 receptor and evidence that degranulation is mediated by the A2B receptor. Mol Pharmacol. 1997;52:846–860. doi: 10.1124/mol.52.5.846. [DOI] [PubMed] [Google Scholar]
  • 135.Ramkumar V, Stiles GL, Beaven MA, Ali H. The A3 adenosine receptor is the unique adenosine receptor which facilitates release of allergic mediators in mast cells. J Biol Chem. 1993;268:16887–16890. [PubMed] [Google Scholar]
  • 136.Fozard JR, Pfannkuche HJ, Schuurman HJ. Mast cell degranulation following adenosine A3 receptor activation in rats. Eur J Pharmacol. 1996;298:293–297. doi: 10.1016/0014-2999(95)00822-5. [DOI] [PubMed] [Google Scholar]
  • 137.Zhong H, Shlykov SG, Molina JG, Sanborn BM, Jacobson MA, Tilley SL, Blackburn MR. Activation of murine lung mast cells by the adenosine A3 receptor. J Immunol. 2003;171:338–345. doi: 10.4049/jimmunol.171.1.338. [DOI] [PubMed] [Google Scholar]
  • 138.Tilley SL, Wagoner VA, Salvatore CA, Jacobson MA, Koller BH. Adenosine and inosine increase cutaneous vasopermeability by activating A3 receptors on mast cells. J Clin Invest. 2000;105:361–367. doi: 10.1172/JCI8253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Tilley SL, Tsai M, Williams CM, Wang ZS, Erikson CJ, Galli SJ, Koller BH. Identification of A3 receptor- and mast cell-dependent and -independent components of adenosine-mediated airway responsiveness in mice. J Immunol. 2003;171:331–337. doi: 10.4049/jimmunol.171.1.331. [DOI] [PubMed] [Google Scholar]
  • 140.Peachell PT, Lichtenstein LM, Schleimer RP. Differential regulation of human basophil and lung mast cell function by adenosine. J Pharmacol Exp Ther. 1991;256:717–726. [PubMed] [Google Scholar]
  • 141.Gomez G, Nardone V, Lotfi-Emran S, Zhao W, Schwartz LB. Intracellular adenosine inhibits IgE-dependent degranulation of human skin mast cells. J Clin Immunol. 2013;33:1349–1359. doi: 10.1007/s10875-013-9950-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Marquardt DL, Walker LL, Heinemann S. Cloning of two adenosine receptor subtypes from mouse bone marrow-derived mast cells. J Immunol. 1994;152:4508–4515. [PubMed] [Google Scholar]
  • 143.Hua X, Chason KD, Jania C, Acosta T, Ledent C, Tilley SL. Gs-coupled adenosine receptors differentially limit antigen-induced mast cell activation. J Pharmacol Exp Ther. 2013;344:426–435. doi: 10.1124/jpet.112.198978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Feoktistov I, Ryzhov S, Goldstein AE, Biaggioni I. Mast cell-mediated stimulation of angiogenesis: cooperative interaction between A2B and A3 adenosine receptors. Circ Res. 2003;92:485–492. doi: 10.1161/01.RES.0000061572.10929.2D. [DOI] [PubMed] [Google Scholar]
  • 145.Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV, Dikov MM, Blackburn MR, Biaggioni I, Feoktistov I. Effect of A2B adenosine receptor gene ablation on proinflammatory adenosine signaling in mast cells. J Immunol. 2008;180:7212–7220. doi: 10.4049/jimmunol.180.11.7212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Hua X, Chason KD, Patel JY, Naselsky WC, Tilley SL. IL-4 amplifies the pro-inflammatory effect of adenosine in human mast cells by changing expression levels of adenosine receptors. PLoS One. 2011;6:e24947. doi: 10.1371/journal.pone.0024947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Keller R. Tissue mast cells in immune reactions. Monogr Allergy. 1966;2:1–144. [PubMed] [Google Scholar]
  • 148.Diamant B, Krüger PG. Histamine release from isolated rat peritoneal mast cells induced by adenosine-5'-triphosphate. Acta Physiol Scand. 1967;71:291–302. doi: 10.1111/j.1748-1716.1967.tb03736.x. [DOI] [PubMed] [Google Scholar]
  • 149.Dahlquist R, Diamant B. Further observations on ATP-induced histamine release from rat mast cells. Acta Pharmacol Toxicol (Copenh) 1970;28:43. [PubMed] [Google Scholar]
  • 150.Sugiyama K. Calcium-dependent histamine release with degranulation from isolated rat mast cells by adenosine 5'-triphosphate. Jpn J Pharmacol. 1971;21:209–226. doi: 10.1254/jjp.21.209. [DOI] [PubMed] [Google Scholar]
  • 151.Krüger G, Bloom D, Diamant B. Structural aspects of histamine release in rat peritoneal mast cells. Effects of adenosine 5'-triphosphate and role of calcium. Int Arch Allergy Appl Immunol. 1974;47:1–13. doi: 10.1159/000231196. [DOI] [PubMed] [Google Scholar]
  • 152.Cockcroft S, Gomperts BD. Activation and inhibition of calcium-dependent histamine secretion by ATP ions applied to rat mast cells. J Physiol. 1979;296:229–243. doi: 10.1113/jphysiol.1979.sp013002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kiernan JA. Effects of known and suspected neurotransmitter substances and of some nucleotides on isolated mast cells. Experientia. 1972;28:653–655. doi: 10.1007/BF01944958. [DOI] [PubMed] [Google Scholar]
  • 154.Bienenstock J, MacQueen G, Sestini P, Marshall JS, Stead RH, Perdue MH. Mast cell/nerve interactions in vitro and in vivo. Am Rev Respir Dis. 1991;143:S55–S58. doi: 10.1164/ajrccm/143.3_Pt_2.S55. [DOI] [PubMed] [Google Scholar]
  • 155.Grosman N, Diamant B. Effects of adenosine-5'-triphosphate (ATP) on rat mast cells: influence on anaphylactic and compound 48/80-induced histamine release. Agents Actions. 1975;5:108–114. doi: 10.1007/BF02027349. [DOI] [PubMed] [Google Scholar]
  • 156.Johansen T, Chakravarty N. The utilization of adenosine triphosphate in rat mast cells during histamine release induced by anaphylactic reaction and compound 48/80. Naunyn Schmiedebergs Arch Pharmacol. 1975;288:243–260. doi: 10.1007/BF00500530. [DOI] [PubMed] [Google Scholar]
  • 157.Bulanova E, Budagian V, Orinska Z, Hein M, Petersen F, Thon L, Adam D, Bulfone-Paus S. Extracellular ATP induces cytokine expression and apoptosis through P2X7 receptor in murine mast cells. J Immunol. 2005;174:3880–3890. doi: 10.4049/jimmunol.174.7.3880. [DOI] [PubMed] [Google Scholar]
  • 158.Tatham PE, Lindau M. ATP-induced pore formation in the plasma membrane of rat peritoneal mast cells. J Gen Physiol. 1990;95:459–476. doi: 10.1085/jgp.95.3.459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Kurashima Y, Amiya T, Nochi T, Fujisawa K, Haraguchi T, Iba H, Tsutsui H, Sato S, Nakajima S, Iijima H, Kubo M, Kunisawa J, Kiyono H. Extracellular ATP mediates mast cell-dependent intestinal inflammation through P2X7 purinoceptors. Nat Commun. 2012;3:1034. doi: 10.1038/ncomms2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Bulanova E, Budagian V, Orinska Z, Koch-Nolte F, Haag F, Bulfone-Paus S. ATP induces P2X7 receptor-independent cytokine and chemokine expression through P2X1 and P2X3 receptors in murine mast cells. J Leukoc Biol. 2009;85:692–702. doi: 10.1189/jlb.0808470. [DOI] [PubMed] [Google Scholar]
  • 161.Wareham K, Vial C, Wykes RC, Bradding P, Seward EP. Functional evidence for the expression of P2X1, P2X4 and P2X7 receptors in human lung mast cells. Br J Pharmacol. 2009;157:1215–1224. doi: 10.1111/j.1476-5381.2009.00287.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Arandjelovic S, McKenney KR, Leming SS, Mowen KA. ATP induces protein arginine deiminase 2-dependent citrullination in mast cells through the P2X7 purinergic receptor. J Immunol. 2012;189:4112–4122. doi: 10.4049/jimmunol.1201098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Qian YX, McCloskey MA. Activation of mast cell K+ channels through multiple G protein-linked receptors. Proc Natl Acad Sci U S A. 1993;90:7844–7848. doi: 10.1073/pnas.90.16.7844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Feng C, Mery AG, Beller EM, Favot C, Boyce JA. Adenine nucleotides inhibit cytokine generation by human mast cells through a G s-coupled receptor. J Immunol. 2004;173:7539–7547. doi: 10.4049/jimmunol.173.12.7539. [DOI] [PubMed] [Google Scholar]
  • 165.Gao ZG, Ding Y, Jacobson KA. UDP-glucose acting at P2Y14 receptors is a mediator of mast cell degranulation. Biochem Pharmacol. 2010;79:873–879. doi: 10.1016/j.bcp.2009.10.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Gao ZG, Wei Q, Jayasekara MP, Jacobson KA. The role of P2Y14 and other P2Y receptors in degranulation of human LAD2 mast cells. Purinergic Signal. 2013;9:31–40. doi: 10.1007/s11302-012-9325-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Haskó G, Szabó C. Regulation of cytokine and chemokine production by transmitters and co-transmitters of the autonomic nervous system. Biochem Pharmacol. 1998;56:1079–1087. doi: 10.1016/s0006-2952(98)00153-1. [DOI] [PubMed] [Google Scholar]
  • 168.Burnstock G. The Autonomic Neuroeffector Junction. In: Robertson D, Low P, Burnstock G, Biaggioni I, editors. Primer on the autonomic nervous system. 2. Amsterdam: Elsevier; 2004. pp. 29–33. [Google Scholar]
  • 169.Burnstock G. Non-synaptic transmission at autonomic neuroeffector junctions. Neurochem Int. 2008;52:14–25. doi: 10.1016/j.neuint.2007.03.007. [DOI] [PubMed] [Google Scholar]
  • 170.Williams RM, Bienenstock J, Stead RH. Mast cells: the neuroimmune connection. Chem Immunol. 1995;61:208–235. [PubMed] [Google Scholar]
  • 171.Kiernan JA. Action of adenosine triphosphate on mast cells in normal and denervated skin. Arch Dermatol Forsch. 1974;251:83–86. doi: 10.1007/BF00561714. [DOI] [PubMed] [Google Scholar]
  • 172.Newson B, Dahlstrom A, Enerback L, Ahlman H. Suggestive evidence for a direct innervation of mucosal mast cells. Neuroscience. 1983;10:565–570. doi: 10.1016/0306-4522(83)90153-7. [DOI] [PubMed] [Google Scholar]
  • 173.Dimitriadou V, Aubineau P, Taxi J, Seylaz J. Ultrastructural evidence for a functional unit between nerve fibers and type II cerebral mast cells in the cerebral vascular wall. Neuroscience. 1987;22:621–630. doi: 10.1016/0306-4522(87)90358-7. [DOI] [PubMed] [Google Scholar]
  • 174.Levine JD, Coderre TJ, Covinsky K, Basbaum AI. Neural influences on synovial mast cell density in rat. J Neurosci Res. 1990;26:301–307. doi: 10.1002/jnr.490260306. [DOI] [PubMed] [Google Scholar]
  • 175.Keller JT, Dimlich RV, Zuccarello M, Lanker L, Strauss TA, Fritts MJ. Influence of the sympathetic nervous system as well as trigeminal sensory fibres on rat dural mast cells. Cephalalgia. 1991;11:215–221. doi: 10.1046/j.1468-2982.1991.1105215.x. [DOI] [PubMed] [Google Scholar]
  • 176.Dimitriadou V, Rouleau A, Trung T, Newlands GJ, Miller HR, Luffau G, Schwartz JC, Garbarg M. Functional relationships between sensory nerve fibers and mast cells of dura mater in normal and inflammatory conditions. Neuroscience. 1997;77:829–839. doi: 10.1016/s0306-4522(96)00488-5. [DOI] [PubMed] [Google Scholar]
  • 177.Gottwald TP, Hewlett BR, Lhotak S, Stead RH. Electrical stimulation of the vagus nerve modulates the histamine content of mast cells in the rat jejunal mucosa. Neuroreport. 1995;7:313–317. [PubMed] [Google Scholar]
  • 178.Suzuki R, Furuno T, Okamoto K, Teshima R, Nakanishi M. ATP plays a role in neurite stimulation with activated mast cells. J Neuroimmunol. 2007;192:49–56. doi: 10.1016/j.jneuroim.2007.09.013. [DOI] [PubMed] [Google Scholar]
  • 179.Nakanishi M, Furuno T. Molecular basis of neuroimmune interaction in an in vitro coculture approach. Cell Mol Immunol. 2008;5:249–259. doi: 10.1038/cmi.2008.31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Arizono N, Matsuda S, Hattori T, Kojima Y, Maeda T, Galli SJ. Anatomical variation in mast cell nerve associations in the rat small intestine, heart, lung, and skin. Similarities of distances between neural processes and mast cells, eosinophils, or plasma cells in the jejunal lamina propria. Lab Invest. 1990;62:626–634. [PubMed] [Google Scholar]
  • 181.Chelmicka-Schorr E, Kwasniewski MN, Czlonkowska A. Sympathetic nervous system modulates macrophage function. Int J Immunopharmacol. 1992;14:841–846. doi: 10.1016/0192-0561(92)90082-v. [DOI] [PubMed] [Google Scholar]
  • 182.Madden KS, Moynihan JA, Brenner GJ, Felten SY, Felten DL. Sympathetic nervous system modulation of the immune system. III. Alterations in T and B cell proliferation and differentiation in vitro following chemical sympathectomy. J Neuroimmunol. 1994;49:77–87. doi: 10.1016/0165-5728(94)90183-x. [DOI] [PubMed] [Google Scholar]
  • 183.Crivellato E, Soldano F, Travan L, Fusaroli P, Mallardi F. Apposition of enteric nerve fibers to plasma cells and immunoblasts in the mouse small bowel. Neurosci Lett. 1998;241:123–126. doi: 10.1016/s0304-3940(98)00004-4. [DOI] [PubMed] [Google Scholar]
  • 184.Genton L, Kudsk KA. Interactions between the enteric nervous system and the immune system: role of neuropeptides and nutrition. Am J Surg. 2003;186:253–258. doi: 10.1016/s0002-9610(03)00210-1. [DOI] [PubMed] [Google Scholar]
  • 185.Lappin D, Whaley K. Cyclic AMP-mediated modulation of the production of the second component of human complement by monocytes. Int Arch Allergy Appl Immunol. 1981;65:85–90. doi: 10.1159/000232742. [DOI] [PubMed] [Google Scholar]
  • 186.Lappin D, Whaley K. Adenosine A2 receptors on human monocytes modulate C2 production. Clin Exp Immunol. 1984;57:454–460. [PMC free article] [PubMed] [Google Scholar]
  • 187.Salmon JE, Brogle N, Brownlie C, Edberg JC, Kimberly RP, Chen BX, Erlanger BF. Human mononuclear phagocytes express adenosine A1 receptors. A novel mechanism for differential regulation of Fcγ receptor function. J Immunol. 1993;151:2775–2785. [PubMed] [Google Scholar]
  • 188.Le Vraux V, Chen YL, Masson I, De Sousa M, Giroud JP, Florentin I, Chauvelot-Moachon L. Inhibition of human monocyte TNF production by adenosine receptor agonists. Life Sci. 1993;52:1917–1924. doi: 10.1016/0024-3205(93)90632-d. [DOI] [PubMed] [Google Scholar]
  • 189.Zhang JG, Hepburn L, Cruz G, Borman RA, Clark KL. The role of adenosine A2A and A2B receptors in the regulation of TNF-α production by human monocytes. Biochem Pharmacol. 2005;69:883–889. doi: 10.1016/j.bcp.2004.12.008. [DOI] [PubMed] [Google Scholar]
  • 190.Link AA, Kino T, Worth JA, McGuire JL, Crane ML, Chrousos GP, Wilder RL, Elenkov IJ. Ligand-activation of the adenosine A2a receptors inhibits IL-12 production by human monocytes. J Immunol. 2000;164:436–442. doi: 10.4049/jimmunol.164.1.436. [DOI] [PubMed] [Google Scholar]
  • 191.Perez-Aso M, Feig JL, Aránzazu M, Cronstein BN. Adenosine A2A receptor and TNF-α regulate the circadian machinery of the human monocytic THP-1 cells. Inflammation. 2013;36:152–162. doi: 10.1007/s10753-012-9530-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Ramakers BP, Riksen NP, Rongen GA, van der Hoeven JG, Smits P, Pickkers P. The effect of adenosine receptor agonists on cytokine release by human mononuclear cells depends on the specific Toll-like receptor subtype used for stimulation. Cytokine. 2006;35:95–99. doi: 10.1016/j.cyto.2006.07.014. [DOI] [PubMed] [Google Scholar]
  • 193.Takahashi HK, Kanke T, Liu K, Yoshino T, Sendo T, Tanaka N, Nishibori M. Adenosine A2A-receptor stimulation inhibits lipopolysaccharide-induced interleukin-18 production in monocytes. J Pharmacol Sci. 2007;104:183–186. doi: 10.1254/jphs.sc0070101. [DOI] [PubMed] [Google Scholar]
  • 194.Hamano R, Takahashi HK, Iwagaki H, Kanke T, Liu K, Yoshino T, Sendo T, Nishibori M, Tanaka N. Stimulation of adenosine A2A receptor inhibits LPS-induced expression of intercellular adhesion molecule 1 and production of TNF-α in human peripheral blood mononuclear cells. Shock. 2008;29:154–159. doi: 10.1097/shk.0b013e31812385da. [DOI] [PubMed] [Google Scholar]
  • 195.Merrill JT, Shen C, Schreibman D, Coffey D, Zakharenko O, Fisher R, Lahita RG, Salmon J, Cronstein BN. Adenosine A1 receptor promotion of multinucleated giant cell formation by human monocytes: a mechanism for methotrexate-induced nodulosis in rheumatoid arthritis. Arthritis Rheum. 1997;40:1308–1315. doi: 10.1002/1529-0131(199707)40:7<1308::AID-ART16>3.0.CO;2-M. [DOI] [PubMed] [Google Scholar]
  • 196.Barbieri D, Abbracchio MP, Salvioli S, Monti D, Cossarizza A, Ceruti S, Brambilla R, Cattabeni F, Jacobson KA, Franceschi C. Apoptosis by 2-chloro-2'-deoxy-adenosine and 2-chloro-adenosine in human peripheral blood mononuclear cells. Neurochem Int. 1998;32:493–504. doi: 10.1016/s0197-0186(97)00129-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Altieri DC, Wiltse WL, Edgington TS. Signal transduction initiated by extracellular nucleotides regulates the high affinity ligand recognition of the adhesive receptor CD11b/CD18. J Immunol. 1990;145:662–670. [PubMed] [Google Scholar]
  • 198.Ventura MA, Thomopoulos P. ADP and ATP activate distinct signaling pathways in human promonocytic U-937 cells differentiated with 1,25-dihydroxy-vitamin D3. Mol Pharmacol. 1995;47:104–114. [PubMed] [Google Scholar]
  • 199.Humphreys BD, Dubyak GR. Induction of the P2z/P2X7 nucleotide receptor and associated phospholipase D activity by lipopolysaccharide and IFN-γ in the human THP-1 monocytic cell line. J Immunol. 1996;157:5627–5637. [PubMed] [Google Scholar]
  • 200.Rassendren F, Buell GN, Virginio C, Collo G, North RA, Surprenant A. The permeabilizing ATP receptor, P2X7. Cloning and expression of a human cDNA. J Biol Chem. 1997;272:5482–5486. doi: 10.1074/jbc.272.9.5482. [DOI] [PubMed] [Google Scholar]
  • 201.Aga M, Johnson CJ, Hart AP, Guadarrama AG, Suresh M, Svaren J, Bertics PJ, Darien BJ. Modulation of monocyte signalling and pore formation in response to agonists of the nucleotide receptor P2X7. J Leukoc Biol. 2002;72:222–232. [PubMed] [Google Scholar]
  • 202.Humphreys BD, Dubyak GR. Modulation of P2X7 nucleotide receptor expression by pro- and anti-inflammatory stimuli in THP-1 monocytes. J Leukoc Biol. 1998;64:265–273. doi: 10.1002/jlb.64.2.265. [DOI] [PubMed] [Google Scholar]
  • 203.Grahames CB, Michel AD, Chessell IP, Humphrey PP. Pharmacological characterization of ATP- and LPS-induced IL-1β release in human monocytes. Br J Pharmacol. 1999;127:1915–1921. doi: 10.1038/sj.bjp.0702732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Warren AY, Harvey L, Shaw RW, Khan RN. Interleukin-1β secretion from cord blood mononuclear cells in vitro involves P2X7 receptor activation. Reprod Sci. 2008;15:189–194. doi: 10.1177/1933719107310710. [DOI] [PubMed] [Google Scholar]
  • 205.Jalilian I, Peranec M, Curtis BL, Seavers A, Spildrejorde M, Sluyter V, Sluyter R. Activation of the damage-associated molecular pattern receptor P2X7 induces interleukin-1β release from canine monocytes. Vet Immunol Immunopathol. 2012;149:86–91. doi: 10.1016/j.vetimm.2012.05.004. [DOI] [PubMed] [Google Scholar]
  • 206.Mehta VB, Hart J, Wewers MD. ATP-stimulated release of interleukin (IL)-1β and IL-18 requires priming by lipopolysaccharide and is independent of caspase-1 cleavage. J Biol Chem. 2001;276:3820–3826. doi: 10.1074/jbc.M006814200. [DOI] [PubMed] [Google Scholar]
  • 207.Gardella S, Andrei C, Ferrera D, Lotti LV, Torrisi MR, Bianchi ME, Rubartelli A. The nuclear protein HMGB1 is secreted by monocytes via a non-classical, vesicle-mediated secretory pathway. EMBO Rep. 2002;3:995–1001. doi: 10.1093/embo-reports/kvf198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Piccini A, Carta S, Tassi S, Lasiglié D, Fossati G, Rubartelli A. ATP is released by monocytes stimulated with pathogen-sensing receptor ligands and induces IL-1β and IL-18 secretion in an autocrine way. Proc Natl Acad Sci U S A. 2008;105:8067–8072. doi: 10.1073/pnas.0709684105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Asgari E, Le Friec G, Yamamoto H, Perucha E, Sacks SS, Köhl J, Cook HT, Kemper C. C3a modulates IL-1β secretion in human monocytes by regulating ATP efflux and subsequent NLRP3 inflammasome activation. Blood. 2013;122:3473–3481. doi: 10.1182/blood-2013-05-502229. [DOI] [PubMed] [Google Scholar]
  • 210.Di Virgilio F. Liaisons dangereuses: P2X7 and the inflammasome. Trends Pharmacol Sci. 2007;28:465–472. doi: 10.1016/j.tips.2007.07.002. [DOI] [PubMed] [Google Scholar]
  • 211.Sluyter R, Shemon AN, Wiley JS. Glu496 to Ala polymorphism in the P2X7 receptor impairs ATP-induced IL-1β release from human monocytes. J Immunol. 2004;172:3399–3405. doi: 10.4049/jimmunol.172.6.3399. [DOI] [PubMed] [Google Scholar]
  • 212.Straub RH, Mayer M, Kreutz M, Leeb S, Schölmerich J, Falk W. Neurotransmitters of the sympathetic nerve terminal are powerful chemoattractants for monocytes. J Leukoc Biol. 2000;67:553–558. doi: 10.1002/jlb.67.4.553. [DOI] [PubMed] [Google Scholar]
  • 213.Kaufmann A, Musset B, Limberg SH, Renigunta V, Sus R, Dalpke AH, Heeg KM, Robaye B, Hanley PJ. "Host tissue damage" signal ATP promotes non-directional migration and negatively regulates toll-like receptor signaling in human monocytes. J Biol Chem. 2005;280:32459–32467. doi: 10.1074/jbc.M505301200. [DOI] [PubMed] [Google Scholar]
  • 214.Akbar GK, Mills DC, Kunapuli SP. Characterization of extracellular nucleotide-induced Mac-1 (αMβ2 integrin) surface expression on peripheral blood leukocytes. Biochem Biophys Res Commun. 1997;233:71–75. doi: 10.1006/bbrc.1997.6396. [DOI] [PubMed] [Google Scholar]
  • 215.Warny M, Aboudola S, Robson SC, Sévigny J, Communi D, Soltoff SP, Kelly CP. P2Y6 nucleotide receptor mediates monocyte interleukin-8 production in response to UDP or lipopolysaccharide. J Biol Chem. 2001;276:26051–26056. doi: 10.1074/jbc.M102568200. [DOI] [PubMed] [Google Scholar]
  • 216.Ben Yebdri F, Kukulski F, Tremblay A, Sévigny J. Concomitant activation of P2Y2 and P2Y6 receptors on monocytes is required for TLR1/2-induced neutrophil migration by regulating IL-8 secretion. Eur J Immunol. 2009;39:2885–2894. doi: 10.1002/eji.200939347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Rizzo R, Ferrari D, Melchiorri L, Stignani M, Gulinelli S, Baricordi OR, Di Virgilio F. Extracellular ATP acting at the P2X7 receptor inhibits secretion of soluble HLA-G from human monocytes. J Immunol. 2009;183:4302–4311. doi: 10.4049/jimmunol.0804265. [DOI] [PubMed] [Google Scholar]
  • 218.Hill LM, Gavala ML, Lenertz LY, Bertics PJ. Extracellular ATP may contribute to tissue repair by rapidly stimulating purinergic receptor X7-dependent vascular endothelial growth factor release from primary human monocytes. J Immunol. 2010;185:3028–3034. doi: 10.4049/jimmunol.1001298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Gu BJ, Saunders BM, Jursik C, Wiley JS. The P2X7-nonmuscle myosin membrane complex regulates phagocytosis of nonopsonized particles and bacteria by a pathway attenuated by extracellular ATP. Blood. 2010;115:1621–1631. doi: 10.1182/blood-2009-11-251744. [DOI] [PubMed] [Google Scholar]
  • 220.Aksamit RR, Backlund PS, Jr, Cantoni GL. Chemotaxis and the synthesis of specific proteins are inhibited by 3-deazaadenosine and other adenosine analogs in a mouse macrophage cell line. J Biol Chem. 1983;258:20–23. [PubMed] [Google Scholar]
  • 221.Riches DW, Watkins JL, Henson PM, Stanworth DR. Regulation of macrophage lysosomal secretion by adenosine, adenosine phosphate esters, and related structural analogues of adenosine. J Leukoc Biol. 1985;37:545–557. doi: 10.1002/jlb.37.5.545. [DOI] [PubMed] [Google Scholar]
  • 222.McWhinney CD, Dudley MW, Bowlin TL, Peet NP, Schook L, Bradshaw M, De M, Borcherding DR, Edwards CK., III Activation of adenosine A3 receptors on macrophages inhibits tumor necrosis factor-α. Eur J Pharmacol. 1996;310:209–216. doi: 10.1016/0014-2999(96)00272-5. [DOI] [PubMed] [Google Scholar]
  • 223.Németh ZH, Leibovich SJ, Deitch EA, Vizi ES, Szabó C, Haskó G. cDNA microarray analysis reveals a nuclear factor-κB-independent regulation of macrophage function by adenosine. J Pharmacol Exp Ther. 2003;306:1042–1049. doi: 10.1124/jpet.103.052944. [DOI] [PubMed] [Google Scholar]
  • 224.Hon WM, Moochhala S, Khoo HE. Adenosine and its receptor agonists potentiate nitric oxide synthase expression induced by lipopolysaccharide in RAW 264.7 murine macrophages. Life Sci. 1997;60:1327–1335. doi: 10.1016/s0024-3205(97)00078-7. [DOI] [PubMed] [Google Scholar]
  • 225.Zídek Z, Kmoníckova E, Holy A. Involvement of adenosine A1 receptors in upregulation of nitric oxide by acyclic nucleotide analogues. Eur J Pharmacol. 2004;501:79–86. doi: 10.1016/j.ejphar.2004.08.031. [DOI] [PubMed] [Google Scholar]
  • 226.Xaus J, Mirabet M, Lloberas J, Soler C, Lluis C, Franco R, Celada A. IFN-γ up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J Immunol. 1999;162:3607–3614. [PubMed] [Google Scholar]
  • 227.Murphree LJ, Sullivan GW, Marshall MA, Linden J. Lipopolysaccharide rapidly modifies adenosine receptor transcripts in murine and human macrophages: role of NF-κB in A2A adenosine receptor induction. Biochem J. 2005;391:575–580. doi: 10.1042/BJ20050888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Elson G, Eisenberg M, Garg C, Outram S, Ferrante CJ, Hasko G, Leibovich SJ. Induction of murine adenosine A2A receptor expression by LPS: analysis of the 5' upstream promoter. Genes Immun. 2013;14:147–153. doi: 10.1038/gene.2012.60. [DOI] [PubMed] [Google Scholar]
  • 229.Kreckler LM, Wan TC, Ge ZD, Auchampach JA. Adenosine inhibits tumor necrosis factor-α release from mouse peritoneal macrophages via A2A and A2B but not the A3 adenosine receptor. J Pharmacol Exp Ther. 2006;317:172–180. doi: 10.1124/jpet.105.096016. [DOI] [PubMed] [Google Scholar]
  • 230.Ezeamuzie CI, Khan I. The role of adenosine A2 receptors in the regulation of TNF-α production and PGE2 release in mouse peritoneal macrophages. Int Immunopharmacol. 2007;7:483–490. doi: 10.1016/j.intimp.2006.12.001. [DOI] [PubMed] [Google Scholar]
  • 231.Chen H, Yang D, Carroll SH, Eltzschig HK, Ravid K. Activation of the macrophage A2b adenosine receptor regulates tumor necrosis factor-α levels following vascular injury. Exp Hematol. 2009;37:533–538. doi: 10.1016/j.exphem.2009.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Buenestado A, Grassin Delyle S, Arnould I, Besnard F, Naline E, Blouquit-Laye S, Chapelier A, Bellamy JF, Devillier P. The role of adenosine receptors in regulating production of tumour necrosis factor-alpha and chemokines by human lung macrophages. Br J Pharmacol. 2010;159:1304–1311. doi: 10.1111/j.1476-5381.2009.00614.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Németh ZH, Lutz CS, Csóka B, Deitch EA, Leibovich SJ, Gause WC, Tone M, Pacher P, Vizi ES, Haskó G. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J Immunol. 2005;175:8260–8270. doi: 10.4049/jimmunol.175.12.8260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Csóka B, Németh ZH, Selmeczy Z, Koscsó B, Pacher P, Vizi ES, Deitch EA, Haskó G. Role of A2A adenosine receptors in regulation of opsonized E. coli-induced macrophage function. Purinergic Signal. 2007;3:447–452. doi: 10.1007/s11302-007-9075-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Csóka B, Németh ZH, Virág L, Gergely P, Leibovich SJ, Pacher P, Sun CX, Blackburn MR, Vizi ES, Deitch EA, Haskó G. A2A adenosine receptors and C/EBPβ are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood. 2007;110:2685–2695. doi: 10.1182/blood-2007-01-065870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Zanin RF, Braganhol E, Bergamin LS, Campesato LF, Filho AZ, Moreira JC, Morrone FB, Sévigny J, Schetinger MR, de Souza Wyse AT, Battastini AM. Differential macrophage activation alters the expression profile of NTPDase and ecto-5'-nucleotidase. PLoS One. 2012;7:e31205. doi: 10.1371/journal.pone.0031205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Soledade CS, Franchini KG, Linden J, Huo Y. Stimulation of adenosine A2A receptor regulates peroxisome proliferator activated receptors in macrophages. FASEB J. 2007;21:A1035. [Google Scholar]
  • 238.De Ponti C, Carini R, Alchera E, Nitti MP, Locati M, Albano E, Cairo G, Tacchini L. Adenosine A2a receptor-mediated, normoxic induction of HIF-1 through PKC and PI-3 K-dependent pathways in macrophages. J Leukoc Biol. 2007;82:392–402. doi: 10.1189/jlb.0107060. [DOI] [PubMed] [Google Scholar]
  • 239.Leibovich SJ, Chen JF, Pinhal-Enfield G, Belem PC, Elson G, Rosania A, Ramanathan M, Montesinos C, Jacobson M, Schwarzschild MA, Fink JS, Cronstein B. Synergistic up-regulation of vascular endothelial growth factor expression in murine macrophages by adenosine A2A receptor agonists and endotoxin. Am J Pathol. 2002;160:2231–2244. doi: 10.1016/S0002-9440(10)61170-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Ernens I, Leonard F, Vausort M, Rolland-Turner M, Devaux Y, Wagner DR. Adenosine up-regulates vascular endothelial growth factor in human macrophages. Biochem Biophys Res Commun. 2010;392:351–356. doi: 10.1016/j.bbrc.2010.01.023. [DOI] [PubMed] [Google Scholar]
  • 241.Velot E, Haas B, Leonard F, Ernens I, Rolland-Turner M, Schwartz C, Longrois D, Devaux Y, Wagner DR. Activation of the adenosine-A3 receptor stimulates matrix metalloproteinase-9 secretion by macrophages. Cardiovasc Res. 2008;80:246–254. doi: 10.1093/cvr/cvn201. [DOI] [PubMed] [Google Scholar]
  • 242.Barczyk K, Ehrchen J, Tenbrock K, Ahlmann M, Kneidl J, Viemann D, Roth J. Glucocorticoids promote survival of anti-inflammatory macrophages via stimulation of adenosine receptor A3. Blood. 2010;116:446–455. doi: 10.1182/blood-2009-10-247106. [DOI] [PubMed] [Google Scholar]
  • 243.Steinberg TH, Newman AS, Swanson JA, Silverstein SC. ATP4− permeabilizes the plasma membrane of mouse macrophages to fluorescent dyes. J Biol Chem. 1987;262:8884–8888. [PubMed] [Google Scholar]
  • 244.Buisman HP, Steinberg TH, Fischbarg J, Silverstein SC, Vogelzang SA, Ince C, Ypey DL, Leijh PC. Extracellular ATP induces a large nonselective conductance in macrophage plasma membranes. Proc Natl Acad Sci U S A. 1988;85:7988–7992. doi: 10.1073/pnas.85.21.7988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Steinberg TH, Silverstein SC. Extracellular ATP4− promotes cation fluxes in the J774 mouse macrophage cell line. J Biol Chem. 1987;262:3118–3122. [PubMed] [Google Scholar]
  • 246.Greenberg S, Di Virgilio F, Steinberg TH, Silverstein SC. Extracellular nucleotides mediate Ca2+ fluxes in J774 macrophages by two distinct mechanisms. J Biol Chem. 1988;263:10337–10343. [PubMed] [Google Scholar]
  • 247.Coutinho-Silva R, Alves LA, Savino W, Persechini PM. A cation non-selective channel induced by extracellular ATP in macrophages and phagocytic cells of the thymic reticulum. Biochim Biophys Acta. 1996;1278:125–130. doi: 10.1016/0005-2736(95)00200-6. [DOI] [PubMed] [Google Scholar]
  • 248.Nakanishi M, Takihara H, Minoru Y, Yagawa K. Extracellular ATP itself elicits superoxide generation in guinea pig peritoneal macrophages. FEBS Lett. 1991;282:91–94. doi: 10.1016/0014-5793(91)80451-8. [DOI] [PubMed] [Google Scholar]
  • 249.Murphy JK, Livingston FR, Gozal E, Torres M, Forman HJ. Stimulation of the rat alveolar macrophage respiratory burst by extracellular adenine nucleotides. Am J Respir Cell Mol Biol. 1993;9:505–510. doi: 10.1165/ajrcmb/9.5.505. [DOI] [PubMed] [Google Scholar]
  • 250.Sung SS, Young JD, Origlio AM, Heiple JM, Kaback HR, Silverstein SC. Extracellular ATP perturbs transmembrane ion fluxes, elevates cytosolic [Ca2+], and inhibits phagocytosis in mouse macrophages. J Biol Chem. 1985;260:13442–13449. [PubMed] [Google Scholar]
  • 251.Murgia M, Pizzo P, Steinberg TH, Di Virgilio F. Characterization of the cytotoxic effect of extracellular ATP in J774 mouse macrophages. Biochem J. 1992;288:897–901. doi: 10.1042/bj2880897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252.Blanchard DK, McMillen S, Djeu JY. IFN-γ enhances sensitivity of human macrophages to extracellular ATP-mediated lysis. J Immunol. 1991;147:2579–2585. [PubMed] [Google Scholar]
  • 253.Pfeilschifter J, Thüring B, Festa F. Extracellular ATP stimulates poly(inositol phospholipid) hydrolysis and eicosanoid synthesis in mouse peritoneal macrophages in culture. Eur J Biochem. 1989;186:509–513. doi: 10.1111/j.1432-1033.1989.tb15236.x. [DOI] [PubMed] [Google Scholar]
  • 254.Murgia M, Hanau S, Pizzo P, Rippa M, Di Virgilio F. Oxidized ATP. An irreversible inhibitor of the macrophage purinergic P2Z receptor. J Biol Chem. 1993;268:8199–8203. [PubMed] [Google Scholar]
  • 255.el-Moatassim C, Dubyak GR. Dissociation of the pore-forming and phospholipase D activities stimulated via P2z purinergic receptors in BAC1.2 F5 macrophages. Product inhibition of phospholipase D enzyme activity. J Biol Chem. 1993;268:15571–15578. [PubMed] [Google Scholar]
  • 256.Hickman SE, El Khoury J, Greenberg S, Schieren I, Silverstein SC. P2Z adenosine triphosphate receptor activity in cultured human monocyte-derived macrophages. Blood. 1994;84:2452–2456. [PubMed] [Google Scholar]
  • 257.Falzoni S, Munerati M, Ferrari D, Spisani S, Moretti S, Di Virgilio F. The purinergic P2Z receptor of human macrophage cells. Characterization and possible physiological role. J Clin Invest. 1995;95:1207–1216. doi: 10.1172/JCI117770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Perregaux D, Gabel CA. Interleukin-1β maturation and release in response to ATP and nigericin. Evidence that potassium depletion mediated by these agents is a necessary and common feature of their activity. J Biol Chem. 1994;269:15195–15203. [PubMed] [Google Scholar]
  • 259.Griffiths RJ, Stam EJ, Downs JT, Otterness IG. ATP induces the release of IL-1 from LPS-primed cells in vivo. J Immunol. 1995;154:2821–2828. [PubMed] [Google Scholar]
  • 260.Ferrari D, Chiozzi P, Falzoni S, dal Susino M, Melchiorri L, Baricordi OR, Di Virgilio F. Extracellular ATP triggers IL-1β release by activating the purinergic P2Z receptor of human macrophages. J Immunol. 1997;159:1451–1458. [PubMed] [Google Scholar]
  • 261.Brough D, Le Feuvre RA, Wheeler RD, Solovyova N, Hilfiker S, Rothwell NJ, Verkhratsky A. Ca2+ stores and Ca2+ entry differentially contribute to the release of IL-1β and IL-1α from murine macrophages. J Immunol. 2003;170:3029–3036. doi: 10.4049/jimmunol.170.6.3029. [DOI] [PubMed] [Google Scholar]
  • 262.Qu Y, Franchi L, Nunez G, Dubyak GR. Nonclassical IL-1β secretion stimulated by P2X7 receptors is dependent on inflammasome activation and correlated with exosome release in murine macrophages. J Immunol. 2007;179:1913–1925. doi: 10.4049/jimmunol.179.3.1913. [DOI] [PubMed] [Google Scholar]
  • 263.Pelegrin P, Barroso-Gutierrez C, Surprenant A. P2X7 receptor differentially couples to distinct release pathways for IL-1β in mouse macrophage. J Immunol. 2008;180:7147–7157. doi: 10.4049/jimmunol.180.11.7147. [DOI] [PubMed] [Google Scholar]
  • 264.Lee BH, Hwang DM, Palaniyar N, Grinstein S, Philpott DJ, Hu J. Activation of P2X7 receptor by ATP plays an important role in regulating inflammatory responses during acute viral infection. PLoS One. 2012;7:e35812. doi: 10.1371/journal.pone.0035812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Petrovski G, Ayna G, Majai G, Hodrea J, Benko S, Mádi A, Fésüs L. Phagocytosis of cells dying through autophagy induces inflammasome activation and IL-1β release in human macrophages. Autophagy. 2011;7:321–330. doi: 10.4161/auto.7.3.14583. [DOI] [PubMed] [Google Scholar]
  • 266.Niemi K, Teirilä L, Lappalainen J, Rajamäki K, Baumann MH, Öörni K, Wolff H, Kovanen PT, Matikainen S, Eklund KK. Serum amyloid A activates the NLRP3 inflammasome via P2X7 receptor and a cathepsin B-sensitive pathway. J Immunol. 2011;186:6119–6128. doi: 10.4049/jimmunol.1002843. [DOI] [PubMed] [Google Scholar]
  • 267.Lucattelli M, Cicko S, Muller T, Lommatzsch M, De Cunto G, Cardini S, Sundas W, Grimm M, Zeiser R, Durk T, Zissel G, Sorichter S, Ferrari D, Di Virgilio F, Virchow JC, Lungarella G, Idzko M. P2X7 receptor signaling in the pathogenesis of smoke-induced lung inflammation and emphysema. Am J Respir Cell Mol Biol. 2011;44:423–429. doi: 10.1165/rcmb.2010-0038OC. [DOI] [PubMed] [Google Scholar]
  • 268.Lammas DA, Stober C, Harvey CJ, Kendrick N, Panchalingam S, Kumararatne DS. ATP-induced killing of mycobacteria by human macrophages is mediated by purinergic P2Z (P2X7) receptors. Immunity. 1997;7:433–444. doi: 10.1016/s1074-7613(00)80364-7. [DOI] [PubMed] [Google Scholar]
  • 269.Kusner DJ, Barton JA. ATP stimulates human macrophages to kill intracellular virulent Mycobacterium tuberculosis via calcium-dependent phagosome-lysosome fusion. J Immunol. 2001;167:3308–3315. doi: 10.4049/jimmunol.167.6.3308. [DOI] [PubMed] [Google Scholar]
  • 270.Kusner DJ, Adams J. ATP-induced killing of virulent Mycobacterium tuberculosis within human macrophages requires phospholipase D. J Immunol. 2000;164:379–388. doi: 10.4049/jimmunol.164.1.379. [DOI] [PubMed] [Google Scholar]
  • 271.Fairbairn IP, Stober CB, Kumararatne DS, Lammas DA. ATP-mediated killing of intracellular mycobacteria by macrophages is a P2X 7-dependent process inducing bacterial death by phagosome-lysosome fusion. J Immunol. 2001;167:3300–3307. doi: 10.4049/jimmunol.167.6.3300. [DOI] [PubMed] [Google Scholar]
  • 272.Placido R, Auricchio G, Falzoni S, Battistini L, Colizzi V, Brunetti E, Di Virgilio F, Mancino G. P2X7 purinergic receptors and extracellular ATP mediate apoptosis of human monocytes/macrophages infected with Mycobacterium tuberculosis reducing the intracellular bacterial viability. Cell Immunol. 2006;244:10–18. doi: 10.1016/j.cellimm.2007.02.001. [DOI] [PubMed] [Google Scholar]
  • 273.Biswas D, Qureshi OS, Lee WY, Croudace JE, Mura M, Lammas DA. ATP-induced autophagy is associated with rapid killing of intracellular mycobacteria within human monocytes/macrophages. BMC Immunol. 2008;9:35. doi: 10.1186/1471-2172-9-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Saunders BM, Fernando SL, Sluyter R, Britton WJ, Wiley JS. A loss-of-function polymorphism in the human P2X7 receptor abolishes ATP-mediated killing of mycobacteria. J Immunol. 2003;171:5442–5446. doi: 10.4049/jimmunol.171.10.5442. [DOI] [PubMed] [Google Scholar]
  • 275.Fernando SL, Saunders BM, Sluyter R, Skarratt KK, Wiley JS, Britton WJ. Gene dosage determines the negative effects of polymorphic alleles of the P2X7 receptor on adenosine triphosphate-mediated killing of mycobacteria by human macrophages. J Infect Dis. 2005;192:149–155. doi: 10.1086/430622. [DOI] [PubMed] [Google Scholar]
  • 276.Shemon AN, Sluyter R, Fernando SL, Clarke AL, Dao-Ung LP, Skarratt KK, Saunders BM, Tan KS, Gu BJ, Fuller SJ, Britton WJ, Petrou S, Wiley JS. A Thr357 to Ser polymorphism in homozygous and compound heterozygous subjects causes absent or reduced P2X7 function and impairs ATP-induced mycobacterial killing by macrophages. J Biol Chem. 2006;281:2079–2086. doi: 10.1074/jbc.M507816200. [DOI] [PubMed] [Google Scholar]
  • 277.Franco-Martínez S, Niño-Moreno P, Bernal-Silva S, Baranda L, Rocha-Meza M, Portales-Cervantes L, Layseca-Espinosa E, González-Amaro R, Portales-Pérez D. Expression and function of the purinergic receptor P2X7 in patients with pulmonary tuberculosis. Clin Exp Immunol. 2006;146:253–261. doi: 10.1111/j.1365-2249.2006.03213.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Chaves SP, Torres-Santos EC, Marques C, Figliuolo VR, Persechini PM, Coutinho-Silva R, Rossi-Bergmann B. Modulation of P2X7 purinergic receptor in macrophages by Leishmania amazonensis and its role in parasite elimination. Microbes Infect. 2009;11:842–849. doi: 10.1016/j.micinf.2009.05.001. [DOI] [PubMed] [Google Scholar]
  • 279.Chaves MM, Marques-da-Silva C, Monteiro AP, Canetti C, Coutinho-Silva R. Leukotriene B4 modulates P2X7 receptor-mediated Leishmania amazonensis elimination in murine macrophages. J Immunol. 2014;192:4765–4773. doi: 10.4049/jimmunol.1301058. [DOI] [PubMed] [Google Scholar]
  • 280.Corrêa G, Marques da Silva C, de Abreu Moreira-Souza AC, Vommaro RC, Coutinho-Silva R. Activation of the P2X7 receptor triggers the elimination of Toxoplasma gondii tachyzoites from infected macrophages. Microbes Infect. 2010;12:497–504. doi: 10.1016/j.micinf.2010.03.004. [DOI] [PubMed] [Google Scholar]
  • 281.Lees MP, Fuller SJ, McLeod R, Boulter NR, Miller CM, Zakrzewski AM, Mui EJ, Witola WH, Coyne JJ, Hargrave AC, Jamieson SE, Blackwell JM, Wiley JS, Smith NC. P2X7 receptor-mediated killing of an intracellular parasite, Toxoplasma gondii, by human and murine macrophages. J Immunol. 2010;184:7040–7046. doi: 10.4049/jimmunol.1000012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Tonetti M, Sturla L, Bistolfi T, Benatti U, De Flora A. Extracellular ATP potentiates nitric oxide synthase expression induced by lipopolysaccharide in RAW 264.7 murine macrophages. Biochem Biophys Res Commun. 1994;203:430–435. doi: 10.1006/bbrc.1994.2200. [DOI] [PubMed] [Google Scholar]
  • 283.Tonetti M, Sturla L, Giovine M, Benatti U, De Flora A. Extracellular ATP enhances mRNA levels of nitric oxide synthase and TNF-α in lipopolysaccharide-treated RAW 264.7 murine macrophages. Biochem Biophys Res Commun. 1995;214:125–130. doi: 10.1006/bbrc.1995.2265. [DOI] [PubMed] [Google Scholar]
  • 284.Denlinger LC, Fisette PL, Garis KA, Kwon G, Vazquez-Torres A, Simon AD, Nguyen B, Proctor RA, Bertics PJ, Corbett JA. Regulation of inducible nitric oxide synthase expression by macrophage purinoreceptors and calcium. J Biol Chem. 1996;271:337–342. doi: 10.1074/jbc.271.1.337. [DOI] [PubMed] [Google Scholar]
  • 285.Hu Y, Fisette PL, Denlinger LC, Guadarrama AG, Sommer JA, Proctor RA, Bertics PJ. Purinergic receptor modulation of lipopolysaccharide signaling and inducible nitric-oxide synthase expression in RAW 264.7 macrophages. J Biol Chem. 1998;273:27170–27175. doi: 10.1074/jbc.273.42.27170. [DOI] [PubMed] [Google Scholar]
  • 286.Sperlágh B, Haskó G, Németh Z, Vizi ES. ATP released by LPS increases nitric oxide production in raw 264.7 macrophage cell line via P2Z/P2X7 receptors. Neurochem Int. 1998;33:209–215. doi: 10.1016/s0197-0186(98)00025-4. [DOI] [PubMed] [Google Scholar]
  • 287.Sommer JA, Fisette PL, Hu Y, Denlinger LC, Guerra AN, Bertics PJ, Proctor RA. Purinergic receptor modulation of LPS-stimulated signaling events and nitric oxide release in RAW 264.7 macrophages. J Endotoxin Res. 1999;5:70–74. [Google Scholar]
  • 288.Chen YJ, Hsu KW, Chen YL. Acute glucose overload potentiates nitric oxide production in lipopolysaccharide-stimulated macrophages: the role of purinergic receptor activation. Cell Biol Int. 2006;30:817–822. doi: 10.1016/j.cellbi.2006.06.003. [DOI] [PubMed] [Google Scholar]
  • 289.Pfeiffer ZA, Guerra AN, Hill LM, Gavala ML, Prabhu U, Aga M, Hall DJ, Bertics PJ. Nucleotide receptor signaling in murine macrophages is linked to reactive oxygen species generation. Free Radic Biol Med. 2007;42:1506–1516. doi: 10.1016/j.freeradbiomed.2007.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 290.Cruz CM, Rinna A, Forman HJ, Ventura AL, Persechini PM, Ojcius DM. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem. 2007;282:2871–2879. doi: 10.1074/jbc.M608083200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291.Lenertz LY, Gavala ML, Hill LM, Bertics PJ. Cell signaling via the P2X7 nucleotide receptor: linkage to ROS production, gene transcription, and receptor trafficking. Purinergic Signal. 2009;5:175–187. doi: 10.1007/s11302-009-9133-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Costa-Junior HM, Mendes AN, Davis GH, da Cruz CM, Ventura AL, Serezani CH, Faccioli LH, Nomizo A, Freire-de-Lima CG, Bisaggio RC, Persechini PM. ATP-induced apoptosis involves a Ca2+-independent phospholipase A2 and 5-lipoxygenase in macrophages. Prostaglandins Other Lipid Mediat. 2009;88:51–61. doi: 10.1016/j.prostaglandins.2008.09.004. [DOI] [PubMed] [Google Scholar]
  • 293.Moore SF, MacKenzie AB. Murine macrophage P2X7 receptors support rapid prothrombotic responses. Cell Signal. 2007;19:855–866. doi: 10.1016/j.cellsig.2006.10.010. [DOI] [PubMed] [Google Scholar]
  • 294.Furlan-Freguia C, Marchese P, Gruber A, Ruggeri ZM, Ruf W. P2X7 receptor signaling contributes to tissue factor-dependent thrombosis in mice. J Clin Invest. 2011;121:2932–2944. doi: 10.1172/JCI46129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Gu BJ, Duce JA, Valova VA, Wong B, Bush AI, Petrou S, Wiley JS. P2X7 receptor-mediated scavenger activity of mononuclear phagocytes toward non-opsonized particles and apoptotic cells is inhibited by serum glycoproteins but remains active in cerebrospinal fluid. J Biol Chem. 2012;287:17318–17330. doi: 10.1074/jbc.M112.340885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296.Gu BJ, Baird PN, Vessey KA, Skarratt KK, Fletcher EL, Fuller SJ, Richardson AJ, Guymer RH, Wiley JS. A rare functional haplotype of the P2RX4 and P2RX7 genes leads to loss of innate phagocytosis and confers increased risk of age-related macular degeneration. FASEB J. 2013;27:1479–1487. doi: 10.1096/fj.12-215368. [DOI] [PubMed] [Google Scholar]
  • 297.Di Virgilio F, Falzoni S, Chiozzi P, Sanz JM, Ferrari D, Buell GN. ATP receptors and giant cell formation. J Leukoc Biol. 1999;66:723–726. doi: 10.1002/jlb.66.5.723. [DOI] [PubMed] [Google Scholar]
  • 298.Chiozzi P, Sanz JM, Ferrari D, Falzoni S, Aleotti A, Buell GN, Collo G, Di Virgilio F. Spontaneous cell fusion in macrophage cultures expressing high levels of the P2Z/P2X7 receptor. J Cell Biol. 1997;138:697–706. doi: 10.1083/jcb.138.3.697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299.Lemaire I, Falzoni S, Leduc N, Zhang B, Pellegatti P, Adinolfi E, Chiozzi P, Di Virgilio F. Involvement of the purinergic P2X7 receptor in the formation of multinucleated giant cells. J Immunol. 2006;177:7257–7265. doi: 10.4049/jimmunol.177.10.7257. [DOI] [PubMed] [Google Scholar]
  • 300.Lemaire I, Falzoni S, Zhang B, Pellegatti P, Di Virgilio F. The P2X7 receptor and Pannexin-1 are both required for the promotion of multinucleated macrophages by the inflammatory cytokine GM-CSF. J Immunol. 2011;187:3878–3887. doi: 10.4049/jimmunol.1002780. [DOI] [PubMed] [Google Scholar]
  • 301.Coutinho-Silva R, Ojcius DC, Gorecki DC, Persechini PM, Bisaggio RC, Dunn PM, Burnstock G. Multiple P2X and P2Y receptor subtypes in mouse J774, spleen and peritoneal macrophages. Biochem Pharmacol. 2005;69:641–655. doi: 10.1016/j.bcp.2004.11.012. [DOI] [PubMed] [Google Scholar]
  • 302.Myrtek D, Müller T, Geyer V, Derr N, Ferrari D, Zissel G, Dürk T, Sorichter S, Luttmann W, Kuepper M, Norgauer J, Di Virgilio F, Virchow JC, Jr, Idzko M. Activation of human alveolar macrophages via P2 receptors: coupling to intracellular Ca2+ increases and cytokine secretion. J Immunol. 2008;181:2181–2188. doi: 10.4049/jimmunol.181.3.2181. [DOI] [PubMed] [Google Scholar]
  • 303.Eschke D, Wüst M, Hauschildt S, Nieber K. Pharmacological characterization of the P2X7 receptor on human macrophages using the patch-clamp technique. Naunyn Schmiedebergs Arch Pharmacol. 2002;365:168–171. doi: 10.1007/s00210-001-0501-2. [DOI] [PubMed] [Google Scholar]
  • 304.Brône B, Moechars D, Marrannes R, Mercken M, Meert T. P2X currents in peritoneal macrophages of wild type and P2X4−/− mice. Immunol Lett. 2007;113:83–89. doi: 10.1016/j.imlet.2007.07.015. [DOI] [PubMed] [Google Scholar]
  • 305.Kawano A, Tsukimoto M, Mori D, Noguchi T, Harada H, Takenouchi T, Kitani H, Kojima S. Regulation of P2X7-dependent inflammatory functions by P2X4 receptor in mouse macrophages. Biochem Biophys Res Commun. 2012;420:102–107. doi: 10.1016/j.bbrc.2012.02.122. [DOI] [PubMed] [Google Scholar]
  • 306.Kawano A, Tsukimoto M, Noguchi T, Hotta N, Harada H, Takenouchi T, Kitani H, Kojima S. Involvement of P2X4 receptor in P2X7 receptor-dependent cell death of mouse macrophages. Biochem Biophys Res Commun. 2012;419:374–380. doi: 10.1016/j.bbrc.2012.01.156. [DOI] [PubMed] [Google Scholar]
  • 307.Hazleton JE, Berman JW, Eugenin EA. Purinergic receptors are required for HIV-1 infection of primary human macrophages. J Immunol. 2012;188:4488–4495. doi: 10.4049/jimmunol.1102482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308.Hanley PJ, Musset B, Renigunta V, Limberg SH, Dalpke AH, Sus R, Heeg KM, Preisig-Müller R, Daut J. Extracellular ATP induces oscillations of intracellular Ca2+ and membrane potential and promotes transcription of IL-6 in macrophages. Proc Natl Acad Sci U S A. 2004;101:9479–9484. doi: 10.1073/pnas.0400733101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309.del Rey A, Renigunta V, Dalpke AH, Leipziger J, Matos JE, Robaye B, Zuzarte M, Kavelaars A, Hanley PJ. Knock-out mice reveal the contributions of P2Y and P2X receptors to nucleotide-induced Ca2+ signaling in macrophages. J Biol Chem. 2006;281:35147–35155. doi: 10.1074/jbc.M607713200. [DOI] [PubMed] [Google Scholar]
  • 310.Qu Y, Misaghi S, Newton K, Gilmour LL, Louie S, Cupp JE, Dubyak GR, Hackos D, Dixit VM. Pannexin-1 is required for ATP release during apoptosis but not for inflammasome activation. J Immunol. 2011;186:6553–6561. doi: 10.4049/jimmunol.1100478. [DOI] [PubMed] [Google Scholar]
  • 311.Kronlage M, Song J, Sorokin L, Isfort K, Schwerdtle T, Leipziger J, Robaye B, Conley PB, Kim HC, Sargin S, Schön P, Schwab A, Hanley PJ. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci Signal. 2010;3:ra55. doi: 10.1126/scisignal.2000588. [DOI] [PubMed] [Google Scholar]
  • 312.Chen BC, Chou CF, Lin WW. Pyrimidinoceptor-mediated potentiation of inducible nitric-oxide synthase induction in J774 macrophages. Role of intracellular calcium. J Biol Chem. 1998;273:29754–29763. doi: 10.1074/jbc.273.45.29754. [DOI] [PubMed] [Google Scholar]
  • 313.Chen BC, Lin WW. Pyrimidinoceptor potentiation of macrophage PGE2 release involved in the induction of nitric oxide synthase. Br J Pharmacol. 2000;130:777–786. doi: 10.1038/sj.bjp.0703375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314.Stokes L, Surprenant A. Purinergic P2Y2 receptors induce increased MCP-1/CCL2 synthesis and release from rat alveolar and peritoneal macrophages. J Immunol. 2007;179:6016–6023. doi: 10.4049/jimmunol.179.9.6016. [DOI] [PubMed] [Google Scholar]
  • 315.Eun SY, Seo J, Park SW, Lee JH, Chang KC, Kim HJ. LPS potentiates nucleotide-induced inflammatory gene expression in macrophages via the upregulation of P2Y2 receptor. Int Immunopharmacol. 2014;18:270–276. doi: 10.1016/j.intimp.2013.11.026. [DOI] [PubMed] [Google Scholar]
  • 316.Bar I, Guns PJ, Metallo J, Cammarata D, Wilkin F, Boeynams JM, Bult H, Robaye B. Knockout mice reveal a role for P2Y6 receptor in macrophages, endothelial cells, and vascular smooth muscle cells. Mol Pharmacol. 2008;74:777–784. doi: 10.1124/mol.108.046904. [DOI] [PubMed] [Google Scholar]
  • 317.Zhang Z, Wang Z, Ren H, Yue M, Huang K, Gu H, Liu M, Du B, Qian M. P2Y6 agonist uridine 5'-diphosphate promotes host defense against bacterial infection via monocyte chemoattractant protein-1-mediated monocytes/macrophages recruitment. J Immunol. 2011;186:5376–5387. doi: 10.4049/jimmunol.1002946. [DOI] [PubMed] [Google Scholar]
  • 318.Sakaki H, Tsukimoto M, Harada H, Moriyama Y, Kojima S. Autocrine regulation of macrophage activation via exocytosis of ATP and activation of P2Y11 receptor. PLoS One. 2013;8:e59778. doi: 10.1371/journal.pone.0059778. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 319.Parkinson FE, Paterson AR, Young JD, Cass CE. Inhibitory effects of propentofylline on [3H]adenosine influx. A study of three nucleoside transport systems. Biochem Pharmacol. 1993;46:891–896. doi: 10.1016/0006-2952(93)90499-m. [DOI] [PubMed] [Google Scholar]
  • 320.Banati RB, Schubert P, Rothe G, Gehrmann J, Rudolphi K, Valet G, Kreutzberg GW. Modulation of intracellular formation of reactive oxygen intermediates in peritoneal macrophages and microglia/brain macrophages by propentofylline. J Cereb Blood Flow Metab. 1994;14:145–149. doi: 10.1038/jcbfm.1994.19. [DOI] [PubMed] [Google Scholar]
  • 321.Schubert P, Rudolphi KA, Fredholm BB, Nakamura Y. Modulation of nerve and glial function by adenosine - role in the development of ischemic damage. Int J Biochem. 1994;26:1227–1236. doi: 10.1016/0020-711x(94)90092-2. [DOI] [PubMed] [Google Scholar]
  • 322.McRae A, Rudolphi KA, Schubert P. Propentosylline depresses amyloid and Alzheimer's CSF microglial antigens after ischaemia. Neuroreport. 1994;5:1193–1196. doi: 10.1097/00001756-199406020-00008. [DOI] [PubMed] [Google Scholar]
  • 323.McRae A, Ling EA, Schubert P, Rudolphi K. Properties of activated microglia and pharmacologic interference by propentofylline. Alzheimer Dis Assoc Disord. 1998;12(Suppl 2):S15–S20. [PubMed] [Google Scholar]
  • 324.Schubert P, Ogata T, Rudolphi K, Marchini C, McRae A, Ferroni S. Support of homeostatic glial cell signaling: a novel therapeutic approach by propentofylline. Ann N Y Acad Sci. 1997;826:337–347. doi: 10.1111/j.1749-6632.1997.tb48484.x. [DOI] [PubMed] [Google Scholar]
  • 325.Tsutsui S, Schnermann J, Noorbakhsh F, Henry S, Yong VW, Winston BW, Warren K, Power C. A1 adenosine receptor upregulation and activation attenuates neuroinflammation and demyelination in a model of multiple sclerosis. J Neurosci. 2004;24:1521–1529. doi: 10.1523/JNEUROSCI.4271-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 326.Färber K, Markworth S, Pannasch U, Nolte C, Prinz V, Kronenberg G, Gertz K, Endres M, Bechmann I, Enjyoji K, Robson SC, Kettenmann H. The ectonucleotidase cd39/ENTPDase1 modulates purinergic-mediated microglial migration. Glia. 2008;56:331–341. doi: 10.1002/glia.20606. [DOI] [PubMed] [Google Scholar]
  • 327.Ohsawa K, Sanagi T, Nakamura Y, Suzuki E, Inoue K, Kohsaka S. Adenosine A3 receptor is involved in ADP-induced microglial process extension and migration. J Neurochem. 2012;121:217–227. doi: 10.1111/j.1471-4159.2012.07693.x. [DOI] [PubMed] [Google Scholar]
  • 328.Orr AG, Orr AL, Li XJ, Gross RE, Traynelis SF. Adenosine A2A receptor mediates microglial process retraction. Nat Neurosci. 2009;12:872–878. doi: 10.1038/nn.2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 329.Yao SQ, Li ZZ, Huang QY, Li F, Wang ZW, Augusto E, He JC, Wang XT, Chen JF, Zheng RY. Genetic inactivation of the adenosine A2A receptor exacerbates brain damage in mice with experimental autoimmune encephalomyelitis. J Neurochem. 2012;123:100–112. doi: 10.1111/j.1471-4159.2012.07807.x. [DOI] [PubMed] [Google Scholar]
  • 330.Kettenmann H, Banati R, Walz W. Electrophysiological behavior of microglia. Glia. 1993;7:93–101. doi: 10.1002/glia.440070115. [DOI] [PubMed] [Google Scholar]
  • 331.Walz W, Ilschner S, Ohlemeyer C, Banati R, Kettenmann H. Extracellular ATP activates a cation conductance and a K+ conductance in cultured microglial cells from mouse brain. J Neurosci. 1993;13:4403–4411. doi: 10.1523/JNEUROSCI.13-10-04403.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 332.Langosch JM, Gebicke-Haerter PJ, Nörenberg W, Illes P. Characterization and transduction mechanisms of purinoceptors in activated rat microglia. Br J Pharmacol. 1994;113:29–34. doi: 10.1111/j.1476-5381.1994.tb16169.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 333.Nörenberg W, Langosch JM, Gebicke-Haerter PJ, Illes P. Characterization and possible function of adenosine 5'-triphosphate receptors in activated rat microglia. Br J Pharmacol. 1994;111:942–950. doi: 10.1111/j.1476-5381.1994.tb14830.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 334.Ferrari D, Villalba M, Chiozzi P, Falzoni S, Ricciardi-Castagnoli P, Di Virgilio F. Mouse microglial cells express a plasma membrane pore gated by extracellular ATP. J Immunol. 1996;156:1531–1539. [PubMed] [Google Scholar]
  • 335.Ferrari D, Chiozzi P, Falzoni S, Hanau S, Di Virgilio F. Purinergic modulation of interleukin-1β release from microglial cells stimulated with bacterial endotoxin. J Exp Med. 1997;185:579–582. doi: 10.1084/jem.185.3.579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Brough D, Le Feuvre RA, Iwakura Y, Rothwell NJ. Purinergic (P2X7) receptor activation of microglia induces cell death via an interleukin-1-independent mechanism. Mol Cell Neurosci. 2002;19:272–280. doi: 10.1006/mcne.2001.1054. [DOI] [PubMed] [Google Scholar]
  • 337.Skaper SD, Facci L, Culbert AA, Evans NA, Chessell I, Davis JB, Richardson JC. P2X7 receptors on microglial cells mediate injury to cortical neurons in vitro. Glia. 2006;54:234–242. doi: 10.1002/glia.20379. [DOI] [PubMed] [Google Scholar]
  • 338.Sanz JM, Chiozzi P, Ferrari D, Colaianna M, Idzko M, Falzoni S, Fellin R, Trabace L, Di Virgilio F. Activation of microglia by amyloid β requires P2X7 receptor expression. J Immunol. 2009;182:4378–4385. doi: 10.4049/jimmunol.0803612. [DOI] [PubMed] [Google Scholar]
  • 339.Tsuda M, Shigemoto-Mogami Y, Koizumi S, Mizokoshi A, Kohsaka S, Salter MW, Inoue K. P2X4 receptors induced in spinal microglia gate tactile allodynia after nerve injury. Nature. 2003;424:778–783. doi: 10.1038/nature01786. [DOI] [PubMed] [Google Scholar]
  • 340.Ohsawa K, Irino Y, Nakamura Y, Akazawa C, Inoue K, Kohsaka S. Involvement of P2X4 and P2Y12 receptors in ATP-induced microglial chemotaxis. Glia. 2007;55:604–616. doi: 10.1002/glia.20489. [DOI] [PubMed] [Google Scholar]
  • 341.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9:1512–1519. doi: 10.1038/nn1805. [DOI] [PubMed] [Google Scholar]
  • 342.Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi BV, Jacobson KA, Kohsaka S, Inoue K. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature. 2007;446:1091–1095. doi: 10.1038/nature05704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 343.Inoue K, Koizumi S, Kataoka A, Tozaki-Saitoh H, Tsuda M. P2Y6-evoked microglial phagocytosis. Int Rev Neurobiol. 2009;85:159–163. doi: 10.1016/S0074-7742(09)85012-5. [DOI] [PubMed] [Google Scholar]
  • 344.Koizumi S, Ohsawa K, Inoue K, Kohsaka S. Purinergic receptors in microglia: functional modal shifts of microglia mediated by P2 and P1 receptors. Glia. 2013;61:47–54. doi: 10.1002/glia.22358. [DOI] [PubMed] [Google Scholar]
  • 345.Berchtold S, Ogilvie AL, Bogdan C, Mühl-Zürbes P, Ogilvie A, Schuler G, Steinkasserer A. Human monocyte derived dendritic cells express functional P2X and P2Y receptors as well as ecto-nucleotidases. FEBS Lett. 1999;458:424–428. doi: 10.1016/s0014-5793(99)01197-7. [DOI] [PubMed] [Google Scholar]
  • 346.Panther E, Idzko M, Herouy Y, Rheinen H, Gebicke-Haerter PJ, Mrowietz U, Dichmann S, Norgauer J. Expression and function of adenosine receptors in human dendritic cells. FASEB J. 2001;15:1963–1970. doi: 10.1096/fj.01-0169com. [DOI] [PubMed] [Google Scholar]
  • 347.Panther E, Corinti S, Idzko M, Herouy Y, Napp M, la SA, Girolomoni G, Norgauer J. Adenosine affects expression of membrane molecules, cytokine and chemokine release, and the T-cell stimulatory capacity of human dendritic cells. Blood. 2003;101:3985–3990. doi: 10.1182/blood-2002-07-2113. [DOI] [PubMed] [Google Scholar]
  • 348.Challier J, Bruniquel D, Sewell AK, Laugel B. Adenosine and cAMP signalling skew human dendritic cell differentiation towards a tolerogenic phenotype with defective CD8+ T-cell priming capacity. Immunology. 2013;138:402–410. doi: 10.1111/imm.12053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 349.Ben Addi A, Lefort A, Hua X, Libert F, Communi D, Ledent C, Macours P, Tilley SL, Boeynaems JM, Robaye B. Modulation of murine dendritic cell function by adenine nucleotides and adenosine: involvement of the A2B receptor. Eur J Immunol. 2008;38:1610–1620. doi: 10.1002/eji.200737781. [DOI] [PubMed] [Google Scholar]
  • 350.Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY, Blackburn MR, Biaggioni I, Carbone DP, Feoktistov I, Dikov MM. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood. 2008;112:1822–1831. doi: 10.1182/blood-2008-02-136325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 351.Wilson JM, Ross WG, Agbai ON, Frazier R, Figler RA, Rieger J, Linden J, Ernst PB. The A2B adenosine receptor impairs the maturation and immunogenicity of dendritic cells. J Immunol. 2009;182:4616–4623. doi: 10.4049/jimmunol.0801279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 352.Mascanfroni ID, Yeste A, Vieira SM, Burns EJ, Patel B, Sloma I, Wu Y, Mayo L, Ben-Hamo R, Efroni S, Kuchroo VK, Robson SC, Quintana FJ. IL-27 acts on DCs to suppress the T cell response and autoimmunity by inducing expression of the immunoregulatory molecule CD39. Nat Immunol. 2013;14:1054–1063. doi: 10.1038/ni.2695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 353.Wilson JM, Kurtz CC, Black SG, Ross WG, Alam MS, Linden J, Ernst PB. The A2B adenosine receptor promotes Th17 differentiation via stimulation of dendritic cell IL-6. J Immunol. 2011;186:6746–6752. doi: 10.4049/jimmunol.1100117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Wei W, Du C, Lv J, Zhao G, Li Z, Wu Z, Haskó G, Xie X. Blocking A2B adenosine receptor alleviates pathogenesis of experimental autoimmune encephalomyelitis via inhibition of IL-6 production and Th17 differentiation. J Immunol. 2013;190:138–146. doi: 10.4049/jimmunol.1103721. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 355.Schnurr M, Toy T, Shin A, Hartmann G, Rothenfusser S, Soellner J, Davis ID, Cebon J, Maraskovsky E. Role of adenosine receptors in regulating chemotaxis and cytokine production of plasmacytoid dendritic cells. Blood. 2004;103:1391–1397. doi: 10.1182/blood-2003-06-1959. [DOI] [PubMed] [Google Scholar]
  • 356.Desrosiers MD, Cembrola KM, Fakir MJ, Stephens LA, Jama FM, Shameli A, Mehal WZ, Santamaria P, Shi Y. Adenosine deamination sustains dendritic cell activation in inflammation. J Immunol. 2007;179:1884–1892. doi: 10.4049/jimmunol.179.3.1884. [DOI] [PubMed] [Google Scholar]
  • 357.Ghaemi Oskouie F, Shameli A, Yang A, Desrosiers MD, Mucsi AD, Blackburn MR, Yang Y, Santamaria P, Shi Y. High levels of adenosine deaminase on dendritic cells promote autoreactive T cell activation and diabetes in nonobese diabetic mice. J Immunol. 2011;186:6798–6806. doi: 10.4049/jimmunol.1004222. [DOI] [PubMed] [Google Scholar]
  • 358.Mizumoto N, Kumamoto T, Robson SC, Sévigny J, Matsue H, Enjyoji K, Takashima A. CD39 is the dominant Langerhans cell-associated ecto-NTPDase: modulatory roles in inflammation and immune responsiveness. Nat Med. 2002;8:358–365. doi: 10.1038/nm0402-358. [DOI] [PubMed] [Google Scholar]
  • 359.Idzko M, Ayata K, Müller T, Dürk T, Grimm M, Baudiss K, Vieira RP, Cicko S, Boehlke C, Zech A, Sorichter S, Pelletier J, Sévigny J, Robson SC. Attenuated allergic airway inflammation in Cd39 null mice. Allergy. 2013;68:472–480. doi: 10.1111/all.12119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 360.Idzko M, Panther E, Bremer HC, Windisch W, Sorichter S, Herouy Y, Elsner P, Mockenhaupt M, Girolomoni G, Norgauer J. Inosine stimulates chemotaxis, Ca2+-transients and actin polymerization in immature human dendritic cells via a pertussis toxin-sensitive mechanism independent of adenosine receptors. J Cell Physiol. 2004;199:149–156. doi: 10.1002/jcp.10431. [DOI] [PubMed] [Google Scholar]
  • 361.Panther E, Dürk T, Ferrari D, Di Virgilio F, Grimm M, Sorichter S, Cicko S, Herouy Y, Norgauer J, Idzko M, Müller T. AMP affects intracellular Ca2+ signaling, migration, cytokine secretion and T cell priming capacity of dendritic cells. PLoS One. 2012;7:e37560. doi: 10.1371/journal.pone.0037560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 362.Liu QH, Bohlen H, Titzer S, Christensen O, Diehl V, Hescheler J, Fleischmann BK. Expression and a role of functionally coupled P2Y receptors in human dendritic cells. FEBS Lett. 1999;445:402–408. doi: 10.1016/s0014-5793(99)00161-1. [DOI] [PubMed] [Google Scholar]
  • 363.Ferrari D, la Sala A, Chiozzi P, Morelli A, Falzoni S, Girolomoni G, Idzko M, Dichmann S, Norgauer J, Di Virgilio F. The P2 purinergic receptors of human dendritic cells: identification and coupling to cytokine release. FASEB J. 2000;14:2466–2476. doi: 10.1096/fj.00-0031com. [DOI] [PubMed] [Google Scholar]
  • 364.Idzko M, Dichmann S, Ferrari D, Di Virgilio F, la Sala A, Girolomoni G, Panther E, Norgauer J. Nucleotides induce chemotaxis and actin polymerization in immature but not mature human dendritic cells via activation of pertussis toxin-sensitive P2y receptors. Blood. 2002;100:925–932. doi: 10.1182/blood.v100.3.925. [DOI] [PubMed] [Google Scholar]
  • 365.Vanderstocken G, Van de Paar E, Robaye B, Di Pietrantonio L, Bondue B, Boeynaems JM, Desmecht D, Communi D. Protective role of P2Y2 receptor against lung infection induced by pneumonia virus of mice. PLoS One. 2012;7:e50385. doi: 10.1371/journal.pone.0050385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Schnurr M, Toy T, Stoitzner P, Cameron P, Shin A, Beecroft T, Davis ID, Cebon J, Maraskovsky E. ATP gradients inhibit the migratory capacity of specific human dendritic cell types: implications for P2Y11 receptor signaling. Blood. 2003;102:613–620. doi: 10.1182/blood-2002-12-3745. [DOI] [PubMed] [Google Scholar]
  • 367.la Sala A, Sebastiani S, Ferrari D, Di Virgilio F, Idzko M, Norgauer J, Girolomoni G. Dendritic cells exposed to extracellular adenosine triphosphate acquire the migratory properties of mature cells and show a reduced capacity to attract type 1 T lymphocytes. Blood. 2002;99:1715–1722. doi: 10.1182/blood.v99.5.1715. [DOI] [PubMed] [Google Scholar]
  • 368.Horckmans M, Marcet B, Marteau F, Bulté F, Maho A, Parmentier M, Boeynaems JM, Communi D. Extracellular adenine nucleotides inhibit the release of major monocyte recruiters by human monocyte-derived dendritic cells. FEBS Lett. 2006;580:747–754. doi: 10.1016/j.febslet.2005.12.091. [DOI] [PubMed] [Google Scholar]
  • 369.Schnurr M, Then F, Galambos P, Scholz C, Siegmund B, Endres S, Eigler A. Extracellular ATP and TNF-α synergize in the activation and maturation of human dendritic cells. J Immunol. 2000;165:4704–4709. doi: 10.4049/jimmunol.165.8.4704. [DOI] [PubMed] [Google Scholar]
  • 370.Wilkin F, Duhant X, Bruyns C, Suarez-Huerta N, Boeynaems JM, Robaye B. The P2Y11 receptor mediates the ATP-induced maturation of human monocyte-derived dendritic cells. J Immunol. 2001;166:7172–7177. doi: 10.4049/jimmunol.166.12.7172. [DOI] [PubMed] [Google Scholar]
  • 371.la Sala A, Ferrari D, Corinti S, Cavani A, Di Virgilio F, Girolomoni G. Extracellular ATP induces a distorted maturation of dendritic cells and inhibits their capacity to initiate Th1 responses. J Immunol. 2001;166:1611–1617. doi: 10.4049/jimmunol.166.3.1611. [DOI] [PubMed] [Google Scholar]
  • 372.Wilkin F, Stordeur P, Goldman M, Boeynaems JM, Robaye B. Extracellular adenine nucleotides modulate cytokine production by human monocyte-derived dendritic cells: dual effect on IL-12 and stimulation of IL-10. Eur J Immunol. 2002;32:2409–2417. doi: 10.1002/1521-4141(200209)32:9<2409::AID-IMMU2409>3.0.CO;2-H. [DOI] [PubMed] [Google Scholar]
  • 373.Marteau F, Gonzalez NS, Communi D, Goldman M, Boeynaems JM, Communi D. Thrombospondin-1 and indoleamine 2,3-dioxygenase are major targets of extracellular ATP in human dendritic cells. Blood. 2005;106:3860–3866. doi: 10.1182/blood-2005-05-1843. [DOI] [PubMed] [Google Scholar]
  • 374.Bles N, Horckmans M, Lefort A, Libert F, Macours P, El Housni H, Marteau F, Boeynaems JM, Communi D. Gene expression profiling defines ATP as a key regulator of human dendritic cell functions. J Immunol. 2007;179:3550–3558. doi: 10.4049/jimmunol.179.6.3550. [DOI] [PubMed] [Google Scholar]
  • 375.Bles N, Di Pietrantonio L, Boeynaems JM, Communi D. ATP confers tumorigenic properties to dendritic cells by inducing amphiregulin secretion. Blood. 2010;116:3219–3226. doi: 10.1182/blood-2010-01-265611. [DOI] [PubMed] [Google Scholar]
  • 376.Marteau F, Communi D, Boeynaems JM, Suarez Gonzalez N. Involvement of multiple P2Y receptors and signaling pathways in the action of adenine nucleotides diphosphates on human monocyte-derived dendritic cells. J Leukoc Biol. 2004;76:796–803. doi: 10.1189/jlb.0104032. [DOI] [PubMed] [Google Scholar]
  • 377.Ben Addi A, Cammarata D, Conley PB, Boeynaems JM, Robaye B. Role of the P2Y12 receptor in the modulation of murine dendritic cell function by ADP. J Immunol. 2010;185:5900–5906. doi: 10.4049/jimmunol.0901799. [DOI] [PubMed] [Google Scholar]
  • 378.Idzko M, Panther E, Sorichter S, Herouy Y, Berod L, Geissler M, Mockenhaupt M, Elsner P, Girolomoni G, Norgauer J. Characterization of the biological activities of uridine diphosphate in human dendritic cells: Influence on chemotaxis and CXCL8 release. J Cell Physiol. 2004;201:286–293. doi: 10.1002/jcp.20070. [DOI] [PubMed] [Google Scholar]
  • 379.Marriott I, Inscho EW, Bost KL. Extracellular uridine nucleotides initiate cytokine production by murine dendritic cells. Cell Immunol. 1999;195:147–156. doi: 10.1006/cimm.1999.1531. [DOI] [PubMed] [Google Scholar]
  • 380.Skelton L, Cooper M, Murphy M, Platt A. Human immature monocyte-derived dendritic cells express the G protein-coupled receptor GPR105 (KIAA0001, P2Y14) and increase intracellular calcium in response to its agonist, uridine diphosphoglucose. J Immunol. 2003;171:1941–1949. doi: 10.4049/jimmunol.171.4.1941. [DOI] [PubMed] [Google Scholar]
  • 381.Shin A, Toy T, Rothenfusser S, Robson N, Vorac J, Dauer M, Stuplich M, Endres S, Cebon J, Maraskovsky E, Schnurr M. P2Y receptor signaling regulates phenotype and IFN-α secretion of human plasmacytoid dendritic cells. Blood. 2008;111:3062–3069. doi: 10.1182/blood-2007-02-071910. [DOI] [PubMed] [Google Scholar]
  • 382.Qu Y, Ramachandra L, Mohr S, Franchi L, Harding CV, Nunez G, Dubyak GR. P2X7 receptor-stimulated secretion of MHC class II-containing exosomes requires the ASC/NLRP3 inflammasome but is independent of caspase-1. J Immunol. 2009;182:5052–5062. doi: 10.4049/jimmunol.0802968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 383.Dubyak GR. P2X7 receptor regulation of non-classical secretion from immune effector cells. Cell Microbiol. 2012;14:1697–1706. doi: 10.1111/cmi.12001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 384.Mutini C, Falzoni S, Ferrari D, Chiozzi P, Morelli A, Baricordi OR, Collo G, Ricciardi-Castagnoli P, Di Virgilio F. Mouse dendritic cells express the P2X7 purinergic receptor: characterization and possible participation in antigen presentation. J Immunol. 1999;163:1958–1965. [PubMed] [Google Scholar]
  • 385.Pizzirani C, Ferrari D, Chiozzi P, Adinolfi E, Sandonà D, Savaglio E, Di Virgilio F. Stimulation of P2 receptors causes release of IL-1β-loaded microvesicles from human dendritic cells. Blood. 2007;109:3856–3864. doi: 10.1182/blood-2005-06-031377. [DOI] [PubMed] [Google Scholar]
  • 386.Weber FC, Esser PR, Müller T, Ganesan J, Pellegatti P, Simon MM, Zeiser R, Idzko M, Jakob T, Martin SF. Lack of the purinergic receptor P2X7 results in resistance to contact hypersensitivity. J Exp Med. 2010;207:2609–2619. doi: 10.1084/jem.20092489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 387.Sluyter R, Wiley JS. Extracellular adenosine 5'-triphosphate induces a loss of CD23 from human dendritic cells via activation of P2X7 receptors. Int Immunol. 2002;14:1415–1421. doi: 10.1093/intimm/dxf111. [DOI] [PubMed] [Google Scholar]
  • 388.Baroni M, Pizzirani C, Pinotti M, Ferrari D, Adinolfi E, Calzavarini S, Caruso P, Bernardi F, Di Virgilio F. Stimulation of P2 (P2X7) receptors in human dendritic cells induces the release of tissue factor-bearing microparticles. FASEB J. 2007;21:1926–1933. doi: 10.1096/fj.06-7238com. [DOI] [PubMed] [Google Scholar]
  • 389.Coutinho-Silva R, Persechini PM, Bisaggio RD, Perfettini JL, Neto AC, Kanellopoulos JM, Motta-Ly I, Dautry-Varsat A, Ojcius DM. P2Z/P2X7 receptor-dependent apoptosis of dendritic cells. Am J Physiol. 1999;276:C1139–C1147. doi: 10.1152/ajpcell.1999.276.5.C1139. [DOI] [PubMed] [Google Scholar]
  • 390.Nihei OK, de Carvalho AC, Savino W, Alves LA. Pharmacologic properties of P2Z/P2X7 receptor characterized in murine dendritic cells: role on the induction of apoptosis. Blood. 2000;96:996–1005. [PubMed] [Google Scholar]
  • 391.Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue M, Yagita H, Ishii N, Evans R, Honda K, Takeda K. ATP drives lamina propria T(H)17 cell differentiation. Nature. 2008;455:808–812. doi: 10.1038/nature07240. [DOI] [PubMed] [Google Scholar]
  • 392.Kusu T, Kayama H, Kinoshita M, Jeon SG, Ueda Y, Goto Y, Okumura R, Saiga H, Kurakawa T, Ikeda K, Maeda Y, Nishimura J, Arima Y, Atarashi K, Honda K, Murakami M, Kunisawa J, Kiyono H, Okumura M, Yamamoto M, Takeda K. Ecto-nucleoside triphosphate diphosphohydrolase 7 controls Th17 cell responses through regulation of luminal ATP in the small intestine. J Immunol. 2013;190:774–783. doi: 10.4049/jimmunol.1103067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 393.Fredholm BB, Sandberg G, Ernström U. Cyclic AMP in freshly prepared thymocyte suspensions. Evidence for stimulation by endogenous adenosine. Biochem Pharmacol. 1978;27:2675–2682. doi: 10.1016/0006-2952(78)90041-2. [DOI] [PubMed] [Google Scholar]
  • 394.Marone G, Plaut M, Lichtenstein LM. Characterization of a specific adenosine receptor on human lymphocytes. J Immunol. 1978;121:2153–2159. [PubMed] [Google Scholar]
  • 395.Schwartz AL, Stern RC, Polmar SH. Demonstration of adenosine receptor on human lymphocytes in vitro and its possible role in the adenosine deaminase-deficient form of severe combined immunodeficiency. Clin Immunol Immunopathol. 1978;9:499–505. doi: 10.1016/0090-1229(78)90146-0. [DOI] [PubMed] [Google Scholar]
  • 396.Fredholm BB, Sandberg G. Inhibition by xanthine derivatives of adenosine receptor-stimulated cyclic adenosine 3',5'-monophosphate accumulation in rat and guinea-pig thymocytes. Br J Pharmacol. 1983;80:639–644. doi: 10.1111/j.1476-5381.1983.tb10053.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 397.Fishman RF, Rubin AL, Novogrodsky A, Stenzel KH. Selective suppression of blastogenesis induced by different mitogens: effect of noncyclic adenosine-containing compounds. Cell Immunol. 1980;54:129–139. doi: 10.1016/0008-8749(80)90196-3. [DOI] [PubMed] [Google Scholar]
  • 398.Bessler H, Djaldetti M, Moroz C. The regulatory role of adenosine-activated T-lymphocyte subset on the immune response in humans. I. Mitogenic response and production of mediators. Cell Immunol. 1982;73:216–229. doi: 10.1016/0008-8749(82)90451-8. [DOI] [PubMed] [Google Scholar]
  • 399.Wolberg G, Zimmerman TP, Hiemstra K, Winston M, Chu LC. Adenosine inhibition of lymphocyte-mediated cytolysis: possible role of cyclic adenosine monophosphate. Science. 1975;187:957–959. doi: 10.1126/science.167434. [DOI] [PubMed] [Google Scholar]
  • 400.Smith GP, Shah T, Webster AD, Peters TJ. Studies on the kinetic properties and subcellular localization of adenosine diphosphatase activity in human peripheral blood lymphocytes. Clin Exp Immunol. 1981;46:321–326. [PMC free article] [PubMed] [Google Scholar]
  • 401.Cuschieri A, Mughal S, Kharbat BA. Ultrastructural localization of adenosine triphosphatase activity in lymphocytes activated in vitro by phytohaemagglutinin. Histochem J. 1982;14:593–607. doi: 10.1007/BF01011892. [DOI] [PubMed] [Google Scholar]
  • 402.Dornand J, Bonnafous JC, Favero J, Gartner A, Mani JC. 5'Nucleotidase and adenosine deaminase activities in human lymphocytes and lymphoblastoid cell lines. Adv Exp Med Biol. 1984;165:261–266. doi: 10.1007/978-1-4757-0390-0_50. [DOI] [PubMed] [Google Scholar]
  • 403.Pechán I, Rendeková V, Niks M, Pechánová E, Krizko J. Adenosine deaminase and 5'-nucleotidase activity of T- and B-lymphocytes of human peripheral blood. Biologia (Bratislava) 1984;39:381–386. [Google Scholar]
  • 404.Barankiewicz J, Dosch HM, Cohen A. Extracellular nucleotide catabolism in human B and T lymphocytes. The source of adenosine production. J Biol Chem. 1988;263:7094–7098. [PubMed] [Google Scholar]
  • 405.Barankiewicz J, Ronlov G, Jimenez R, Gruber HE. Selective adenosine release from human B but not T lymphoid cell line. J Biol Chem. 1990;265:15738–15743. [PubMed] [Google Scholar]
  • 406.Filippini A, Taffs RE, Agui T, Sitkovsky MV. Ecto-ATPase activity in cytolytic T-lymphocyte. s Protection from the cytolytic effects of extracellular ATP. J Biol Chem. 1990;265:334–340. [PubMed] [Google Scholar]
  • 407.Langston HP, Ke Y, Gewirtz AT, Dombrowski KE, Kapp JA. Secretion of IL-2 and IFN-γ, but not IL-4, by antigen-specific T cells requires extracellular ATP. J Immunol. 2003;170:2962–2970. doi: 10.4049/jimmunol.170.6.2962. [DOI] [PubMed] [Google Scholar]
  • 408.Horenstein AL, Chillemi A, Zaccarello G, Bruzzone S, Quarona V, Zito A, Serra S, Malavasi F. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology. 2013;2:e26246. doi: 10.4161/onci.26246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 409.Huang S, Apasov S, Koshiba M, Sitkovsky M. Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood. 1997;90:1600–1610. [PubMed] [Google Scholar]
  • 410.Koshiba M, Kojima H, Huang S, Apasov S, Sitkovsky MV. Memory of extracellular adenosine A2A purinergic receptor-mediated signaling in murine T cells. J Biol Chem. 1997;272:25881–25889. doi: 10.1074/jbc.272.41.25881. [DOI] [PubMed] [Google Scholar]
  • 411.Varani K, Gessi S, Dalpiaz A, Ongini E, Borea PA. Characterization of A2A adenosine receptors in human lymphocyte membranes by [3H]-SCH 58261 binding. Br J Pharmacol. 1997;122:386–392. doi: 10.1038/sj.bjp.0701378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 412.Mirabet M, Herrera C, Cordero OJ, Mallol J, Lluis C, Franco R. Expression of A2B adenosine receptors in human lymphocytes: their role in T cell activation. J Cell Sci. 1999;112:491–502. doi: 10.1242/jcs.112.4.491. [DOI] [PubMed] [Google Scholar]
  • 413.Koshiba M, Rosin DL, Hayashi N, Linden J, Sitkovsky MV. Patterns of A2A extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. Flow cytometry studies with anti-A2A receptor monoclonal antibodies. Mol Pharmacol. 1999;55:614–624. [PubMed] [Google Scholar]
  • 414.Gessi S, Varani K, Merighi S, Cattabriga E, Avitabile A, Gavioli R, Fortini C, Leung E, Mac LS, Borea PA. Expression of A3 adenosine receptors in human lymphocytes: up-regulation in T cell activation. Mol Pharmacol. 2004;65:711–719. doi: 10.1124/mol.65.3.711. [DOI] [PubMed] [Google Scholar]
  • 415.Takahashi HK, Iwagaki H, Hamano R, Kanke T, Liu K, Sadamori H, Yagi T, Yoshino T, Sendo T, Tanaka N, Nishibori M. Effect of adenosine receptor subtypes stimulation on mixed lymphocyte reaction. Eur J Pharmacol. 2007;564:204–210. doi: 10.1016/j.ejphar.2007.02.005. [DOI] [PubMed] [Google Scholar]
  • 416.Yang A, Mucsi AD, Desrosiers MD, Chen JF, Schnermann JB, Blackburn MR, Shi Y. Adenosine mediated desensitization of cAMP signaling enhances T-cell responses. Eur J Immunol. 2010;40:449–459. doi: 10.1002/eji.200939586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 417.Armstrong JM, Chen JF, Schwarzschild MA, Apasov S, Smith PT, Caldwell C, Chen P, Figler H, Sullivan G, Fink S, Linden J, Sitkovsky M. Gene dose effect reveals no Gs-coupled A2A adenosine receptor reserve in murine T-lymphocytes: studies of cells from A2A-receptor-gene-deficient mice. Biochem J. 2001;354:123–130. doi: 10.1042/0264-6021:3540123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 418.Lappas CM, Rieger JM, Linden J. A2A adenosine receptor induction inhibits IFN-γ production in murine CD4+ T cells. J Immunol. 2005;174:1073–1080. doi: 10.4049/jimmunol.174.2.1073. [DOI] [PubMed] [Google Scholar]
  • 419.Csóka B, Himer L, Selmeczy Z, Vizi ES, Pacher P, Ledent C, Deitch EA, Spolarics Z, Németh ZH, Haskó G. Adenosine A2A receptor activation inhibits T helper 1 and T helper 2 cell development and effector function. FASEB J. 2008;22:3491–3499. doi: 10.1096/fj.08-107458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 420.Apasov SG, Chen JF, Smith PT, Schwarzschild MA, Fink JS, Sitkovsky MV. Study of A2A adenosine receptor gene deficient mice reveals that adenosine analogue CGS 21680 possesses no A2A receptor-unrelated lymphotoxicity. Br J Pharmacol. 2000;131:43–50. doi: 10.1038/sj.bjp.0703532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 421.Himer L, Csóka B, Selmeczy Z, Koscsó B, Pócza T, Pacher P, Németh ZH, Deitch EA, Vizi ES, Cronstein BN, Haskó G. Adenosine A2A receptor activation protects CD4+ T lymphocytes against activation-induced cell death. FASEB J. 2010;24:2631–2640. doi: 10.1096/fj.10-155192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 422.Cekic C, Sag D, Day YJ, Linden J. Extracellular adenosine regulates naive T cell development and peripheral maintenance. J Exp Med. 2013;210:2693–2706. doi: 10.1084/jem.20130249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 423.Chimote AA, Hajdu P, Kucher V, Boiko N, Kuras Z, Szilagyi O, Yun YH, Conforti L. Selective inhibition of KCa3.1 channels mediates adenosine regulation of the motility of human T cells. J Immunol. 2013;191:6273–6280. doi: 10.4049/jimmunol.1300702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 424.Takedachi M, Qu D, Ebisuno Y, Oohara H, Joachims ML, McGee ST, Maeda E, McEver RP, Tanaka T, Miyasaka M, Murakami S, Krahn T, Blackburn MR, Thompson LF. CD73-generated adenosine restricts lymphocyte migration into draining lymph nodes. J Immunol. 2008;180:6288–6296. doi: 10.4049/jimmunol.180.9.6288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 425.Ring S, Oliver SJ, Cronstein BN, Enk AH, Mahnke K. CD4+CD25+ regulatory T cells suppress contact hypersensitivity reactions through a CD39, adenosine-dependent mechanism. J Allergy Clin Immunol. 2009;123:1287–1296. doi: 10.1016/j.jaci.2009.03.022. [DOI] [PubMed] [Google Scholar]
  • 426.Antonioli L, Pacher P, Vizi ES, Haskó G. CD39 and CD73 in immunity and inflammation. Trends Mol Med. 2013;19:355–367. doi: 10.1016/j.molmed.2013.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 427.Ernst PB, Garrison JC, Thompson LF. Much ado about adenosine: adenosine synthesis and function in regulatory T cell biology. J Immunol. 2010;185:1993–1998. doi: 10.4049/jimmunol.1000108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 428.Whiteside TL, Mandapathil M, Schuler P. The role of the adenosinergic pathway in immunosuppression mediated by human regulatory T cells (Treg) Curr Med Chem. 2011;18:5217–5223. doi: 10.2174/092986711798184334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 429.Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, Giometto R, Hopner S, Centonze D, Bernardi G, Dell'Acqua ML, Rossini PM, Battistini L, Rotzschke O, Falk K. Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood. 2007;110:1225–1232. doi: 10.1182/blood-2006-12-064527. [DOI] [PubMed] [Google Scholar]
  • 430.Kobie JJ, Shah PR, Yang L, Rebhahn JA, Fowell DJ, Mosmann TR. T regulatory and primed uncommitted CD4 T cells express CD73, which suppresses effector CD4 T cells by converting 5'-adenosine monophosphate to adenosine. J Immunol. 2006;177:6780–6786. doi: 10.4049/jimmunol.177.10.6780. [DOI] [PubMed] [Google Scholar]
  • 431.Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, Chen JF, Enjyoji K, Linden J, Oukka M, Kuchroo VK, Strom TB, Robson SC. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med. 2007;204:1257–1265. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 432.Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, Lang S, Jackson EK, Gorelik E, Whiteside TL. Generation and accumulation of immunosuppressive adenosine by human CD4 + CD25highFOXP3+ regulatory T cells. J Biol Chem. 2010;285:7176–7186. doi: 10.1074/jbc.M109.047423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 433.Dwyer KM, Hanidziar D, Putheti P, Hill PA, Pommey S, McRae JL, Winterhalter A, Doherty G, Deaglio S, Koulmanda M, Gao W, Robson SC, Strom TB. Expression of CD39 by human peripheral blood CD4+ CD25+ T cells denotes a regulatory memory phenotype. Am J Transplant. 2010;10:2410–2420. doi: 10.1111/j.1600-6143.2010.03291.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 434.Mandler R, Birch RE, Polmar SH, Kammer GM, Rudolph SA. Abnormal adenosine-induced immunosuppression and cAMP metabolism in T lymphocytes of patients with systemic lupus erythematosus. Proc Natl Acad Sci U S A. 1982;79:7542–7546. doi: 10.1073/pnas.79.23.7542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 435.Schultz LA, Kammer GM, Rudolph SA. Characterization of the human T lymphocyte adenosine receptor: comparison of normal and systemic lupus erythematosus cells. FASEB J. 1988;2:244–250. doi: 10.1096/fasebj.2.3.3258258. [DOI] [PubMed] [Google Scholar]
  • 436.Yang Z, Day YJ, Toufektsian MC, Ramos SI, Marshall M, Wang XQ, French BA, Linden J. Infarct-sparing effect of A2A -adenosine receptor activation is due primarily to its action on lymphocytes. Circulation. 2005;111:2190–2197. doi: 10.1161/01.CIR.0000163586.62253.A5. [DOI] [PubMed] [Google Scholar]
  • 437.Yang Z, Day YJ, Toufektsian MC, Xu Y, Ramos SI, Marshall MA, French BA, Linden J. Myocardial infarct-sparing effect of adenosine A2A receptor activation is due to its action on CD4+ T lymphocytes. Circulation. 2006;114:2056–2064. doi: 10.1161/CIRCULATIONAHA.106.649244. [DOI] [PubMed] [Google Scholar]
  • 438.Sevigny CP, Li L, Awad AS, Huang L, McDuffie M, Linden J, Lobo PI, Okusa MD. Activation of adenosine 2A receptors attenuates allograft rejection and alloantigen recognition. J Immunol. 2007;178:4240–4249. doi: 10.4049/jimmunol.178.7.4240. [DOI] [PubMed] [Google Scholar]
  • 439.Han KL, Thomas SV, Koontz SM, Changpriroa CM, Ha SK, Malech HL, Kang EM. Adenosine A2A receptor agonist-mediated increase in donor-derived regulatory T cells suppresses development of graft-versus-host disease. J Immunol. 2013;190:458–468. doi: 10.4049/jimmunol.1201325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 440.Wang L, Fan J, Chen S, Zhang Y, Curiel TJ, Zhang B. Graft-versus-host disease is enhanced by selective CD73 blockade in mice. PLoS One. 2013;8:e58397. doi: 10.1371/journal.pone.0058397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 441.Li N, Mu L, Wang J, Zhang J, Xie X, Kong Q, Tang W, Yao X, Liu Y, Wang L, Wang G, Wang D, Jin L, Sun B, Li H. Activation of the adenosine A2A receptor attenuates experimental autoimmune myasthenia gravis severity. Eur J Immunol. 2012;42:1140–1151. doi: 10.1002/eji.201142088. [DOI] [PubMed] [Google Scholar]
  • 442.Alam MS, Kurtz CC, Rowlett RM, Reuter BK, Wiznerowicz E, Das S, Linden J, Crowe SE, Ernst PB. CD73 is expressed by human regulatory T helper cells and suppresses proinflammatory cytokine production and Helicobacter felis-induced gastritis in mice. J Infect Dis. 2009;199:494–504. doi: 10.1086/596205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 443.Németh ZH, Csóka B, Wilmanski J, Xu D, Lu Q, Ledent C, Deitch EA, Pacher P, Spolarics Z, Haskó G. Adenosine A2A receptor inactivation increases survival in polymicrobial sepsis. J Immunol. 2006;176:5616–5626. doi: 10.4049/jimmunol.176.9.5616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 444.Häusler SF, Montalbán del Barrio I, Strohschein J, Anoop CP, Engel JB, Hönig A, Ossadnik M, Horn E, Fischer B, Krockenberger M, Heuer S, Seida AA, Junker M, Kneitz H, Kloor D, Klotz KN, Dietl J, Wischhusen J. Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine-generating enzymes responsible for adenosine receptor 2A-dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunol Immunother. 2011;60:1405–1418. doi: 10.1007/s00262-011-1040-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 445.Nikolova M, Carriere M, Lelievre J, Muhtarova M, Bensussan A, Lévy Y. Regulatory T cells inhibit CD8 T cell proliferation in HIV-1 infection through CD39/adenosine pathway. Retrovirology. 2009;6:O20. [Google Scholar]
  • 446.Nikolova M, Carriere M, Jenabian MA, Limou S, Younas M, Kök A, Huë S, Seddiki N, Hulin A, Delaneau O, Schuitemaker H, Herbeck JT, Mullins JI, Muhtarova M, Bensussan A, Zagury JF, Lelievre JD, Lévy Y. CD39/adenosine pathway is involved in AIDS progression. PLoS Pathog. 2011;7:e1002110. doi: 10.1371/journal.ppat.1002110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 447.Sakowicz-Burkiewicz M, Kocbuch K, Grden M, Maciejewska I, Szutowicz A, Pawelczyk T. Impact of adenosine receptors on immunoglobulin production by human peripheral blood B lymphocytes. J Physiol Pharmacol. 2012;63:661–668. [PubMed] [Google Scholar]
  • 448.Saze Z, Schuler PJ, Hong CS, Cheng D, Jackson EK, Whiteside TL. Adenosine production by human B cells and B cell-mediated suppression of activated T cells. Blood. 2013;122:9–18. doi: 10.1182/blood-2013-02-482406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 449.Wilkinson JH, Robinson JM. Effect of ATP on release of intracellular enzymes from damaged cells. Nature. 1974;249:662–663. doi: 10.1038/249662a0. [DOI] [PubMed] [Google Scholar]
  • 450.Hallak GJ, Wilkinson JH. Action of adenosine phosphates on the release of intracellular lactate dehydrogenase from human and rat lymphocytes. Enzyme. 1977;22:361–369. doi: 10.1159/000458820. [DOI] [PubMed] [Google Scholar]
  • 451.Wolberg G, Zimmerman TP, Duncan GS, Singer KH, Elion GB. Inhibition of lymphocyte-mediated cytolysis by adenosine analogs. Biochemical studies concerning mechanism of action. Biochem Pharmacol. 1978;27:1487–1495. doi: 10.1016/0006-2952(78)90105-3. [DOI] [PubMed] [Google Scholar]
  • 452.Ikehara S, Pahwa RN, Lunzer DG, Good RA, Modak MJ. Adenosine 5'-triphosphate- (ATP) mediated stimulation and suppression of DNA synthesis in lymphoid cells. I. Characterization of ATP responsive cells in mouse lymphoid organs. J Immunol. 1981;127:1834–1838. [PubMed] [Google Scholar]
  • 453.Gregory SH, Kern M. Mitogenic response of T-cell subclasses to agarose-linked and to free ribonucleotides. Immunology. 1981;42:451–457. [PMC free article] [PubMed] [Google Scholar]
  • 454.Di Virgilio F, Bronte V, Collavo D, Zanovello P. Responses of mouse lymphocytes to extracellular adenosine 5'-triphosphate (ATP). Lymphocytes with cytotoxic activity are resistant to the permeabilizing effects of ATP. J Immunol. 1989;143:1955–1960. [PubMed] [Google Scholar]
  • 455.Zheng LM, Zychlinsky A, Liu CC, Ojcius DM, Young JD. Extracellular ATP as a trigger for apoptosis or programmed cell death. J Cell Biol. 1991;112:279–288. doi: 10.1083/jcb.112.2.279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 456.Lin J, Krishnaraj R, Kemp RG. Exogenous ATP enhances calcium influx in intact thymocytes. J Immunol. 1985;135:3403–3410. [PubMed] [Google Scholar]
  • 457.el-Moatassim C, Dornand J, Mani JC. Extracellular ATP increases cytosolic free calcium in thymocytes and initiates the blastogenesis of the phorbol 12-myristate 13-acetate-treated medullary population. Biochim Biophys Acta. 1987;927:437–444. doi: 10.1016/0167-4889(87)90110-8. [DOI] [PubMed] [Google Scholar]
  • 458.Ross PE, Ehring GR, Cahalan MD. Dynamics of ATP-induced calcium signaling in single mouse thymocytes. J Cell Biol. 1997;138:987–998. doi: 10.1083/jcb.138.5.987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 459.Padeh S, Cohen A, Roifman CM. ATP-induced activation of human B lymphocytes via P2-purinoceptors. J Immunol. 1991;146:1626–1632. [PubMed] [Google Scholar]
  • 460.Wiley JS, Chen R, Wiley MJ, Jamieson GP. The ATP4− receptor-operated ion channel of human lymphocytes: inhibition of ion fluxes by amiloride analogs and by extracellular sodium ions. Arch Biochem Biophys. 1992;292:411–418. doi: 10.1016/0003-9861(92)90010-t. [DOI] [PubMed] [Google Scholar]
  • 461.Wiley JS, Chen R, Jamieson GP. The ATP4− receptor-operated channel (P2Z class) of human lymphocytes allows Ba2+ and ethidium+ uptake: inhibition of fluxes by suramin. Arch Biochem Biophys. 1993;305:54–60. doi: 10.1006/abbi.1993.1392. [DOI] [PubMed] [Google Scholar]
  • 462.Ferrari D, Munerati M, Melchiorri L, Hanau S, Di Virgilio F, Baricordi OR. Responses to extracellular ATP of lymphoblastoid cell lines from Duchenne muscular dystrophy patients. Am J Physiol. 1994;267:C886–C892. doi: 10.1152/ajpcell.1994.267.4.C886. [DOI] [PubMed] [Google Scholar]
  • 463.Wiley JS, Chen JR, Snook MB, Jamieson GP. The P2Z-purinoceptor of human lymphocytes: actions of nucleotide agonists and irreversible inhibition by oxidized ATP. Br J Pharmacol. 1994;112:946–950. doi: 10.1111/j.1476-5381.1994.tb13172.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 464.Gargett CE, Wiley JS. The isoquinoline derivative KN-62 a potent antagonist of the P2Z-receptor of human lymphocytes. Br J Pharmacol. 1997;120:1483–1490. doi: 10.1038/sj.bjp.0701081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 465.Bretschneider F, Klapperstück M, Löhn M, Markwardt F. Nonselective cationic currents elicited by extracellular ATP in human B-lymphocytes. Pflugers Arch. 1995;429:691–698. doi: 10.1007/BF00373990. [DOI] [PubMed] [Google Scholar]
  • 466.Markwardt F, Löhn M, Böhm T, Klapperstück M. Purinoceptor-operated cationic channels in human B lymphocytes. J Physiol. 1997;498:143–151. doi: 10.1113/jphysiol.1997.sp021847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 467.Chused TM, Apasov S, Sitkovsky M. Murine T lymphocytes modulate activity of an ATP-activated P2Z-type purinoceptor during differentiation. J Immunol. 1996;157:1371–1380. [PubMed] [Google Scholar]
  • 468.Baricordi OR, Ferrari D, Melchiorri L, Chiozzi P, Hanau S, Chiari E, Rubini M, Di Virgilio F. An ATP-activated channel is involved in mitogenic stimulation of human T lymphocytes. Blood. 1996;87:682–690. [PubMed] [Google Scholar]
  • 469.Baricordi OR, Melchiorri L, Adinolfi E, Falzoni S, Chiozzi P, Buell G, Di Virgilio F. Increased proliferation rate of lymphoid cells transfected with the P2X7 ATP receptor. J Biol Chem. 1999;274:33206–33208. doi: 10.1074/jbc.274.47.33206. [DOI] [PubMed] [Google Scholar]
  • 470.Schenk U, Westendorf AM, Radaelli E, Casati A, Ferro M, Fumagalli M, Verderio C, Buer J, Scanziani E, Grassi F. Purinergic control of T cell activation by ATP released through pannexin-1 hemichannels. Sci Signal. 2008;1:ra6. doi: 10.1126/scisignal.1160583. [DOI] [PubMed] [Google Scholar]
  • 471.Tokunaga A, Tsukimoto M, Harada H, Moriyama Y, Kojima S. Involvement of SLC17A9-dependent vesicular exocytosis in the mechanism of ATP release during T cell activation. J Biol Chem. 2010;285:17406–17416. doi: 10.1074/jbc.M110.112417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 472.Yip L, Woehrle T, Corriden R, Hirsh M, Chen Y, Inoue Y, Ferrari V, Insel PA, Junger WG. Autocrine regulation of T-cell activation by ATP release and P2X7 receptors. FASEB J. 2009;23:1685–1693. doi: 10.1096/fj.08-126458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 473.Yu T, Junger WG, Yuan C, Jin A, Zhao Y, Zheng X, Zeng Y, Liu J. Shockwaves increase T-cell proliferation and IL-2 expression through ATP release, P2X7 receptors, and FAK activation. Am J Physiol Cell Physiol. 2010;298:C457–C464. doi: 10.1152/ajpcell.00342.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 474.Junger WG. Immune cell regulation by autocrine purinergic signalling. Nat Rev Immunol. 2011;11:201–212. doi: 10.1038/nri2938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 475.Wang CM, Ploia C, Anselmi F, Sarukhan A, Viola A. Adenosine triphosphate acts as a paracrine signaling molecule to reduce the motility of T cells. EMBO J. 2014;33:1354–1364. doi: 10.15252/embj.201386666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 476.Jamieson GP, Snook MB, Thurlow PJ, Wiley JS. Extracellular ATP causes of loss of L-selectin from human lymphocytes via occupancy of P2Z purinocepters. J Cell Physiol. 1996;166:637–642. doi: 10.1002/(SICI)1097-4652(199603)166:3<637::AID-JCP19>3.0.CO;2-3. [DOI] [PubMed] [Google Scholar]
  • 477.Gu B, Bendall LJ, Wiley JS. Adenosine triphosphate-induced shedding of CD23 and L-selectin (CD62L) from lymphocytes is mediated by the same receptor but different metalloproteases. Blood. 1998;92:946–951. [PubMed] [Google Scholar]
  • 478.Elliott JI, Surprenant A, Marelli-Berg FM, Cooper JC, Cassady-Cain RL, Wooding C, Linton K, Alexander DR, Higgins CF. Membrane phosphatidylserine distribution as a non-apoptotic signalling mechanism in lymphocytes. Nat Cell Biol. 2005;7:808–816. doi: 10.1038/ncb1279. [DOI] [PubMed] [Google Scholar]
  • 479.Moon H, Na HY, Chong KH, Kim TJ. P2X7 receptor-dependent ATP-induced shedding of CD27 in mouse lymphocytes. Immunol Lett. 2006;102:98–105. doi: 10.1016/j.imlet.2005.08.004. [DOI] [PubMed] [Google Scholar]
  • 480.Sengstake S, Boneberg EM, Illges H. CD21 and CD62L shedding are both inducible via P2X7Rs. Int Immunol. 2006;18:1171–1178. doi: 10.1093/intimm/dxl051. [DOI] [PubMed] [Google Scholar]
  • 481.Scheuplein F, Schwarz N, Adriouch S, Krebs C, Bannas P, Rissiek B, Seman M, Haag F, Koch-Nolte F. NAD+ and ATP released from injured cells induce P2X7-dependent shedding of CD62L and externalization of phosphatidylserine by murine T cells. J Immunol. 2009;182:2898–2908. doi: 10.4049/jimmunol.0801711. [DOI] [PubMed] [Google Scholar]
  • 482.Foster JG, Carter E, Kilty I, MacKenzie AB, Ward SG. Mitochondrial superoxide generation enhances P2X7R-mediated loss of cell surface CD62L on naive human CD4+ T lymphocytes. J Immunol. 2013;190:1551–1559. doi: 10.4049/jimmunol.1201510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 483.Apasov SG, Koshiba M, Chused TM, Sitovsky MV. Effects of extracellular ATP and adenosine on different thymocyte subsets. Possible role of ATP-gated channels and G protein-coupled purinergic receptor. J Immunol. 1997;158:5095–5105. [PubMed] [Google Scholar]
  • 484.Nagy PV, Fehér T, Morga S, Matkó J. Apoptosis of murine thymocytes induced by extracellular ATP is dose- and cytosolic pH-dependent. Immunol Lett. 2000;72:23–30. doi: 10.1016/s0165-2478(00)00168-1. [DOI] [PubMed] [Google Scholar]
  • 485.Lépine S, Le Stunff H, Lakatos B, Sulpice JC, Giraud F. ATP-induced apoptosis of thymocytes is mediated by activation of P2X7 receptor and involves de novo ceramide synthesis and mitochondria. Biochim Biophys Acta. 2006;1761:73–82. doi: 10.1016/j.bbalip.2005.10.001. [DOI] [PubMed] [Google Scholar]
  • 486.Aswad F, Dennert G. P2X7 receptor expression levels determine lethal effects of a purine based danger signal in T lymphocytes. Cell Immunol. 2006;243:58–65. doi: 10.1016/j.cellimm.2006.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 487.Tsukimoto M, Maehata M, Harada H, Ikari A, Takagi K, Degawa M. P2X7 receptor-dependent cell death is modulated during murine T cell maturation and mediated by dual signaling pathways. J Immunol. 2006;177:2842–2850. doi: 10.4049/jimmunol.177.5.2842. [DOI] [PubMed] [Google Scholar]
  • 488.Schenk U, Frascoli M, Proietti M, Geffers R, Traggiai E, Buer J, Ricordi C, Westendorf AM, Grassi F. ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci Signal. 2011;4:ra12. doi: 10.1126/scisignal.2001270. [DOI] [PubMed] [Google Scholar]
  • 489.Cappelli C, López X, Labra Y, Montoya M, Fernández R, Imarai M, Rojas JL, Miranda D, Escobar A, Acuña-Castillo C. Polymyxin B increases the depletion of T regulatory cell induced by purinergic agonist. Immunobiology. 2012;217:307–315. doi: 10.1016/j.imbio.2011.10.006. [DOI] [PubMed] [Google Scholar]
  • 490.Aswad F, Kawamura H, Dennert G. High sensitivity of CD4 + CD25+ regulatory T cells to extracellular metabolites nicotinamide adenine dinucleotide and ATP: a role for P2X7 receptors. J Immunol. 2005;175:3075–3083. doi: 10.4049/jimmunol.175.5.3075. [DOI] [PubMed] [Google Scholar]
  • 491.Seman M, Adriouch S, Scheuplein F, Krebs C, Freese D, Glowacki G, Deterre P, Haag F, Koch-Nolte F. NAD-induced T cell death: ADP-ribosylation of cell surface proteins by ART2 activates the cytolytic P2X7 purinoceptor. Immunity. 2003;19:571–582. doi: 10.1016/s1074-7613(03)00266-8. [DOI] [PubMed] [Google Scholar]
  • 492.Kawamura H, Aswad F, Minagawa M, Malone K, Kaslow H, Koch-Nolte F, Schott WH, Leiter EH, Dennert G. P2X7 receptor-dependent and -independent T cell death is induced by nicotinamide adenine dinucleotide. J Immunol. 2005;174:1971–1979. doi: 10.4049/jimmunol.174.4.1971. [DOI] [PubMed] [Google Scholar]
  • 493.Adriouch S, Hubert S, Pechberty S, Koch-Nolte F, Haag F, Seman M. NAD+ released during inflammation participates in T cell homeostasis by inducing ART2-mediated death of naive T cells in vivo. J Immunol. 2007;179:186–194. doi: 10.4049/jimmunol.179.1.186. [DOI] [PubMed] [Google Scholar]
  • 494.Hubert S, Rissiek B, Klages K, Huehn J, Sparwasser T, Haag F, Koch-Nolte F, Boyer O, Seman M, Adriouch S. Extracellular NAD+ shapes the Foxp3+ regulatory T cell compartment through the ART2-P2X7 pathway. J Exp Med. 2010;207:2561–2568. doi: 10.1084/jem.20091154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 495.Trabanelli S, Ocadlíková D, Gulinelli S, Curti A, Salvestrini V, Vieira RP, Idzko M, Di Virgilio F, Ferrari D, Lemoli RM. Extracellular ATP exerts opposite effects on activated and regulatory CD4+ T cells via purinergic P2 receptor activation. J Immunol. 2012;189:1303–1310. doi: 10.4049/jimmunol.1103800. [DOI] [PubMed] [Google Scholar]
  • 496.Adinolfi E, Callegari MG, Ferrari D, Bolognesi C, Minelli M, Wieckowski MR, Pinton P, Rizzuto R, Di Virgilio F. Basal activation of the P2X7 ATP receptor elevates mitochondrial calcium and potential, increases cellular ATP levels, and promotes serum-independent growth. Mol Biol Cell. 2005;16:3260–3272. doi: 10.1091/mbc.E04-11-1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 497.Santos AA, Jr, Rodrigues-Junior V, Zanin RF, Borges TJ, Bonorino C, Coutinho-Silva R, Takyia CM, Santos DS, Campos MM, Morrone FB. Implication of purinergic P2X7 receptor in M. tuberculosis infection and host interaction mechanisms: a mouse model study. Immunobiology. 2013;218:1104–1112. doi: 10.1016/j.imbio.2013.03.003. [DOI] [PubMed] [Google Scholar]
  • 498.Lang PA, Merkler D, Funkner P, Shaabani N, Meryk A, Krings C, Barthuber C, Recher M, Brück W, Häussinger D, Ohashi PS, Lang KS. Oxidized ATP inhibits T-cell-mediated autoimmunity. Eur J Immunol. 2010;40:2401–2408. doi: 10.1002/eji.200939838. [DOI] [PubMed] [Google Scholar]
  • 499.Chen YG, Scheuplein F, Driver JP, Hewes AA, Reifsnyder PC, Leiter EH, Serreze DV. Testing the role of P2X7 receptors in the development of type 1 diabetes in nonobese diabetic mice. J Immunol. 2011;186:4278–4284. doi: 10.4049/jimmunol.1003733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 500.Vergani A, Fotino C, D'Addio F, Tezza S, Podetta M, Gatti F, Chin M, Bassi R, Molano RD, Corradi D, Gatti R, Ferrero ME, Secchi A, Grassi F, Ricordi C, Sayegh MH, Maffi P, Pileggi A, Fiorina P. Effect of the purinergic inhibitor oxidized ATP in a model of islet allograft rejection. Diabetes. 2013;62:1665–1675. doi: 10.2337/db12-0242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 501.Vergani A, Tezza S, D'Addio F, Fotino C, Liu K, Niewczas M, Bassi R, Molano RD, Kleffel S, Petrelli A, Soleti A, Ammirati E, Frigerio M, Visner G, Grassi F, Ferrero ME, Corradi D, Abdi R, Ricordi C, Sayegh MH, Pileggi A, Fiorina P. Long-term heart transplant survival by targeting the ionotropic purinergic receptor P2X7. Circulation. 2013;127:463–475. doi: 10.1161/CIRCULATIONAHA.112.123653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 502.Heiss K, Jänner N, Mähnss B, Schumacher V, Koch-Nolte F, Haag F, Mittrücker HW. High sensitivity of intestinal CD8+ T cells to nucleotides indicates P2X7 as a regulator for intestinal T cell responses. J Immunol. 2008;181:3861–3869. doi: 10.4049/jimmunol.181.6.3861. [DOI] [PubMed] [Google Scholar]
  • 503.Wilhelm K, Ganesan J, Müller T, Dürr C, Grimm M, Beilhack A, Krempl CD, Sorichter S, Gerlach UV, Jüttner E, Zerweck A, Gärtner F, Pellegatti P, Di Virgilio F, Ferrari D, Kambham N, Fisch P, Finke J, Idzko M, Zeiser R. Graft-versus-host disease is enhanced by extracellular ATP activating P2X7R. Nat Med. 2010;16:1434–1438. doi: 10.1038/nm.2242. [DOI] [PubMed] [Google Scholar]
  • 504.Chen L, Brosnan CF. Exacerbation of experimental autoimmune encephalomyelitis in P2X7R−/− mice: evidence for loss of apoptotic activity in lymphocytes. J Immunol. 2006;176:3115–3126. doi: 10.4049/jimmunol.176.5.3115. [DOI] [PubMed] [Google Scholar]
  • 505.Sharp AJ, Polak PE, Simonini V, Lin SX, Richardson JC, Bongarzone ER, Feinstein DL. P2x7 deficiency suppresses development of experimental autoimmune encephalomyelitis. J Neuroinflammation. 2008;5:33. doi: 10.1186/1742-2094-5-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 506.Freedman BD, Liu QH, Gaulton G, Kotlikoff MI, Hescheler J, Fleischmann BK. ATP-evoked Ca2+ transients and currents in murine thymocytes: possible role for P2X receptors in death by neglect. Eur J Immunol. 1999;29:1635–1646. doi: 10.1002/(SICI)1521-4141(199905)29:05<1635::AID-IMMU1635>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
  • 507.Woehrle T, Yip L, Elkhal A, Sumi Y, Chen Y, Yao Y, Insel PA, Junger WG. Pannexin-1 hemichannel-mediated ATP release together with P2X1 and P2X4 receptors regulate T-cell activation at the immune synapse. Blood. 2010;116:3475–3484. doi: 10.1182/blood-2010-04-277707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 508.Loomis WH, Namiki S, Ostrom RS, Insel PA, Junger WG. Hypertonic stress increases T cell interleukin-2 expression through a mechanism that involves ATP release, P2 receptor, and p38 MAPK activation. J Biol Chem. 2003;278:4590–4596. doi: 10.1074/jbc.M207868200. [DOI] [PubMed] [Google Scholar]
  • 509.Woehrle T, Yip L, Manohar M, Sumi Y, Yao Y, Chen Y, Junger WG. Hypertonic stress regulates T cell function via pannexin-1 hemichannels and P2X receptors. J Leukoc Biol. 2010;88:1181–1189. doi: 10.1189/jlb.0410211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 510.Frascoli M, Marcandalli J, Schenk U, Grassi F. Purinergic P2X7 receptor drives T cell lineage choice and shapes peripheral γδ cells. J Immunol. 2012;189:174–180. doi: 10.4049/jimmunol.1101582. [DOI] [PubMed] [Google Scholar]
  • 511.Manohar M, Hirsh MI, Chen Y, Woehrle T, Karande AA, Junger WG. ATP release and autocrine signaling through P2X4 receptors regulate γδ T cell activation. J Leukoc Biol. 2012;92:787–794. doi: 10.1189/jlb.0312121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 512.Koshiba M, Apasov S, Sverdlov V, Chen P, Erb L, Turner JT, Weisman GA, Sitkovsky MV. Transient up-regulation of P2Y2 nucleotide receptor mRNA expression is an immediate early gene response in activated thymocytes. Proc Natl Acad Sci U S A. 1997;94:831–836. doi: 10.1073/pnas.94.3.831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 513.Somers GR, Hammet FM, Trute L, Southey MC, Venter DJ. Expression of the P2Y6 purinergic receptor in human T cells infiltrating inflammatory bowel disease. Lab Invest. 1998;78:1375–1383. [PubMed] [Google Scholar]
  • 514.Tsukimoto M, Tokunaga A, Harada H, Kojima S. Blockade of murine T cell activation by antagonists of P2Y6 and P2X7 receptors. Biochem Biophys Res Commun. 2009;384:512–518. doi: 10.1016/j.bbrc.2009.05.011. [DOI] [PubMed] [Google Scholar]
  • 515.Giannattasio G, Ohta S, Boyce JR, Xing W, Balestrieri B, Boyce JA. The purinergic G protein-coupled receptor 6 inhibits effector T cell activation in allergic pulmonary inflammation. J Immunol. 2011;187:1486–1495. doi: 10.4049/jimmunol.1003669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 516.Scrivens M, Dickenson JM. Functional expression of the P2Y14 receptor in murine T-lymphocytes. Br J Pharmacol. 2005;146:435–444. doi: 10.1038/sj.bjp.0706322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 517.Duhant X, Schandené L, Bruyns C, Gonzalez NS, Goldman M, Boeynaems JM, Communi D. Extracellular adenine nucleotides inhibit the activation of human CD4+ T lymphocytes. J Immunol. 2002;169:15–21. doi: 10.4049/jimmunol.169.1.15. [DOI] [PubMed] [Google Scholar]
  • 518.Priebe T, Platsoucas CD, Nelson JA. Adenosine receptors and modulation of natural killer cell activity by purine nucleosides. Cancer Res. 1990;50:4328–4331. [PubMed] [Google Scholar]
  • 519.Raskovalova T, Lokshin A, Huang X, Jackson EK, Gorelik E. Adenosine-mediated inhibition of cytotoxic activity and cytokine production by IL-2/NKp46-activated NK cells: involvement of protein kinase A isozyme I (PKA I) Immunol Res. 2006;36:91–99. doi: 10.1385/IR:36:1:91. [DOI] [PubMed] [Google Scholar]
  • 520.Lappas CM, Day YJ, Marshall MA, Engelhard VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J Exp Med. 2006;203:2639–2648. doi: 10.1084/jem.20061097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 521.Nowak M, Lynch L, Yue S, Ohta A, Sitkovsky M, Balk SP, Exley MA. The A2aR adenosine receptor controls cytokine production in iNKT cells. Eur J Immunol. 2010;40:682–687. doi: 10.1002/eji.200939897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 522.Subramanian M, Kini R, Madasu M, Ohta A, Nowak M, Exley M, Sitkovsky M, Ohta A. Extracellular adenosine controls NKT-cell-dependent hepatitis induction. Eur J Immunol. 2014;44:1119–1129. doi: 10.1002/eji.201343866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 523.Wallace KL, Linden J. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood. 2010;116:5010–5020. doi: 10.1182/blood-2010-06-290643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 524.Field JJ, Lin G, Okam MM, Majerus E, Keefer J, Onyekwere O, Ross A, Campigotto F, Neuberg D, Linden J, Nathan DG. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood. 2013;121:3329–3334. doi: 10.1182/blood-2012-11-465963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 525.Lin G, Field JJ, Yu JC, Ken R, Neuberg D, Nathan DG, Linden J. NF-κB is activated in CD4+ iNKT cells by sickle cell disease and mediates rapid induction of adenosine A2A receptors. PLoS One. 2013;8:e74664. doi: 10.1371/journal.pone.0074664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 526.Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A. 2013;110:14711–14716. doi: 10.1073/pnas.1308209110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 527.Harish A, Hohana G, Fishman P, Arnon O, Bar-Yehuda S. A3 adenosine receptor agonist potentiates natural killer cell activity. Int J Oncol. 2003;23:1245–1249. [PubMed] [Google Scholar]
  • 528.Jeffe F, Stegmann KA, Broelsch F, Manns MP, Cornberg M, Wedemeyer H. Adenosine and IFN-α synergistically increase IFN-γ production of human NK cells. J Leukoc Biol. 2009;85:452–461. doi: 10.1189/jlb.0108046. [DOI] [PubMed] [Google Scholar]
  • 529.Henriksson T. Inhibition of natural killing by adenosine ribonucleotides. Immunol Lett. 1983;7:171–176. doi: 10.1016/0165-2478(83)90066-4. [DOI] [PubMed] [Google Scholar]
  • 530.Schmidt A, Ortaldo JR, Herberman RB. Inhibition of human natural killer cell reactivity by exogenous adenosine 5'-triphosphate. J Immunol. 1984;132:146–150. [PubMed] [Google Scholar]
  • 531.Krishnaraj R. Negative modulation of human NK cell activity by purinoceptors. 1. Effect of exogenous adenosine triphosphatea. Cell Immunol. 1992;141:306–322. doi: 10.1016/0008-8749(92)90150-n. [DOI] [PubMed] [Google Scholar]
  • 532.Krishnaraj R. Negative modulation of human NK cell activity by purinoceptors. 2. Age-associated, gender-specific partial loss of sensitivity to ATP. Cell Immunol. 1992;144:11–21. doi: 10.1016/0008-8749(92)90221-a. [DOI] [PubMed] [Google Scholar]
  • 533.Gorini S, Callegari G, Romagnoli G, Mammi C, Mavilio D, Rosano G, Fini M, Di Virgilio F, Gulinelli S, Falzoni S, Cavani A, Ferrari D, la Sala A. ATP secreted by endothelial cells blocks CX3CL 1-elicited natural killer cell chemotaxis and cytotoxicity via P2Y11 receptor activation. Blood. 2010;116:4492–4500. doi: 10.1182/blood-2009-12-260828. [DOI] [PubMed] [Google Scholar]
  • 534.Kawamura H, Aswad F, Minagawa M, Govindarajan S, Dennert G. P2X7 receptors regulate NKT cells in autoimmune hepatitis. J Immunol. 2006;176:2152–2160. doi: 10.4049/jimmunol.176.4.2152. [DOI] [PubMed] [Google Scholar]
  • 535.Beldi G, Wu Y, Banz Y, Nowak M, Miller L, Enjyoji K, Haschemi A, Yegutkin GG, Candinas D, Exley M, Robson SC. Natural killer T cell dysfunction in CD39-null mice protects against concanavalin A-induced hepatitis. Hepatology. 2008;48:841–852. doi: 10.1002/hep.22401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 536.Nowak-Machen M, Schmelzle M, Hanidziar D, Junger W, Exley M, Otterbein L, Wu Y, Csizmadia E, Doherty G, Sitkovsky M, Robson SC. Pulmonary natural killer T cells play an essential role in mediating hyperoxic acute lung injury. Am J Respir Cell Mol Biol. 2013;48:601–609. doi: 10.1165/rcmb.2012-0180OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 537.Di Virgilio F, Ferrari D, Chiozzi P, Falzoni S, Sanz JM, dal Susino M, Mutini C, Hanau S, Baricordi OR. Purinoceptor function in the immune system. Drug Dev Res. 1996;39:319–329. [Google Scholar]
  • 538.Di Virgilio F, Chiozzi P, Ferrari D, Falzoni S, Sanz JM, Morelli A, Torboli M, Bolognesi G, Baricordi OR. Nucleotide receptors: an emerging family of regulatory molecules in blood cells. Blood. 2001;97:587–600. doi: 10.1182/blood.v97.3.587. [DOI] [PubMed] [Google Scholar]
  • 539.Di Virgilio F, Borea PA, Illes P. P2 receptors meet the immune system. Trends Pharmacol Sci. 2001;22:5–7. doi: 10.1016/s0165-6147(00)01574-1. [DOI] [PubMed] [Google Scholar]
  • 540.Smith PT, Armstrong J, Koshiba M, Huang S, Apasov S, Sitkovsky M. Studies of expression and possible functional role of purinergic receptors in cell-mediated immunity: Experimental approaches, controls, and caveats. Drug Dev Res. 1998;45:229–244. [Google Scholar]
  • 541.Burnstock G. Overview of P2 receptors: possible functions in immune cells. Drug Dev Res. 2001;53:53–59. [Google Scholar]
  • 542.Di Virgilio F. Dr. Jekyll/Mr. Hyde: the dual role of extracellular ATP. J Auton Nerv Syst. 2000;81:59–63. doi: 10.1016/s0165-1838(00)00114-4. [DOI] [PubMed] [Google Scholar]
  • 543.Di Virgilio F. Purinergic signalling in the immune system. A brief update. Purinergic Signal. 2007;3:1–3. doi: 10.1007/s11302-006-9048-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 544.Sitkovsky MV. Extracellular purines and their receptors in immunoregulation. Review of recent advances. Nihon Ika Daigaku Zasshi. 1998;65:351–357. doi: 10.1272/jnms1923.65.351. [DOI] [PubMed] [Google Scholar]
  • 545.Armstrong S, Korcok J, Sims SM, Dixon SJ. Activation of transcription factors by extracellular nucleotides in immune and related cell types. Purinergic Signal. 2007;3:59–69. doi: 10.1007/s11302-006-9037-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 546.Myrtek D, Idzko M. Chemotactic activity of extracellular nucleotideson human immune cells. Purinergic Signal. 2007;3:5–11. doi: 10.1007/s11302-006-9032-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 547.Di Virgilio F, Ceruti S, Bramanti P, Abbracchio MP. Purinergic signalling in inflammation of the central nervous system. Trends Neurosci. 2009;32:79–87. doi: 10.1016/j.tins.2008.11.003. [DOI] [PubMed] [Google Scholar]
  • 548.Zeiser R, Penack O, Holler E, Idzko M. Danger signals activating innate immunity in graft-versus-host disease. J Mol Med (Berl) 2011;89:833–845. doi: 10.1007/s00109-011-0767-x. [DOI] [PubMed] [Google Scholar]
  • 549.Vitiello L, Gorini S, Rosano G, la Sala A. Immunoregulation through extracellular nucleotides. Blood. 2012;120:511–518. doi: 10.1182/blood-2012-01-406496. [DOI] [PubMed] [Google Scholar]
  • 550.Jacob F, Novo CP, Bachert C, Crombruggen K. Purinergic signaling in inflammatory cells: P2 receptor expression, functional effects, and modulation of inflammatory responses. Purinergic Signall. 2013;9:285–306. doi: 10.1007/s11302-013-9357-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 551.Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling during inflammation. Nature. 2014;509:310–317. doi: 10.1038/nature13085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 552.Kammer GM (1986) Adenosine: emerging role as an immunomodifying agent. J Lab Clin Sept:255–256 [PubMed]
  • 553.Gessi S, Varani K, Merighi S, Ongini E, Borea PA. A2A adenosine receptors in human peripheral blood cells. Br J Pharmacol. 2000;129:2–11. doi: 10.1038/sj.bjp.0703045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 554.Haskó G, Deitch EA, Szabo C, Nemeth ZH, Vizi ES. Adenosine: a potential mediator of immunosuppression in multiple organ failure. Curr Opin Pharmacol. 2002;2:440–444. doi: 10.1016/s1471-4892(02)00172-8. [DOI] [PubMed] [Google Scholar]
  • 555.Haskó G, Cronstein BN. Adenosine: an endogenous regulator of innate immunity. Trends Immunol. 2004;25:33–39. doi: 10.1016/j.it.2003.11.003. [DOI] [PubMed] [Google Scholar]
  • 556.McCallion K, Harkin DW, Gardiner KR. Role of adenosine in immunomodulation: review of the literature. Crit Care Med. 2004;32:273–277. doi: 10.1097/01.CCM.0000098026.12020.45. [DOI] [PubMed] [Google Scholar]
  • 557.Sitkovsky MV, Ohta A. The 'danger' sensors that STOP the immune response: the A2 adenosine receptors? Trends Immunol. 2005;26:299–304. doi: 10.1016/j.it.2005.04.004. [DOI] [PubMed] [Google Scholar]
  • 558.Kumar V, Sharma A. Adenosine: an endogenous modulator of innate immune system with therapeutic potential. Eur J Pharmacol. 2009;616:7–15. doi: 10.1016/j.ejphar.2009.05.005. [DOI] [PubMed] [Google Scholar]
  • 559.Ohta A, Sitkovsky M. The adenosinergic immunomodulatory drugs. Curr Opin Pharmacol. 2009;9:501–506. doi: 10.1016/j.coph.2009.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 560.Drygiannakis I, Ernst PB, Lowe D, Glomski IJ. Immunological alterations mediated by adenosine during host-microbial interactions. Immunol Res. 2011;50:69–77. doi: 10.1007/s12026-011-8207-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 561.Ramakers BP, Riksen NP, van der Hoeven JG, Smits P, Pickkers P. Modulation of innate immunity by adenosine receptor stimulation. Shock. 2011;36:208–215. doi: 10.1097/SHK.0b013e318225aee4. [DOI] [PubMed] [Google Scholar]
  • 562.Antonioli L, Blandizzi C, Pacher P, Haskó G. Immunity, inflammation and cancer: a leading role for adenosine. Nat Rev Cancer. 2013;13:842–857. doi: 10.1038/nrc3613. [DOI] [PubMed] [Google Scholar]
  • 563.Antonioli L, Csóka B, Fornai M, Colucci R, Kókai E, Blandizzi C, Haskó G. Adenosine and inflammation: what's new on the horizon? Drug Discov Today. 2014;19:1051–1068. doi: 10.1016/j.drudis.2014.02.010. [DOI] [PubMed] [Google Scholar]
  • 564.Salmi M, Jalkanen S. Cell-surface enzymes in control of leukocyte trafficking. Nat Rev Immunol. 2005;5:760–771. doi: 10.1038/nri1705. [DOI] [PubMed] [Google Scholar]
  • 565.Dwyer KM, Deaglio S, Gao W, Friedman D, Strom TB, Robson SC. CD39 and control of cellular immune responses. Purinergic Signal. 2007;3:171–180. doi: 10.1007/s11302-006-9050-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 566.Beavis PA, Stagg J, Darcy PK, Smyth MJ. CD73: a potent suppressor of antitumor immune responses. Trends Immunol. 2012;33:231–237. doi: 10.1016/j.it.2012.02.009. [DOI] [PubMed] [Google Scholar]
  • 567.Junger WG. Purinergic regulation of neutrophil chemotaxis. Cell Mol Life Sci. 2008;65:2528–2540. doi: 10.1007/s00018-008-8095-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 568.Grassi F. Purinergic control of neutrophil activation. J Mol Cell Biol. 2010;2:176–177. doi: 10.1093/jmcb/mjq014. [DOI] [PubMed] [Google Scholar]
  • 569.Barletta KE, Ley K, Mehrad B. Regulation of neutrophil function by adenosine. Arterioscler Thromb Vasc Biol. 2012;32:856–864. doi: 10.1161/ATVBAHA.111.226845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 570.Ferrari D, la Sala A, Panther E, Norgauer J, Di Virgilio F, Idzko M. Activation of human eosinophils via P2 receptors: novel findings and future perspectives. J Leukoc Biol. 2006;79:7–15. doi: 10.1189/jlb.0505286. [DOI] [PubMed] [Google Scholar]
  • 571.Bulanova E, Bulfone-Paus S. P2 receptor-mediated signaling in mast cell biology. Purinergic Signal. 2010;6:3–17. doi: 10.1007/s11302-009-9173-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 572.Skaper SD, Giusti P, Facci L. Microglia and mast cells: two tracks on the road to neuroinflammation. FASEB J. 2012;26:3103–3117. doi: 10.1096/fj.11-197194. [DOI] [PubMed] [Google Scholar]
  • 573.Weisman GA, Erb L, Garrad RC, Theiss PM, Santiago-Pérez LI, Flores RV, Santos-Berríos C, Méndez Y, González FA. P2Y nucleotide receptors in the immune system: Signaling by a P2Y2 receptor in U937 monocytes. Drug Dev Res. 1998;45:222–228. [Google Scholar]
  • 574.Haskó G, Pacher P. Regulation of macrophage function by adenosine. Arterioscler Thromb Vasc Biol. 2012;32:865–869. doi: 10.1161/ATVBAHA.111.226852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 575.Inoue K. Purinergic systems in microglia. Cell Mol Life Sci. 2008;65:3074–3080. doi: 10.1007/s00018-008-8210-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 576.Burnstock G, Verkhratsky A. Purinergic signalling and the nervous system. Heidelberg/Berlin: Springer; 2012. pp. 1–715. [Google Scholar]
  • 577.Ferrari D, Gorini S, Callegari G, la Sala A. Shaping immune responses through the activation of dendritic cells' P2 receptors. Purinergic Signal. 2007;3:99–107. doi: 10.1007/s11302-006-9024-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 578.Salter RD, Watkins SC. Dendritic cell altered states: what role for calcium? Immunol Rev. 2009;231:278–288. doi: 10.1111/j.1600-065X.2009.00806.x. [DOI] [PubMed] [Google Scholar]
  • 579.Kefford RF, Fox RM. Purinogenic lymphocytotoxicity: clues to a wider chemotherapeutic potential for the adenosine deaminase inhibitors. Cancer Chemother Pharmacol. 1983;10:73–78. doi: 10.1007/BF00446213. [DOI] [PubMed] [Google Scholar]
  • 580.Marone G, Vigorita S, Triggiani M, Condorelli M. Adenosine receptors on human lymphocytes. Adv Exp Med Biol. 1986;195:7–14. doi: 10.1007/978-1-4684-1248-2_2. [DOI] [PubMed] [Google Scholar]
  • 581.Goding JW, Howard MC. Ecto-enzymes of lymphoid cells. Immunol Rev. 1998;161:5–10. doi: 10.1111/j.1600-065x.1998.tb01567.x. [DOI] [PubMed] [Google Scholar]
  • 582.Gessi S, Varani K, Merighi S, Fogli E, Sacchetto V, Benini A, Leung E, Mac-Lennan S, Borea PA. Adenosine and lymphocyte regulation. Purinergic Signal. 2007;3:109–116. doi: 10.1007/s11302-006-9042-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 583.Leavy O. Regulatory T cells: Adding adenosine to the mix. Nat Rev Immunol. 2007;7:493. [Google Scholar]
  • 584.Zarek PE, Powell JD. Adenosine and anergy. Autoimmunity. 2007;40:425–432. doi: 10.1080/08916930701464939. [DOI] [PubMed] [Google Scholar]
  • 585.Sitkovsky M, Lukashev D, Deaglio S, Dwyer K, Robson SC, Ohta A. Adenosine A2A receptor antagonists: blockade of adenosinergic effects and T regulatory cells. Br J Pharmacol. 2008;153(Suppl 1):S457–S464. doi: 10.1038/bjp.2008.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 586.Linden J, Cekic C. Regulation of lymphocyte function by adenosine. Arterioscler Thromb Vasc Biol. 2012;32:2097–2103. doi: 10.1161/ATVBAHA.111.226837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 587.Di Virgilio F, Vishwanath V, Ferrari D (2001) On the role of the P2X7 receptor in the immune system. In: Abbracchio MP, Williams M (eds) Handbook of Experimental Pharmacology, Volume 151/II. Purinergic and Pyrimidinergic Signalling II - Cardiovascular, Respiratory, Immune, Metabolic and Gastrointestinal Tract Function. Springer-Verlag, Berlin, pp 356–374
  • 588.Haag F, Adriouch S, Brass A, Jung C, Möller S, Scheuplein F, Bannas P, Seman M, Koch-Nolte F. Extracellular NAD and ATP: Partners in immune cell modulation. Purinergic Signal. 2007;3:71–81. doi: 10.1007/s11302-006-9038-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 589.Costa-Junior HM, Marques-da-Silva C, Vieira FS, Monção-Ribeiro LC, Coutinho-Silva R. Lipid metabolism modulation by the P2X7 receptor in the immune system and during the course of infection: new insights into the old view. Purinergic Signal. 2011;7:381–392. doi: 10.1007/s11302-011-9255-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 590.Yamada T, Chakrabarty AM. ATP-utilizing enzymes, purinergic receptor modulation, cupredoxins and mammalian cell death. In: Ramos JL, editor. Pseudomonas. New York: Kluwer Academic/Plenum Publishers; 2004. pp. 47–67. [Google Scholar]
  • 591.Coutinho-Silva R, Monteiro da Cruz C, Persechini PM, Ojcius DM. The role of P2 receptors in controlling infections by intracellular pathogens. Purinergic Signal. 2007;3:83–90. doi: 10.1007/s11302-006-9039-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 592.Coutinho-Silva R, Corrêa G, Sater AA, Ojcius DM. The P2X7 receptor and intracellular pathogens: a continuing struggle. Purinergic Signal. 2009;5:197–204. doi: 10.1007/s11302-009-9130-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 593.Wewers MD, Sarkar A. P2X7 receptor and macrophage function. Purinergic Signal. 2009;5:189–195. doi: 10.1007/s11302-009-9131-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 594.Bald D, Koul A. Respiratory ATP synthesis: the new generation of mycobacterial drug targets? FEMS Microbiol Lett. 2010;308:1–7. doi: 10.1111/j.1574-6968.2010.01959.x. [DOI] [PubMed] [Google Scholar]

Articles from Purinergic Signalling are provided here courtesy of Springer

RESOURCES