Nucleotides are emerging as an ubiquitous family of extracellular signaling molecules. It has been known for many years that adenosine diphosphate is a potent platelet aggregating factor, but it is now clear that virtually every circulating cell is responsive to nucleotides. Effects as different as proliferation or differentiation, chemotaxis, release of cytokines or lysosomal constituents, and generation of reactive oxygen or nitrogen species are elicited upon stimulation of blood cells with extracellular adenosine triphosphate (ATP). These effects are mediated through a specific class of plasma membrane receptors called purinergic P2 receptors that, according to the molecular structure, are further subdivided into 2 subfamilies: P2Y and P2X. ATP and possibly other nucleotides are released from damaged cells or secreted via nonlytic mechanisms. Thus, during inflammation or vascular damage, nucleotides may provide an important mechanism involved in the activation of leukocytes and platelets. However, the cell physiology of these receptors is still at its dawn, and the precise function of the multiple P2X and P2Y receptor subtypes remains to be understood.

In 1978 the existence of plasma membrane receptors for extracellular nucleotides, the P2 purinergic receptors, was formally recognized.1 At that time, this identification was only based on pharmacologic and functional evidence and on the prophetic intuition of Geoff Burnstock. To date, 12 mammalian P2 receptors have been cloned, characterized, and recognized as responsible for the diverse cellular responses to stimulation with extracellular nucleotides.2,3 The P2 receptor family also includes receptors for extracellular pyrimidines. The new classification based on the molecular structure is rapidly replacing the previous one (P2Y, P2X, P2U, P2T, and P2Z) based on the pharmacologic profile,4 although doubts remain on whether functional responses of the native P2Z receptor of immune cells can be entirely explained by the cloned P2X7 subunit. A similar uncertainty also concerns the platelet P2T receptor, which is likely to arise from the combination of P2Y and P2X-dependent responses.2,5 Extracellular effects of nucleotides were initially recognized in smooth muscle contraction, neurotransmission, regulation of cardiac function, and platelet aggregation.6However, over the last 10 years it has become clear that the intercellular mediator role of these molecules is widespread, and blood cells have emerged as one of the most interesting targets.

Contrary to a widely held opinion, adenosine triphosphate (ATP) and possibly also uridine triphosphate (UTP) are often released into the extracellular environment via nonlytic mechanisms7-12 and also more frequently as a consequence of cell damage or acute cell death. Furthermore, platelet-dense granules are a relevant source of secreted ATP.13,14 Once in the pericellular environment, ATP can serve as a ligand for P2 receptors or be quickly hydrolyzed by powerful ubiquitous ecto-ATPases and ectonucleotidases.15-18 ATP can also be used as a phosphate donor by poorly characterized ectokinases.19Thus, ATP possesses all the properties of a bona fide fast-acting intercellular messenger: (a) it is released in a controlled fashion, (b) ligates specific plasma membrane receptors coupled to intracellular signal transduction, and (c) is quickly degraded to terminate its action.

Outside excitable tissues, P2 receptors have an obvious relevance in platelet aggregation, but immunity and inflammation are providing some of the most exciting developments in this evolving field. A few reviews covering different aspects of P2 receptor distribution and function in hemopoietic cells have appeared and have been an invaluable source of information for the present work.20-26 

According to the International Union of Pharmacology (IUPHAR) Committee on Receptor Nomenclature and Drug Classification,27 receptors for extracellular nucleotides are termed P2 receptors (this nomenclature replaces the older “P2-purinoceptor”). P2 receptors are divided into 2 subfamilies: G protein–coupled (P2Y) and ligand-gated ion channels (P2X).3,28-30 Current P2Y/P2X nomenclature is based on the molecular structure and has replaced the previous one based on pharmacologic and functional criteria. In mammalian cells, 5 P2Y (P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11) and 7 P2X (P2X1-7) receptors have been cloned and characterized pharmacologically2 (Table 1). P2Y5, P2Y7, P2Y9, and P2Y10 have been purged from this sequence because they are primarily non-nucleotide receptors (although they may also bind extracellular nucleotides). A p2y3 (lower case to indicate that it has not been cloned from mammals) receptor has been cloned from chick brain and suggested to be a homologue of the mammalian P2Y6.2 P2Y8 has so far only been cloned from Xenopus neural plate; thus it is not included in the list of mammalian receptors. The adenosine diphosphate (ADP)-activated, G protein–coupled receptor of platelets that triggers inhibition of stimulated adenylate cyclase has not yet been cloned; thus it is recommended that this receptor should be given in italics: P2Y ADP.2 

Table 1.

P2Y and P2X receptor subtypes

P2YAmino acid numberP2XAmino acid number
P2Y1 362 P2X1 399 
P2Y2 373 P2X2 472, 401 
p2y3* 328 P2X3 397 
P2Y4 352 P2X4 388, 329 
P2Y6 379 P2X5 455 
P2Y11 371 P2X6 379 
  P2X7 595 
P2YAmino acid numberP2XAmino acid number
P2Y1 362 P2X1 399 
P2Y2 373 P2X2 472, 401 
p2y3* 328 P2X3 397 
P2Y4 352 P2X4 388, 329 
P2Y6 379 P2X5 455 
P2Y11 371 P2X6 379 
  P2X7 595 
*

p2y3 was cloned from chick brain and may be the chick homologue of the mammalian P2Y6.

P2X2 and P2X4 are present in two splice variants.

P2Y receptors are 7-membrane–spanning proteins, numbering from 328 to 379 amino acids, for a molecular mass of 41 to 53 kd after glycosylation.2,31,32 The aminoterminal domain faces the extracellular environment, and the carboxyterminal is on the cytoplasmic side of the plasma membrane (Figure1). Signal transduction occurs via the classical pathways triggered by most 7-membrane–spanning receptors: activation of phospholipase C and/or stimulation/inhibition of adenylate cyclase. All of the P2Y receptors are activated by ATP, but at 2 of them, P2Y4 and P2Y6, UTP is more potent,33-36 and at P2Y2 ATP and UTP are equipotent.31 At P2Y1, UTP is inactive and ADP is reported to be equipotent or even more potent than ATP37,38; at P2Y11 ATP is more potent than ADP and UTP is inactive.39 With respect to the signal transduction pathway, P2Y1 and P2Y2 are coupled to stimulation of phospholipase C-β and inhibition of adenylate cyclase via Gq/11 and Gi proteins, respectively.2 There are reports suggesting that P2Y2 can also trigger stimulation of phospholipase D and breakdown of phosphatidylcholine, but the mechanism is unclear.40 P2Y4 and P2Y6 seem to only couple to phosphoinositide breakdown, whereas P2Y11rather surprisingly stimulates activation of both the phosphoinositide and the adenylate cyclase pathways.

Fig. 1.

Membrane topology of P2Y and P2X receptor subunits.

(A) P2Y receptors (i) are typical 7-membrane–spanning receptors made of a single polypeptide chain, with the N- and C-termini on the external and cytoplasmic side of the plasma membrane, respectively. P2X receptors (ii) are formed by subunits that span the plasma membrane twice and have both the N- and C-termini on the cytoplasmic side. The P2X7 subunit differs from the other members of the P2X subfamily (P2X1-P2X6) in the extended carboxyterminal tail (iii). (B) It is hypothesized that P2X7 receptor is generated by the aggregation of an unknown number of subunits (maybe 6) to form an ATP-activated channel. Recruitment of additional subunits causes formation of a nonselective pore (also see Figure 2).

Fig. 1.

Membrane topology of P2Y and P2X receptor subunits.

(A) P2Y receptors (i) are typical 7-membrane–spanning receptors made of a single polypeptide chain, with the N- and C-termini on the external and cytoplasmic side of the plasma membrane, respectively. P2X receptors (ii) are formed by subunits that span the plasma membrane twice and have both the N- and C-termini on the cytoplasmic side. The P2X7 subunit differs from the other members of the P2X subfamily (P2X1-P2X6) in the extended carboxyterminal tail (iii). (B) It is hypothesized that P2X7 receptor is generated by the aggregation of an unknown number of subunits (maybe 6) to form an ATP-activated channel. Recruitment of additional subunits causes formation of a nonselective pore (also see Figure 2).

Close modal

Investigation of P2Y receptors has been severely hindered by the lack of specific antibodies, whether polyclonal or monoclonal. Likewise, few selective agonists, besides naturally occurring nucleotides, or antagonists are available. A widely used P2Y antagonist is suramin,41 a drug originally developed for the treatment of tripanosomiasis. However, suramin does not discriminate between P2Y and P2X and has been reported to inhibit other receptors such as the nicotinic, glutamate, GABA, and 5-hydroxytryptamine receptors as well as the activity of diverse growth factors.2 Reactive blue 2, trypan blue, and reactive red have also been used as P2Y antagonists, but they also block P2X-dependent responses.2Recently Harden and coworkers have introduced a number of nucleotide analogues as competitive P2Y1antagonists.42,43 Pyridoxal phosphate (P5P) and pyridoxalphosphate-6-azophenyl 2′,4′-disulfonic acid (PPADS) are also sometimes used to inhibit P2Y-dependent responses, but they are more widely employed to block P2X receptors.

P2X receptors are ATP-gated ion channels—originally cloned and characterized in excitable cells44,45 and then shown to be nearly ubiquitous24,46,47—that mediate fast permeability changes to monovalent and divalent cations (Na+, K+, and Ca++). One of the members of this subfamily, P2X7, has sparked vivid interest for its peculiar ability to undergo a progressive increase in size that leads to the generation of a nonselective membrane pore22,48-50(Figures 1 and 2). All P2X receptors are likely to be multimeric structures of which 7 basic subunits have been cloned. The subunit composition of the native receptors has been resolved only in one case: rat dorsal ganglia that express a P2X2/P2X3 heteromer.51 It is not known how P2X receptors assemble for most cell types. Subunit stoichiometry is likewise unknown but for P2X1 and P2X3, which have been shown to assemble as trimers or hexamers.52 Whether other P2X receptors also assemble according to the same stoichiometry is not known. P2X receptors range from 379 to 595 amino acids and are thought to have 2 transmembrane hydrophobic domains separated by a bulky extracellular region harbouring 10 cysteines and 2 to 6 N-linked glycosylation sites.2,30,44,45 The aminotermini and carboxytermini are both on the cytoplasmic side of the plasma membrane. This tertiary structure and membrane topology is reminiscent of that of other ion channels such as the epithelial amiloride-sensitive Na+channel (ENaC), the degenerins cloned from Caenorhabditis elegans, and the inward rectifying K+ channel (Kir).30 Signal transduction occurs via fast Na+ and Ca++ influx and K+ efflux, leading to depolarization of the plasma membrane and an increase in the concentration of cytosolic Ca++([Ca++]i). It is likely that the drastic upset in intracellular ion homeostasis caused by P2X receptor opening activates several additional intracellular messengers and enzyme pathways, but few studies are available on this novel and exciting field of P2X receptor biochemistry. Electrophysiologic investigation of recombinant P2X receptor subunits transfected into mammalian recipient cells has allowed identification of fast desensitizing and slowly desensitizing (or nondesensitizing) P2X receptors.53 

Fig. 2.

Permeability transition of P2X7 receptor.

A transient stimulation with ATP causes the opening of the P2X7 channel and the concomitant Ca++ and Na+ influx and K+ efflux. However, upon sustained stimulation with ATP, the P2X7 receptor undergoes a transition that generates a reversible membrane pore permeable to several low-molecular-weight hydrophylic solutes (Fura-2/FA, Fura-2 free acid; LY, lucifer yellow; EB, ethidium bromide) and to nucleotides.

Fig. 2.

Permeability transition of P2X7 receptor.

A transient stimulation with ATP causes the opening of the P2X7 channel and the concomitant Ca++ and Na+ influx and K+ efflux. However, upon sustained stimulation with ATP, the P2X7 receptor undergoes a transition that generates a reversible membrane pore permeable to several low-molecular-weight hydrophylic solutes (Fura-2/FA, Fura-2 free acid; LY, lucifer yellow; EB, ethidium bromide) and to nucleotides.

Close modal

Although still on a limited basis, a few anti-P2X antibodies were made available over the last 2 years by single laboratories or commercial sources. Polyclonal antibodies against P2X1, P2X4, and P2X7 can be obtained from at least 2 companies; in a few laboratories sera against all the members of the subfamily have been raised.53,46,49,54 One monoclonal antibody selective for the human P2X7 receptor has been produced and characterized by Buell and colleagues.55 Interestingly, this monoclonal antibody, which recognizes an as yet to be identified epitope on the extracellular domain, inhibits activation of human macrophages by 3′-O-(4-benzoyl)benzoyl-ATP (BzATP), a P2X7agonist.55 

The unique naturally occurring agonist of P2X receptors is ATP, albeit diadenosine polyphosphates, such as P,1P4-diadenosine tetraphosphate (Ap4A) and P,1P6-diadenosine hexaphosphate (Ap6A), are active at P2X12, and UTP has been reported to be an agonist at P2X3 as well as P2X1.56,57 There is an ongoing debate, initiated by the pioneeristic experiments of Cockcroft and Gomperts in mast cells,58,59 on whether P2X receptors recognize the bianionic (ATP2−) or tetra-anionic (ATP4−) form of the nucleotide. In physiologic solutions, the free acid ATP4− is complexed by Mg++, Ca++, or H+ to yield various mixtures of MgATP2−, CaATP2−, and HATP3−, whereas a small amount (1%-10%, depending on the divalent cation concentration and the pH) is present as the fully dissociated tetra-anion. Removal of Mg++ and Ca++ and alkalinization of the medium increases the apparent affinity of ATP and BzATP for the native P2Z receptor of mast cells and other cell types58-61 and for the recombinant P2X7 receptor,48,62 but addition of Mg++ quickly terminates stimulation. Interpretation of the effects of divalent cation removal is complicated by the concomitant inhibition of ecto-ATPase/ectonucleotidase activity, because these enzymes require the presence of divalent cations. Slowing of hydrolytic activity prolongs the half-life of the nucleotide, thus increasing its apparent potency. This may explain the finding that the potency of stable ATP analogues, such as α, β-methylene ATP (α, β MeATP), is unaffected by Ca++ and Mg++removal.63 This ATP analogue is used to pharmacologically discriminate P2X receptor subtypes: P2X1 and P2X3 are sensitive to low (0.5-5 μM) concentrations, and P2X2 and P2X4-7 are activated by high (>100 μM) doses.2 An often neglected finding is the high potency of BzATP at all P2X—not just P2X7—receptors and its agonist activity at P2Y receptors, a feature that makes this ATP analogue one of the most useful tools for the study of native and recombinant P2X receptors.53 

Better antagonists, with better-characterized activity, are available at P2X than at P2Y receptors. PPADS is a noncompetitive inhibitor of most P2X receptors53 and, depending on the experimental conditions, may act irreversibly. Oxidized ATP (oATP) was introduced 7 years ago as a selective P2Z (P2X7) inhibitor,64 but it is likely to show the same P2X antagonist selectivity of PPADS, although no detailed investigation has been carried out. At the effective concentrations (100-300 μM), oATP shows little or no inhibitory activity at P2Y receptors and at ectonucleotidases.64 Action of oATP on ectokinases has not been tested in depth; thus it cannot be excluded that some effects of this compound may be due to inhibition of ectophosphorylation.65 PPADS and oATP likely share the same mechanism of action. Both compounds have aldehyde groups (1 PPADS, 2 oATPs) that can react with unprotonated lysines to form Schiff's bases. It is assumed that they preferentially modify lysine residues in the vicinity of the ATP binding site, but this assumption is yet to be proved. Although PPADS has been used as a P2 blocker for some time, it was only after the introduction of oATP that attention has been paid to the time-dependent and irreversible inhibitory effect of this P5P derivative. Time-dependent and irreversible block is extensively documented for oATP at the P2X7 receptor: A 1- to 2-hour preincubation with this inhibitor, even if followed by extensive rinsing, makes all cells so far investigated fully refractory to ATP stimulation via the P2X7receptor.64,66-69 Refractoriness lasts several hours, until new receptors are inserted into the plasma membrane.

More recently, Wiley and colleagues have introduced another powerful blocker of P2X7, compound 1-[N,0-bis(5-isoquinolinesulphonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine (KN-62).70 This molecule was originally used as an inhibitor of the calcium calmodulin–dependent kinase71and made its first appearance in the purinergic field in a study by Blanchard et al72 aimed at investigating the role of the P2Z receptor in cell-mediated cytotoxicity. KN-62 acts as a competitive inhibitor at nanomolar concentrations and shows a striking species specificity: It is active only at the human and not at the rat or mouse P2X7 receptor.73 KN-62 is a very useful tool for short-term studies, but modification of long-term responses should be interpreted with caution because of concomitant inhibition of calcium calmodulin kinase. Surprenant and colleagues74have recently shown that Coomassie Brilliant Blue G selectively blocks rat P2X7 with nanomolar affinity.

Early studies by Steinberg and Silverstein showed that the J774 mouse macrophage cell line expressed a plasma membrane receptor selectively activated by ATP and a few analogues.60,75Stimulation of this receptor triggered the same reversible increase in plasma membrane permeability to low-molecular-mass solutes originally described by Cockcroft and Gomperts in rat mast cells.58,59 An intriguing finding of these studies was that stimulation of the ATP-permeabilizing receptor eventually led to cell death.75 This incidental observation stirred interest in the possible physiologic meaning of ATP-dependent cytotoxicity and fostered subsequent studies on the role of P2 receptors in the immune system. At about the same time, Greenberg et al demonstrated that J774 macrophages also expressed P2Y-like receptors coupled to Ca++ mobilization via a mechanism other than the ATP-permeabilizing receptor.76 This was made possible by the selection by Steinberg and colleagues75 of ATP-resistant J774 macrophage clones later shown to lack the P2X7 receptor.77,78 

According to the nomenclature proposed by Gordon,4 the macrophage-permeabilizing receptor was named P2Z, analogously to the mast cell and lymphocyte ATP receptor. The receptor responsible for ATP-dependent permeabilization has been referred to as P2Z until very recently and, even after the cloning of P2X7 and the demonstration that its transfection confers susceptibility to ATP-dependent permeabilization, some investigators prefer the P2Z nomenclature to indicate the native ATP-permeabilizing receptor, because it is not clear whether P2X7 is the only constitutive subunit or, rather, the native P2Z receptor is formed by the assembly of P2X7 in association with other P2X subtypes. However, because P2X7 reproduces all known effects of the native P2Z and cells resisting ATP-mediated permeabilization lack P2X7, we will assume heretofore that the macrophage P2Z and P2X7 receptors are the same molecule. As seen below, the picture is more complex in lymphocytes and other cells that do not undergo the typical ATP-dependent permeabilization, although they may express P2X7.

All murine macrophage lines so far investigated express P2Y receptors coupled to release of Ca++ from intracellular stores and IP3 generation, but the individual subtypes have not been investigated in detail. Functional and molecular expression of P2X7 has been shown in some murine cell lines and in mouse and rat peritoneal macrophages.60,75,79-83Monocyte-derived human macrophages are susceptible to ATP-mediated permeabilization and express P2X7.66,84,85Among human macrophage lines, THP-1 and U937 cells express P2Y receptors (P2Y2, P2Y4, and P2Y6),5,86-88 but only the THP-1 monocytic cell line has been reported to express P2X7 to a significant level.88 However, P2X7 receptor expression can differ significantly among cell batches propagated in different laboratories. Monocytes freshly isolated from peripheral blood express P2Y receptors but lack P2X7, whether investigated at the molecular or at the functional level. Although a few studies are available, it is generally agreed that, at the most, 15% to 17% of human monocytes undergo the plasma membrane permeability transitions diagnostic of P2X7 expression when stimulated with ATP.66,84 There appears to be an inverse correlation between P2Y2 and P2X7 expression during monocyte to macrophage maturation: P2Y2 messenger RNA (mRNA) declines while P2X7 mRNA increases.89 Up-regulation of P2X7 and acquisition of P2X7-dependent responses are detectable within 24 hours of seeding human monocytes on plastic dishes. Up-regulation of P2X7 and down-regulation of P2Y2 by the inflammatory mediators interferon-γ and tumor necrosis factor (TNF)-α and by lipopolysaccharide (LPS) have been reported.66,89 In addition, phorbol myristate acetate causes a decrease in P2Y2 mRNA in THP-1 cells.90 

The first report on the effect of exogenous nucleotides on macrophage function was a paper by Cohn and Parks.91 In this study the authors showed that addition of adenine nucleotides to a mouse macrophage culture resulted in a dramatic increase in pinocytic vesicle formation. After this early study, exogenous nucleotides as a stimulant for macrophages were basically neglected for several years and resurrected only in 1985 by Silverstein and coworkers,92 who reported that extracellular ATP inhibited Fc receptor–mediated phagocytosis and at the same time caused influx of Na+, efflux of K+, and an increase in [Ca++]i. In this study it was also for the first time suggested that macrophages expressed receptors specific for ATP. The possibility that these ATP effects could be due to ATP hydrolysis by plasma membrane ecto-ATPase was ruled out by subsequent papers by Steinberg and Silverstein60,75,76 that reported an in-depth characterization of the macrophage-permeabilizing ATP receptor. It was also soon clear that the ATP receptor coupled to release of Ca++ from intracellular stores (P2Y) and the ATP-permeabilizing (P2Z/P2X7) receptor were 2 separate entities with widely different nucleotide selectivity and affinity and likely involved in different responses.76 In J774 macrophages, the concentration of ATP giving one half of the maximal response (EC50) for Ca++ release from intracellular stores (and which therefore reflects activation of P2Y) is in the range of 50 to 70 μM. In microelectrode impalement experiments, the ATP EC50 for depolarization, presumably reflecting opening of P2X7, was reported to be between 250 and 400 μM,93 but a lower EC50 was reported for P2X7-triggered Ca++ rise in thioglycollate-elicited mouse peritoneal macrophages.94However, determinations based on the measurement of uptake of fluorescent markers give higher EC50 (1.0-1.5 mM ATP) for the activation of the native mouse P2X7receptor.76,95 The UTP EC50 for Ca++ release from intracellular stores is between 300 and 500 nM 76 and thus much lower than the ATP EC50.76,94 This suggests that macrophages express P2Y4 or P2Y6 or an endogenous yet to be identified uridine nucleotide–specific receptor. Therefore, it is clear that should ATP release occur in a tissue, macrophage P2Y receptors are likely to be activated more easily and more frequently than P2X7.

An early and, with hindsight, obvious proposal was that macrophages and, in general, inflammatory cells, could use P2Y receptors as very sensitive sensors of cell and tissue damage.76 After all, mammalian cells contain huge amounts (5-10 mM) of ATP in their cytosol; thus, any event that causes even a transient break in the plasma membrane will cause release of ATP into the pericellular environment. Furthermore, it is becoming apparent that frank cell injury or death might not even be necessary for ATP release because shear stress forces and stretching are also powerful stimuli for ATP leakage.8-12 J774 macrophages chemotact in response to micromolar concentrations of ADP but, rather intriguingly, not of UTP.96 Human macrophages in the vicinity of dying K562 cells have been shown in vitro to undergo an increase in [Ca++]i that can be closely mimicked by the addition of cell lysate or of ATP at micromolar doses.97Precedent treatment with the cell lysate made the macrophages refractory to the subsequent application of ATP, suggesting, although not proving, that a substance contained in the lysate and ATP might converge on the same receptor. Thus it can be hypothesized that ATP and other intracellular nucleotides function as early alarm signals that alert macrophages of even minor cell and tissue damage (a response could be elicited with as little as 100 nM ATP) (Figure3).

Fig. 3.

Fate of released ATP: possible role in leukocyte chemotaxis.

(A) The intracellular ATP concentration is in the 5 to 10 mM range; thus, an ATP gradient capable of driving leukocyte chemotaxis by acting at P2Y receptors is likely to occur at sites of cell or tissue damage. (B) ATP released into the pericellular milieu can either ligate P2Y or P2X receptors or be hydrolyzed by plasma membrane ecto-ATPases or ecto-ATP diphosphohydrolase (CD39). Hydrolysis of AMP by 5′-nucleotidase generates adenosine that activates P1 purinergic receptors.

Fig. 3.

Fate of released ATP: possible role in leukocyte chemotaxis.

(A) The intracellular ATP concentration is in the 5 to 10 mM range; thus, an ATP gradient capable of driving leukocyte chemotaxis by acting at P2Y receptors is likely to occur at sites of cell or tissue damage. (B) ATP released into the pericellular milieu can either ligate P2Y or P2X receptors or be hydrolyzed by plasma membrane ecto-ATPases or ecto-ATP diphosphohydrolase (CD39). Hydrolysis of AMP by 5′-nucleotidase generates adenosine that activates P1 purinergic receptors.

Close modal

The [Ca++]i rise could also be exploited by the macrophages for the potentiation of antimicrobial defense mechanisms. Nucleotides by themselves are unable to activate the macrophage NADPH oxidase but enhance superoxide generation stimulated by phagocytosable particles.98 It is conceivable that P2 receptors could also be used as an amplification system to spread the alarm by generating additional inflammatory mediators. In murine and human macrophages, extracellular ATP triggers release of TNF-α and interleukin (IL)-1β. P2 receptors involved in TNF-α release have not been identified and there is accordingly little insight into the molecular mechanism involved. An early report in Raw 264.7 murine macrophages99 suggests that ATP might act by enhancing the level of TNF-α mRNA, and this would not require priming by other proinflammatory factors, but a much more detailed investigation is clearly needed.

Participation of P2 receptors in IL-1β secretion is more firmly established and dissected mechanistically. It has been known for a while that LPS-dependent release of the proinflammatory cytokine IL-1β from macrophages and microglial cells, in contrast to peripheral blood monocytes, is a slow and inefficient process that leads to extracellular accumulation of minute amounts of this cytokine and mainly of the high-molecular-weight (31-34 kd), uncleaved, biologically inactive, procytokine form.100,101 This finding has led many investigators to postulate that a second stimulus is needed to trigger processing and secretion of the cytokine in its low-molecular-weight (17 kd) biologically active form, but the identity of this second stimulus has remained elusive. In 1991 Hogquist and colleagues observed that extracellular ATP triggered IL-1β processing and release,102 and in 1992 Gabel and coworkers reported that mature IL-1β formation could be induced by the K+ionophore nigericin.101 What is in common between nigericin and ATP? Perregaux and coworkers101 reasoned that both nigericin and ATP decrease intracellular K+levels60 and that perhaps this ionic perturbation was needed to activate the enzyme that cleaves pro–IL-1β into mature IL-1β, ie, IL-1β–converting enzyme (ICE), also known as caspase-1.103 Later studies fulfilled this prediction because ATP was shown to trigger IL-1β release via a nonlytic mechanism in many different mononuclear phagocytic cells, and release was inhibited by procedures that prevented K+ efflux85,104-106 (Figure 4). In support of a key role for K+ in ICE activation, Cheneval et al107 have shown that a reduction in the K+concentration leads to proteolytic cleavage of isolated recombinant ICE. Interestingly, although proteolytic activation of the isolated enzyme could be induced by a reduction in the concentration of other cations besides K+, autoprocessing of cytoplasmic ICE showed an absolute requirement for K+depletion.107 That ATP acts via ICE is also demonstrated by the ability of a specific ICE inhibitor, the tetrapeptide YVAD (Tyr-Val-Ala-Asp), to block ATP-dependent IL-1β maturation.106,108 Furthermore, macrophages isolated from mice deficient in ICE were unable to process pro–IL-1β upon challenge with LPS plus ATP.109 Finally, involvement of P2X7 in ATP-mediated ICE activation is supported by (a) agonist and antagonist profile of cytokine release,85 (b) blockade by a specific anti-P2X7 monoclonal antibody,55 and (c) detection of ICE proteolytic fragments (p20 and p10) in ATP-stimulated microglial cells.106,110 There are no clues as to how a decrease in K+ concentration may activate ICE autoprocessing; nevertheless, K+ provides a straightforward link between P2X7 and ICE because opening of the P2X7 channel/pore provides a very fast pathway for K+ efflux.60,104 It would be interesting to test whether the same K+-based mechanism of activation also applies to other caspases and how this may be involved in apoptosis.111 

Fig. 4.

Model for P2X7-mediated ICE/caspase-1 activation and IL-1β maturation.

Stimulation with LPS triggers IL-1β gene transcription and pro–IL-1β accumulation. Opening of the P2X7 pore by extracellular ATP causes a large K+ efflux that triggers proteolytic activation of ICE and cleavage of pro–IL-1β. Mature IL-1β is then secreted through an unknown pathway.

Fig. 4.

Model for P2X7-mediated ICE/caspase-1 activation and IL-1β maturation.

Stimulation with LPS triggers IL-1β gene transcription and pro–IL-1β accumulation. Opening of the P2X7 pore by extracellular ATP causes a large K+ efflux that triggers proteolytic activation of ICE and cleavage of pro–IL-1β. Mature IL-1β is then secreted through an unknown pathway.

Close modal

In human monocytes, ATP is a powerful stimulus not only for caspase-1 activation but also for the externalization of mature caspase-1 subunits.112 The meaning of this novel observation is elusive, but it may point to a possible function of activated caspase-1 either in the extracellular space or on the outer leaflet of the plasma membrane. In addition, ATP might trigger IL-1β release by alternative mechanisms, eg, by inducing exocytosis of IL-1β–containing specialized vesicles (late endosomes or lysosomes), as recently suggested by Rubartelli and coworkers.113 It is possible that the LPS signal for IL-1β release consists, at least in part, in an autocrine/paracrine stimulation mediated by ATP secretion, as suggested by studies in human and mouse macrophages and mouse microglia 9,114 

Participation of P2X7 in LPS-dependent activation of immune cells might have very interesting and far-reaching practical applications in the treatment of sepsis caused by gram-negative bacteria. In 1994 Proctor and colleagues115 showed that the ATP analogue, 2-methylthio-ATP (2-MeS-ATP), inhibited endotoxin-stimulated release of toxic mediators such as TNF-α and IL-1β and protected mice from endotoxin-induced death. Interpretation of this early experiment is not straightforward, because 2-MeS-ATP is an agonist at P2Y as well as P2X purinoceptors2,3; however, at P2X7 2-MeS-ATP acts as a partial agonist and thus it is conceivable that it might antagonize P2X7stimulation by locally secreted ATP and reduce LPS-dependent TNF-α and IL-1β release. Altogether, these observations suggest that P2X7 (and maybe other P2 receptors) take part in some crucial but as yet unknown steps in the complex chain of events leading to septic shock, either as a component of a paracrine/autocrine loop9 or as a binding site for LPS.115,116 

Stimulation with extracellular nucleotides also switches on the inducible nitric oxide synthase (iNOS),116-118 a key enzyme for the bactericidal activity of macrophages. Nucleotides per se are ineffective, but coexposure to low doses of ATP (or UTP) and LPS produces a much higher stimulation of iNOS activity compared with LPS alone. In murine Raw 264.7 macrophages a prolonged (18 hours) incubation was needed to elicit nitrite release, suggesting that P2 stimulation acted by increasing iNOS gene expression rather than by increasing enzyme activity. Other data suggest that P2 receptors are involved in NO generation in a rather more complex fashion. Denlinger and coworkers showed that pretreatment with 2-MeS-ATP prevented iNOS expression and NO generation due to the subsequent addition of LPS,117 raising the issue of the possible participation of P2 receptors in LPS-dependent signaling.116,117 In addition, it has been recently shown that NO production due toMycobacterium tuberculosis infection also occurs in P2X7 knockout mice and it is inhibited by P2 blockers,119 thus pointing to the participation of other P2X and P2Y receptors. There are an increasing number of papers suggesting that P2 receptors (namely P2X7) might have a role in endotoxin- or parasite-mediated macrophage stimulation. Besides the studies carried out in our laboratory showing that incubation of macrophages or microglia with oATP or apyrase inhibited LPS-dependent IL-1β release,9 other groups have reported that LPS-dependent NO release and nuclear factor (NF)-κB and mitogen-associated protein kinase (MAPK) activation are profoundly inhibited by oATP or by PPADS.116 MAPK in Raw 264.7 macrophages can also be stimulated via P2Y, but the putative purinergic receptor involved in LPS-dependent activation does not seem to be a member of the P2Y family because oATP or PPADS, which block LPS-dependent stimulation, do not affect MAPK stimulation by UTP.116 In the light of the report that ATP triggers NF-κB activation via P2X7 and that this activation is blocked by oATP,110 it is likely that the P2 receptor that participates in LPS-dependent macrophage activation is P2X7.

A common event observed in many reactions involving mononuclear phagocytes is multinucleation: often during chronic inflammatory reactions macrophages differentiate into epithelioid cells that eventually fuse into large polykarions named multinucleated giant cells (MGCs).120 Furthermore, in the bone, osteoclast precursors normally fuse to generate large elements with increasing bone resorption activity. MGCs are a common finding of widespread infectious diseases such as tuberculosis, but little is known about the molecular mechanism underlying fusion. In 1995, Falzoni et al66suggested that the P2X7 receptor could be involved in MGC formation. Monocyte-derived human macrophages can be induced to fuse in vitro by incubation with concanavalin A or phytohemagglutinin, provided that contaminating lymphocytes are also present.121Pretreatment with oATP fully inhibits this process, although other responses such as concanavalin A–dependent [Ca++]i changes, chemotaxis, or expression of plasma membrane molecules thought to take part in cell fusion (eg, CD11a, CD18, and CD54) are unaffected.66 We have extended these studies to J774 macrophages and selected several clones that either express P2X7 to a very high level (P2X7plus) or lack it altogether (P2X7less). P2X7plus cells spontaneously fuse in culture to form MGCs of different size and shape, containing from a few to 20 or more nuclei.77 A monoclonal antibody raised against the P2X7 outer domain prevents fusion of human macrophages in culture.122,123 

The participation in ICE activation and IL-1β release, and in MGC formation establishes an intriguing link between the P2X7 receptor and chronic inflammation. Experiments from Molloy et al124 and Lammas et al125 further strengthen this link. Both groups investigated the effect of extracellular ATP on macrophage cultures infected withMycobacterium bovis (bacille Calmette-Guérin) and reported that P2X7 activation caused killing of the phagocyte as well as of the intracellular pathogen. The mechanism involved is unknown, but a recent paper suggests that it might require activation of phospholipase D.126 Another possibility is that the known vesiculating activity of ATP91,127,128affects viability of the intracellular pathogen by increasing phagosome-lysosome fusion, as suggested by some of the electron microscopy images reported by Molloy et al.124 In macrophages, stimulation of a phosphatidylcholine-selective phospholipase D by P2X7 agonists was reported as early as 1992 83 and shown to be independent of pore formation and of the ensuing Ca++ influx.129 The mechanism by which an activated phospholipase D might partake in parasite killing is unknown but might be related to the enhanced rate of vesicle fusion observed in ATP-stimulated phagocytes. Ability of macrophages to eliminate intracellular parasites is enhanced upon activation with interferon-γ125; thus it might not be a coincidence that this cytokine and other proinflammatory factors also up-regulate P2X7 expression.66,130 It is also intriguing that to kill the intraphagosomal parasite, ATP concentrations cytotoxic for the macrophage need to be used or, in other words, parasite killing appears to be a consequence of macrophage death, as if intracellular parasite elimination via P2X7 was not a primary function of the receptor but rather an accessory consequence of its primary cytotoxic activity.

That extracellular ATP is a potent cytotoxic factor for macrophages was immediately apparent as soon as a thorough investigation of ATP receptors was started in these cells, and P2X7 was quickly identified as the culprit. Initially in Silverstein's and later in our laboratory, murine macrophage clones were selected that showed an almost absolute refractoriness to ATP-mediated cytotoxicity.60,75,76,95 These cells showed a normal mobilization of Ca++ from intracellular stores in response to ATP, but no permeabilization of the plasma membrane, and accordingly lacked reactivity to anti-P2X7 antibodies.77Blockade of the P2X7 receptor by oATP or KN-62 abrogated ATP-dependent cytotoxicity. The role of P2 receptors in cytotoxicity is usually tested in the presence of exogenous ATP, but it cannot be excluded that ATP spontaneously released by cell monolayers may provide a chronic cytolytic stimulus by acting as an autocrine/paracrine factor. We have tested this hypothesis in P2X7plus J774 macrophage cultures grown to confluence. These macrophage clones show an unusually high rate of spontaneous cell death that can be significantly reduced by pretreatment with oATP or coincubation with apyrase or hexokinase.131 In contrast to the P2X7plus clones, the P2X7less cells have a low rate of spontaneous death that is not affected by the presence of P2X7 blockers or ATP-hydrolyzing enzymes. The mechanism of ATP-dependent death can be either necrosis or apoptosis, depending on the length of incubation in the presence of the nucleotide and the dose. In our hands, ATP-pulsed J774 macrophages appear to die mostly by colloido-osmotic lysis; on the contrary, monocyte-derived human macrophages, which incidentally are more resistant to ATP-mediated cytotoxicity, are prone to die by apoptosis.124,125 It is possible that the propensity of these cells to die by apoptosis is related to their lower susceptibility to ATP because we have previously observed132 that, to set in motion the complex machinery involved in apoptosis, a certain amount of time is needed that is clearly unavailable in those cells that are so sensitive to ATP as to decease quickly. An in-depth investigation of the apoptotic pathways triggered by ATP in macrophages has not been yet carried out, but we know from work in microglial cells that caspase-1, -3, and -8 are activated with the subsequent cleavage of the caspase substrates PARP (poly [ADP-ribose] polymerase) and lamin B.106,133In addition, the crucial transcription factors, NF-κB and NFAT (nuclear factor of activated T cells), are also stimulated.110,134 

Dendritic cells are a newcomer in the purinergic field. It has been known for a while that epidermal Langerhans' cells posses a powerful plasma membrane formalin-resistant ecto-ATPase that has been used as a histochemical marker,135 but their physiologic function was never understood. In 1993 Girolomoni and coworkers136 demonstrated that human epidermal Langerhans' cells can be permeabilized by ATP, albeit to a lesser degree than human keratinocytes or J774 macrophages. However, inhibition of ecto-ATPase greatly enhanced sensitivity to ATP, and this led these authors to suggest that one of the possible physiologic functions of this ectoenzyme was protection of Langerhans' cells against the noxious effects of extracellular ATP. Scattered reports have then followed suggesting that phagocytic cells of the thymus reticulum express a P2X7-like ATP-permeabilizing receptor,137 but only during the last few years has a systematic study of these receptors been carried out in human and mouse dendritic cells.138-143 

Human dendritic cells were found to express mRNA for P2X1, P2X4, P2X5, and P2X7 as well as for P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11 receptors.140 Immunohistochemistry with an anti-P2X7 monoclonal antibody performed in human tonsils shows that a cell population of the marginal zone identified as dendritic cells heavily expresses P2X7.55Scanty pharmacologic data suggest that at least P2Y1, P2Y2, and P2Y4 are functional and mediate a Ca++ signal in these cells.139P2X7 functions have been investigated in detail in human and mouse dendritic cells, and available evidence suggests that this receptor mediates cytokine release and might also particpate in antigen presentation.141,142 During their investigation of P2Y receptors in human dendritic cells, Liu et al139 observed that dendritic cells redirect their dendrites toward a nearby patch pipette leaking ATP, an incidental finding that might suggest that dendritic cells, like other mononuclear phagocytes, exhibit a P2Y-mediated chemotactic response to ATP. In addition, it has been shown that stimulation with UTP or uridine diphosphate (but surprisingly not with ATP) provided a potent stimulus for the cytokine gene transcription and secretion.144 Given the high expression of P2X7, it is not surprising that dendritic cells are exceedingly sensitive to the cytotoxic activity of ATP and readily die by apoptosis.141,145 Whether this may have relevance in the overall process of modulation of the immune response is presently unknown.

Lymphocyte responsivity to nucleotides has been known for many years: In 1978 Gregory and Kern reported that extracellular ATP stimulated proliferation of mouse thymocytes146; Fishman et al in 1980 observed that in human peripheral lymphocytes ATP suppressed proliferation,147 presumably via generation of adenosine. In 1981 Ikehara et al148 in some way reconcilied these contrasting observations by showing that ATP stimulation of DNA synthesis was observed in lymphoid cells from the thymus and inhibition in cells from spleen, lymph nodes, and peripheral blood. These early observations were followed by a few other studies that overall were of little help in building a coherent picture of the responses of lymphoid cells to extracellular nucleotides, and they remained basically anecdotal.149,150 It was not until the end of the 1980s that a systematic approach to the study of purinergic receptor expression and function in lymphocytes was started.151-157 

Human B lymphocytes express P2Y receptors, as indicated by the ability of ATP and many other nucleotides (UTP, GTP, CTP, ITP, ADP, adenosine 5′-O-(3′-thiotriphosphate), ATPγS) to trigger Ca++ release from intracellular stores.158Human B lymphocytes also express P2X receptors, although of which particular subtype is still uncertain. Pharmacologic, electrophysiologic, and reverse transcriptase–polymerase chain reaction (RT-PCR) data suggest that the P2X7 receptor is present in these cells and might be up-regulated in chronic leukocytic leukemia cells (quite intriguingly it appears to be also up-regulated in lymphoblastoid cells from patients with Duchenne dystrophy).154,159-161 Rather surprisingly, however, B lymphocytes do not undergo the typical increase in permeability to aqueous solutes up to 900 d, suggesting that a pore of a smaller size, permeable to molecules up to 320 d, is generated by ATP: although ethidium bromide (314 d) is admitted, propidium bromide (414 d) is excluded.159-162 Furthermore, B lymphocytes are also poorly susceptible to ATP-mediated cytotoxicity. Human peripheral T lymphocytes lack P2Y receptors according to functional and pharmacologic studies but express a P2X-like ATP-activated channel.163 Unpublished data from our laboratory show that these cells express at the mRNA level P2X1, P2X4, and P2X7, although a significant variability is observed among samples from different subjects. ATP and BzATP cause in T lymphocytes a large influx of Na+ and Ca++ from the extracellular medium that is fully prevented by pretreatment with oATP. Like in B lymphocytes, ATP treatment of T lymphocytes generates a pore smaller than that seen in macrophages or in HEK293 cells transfected with the recombinant P2X7.48,62 This might be due to the assembly of P2X1 or P2X4 subunits together with P2X7 into the receptor complex, but this is purely speculative.

Expression of P2 receptors in mouse lymphocytes has been more thoroughly investigated. RT-PCR data show that murine thymocytes express the message for P2X1, P2Y1, and P2Y2 receptors and accordingly undergo Ca++release from intracellular stores and an increase in plasma membrane permeability to external cations when challenged with ATP.164-166 Steroid hormones or cross-linking of T-cell receptor (TCR) by anti-TCR monoclonal antibody causes a transient enhancement of P2Y2 mRNA, suggesting that this could be an early event in response to a variety of activatory stimuli.167 Sensitivity to ATP in thymocytes changes depending on the stage of maturation: CD4+CD8TCRhigh thymocytes were found to be very sensitive to ATP-mediated lysis166; large double-positive proliferating thymocytes were much less responsive compared with those terminally differentiated.168 As in the case of human lymphocytes, the cut-off of the ATP-gated plasma membrane channel for mouse lymphocytes appears to be slightly over 300 d (ethidium is admitted, propidium is excluded).166 Functional and pharmacologic data support expression of P2X7 by mouse lymphocytes,155,156,166 but there is little molecular evidence to corroborate this claim. At variance with human lymphocytes, mouse thymocytes are readily killed by ATP.155,156 In double-positive (CD4+CD8+) immature thymocyte populations, expression of P2X1 correlated with susceptibility to undergo apoptosis upon dexamethasone treatment,164 and incubation in the presence of apyrase blocked the process, as if dexamethasone induced P2X1activation via autocrine/paracrine release of ATP. Ability of extracellular ATP to cause apoptosis of mouse cell lines was initially documented in P-815 mastocytoma and YAC lymphoid cells and subsequentely extended to thymocytes.132,169 Among mouse lymphocytes, cytotoxic T-lymphocyte clones, cytotoxic T lymphocytes from peritoneal exudates, and lymphokine-activated killer cells turned out to be fully insensitive to ATP.132,155 Whether this is due to lack of P2 purinergic receptors or to high expression of ecto-ATPase/ectonucleotidases it is not known. The potent cytotoxic activity of extracellular ATP and the remarkable resistance of cytotoxic T lymphocytes and lymphokine-activated killer cells instigated speculations on the possible participation of ATP receptors in target cell killing, as a pathway alternative or parallel to perforin or lymphotoxin.156,157,170However, it has been very difficult to provide sound experimental support to this hypothesis.171 

The role of P2X receptors in the control of lymphocyte proliferation could be more complex than just being effectors of cell death. We have recently examined the effect of transduction of P2X7less human B-lymphoid cells with the P2X7 receptor complementary DNA and have surprisingly found that its expression confers a proliferation advantage in the absence of serum.172 We have not yet dissected the biochemical mechanism underlying the enhanced growth rate of the P2X7 transfectants, but we believe that it involves autocrine stimulation by ATP, because incubation with apyrase or hexokinase or pretreatment with oATP abrogated proliferation of P2X7 transfectants in the absence of serum.172 Whether these observations are relevant for tumors arising from hemopoietic cells is under investigation.

Stimulation with extracellular ATP causes shedding of the cell adhesion molecule L-selectin (CD62L) and the low-affinity receptor for IgE (CD23) from B chronic lymphocytic leukemic cells.173,174These cells express P2X7, and agonist/antagonist studies suggest that the receptor involved is P2X7.162CD23 and L-selectin are normally found in high amounts in sera from patients with B chronic lymphocytic leukemia, and this could be due to ATP-dependent shedding via P2X7 stimulation.

Scattered evidence for a role of extracellular nucleotides in granulocyte responses has been present for a while,175,176but a systematic investigation was only started at the end of the 1980s.177-181 Most studies concentrated on neutrophils, showing that ATP was able to trigger an increase in [Ca++]i, stimulation of phosphoinositide breakdown, superoxide anion generation, and granule exocytosis (both specific and azurophilic).182,183 In human neutrophils, ATP and UTP were reported to be equipotent for both the [Ca++]i increase and superoxide anion formation,178,179 and ATP was also shown to stimulate phospholipase C and diacylglycerol generation as well as protein kinase C activity.183,184 It is of great interest in the light of the proposed proinflammatory role of extracellular ATP that this nucleotide also increases membrane expression of CD11b/CD18 and adhesion to albumin-coated latex beads.185 Because ATP is released by the endothelium and its local concentration is likely to increase during inflammation as a consequence of inactivation of ecto-ATPases by oxygen radicals,186 up-regulation of adhesion molecules by this nucleotide could be of relevance for leukocyte migration across the vessel wall. ATP also enhances the adhesion between neutrophils and pulmonary endothelial cells, a mechanism that might be relevant in syndromes such as adult respiratory distress syndrome and septic shock.187,188 Of course, it cannot be excluded that the ATP effect could be at least in part mediated by its hydrolysis to adenosine,189 but recent data suggest that ecto-ATPase activity has an inhibitory rather than stimulatory effect on granulocyte-endothelium interaction.190 Dubyak and coworkers reported that chronic stimulation with ATPγS or UTP could drive differentiation of myelomonocytic progenitor cells (HL-60 and U937).191 Later studies showed that myeloid progenitors express P2Y1 at earlier and P2Y2 at later stages of differentiation.192 The myeloid progenitors HL-60 cells also express P2 Y11.39 

P2 subtype expression has not been thoroughly investigated in neutrophils, mainly because of the lack of selective antibodies. RT-PCR data show that human polymorphonuclear granulocytes express P2Y4 and P2Y6 but not P2Y1 or P2Y2 receptors.5 Among P2X receptors, the presence of P2X7 was shown by Northern blotting and immunocytochemistry.49 It has been suggested that human neutrophils might express receptors for diadenosine polyphosphates,193 but evidence for this is preliminary. Besides neutrophils, eosinophils also express P2 receptors coupled to [Ca++]i increases, actin reorganization, and stimulation of the NADPH oxidase.194,195 Interestingly, eosinophils show locomotive activity in response to ATP, ADP, and GTP.194 No data are available as to the P2 subtypes expressed.

ADP is one of the best-known activators of platelet aggregation,25,196-198 but the receptors involved have been, at least partially, identified only during the last 5 years. Stimulation with ADP causes release of Ca++from intracellular stores, Ca++ influx, phospholipase C activation, inhibition of stimulated adenylate cyclase, shape change, activation of fibrinogen receptors, and aggregation.199-201 ATP and ATP analogues are potent inhibitors of these responses. It has also been shown that ADP causes granule release and thromboxane A2production.202,203 It was initially thought that these effects were mediated by only one receptor named P2T; however, later studies led to the molecular and pharmacologic characterization in platelets of at least 2 of the known members of the P2 family: P2X1204-207 and P2Y1.208,209 With the availability of more selective platelet P2Y1 and P2X1 agonists and antagonist, it is becoming evident that the view that these are the P2 receptors solely responsible of ADP-mediated platelet activation is an oversimplification. It is clear that it is possible to block ADP-mediated inhibition of stimulated adenylate cyclase activity without decreasing the ADP-dependent [Ca++]irise. Thus it is postulated that ADP-triggered platelet activation is mediated by 3 receptors: One not yet cloned receptor (P2TAC) is coupled to inhibition of stimulated adenylate cyclase activity; a second (P2Y1) to phospholipase C activation, InsP3 formation, and Ca++ release from intracellular stores; and a third one (P2X1) to fast Ca++ influx across the plasma membrane.210,211 According to this proposal, the P2TAC receptor would coincide with the plateletP2YADP (written in italics to signify that it is not yet cloned) receptor of the nomenclature established by IUPHAR.2,27 Development of selective platelet P2 receptor antagonists has progressed further than in other cell types, and some have already reached clinical applications. Two thienopyridine compounds, ticlopidine and clopidogrel, inhibit ADP-triggered platelet aggregation presumably by selectively blockingP2YADP. The limited structure-relationship analysis so far carried out suggests that 2-alkylthio–substituted analogues of ATP and AMP (eg, 2-MeS-ATP; 2-methylthioadenylyl 5′-(β,γ-methylene)-diphosphonate [2-MeS-β,γ-Me-ATP]; 2-propylthioadenylyl 5′-(β,γ-difluoromethylene)-diphosphonate [ARL 66096]; and 2-propylthioadenylyl 5′-(β,γ-dichloromethylene)-diphosphonate [ARL 67085]) are selective competitive antagonists at P2YADP, and 3′-substituted AMP analogues (eg, adenosine 3′-phosphate 5′-phosphosulphate [A3P5PS]) are selective P2Y1antagonists.212 The pharmacology of the platelet P2Y1 receptor was clarified when only high-performance liquid chromatography–purified nucleotides were used and care was taken to avoid degradation of triphosphate analogues to the corresponding diphosphates. These precautions are seldom taken in the analysis of nucleotide effects in other cells, and this may lead to a re-evaluation of the agonist activity of ATP in other cell models. It is likely that full platelet activation requires stimulation and cooperative signaling of all 3 receptors, but the initial data from knockout mice suggest a central role for P2Y1, because P2Y1-deficient animals showed increased bleeding time and reduced collagen- and ADP-induced thromboembolism.213Interestingly, ADP-mediated adenylate cyclase inhibition was not reduced in platelets from the p2y1−/− mice. A P2X1-deficient (P2X1−/−) mouse is also available,214 but no data on platelet function in this animal have been published. A patient was described who was affected by what appears to be a selective deficit inP2YADP receptor expression215 and, on the other hand, expression of P2Y1 (and P2X1) was found to be normal in a patient affected by a severe deficiency of ADP-triggered platelet activation.216Presence of a functional ATP-activated P2X1 receptor raises intriguing questions on the interplay among different P2 receptors in platelet physiology, because ATP or ATP analogues were never shown to cause platelet activation. It might be that the P2X1receptor is chronically desensitized in vivo due to continuous leakage of ATP/ADP from blood or endothelial cells, but this issue clearly needs further scrutiny.217 Injury to blood cells or to the vessel wall releases ATP that is quickly dephosphorylated to ADP by ecto-ATPases expressed on the endothelium. Furthermore, platelets themselves are a major source of ATP and ADP that are stored within dense granules to a concentration of about 1 M. Thus, ADP-triggered secretion activates an autocatalytic cycle of autocrine/paracrine stimulation by released nucleotides.218 Release of ATP from platelets can also feed back on the endothelial cells, inducing secretion of other factors involved in hemostasis and inflammation, such as von Willebrand factor.219 The key role of extracellular ATP and P2 receptors in hemostasis has been underscored by the surprising phenotype of cd39/ATP diphosphohydrolase knockout (cd39−/−) mice.220 It was expected that these mice showed a thrombotic diathesis due to enhanced platelet aggregation, because CD39 has been considered an inhibitor of platelet activation. On the contrary,cd39−/−mice displayed prolonged bleeding times and failure to aggregate. These deficits were shown to be due to P2Y1 receptor desensitization dependent on an increased accumulation of extracellular ATP and were largely corrected by apyrase.

Effects of extracellular ATP on erythrocytes were initially reported in 1972 by Parker and Snow,221 who showed that this nucleotide caused Na+ influx and K+ efflux paralleled by an increase in water content. As later demonstrated in other cell types, ion fluxes were prevented by Mg2+ or hexokinase plus glucose and potentiated by ethylenediaminetetraacetic acid. All other nucleotides tested were ineffective. An increase in plasma membrane permeability of erythrocytes was also reported by Trams,222 who showed a dramatic accumulation of extracellular adenylates in the presence of extracellular ATP. These authors concluded that ATP caused a permeability change in erythrocyte plasma membrane that allowed for leakage of cytoplasmic ATP (“ATP-induced ATP release”). These data would suggest the expression by erythrocytes of a P2X7-like receptor, but no further characterization of this phenomenon was carried out. Release of ATP under hypoxic conditions has also been reported,223but the pathway involved was not elucidated. At variance with P2X, erythrocyte P2Y receptors are more thoroughly characterized. Avian red blood cells express a typical P2Y1 receptor coupled to phospholipase β activation via a G protein of the Gqfamily.224,225 Erythrocytes are an ideal “integrator unit” in the blood because they express P2 receptors and at the same time readily release ATP. These properties, on the one hand, make these cells sensitive to ATP released by other blood elements (eg, platelets) and, on the other hand, endow them with the ability to modulate the function of circulating or endothelial cells by secreting large amounts of this nucleotide. It has been proposed that ATP release from erythrocytes could contribute to regulation of local blood flow by acting at P2Y receptors on vascular endothelium.226,227ATP has a well-known NO releasing activity; thus, under ischemic conditions, when release from erythrocytes is maximal, ATP could be one of the local factors that counteract the decreased blood flow by inducing vasodilatation.

According to the few available studies, all hemopoietic precursors isolated from mouse bone marrow, as opposed to stromal cells, are highly sensitive to the cytotoxic effect of ATP.228,229This phenotypic property has made available a very efficient procedure for the isolation of highly purified marrow stromal cells or the deletion of hemopoietic cell precursors. The cytotoxic mechanisms appear to be dependent on the known pore-forming ability of ATP mediated by P2X7 activation and can be significantly enhanced by including in the reaction medium a low-molecular-weight nonpermeant poisonous agent such as potassium thiocyanate.228,229 This procedure might turn out helpful for the local treatment of tumors of hemopoietic origin.

For many years it was thought that receptors for extracellular nucleotides had a physiologic role only in excitable tissues; however, it is now increasingly clear that they are widespread and involved in signal transduction in several other tissues, including blood cells (Table 2). Drugs based onP2YADP antagonism are already in use as antithrombotic agents, and P2Y1 blockers are being developed for this same purpose. Besides thrombosis, another promising field of application of P2 agonist/antagonist is inflammation. Ability of P2 receptors to mediate chemotaxis (via P2Y), or cytotoxic responses and cytokine secretion (via P2X7), opens an entirely new perspective for the development of anti-inflammatory drugs. Chronic inflammatory diseases might be one of the first targets for the clinical application of selective P2X7 antagonists. These compounds might prove beneficial to reduce IL-1β release and granuloma formation. Finally, high expression of P2X7 by lymphocytic leukemia cells, and its participation in the control of cell death and proliferation, suggests a novel and as yet fully unexplored approach to the treatment of lymphoproliferative disorders.

Table 2.

P2 receptors expressed by blood cells

Cell typeP2YP2XReference no.
Rat/mouse peritonel macrophages P2Y P2X7 49,77, 79-82, 94  
BAC1.2F5 macrophages P2Y P2X7-like 83  
RAW 264.7 macrophages P2Y P2X7-like 81, 116  
J774 macrophages P2Y P2X7 60,75-77  
THP-1 macrophages P2Y2 P2X7 5,88  
KG-1 myeloblastic cells P2Y1  192  
HL-60 myeloid cells P2Y2, P2Y11 P2X1 39, 192, 232  
U937 monocytes P2Y2, P2Y6  5  
Human monocytes P2Y1, P2Y2, P2Y4, P2Y6  5, 89  
Human macrophages P2Y P2X7 77, 79, 89  
FSDC P2Y P2X7 141  
Mouse dendritic cells*  P2X7 141  
Human dendritic cells P2Y1, P2Y2, P2Y4, P2Y6, P2Y11 P2X1, P2X4, P2X5, P2X7 139, 140, 142  
Human Langerhans' cells  P2X7-like 136  
Human dendritic cells2-153  P2X7 55, 137  
P-815 mastocytoma  P2X7-like 132  
YAC lymphoma cells  P2X7-like 132  
Mouse lymphocytes  P2X7 49, 155, 166  
Murine thymocytes P2Y1, P2Y2 P2X1 164, 165, 167,168  
Human B lymphocytes P2Y P2X7 154, 158, 160, 161,162  
Human T lymphocytes  P2X1, P2X4, P2X7 163, O.R.B. et al, unpublished data, 1999  
Human PMN P2Y4, P2Y6 P2X7 5, 49  
Human platelets P2Y1 P2X1 204-209  
Erythrocytes P2Y1 P2X7-like 224,225  
RBL and rat mast cells P2Y P2X7-like 58, 59, 230,231  
Mouse hemopoietic precursors  P2X7-like 228,229  
Cell typeP2YP2XReference no.
Rat/mouse peritonel macrophages P2Y P2X7 49,77, 79-82, 94  
BAC1.2F5 macrophages P2Y P2X7-like 83  
RAW 264.7 macrophages P2Y P2X7-like 81, 116  
J774 macrophages P2Y P2X7 60,75-77  
THP-1 macrophages P2Y2 P2X7 5,88  
KG-1 myeloblastic cells P2Y1  192  
HL-60 myeloid cells P2Y2, P2Y11 P2X1 39, 192, 232  
U937 monocytes P2Y2, P2Y6  5  
Human monocytes P2Y1, P2Y2, P2Y4, P2Y6  5, 89  
Human macrophages P2Y P2X7 77, 79, 89  
FSDC P2Y P2X7 141  
Mouse dendritic cells*  P2X7 141  
Human dendritic cells P2Y1, P2Y2, P2Y4, P2Y6, P2Y11 P2X1, P2X4, P2X5, P2X7 139, 140, 142  
Human Langerhans' cells  P2X7-like 136  
Human dendritic cells2-153  P2X7 55, 137  
P-815 mastocytoma  P2X7-like 132  
YAC lymphoma cells  P2X7-like 132  
Mouse lymphocytes  P2X7 49, 155, 166  
Murine thymocytes P2Y1, P2Y2 P2X1 164, 165, 167,168  
Human B lymphocytes P2Y P2X7 154, 158, 160, 161,162  
Human T lymphocytes  P2X1, P2X4, P2X7 163, O.R.B. et al, unpublished data, 1999  
Human PMN P2Y4, P2Y6 P2X7 5, 49  
Human platelets P2Y1 P2X1 204-209  
Erythrocytes P2Y1 P2X7-like 224,225  
RBL and rat mast cells P2Y P2X7-like 58, 59, 230,231  
Mouse hemopoietic precursors  P2X7-like 228,229  

P2 receptor expression is based on functional and pharmacologic evidence, mRNA detection by reverse transcriptase–polymerase chain reaction, or reactivity with specific antibodies. For P2Y receptors, lack of a subscript indicates that, although functional and pharmacologic data show expression of P2Y receptors, the individual P2Y subtypes have not been yet identified. For P2X receptors, “P2X7-like” means that functional and pharmacologic evidence strongly suggest expression of P2X7, but molecular data are missing. Failure to list a P2Y or P2X receptor for a given cell type means that there is lack of evidence for its expression, whether at the functional, pharmacologic, or molecular level.

FSCD indicates fetal skin–derived dendritic cell; PMN, polimorphonuclear; RBL, rat basophilic leukemia.

*

Derived from bone marrow.

Derived from blood precursors.

Derived from epidermis.

F2-153

Derived from tonsils and lymph nodes.

Supported by grants by the Italian Ministry of Scientific Research (Cofin and 60%), the National Research Council of Italy (Target Project on Biotechnology), the Italian Association for Cancer Research, and Telethon of Italy. J.M.S. supported by a fellowship from the European Community (training grant BMH4-98-5146).

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Burnstock
G
A basis for distinguishing two types of purinergic receptors.
Cell Membrane Receptors for Drugs and Hormones: A Multidisciplinary Approach.
Straub
RW
Bolis
L
1978
107
119
Raven Press
New York, NY
2
Ralevic
V
Burnstock
G
Receptors for purines and pyrimidines.
Pharmacol Rev.
50
1998
413
492
3
Abbracchio
MP
Burnstock
G
Purinoceptors: are there families of P2X and P2Y purinoceptors?
Pharmacol Ther.
64
1994
445
475
4
Gordon
JL
Extracellular ATP: effects, sources and fate.
Biochem J.
233
1986
309
319
5
Jin
J
Dasari
VR
Sistare
FD
Kunapuli
SP
Distribution of P2Y receptor subtypes on hematopoietic cells.
Br J Pharmacol.
123
1998
789
794
6
Burnstock
G
The past, present and future of purine nucleotides as signalling molecules.
Neuropharmacology.
36
1997
1127
1139
7
Filippini
A
Taffs
RE
Sitkovsky
MV
Extracellular ATP in T-lymphocyte activation: possible role in effector functions.
Proc Natl Acad Sci U S A.
87
1990
8267
8271
8
Jorgensen
NR
Geist
ST
Civitelli
R
Steinberg
TH
ATP and gap junction-dependent intercellular calcium signalling in osteoblastic cells.
J Cell Biol.
139
1997
497
506
9
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.
185
1997
579
582
10
Mitchell
CH
Carrè
DA
McGlinn
AM
Stone
RA
Civan
MM
A release mechanism for stored ATP in ocular ciliary epithelial cells.
Proc Natl Acad Sci U S A.
95
1998
7174
7178
11
Cotrina
ML
Lin
JH
Alves-Rodriguez
A
et al
Connexins regulate calcium signalling by controlling ATP release.
Proc Natl Acad Sci U S A.
95
1998
15735
15740
12
Jiang
Q
Mak
D
Devidas
S
et al
Cystic fibrosis transmembrane conductance regulator-associated ATP release is controlled by a chloride sensor.
J Cell Biol.
143
1998
645
657
13
Holmsen
H
Storm
E
Day
HJ
Determination of ATP and ADP in blood platelets.
Anal Biochem.
46
1972
489
501
14
Meyers
KM
Holmsen
H
Seachord
CL
Comparative study of platelet dense granule constituents.
Am J Physiol.
243
1982
R454
R461
15
Kaczmarek
E
Koziak
K
Sevigny
J
et al
Identification and characterization of CD39 vascular ATP diphosphohydrolase.
J Biol Chem.
271
1996
33116
33122
16
Wang
TF
Guidotti
G
CD39 is an ecto-(Ca2+, Mg2+)-apyrase.
J Biol Chem.
271
1996
9898
9901
17
Zimmermann
H
Braun
N
Ecto-nucleotidases: molecular structures, catalytic properties and functional roles in the nervous system.
Prog Brain Res.
120
1999
371
385
18
Beaudoin
AR
Grondin
G
Gendron
FP
Immunolocalization of ATP diphosphohydrolase in pig and mouse brains and sensory organs of the mouse.
Prog Brain Res.
120
1999
387
395
19
Redegeld
FA
Caldwell
CC
Sitkovsky
MV
Ecto-protein kinases: ecto-domain phosphorylation as a novel target for pharmacological manipulation?
Trends Pharmacol Sci.
20
1999
453
459
20
Dubyak
GR
Signal transduction by P2-purinergic receptors for extracellular ATP.
Am J Respir Cell Mol Biol.
4
1991
295
300
21
Dubyak
GR
el-Moatassim
C
Signal transduction via P2-purinergic receptors for extracellular ATP and other nucleotides.
Am J Physiol.
265
1993
C577
C606
22
Di Virgilio
F
The P2Z purinoceptor: an intriguing role in immunity, inflammation and cell death.
Immunol Today.
16
1995
524
528
23
Fredholm
BB
Purines and neutrophil leukocytes.
Gen Pharmacol.
28
1997
345
350
24
Di Virgilio
F
Chiozzi
P
Falzoni
S
et al
Cytolytic P2X purinoceptors.
Cell Death Differ.
5
1998
191
199
25
Kunapuli
SP
Daniel
JL
P2 receptor subtypes in the cardiovascular system.
Biochem J.
336
1998
513
523
26
Hourani
S
Hall
DA
P2T purinoceptors: ADP receptors on platelets.
Ciba Found Symp.
198
1996
52
69
27
IUPHAR Compendium on Receptor Characterization and Classification. International Union of Pharmacology.
1998
IUPHAR Media
London
28
Burnstock
G
Kennedy
C
Is there a basis for distinguishing two types of P2-purinoceptor?
Gen Pharmacol.
16
1985
433
440
29
Barnard
EA
Burnstock
G
Webb
TE
G protein-coupled receptors for ATP and other nucleotides: a new receptor family.
Trends Pharmacol Sci.
15
1994
67
70
30
North
RA
Families of ion channels with two hydrophobic segments.
Curr Opin Cell Biol.
8
1996
474
483
31
Lustig
KD
Shiau
AK
Brake
AJ
Julius
D
Expression cloning of an ATP receptor from mouse neuroblastoma cells.
Proc Natl Acad Sci U S A.
90
1993
5113
5117
32
Webb
TE
Simon
J
Krishek
BJ
et al
Cloning and functional expression of a brain G-protein-coupled ATP receptor.
FEBS Lett.
324
1993
219
225
33
Communi
D
Motte
S
Boeynaems
J-M
Pirotton
S
Pharmacological characterization of the human P2Y4 receptor.
Eur J Pharmacol.
317
1996
383
389
34
Charlton
SJ
Brown
CA
Weisman
GA
Turner
JT
Erb
L
Boarder
ML
Cloned and transfected P2Y4 receptors: characterization of suramin and PPADS-insensitive response to UTP.
Br J Pharmacol.
119
1996
1301
1303
35
Chang
K
Hanaoka
K
Kumada
M
Takuwa
Y
Molecular cloning and functional analysis of a novel P2 nucleotide receptor.
J Biol Chem.
270
1995
26152
26158
36
Communi
D
Parmentier
M
Boeynaems
JM
Cloning, functional expression and tissue distribution of the human P2Y6 receptor.
Biochem Biophys Res Commun.
222
1996
303
308
37
Henderson
DJ
Elliot
DG
Smith
GM
Webb
TE
Dainty
IA
Cloning and characterization of a bovine P2Y receptor.
Biochem Biophys Res Commun.
212
1995
648
656
38
Ayyanathan
K
Webb
TE
Sandhu
AK
Athwal
RS
Barnard
EA
Kunapuli
SP
Cloning and chromosomal localization of the human P2Y1 purinoceptor.
Biochem Biophys Res Commun.
218
1996
783
788
39
Communi
D
Govaerts
C
Parmentier
M
Boeynaems
JM
Cloning of a human purinergic P2Y receptor coupled to phospholipase and adenylyl cyclase.
J Biol Chem.
272
1997
31969
31973
40
Purkiss
JR
Boarder
MR
Stimulation of phosphatidate synthesis in endothelial cells in response to P2-receptor activation. Evidence for phospholipase C and phospholipase D involvement, phosphatidate and diacylglycerol interconversion and the role of protein kinase C.
Biochem J.
287
1992
31
36
41
Dunn
PM
Blakeley
AGH
Suramin: a reversible P2-purinoceptor antagonist in the mouse vas deferens.
Br J Pharmacol.
93
1988
243
245
42
Boyer
JL
Romero-Avila
T
Schachter
JR
Harden
TK
Identification of competitive antagonists of the P2Y1 receptor.
Mol Pharmacol.
50
1996
1323
1329
43
Kim
YC
Gallo-Rodriguez
C
Jang
SY
et al
Acyclic analogues of deoxyadenosine 3′,5′-bisphosphates as P2Y1 receptor antagonists.
J Med Chem.
43
2000
746
755
44
Brake
AJ
Wagenbach
MJ
Julius
D
New structural motif for ligand-gated ion channels defined by an ionotropic ATP receptor.
Nature.
371
1994
519
523
45
Valera
S
Hussy
N
Evans
RJ
et al
A new class of ligand-gated ion channels defined by P2X receptor for extracellular ATP.
Nature.
371
1994
516
519
46
Buell
G
Collo
G
Rassendren
F
P2X receptors: an emerging channel family.
Eur J Neurosci.
8
1996
2221
2228
47
Soto
F
Garcia-Guzman
M
Stuhmer
W
Cloned ligand-gated channels activated by extracellular ATP (P2X receptors).
J Membr Biol.
160
1997
91
100
48
Surprenant
A
Rassendren
F
Kawashima
E
North
RA
Buell
G
The cytolytic P2Z receptor for extracellular ATP identified as a P2X receptor (P2X7).
Science.
272
1996
735
738
49
Collo
G
Neidhart
S
Kawashima
E
Kosco-Vilbois
M
North
RA
Buell
G
Tissue distribution of the P2X7 receptor.
Neuropharmacology.
36
1997
1277
1283
50
Di Virgilio
F
Falzoni
S
Mutini
C
Sanz
JM
Chiozzi
P
Purinergic P2X7 receptor: a pivotal role in inflammation and immunomodulation.
Drug Dev Res.
45
1998
207
213
51
Lewis
C
Neidhart
S
Holy
C
North
RA
Buell
G
Surprenant
A
Co-expression of P2X2 and P2X3 receptor subunits can account for ATP-gated currents in sensory neurons.
Nature.
377
1995
432
435
52
Nicke
A
Baumert
HG
Rettinger
J
et al
P2X1 and P2X3 form stable trimers: a novel structural motif of ligand-gated ion channels.
EMBO J.
17
1998
3016
3028
53
Evans
RJ
Surprenant
A
North
RA
P2X receptors
Turner JT, Weisman GA, Fedan. JS, eds. The P2 Nucleotide Receptors.
1998
43
61
Humana Press
Totowa, NJ
54
Afework
M
Burnstock
G
Distribution of P2X receptors in the rat adrenal gland.
Cell Tissue Res.
298
1999
449
456
55
Buell
G
Chessell
IP
Michel
D
et al
Blockade of human P2X7 receptor function with a monoclonal antibody.
Blood.
92
1998
3521
3528
56
Chen
C-C
Akopian
AN
Sivilotti
L
Colquhoun
D
Burnstock
G
Wood
JN
A P2X purinoceptor expressed by a subset of sensory neurons.
Nature.
377
1995
428
431
57
Hashimoto
M
Kokubun
S
Contribution of P2-purinoceptors to neurogenic contraction of rat urinary bladder smooth muscle.
Br J Pharmacol.
115
1995
636
640
58
Cockcroft
S
Gomperts
BD
ATP induces nucleotide permeability in rat mast cells.
Nature.
279
1979
541
542
59
Cockcroft
S
Gomperts
BD
The ATP4− receptor of rat mast cells.
Biochem J.
188
1980
789
798
60
Steinberg
TH
Silverstein
SC
Extracellular ATP4− promotes cation fluxes in the J774 mouse macrophage cell line.
J Biol Chem.
262
1987
3118
3122
61
Pizzo
P
Zanovello
P
Bronte
V
Di Virgilio
F
Extracellular ATP causes lysis of mouse thymocytes and activates a plasma membrane ion channel.
Biochem J.
274
1991
139
144
62
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.
272
1997
5482
5486
63
Trezise
DJ
Bell
NJ
Kennedy
I
Humphrey
PPA
Effects of divalent cations on the potency of ATP and related agonists in the rat isolated vagus nerve: implications for P2 purinoceptor classification.
Br J Pharmacol.
113
1994
463
470
64
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.
268
1993
8199
8203
65
Redegeld
FA
Smith
P
Apasov
S
Sitkovsky
MV
Phosphorylation of T-lymphocyte plasma membrane-associated proteins by ectoprotein kinases: implications for a possible role for ectophosphorylation in T-cell effector functions.
Biochim Biophys Acta.
1328
1997
151
165
66
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.
95
1995
1207
1216
67
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.
112
1994
946
950
68
Schulze-Lohoff
E
Hugo
C
Rost
S
et al
Extracellular ATP causes apoptosis and necrosis of cultured mesangial cells via P2Z/P2X7 receptors.
Am J Physiol.
275
1998
F962
F971
69
Solini
A
Chiozzi
P
Morelli
A
Fellin
R
Di Virgilio
F
Human primary fibroblasts in vitro express a purinergic P2X7 receptor coupled to ion fluxes, microvesicle formation and IL-6 release.
J Cell Sci.
112
1999
297
305
70
Gargett
CE
Wiley
JS
The isoquinoline derivative KN-62: a potent antagonist of the P2Z-receptor of human lymphocytes.
Br J Pharmacol.
120
1997
1483
1490
71
Kato
M
Hagiwara
M
Nimura
Y
Shionoya
S
Hidaka
H
Purification and characterization of calcium-calmodulin kinase II from human parathyroid glands.
J Endocrinol.
131
1991
155
162
72
Blanchard
DK
Wei
S
Duan
C
Pericle
F
Diaz
JI
Djeu
JY
Role of extracellular adenosine triphosphate in the cytotoxic T lymphocyte-mediated lysis of antigen presenting cells.
Blood.
85
1995
3173
3178
73
Humphreys
BD
Virginio
C
Surprenant
A
Rice
J
Dubyak
GR
Isoquinolines as antagonists of the P2X7 nucleotide receptor: high selectivity for the human versus rat receptor homologues.
Mol Pharmacol.
54
1998
22
32
74
Jiang
LH
Mackenzie
AB
North
RA
Surprenant
A
Brilliant blue G selecticely blocks ATP-gated rat P2X7 receptors.
Mol Pharmacol.
58
2000
82
88
75
Steinberg
TH
Newman
AS
Swanson
JA
Silverstein
SC
ATP4− permeabilizes the plasma membrane of mouse macrophages to fluorescent dyes.
J Biol Chem.
262
1987
8884
8888
76
Greenberg
S
Di Virgilio
F
Steinberg
TH
et al
Extracellular nucleotides mediate Ca2+ fluxes in J774 macrophages by two distinct mechanisms.
J Biol Chem.
263
1988
10337
10343
77
Chiozzi
P
Sanz
JM
Ferrari
D
et al
Spontaneous cell fusion in macrophage cultures expressing high levels of the P2Z/P2X7 receptor.
J Cell Biol.
138
1997
697
706
78
Ferrari
D
Chiozzi
P
Falzoni
S
et al
ATP-mediated cytotoxicity in microglial cells.
Neuropharmacology.
36
1997
1295
1301
79
Di Virgilio
F
Meyer
BC
Greenberg
S
Silverstein
SC
Fc receptor-mediated phagocytosis occurs in macrophages at exceedingly low cytosolic Ca2+ levels.
J Cell Biol.
106
1988
657
666
80
Naumov
AP
Kaznacheyeva
EV
Kiselyov
KI
Kuryshev
YA
Mamin
AG
Mozhayeva
GN
ATP-activated inward currents and calcium-permeable channels in rat macrophage plasma membranes.
J Physiol.
486
1995
323
337
81
Lin
WW
Lee
YT
Pyrimidinoceptor-mediated activation of phospholipase C and phospholipase A2 in RAW 264.7 macrophages.
Br J Pharmacol.
119
1996
261
268
82
Ichinose
M
Modulation of phagocytosis by P2-purinergic receptors in mouse peritoneal macrophages.
Jpn J Physiol.
45
1995
707
721
83
el-Moatassim
C
Dubyak
GR
A novel pathway for the activation of phospholipase D by P2Z purinergic receptors in BAC1.2F5 macrophages.
J Biol Chem.
267
1992
23664
23673
84
Hickman
SE
El Khoury
J
Greenberg
S
Schieren
I
Silverstein
SC
P2Z adenosine triphosphate receptor activity in cultured human monocyte-derived macrophages.
Blood.
84
1994
2452
2456
85
Ferrari
D
Chiozzi
P
Falzoni
S
et al
Extracellular ATP triggers IL-1β release by activating the purinergic P2Z receptor of human macrophages.
J Immunol.
159
1997
1451
1458
86
Cowen
DS
Lazarus
HM
Shurin
SB
Stoll
SE
Dubyak
GR
Extracellular adenosine triphosphate activates calcium mobilization in human phagocytic leukocytes and neutrophil/monocyte progenitor cells.
J Clin Invest.
83
1989
1651
1660
87
Fredholm
BB
Abbracchio
MP
Burnstock
G
et al
Towards a revised nomenclature for P1 and P2 receptors.
Trends Pharmacol Sci.
18
1997
79
82
88
Humphreys
BD
Dubyak
GR
Induction of the P2z/P2X7 nucleotide receptor and associated phospholipase D activity by lipopolysaccharide and IFNγ in the human THP-1 monocytic cell line.
J Immunol.
157
1996
5627
5637
89
Dubyak
GR
Clifford
EE
Humphreys
BD
Kertesy
SB
Martin
KA
Expression of multiple ATP receptor subtypes during the differentiation and inflammatory activation of myeloid leukocytes.
Drug Dev Res.
39
1996
269
278
90
Martin
KA
Kertesy
SB
Dubyak
GR
Down-regulation of P2U-purinergic nucleotide receptor messenger RNA expression during in vitro differentiation of human myeloid leukocytes by phorbol esters or inflammatory activators.
Mol Pharmacol.
51
1997
97
108
91
Cohn
ZA
Parks
E
The regulation of pinocytosis in mouse macrophages, III: the induction of vesicle formation by nucleosides and nucleotides.
J Exp Med.
125
1967
457
466
92
Sung
SS
Young
JD
Origlio
AM
Heiple
JM
Kaback
HR
Silverstein
SC
Extracellular ATP perturbs transmembrane ion fluxes, elevates cytosolic [Ca2+]i, and inhibits phagocytosis in mouse macrophages.
J Biol Chem.
260
1985
13442
13449
93
Buisman
HP
Steinberg
TH
Fischbarg
J
et al
Extracellular ATP induces a large nonselective conductance in macrophage plasma membranes.
Proc Natl Acad Sci U S A.
85
1988
7988
7992
94
Alonso-Torre
SR
Trautmann
A
Calcium responses elicited by nucleotides in macrophages. Interaction between two receptor subtypes.
J Biol Chem.
268
1993
18640
18647
95
Murgia
M
Pizzo
P
Steinberg
TH
Di Virgilio
F
Characterization of the cytotoxic effect of extracellular ATP in J774 mouse macrophages.
Biochem J.
288
1992
897
901
96
McCloskey
MA
Fan
Y
Luther
S
Chemotaxis of rat mast cells toward adenine nucleotides.
J Immunol.
163
1999
970
977
97
Oshimi
Y
Miyazaki
S
Oda
S
ATP-induced Ca2+ response mediated by P2U and P2Y purinoceptors in human macrophages: signalling from dying cells to macrophages.
Immunology.
98
1999
220
227
98
Schmid-Antomarchi
H
Schmid-Alliana
A
Romey
G
et al
Extracellular ATP and UTP control the generation of reactive oxygen intermediates in human macrophages through the opening of a charybdotoxin-sensitive Ca2+-dependent K+ channel.
J Immunol.
159
1997
6209
6215
99
Tonetti
M
Sturla
L
Giovine
M
Benatti
U
De Flora
A
Extracellular ATP enhances mRNA levels of nitric oxice synthase and TNFα in lipopolysaccharide-treated RAW 264.7 murine macrophages.
Biochem Biophys Res Commun.
214
1995
125
130
100
Hogquist
KA
Unanue
ER
Chaplin
DD
Release of IL-1 from mononuclear phagocytes.
J Immunol.
147
1991
2181
2186
101
Perregaux
D
Barberia
J
Lanzetti
AJ
Geoghegan
KF
Carty
TJ
Gabel
CA
IL-1β maturation: evidence that mature cytokine formation can be induced specifically by nigericin.
J Immunol.
149
1992
1294
1303
102
Hogquist
KA
Nett
MA
Unanue
ER
Chaplin
DD
Interleukin 1 is processed and released during apoptosis.
Proc Natl Acad Sci U S A.
88
1991
8485
8489
103
Budihardo
I
Oliver
H
Lutter
M
Luo
X
Wang
X
Biochemical pathways of caspase activation during apoptosis.
Annu Rev Cell Dev Biol.
15
1999
269
290
104
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.
269
1994
15195
15203
105
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.
156
1996
1531
1539
106
Sanz
JM
Di Virgilio
F
Kinetics and mechanism of ATP-dependent IL-1β release from microglial cells.
J Immunol.
164
2000
4893
4898
107
Cheneval
D
Ramage
P
Kastelic
T
et al
Increased mature interleukin-1β secretion from THP-1 cells induced by nigericin is a result of activation of p45 IL-1β-converting enzyme processing.
J Biol Chem.
273
1998
17846
17851
108
Perregaux
DG
Gabel
CA
Post-translational processing of murine IL-1: evidence that ATP-induced release of IL-1α and IL-1β occurs via a similar mechanism.
J Immunol.
160
1998
2469
2477
109
Li
P
Allen
H
Banerjee
S
et al
Mice deficient in IL-1β-convering enzyme are defective in production of mature IL-1β and resistant to endotoxic shock.
Cell.
80
1995
401
411
110
Ferrari
D
Wesselborg
S
Bauer
MKA
Schulze-Osthoff
K
Extracellular ATP activates transcription factor NF-κB through the P2Z purinoceptor by selectively targeting NF-κB p65.
J Cell Biol.
139
1997
1635
1643
111
Dallaporta
B
Marchetti
P
de Pablo
MA
et al
Plasma membrane potential in thymocyte apoptosis.
J Immunol.
162
1999
6534
6542
112
Laliberte
RE
Eggler
J
Gabel
CA
ATP treatment of human monocytes promotes caspase-1 maturation and externalization.
J Biol Chem.
274
1999
36944
36951
113
Andrei
C
Dazzi
C
Lotti
L
Torrisi
MR
Chimini
G
Rubartelli
A
The secretory route of the leaderless protein interleukin 1β involved exocytosis of the endolysosome-related vesicles.
Mol Biol Cell.
10
1999
1463
1475
114
Sperlagh
B
Hasko
G
Nemeth
Z
Vizi
ES
ATP released by LPS increases nitric oxide production in Raw 264.7 macrophage cell line via P2Z/P2X7 receptors.
Neurochem Int.
33
1998
209
215
115
Proctor
RA
Denlinger
LC
Leventhal
PS
et al
Protection of mice from endotoxic death by 2-methylthio-ATP.
Proc Natl Acad Sci U S A.
91
1994
6017
6020
116
Hu
Y
Fisette
PL
Denlinger
LC
et al
Purinergic receptor modulation of lipopolysaccharide signalling and inducible nitric-oxide synthase expression in Raw 264.7 macrophages.
J Biol Chem.
273
1998
27170
27175
117
Denlinger
LC
Fisette
PL
Garis
KA
et al
Regulation of inducible nitric oxide synthase expression by macrophage purinoreceptors and calcium.
J Biol Chem.
271
1996
337
342
118
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.
203
1994
430
435
119
Sikora
A
Liu
J
Brosnan
C
Buell
G
Chessel
I
Bloom
BR
Cutting edge: purinergic signaling regulates radical-mediated bacterial killing mechanisms in macrophages through a P2X7-independent mechanism.
J Immunol.
163
1999
558
561
120
Fais
S
Burgio
VL
Capobianchi
MR
Gessani
S
Pallone
F
Belardelli
F
The biological relevance of polykarions in the immune response.
Immunol Today.
18
1997
522
527
121
Takashima
T
Ohnishi
K
Tsuyuguchi
I
Kishimoto
S
Differential regulation of formation of multinucleated giant cells from concanavalin A–stimulated human blood monocytes by IFN-γ and IL-4.
J Immunol.
150
1993
3002
3010
122
Di Virgilio
F
Falzoni
S
Chiozzi
P
Sanz
JM
Ferrari
D
Buell
GN
ATP receptors and giant cell formation.
J Leukoc Biol.
66
1999
723
726
123
Falzoni
S
Chiozzi
P
Ferrari
D
Buell
G
Di Virgilio
F
P2X7 receptor and polykarion formation.
Mol Biol Cell.
11
2000
3169
3176
124
Molloy
A
Laochumroonvorapong
P
Kaplan
G
Apoptosis, but not necrosis, of infected monocytes is coupled with killing of intracellular bacillus Calmette-Guerin.
J Exp Med.
180
1994
1499
1509
125
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.
7
1997
433
444
126
Kusner
DJ
Adams
J
ATP-induced killing of virulent Mycobacterium tuberculosis within human macrophages requires phospholipase D.
J Immunol.
164
2000
379
388
127
Steinberg
TH
Buisman
HP
Greenberg
S
Di Virgilio
F
Silverstein
SC
Effects of extracellular ATP on mononuclear phagocytes.
Ann N Y Acad Sci.
603
1990
120
129
128
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.
1278
1996
125
130
129
el-Moatassim
C
Dubyak
GR
Dissociation of the pore-forming and phospholipase D activities stimulated via P2Z purinergic receptors in BAC1.2F5 macrophages. Product inhibition of phospholipase D enzyme activity.
J Biol Chem.
268
1993
15571
15581
130
Humphreys
BD
Dubyak
GR
Modulation of P2X7 nucleotide receptor expression by pro- and anti-inflammatory stimuli in THP-1 monocytes.
J Leukoc Biol.
64
1998
265
273
131
Chiozzi
P
Murgia
M
Falzoni
S
Ferrari
D
Di Virgilio
F
Role of the purinergic P2Z receptor in spontaneous cell death in J774 macrophage cultures.
Biochem Biophys Res Commun.
218
1996
176
181
132
Zanovello
P
Bronte
V
Rosato
A
Pizzo
P
Di Virgilio
F
Responses of mouse lymphocytes to extracellular ATP. II. Extracellular ATP causes cell type-dependent lysis and DNA fragmentation.
J Immunol.
145
1990
1545
1550
133
Ferrari
D
Los
M
Bauer
MK
Vandenabeele
P
Wesselborg
S
Schulze-Osthoff
K
P2Z purinoceptor ligation induces activation of caspases with distinct roles in apoptotic and necrotic alterations of cell death.
FEBS Lett.
447
1999
71
75
134
Ferrari
D
Stroh
C
Schulze-Osthoff
K
P2X7/P2Z purinoceptor-mediated activation of transcription factor NFAT in microglial cells.
J Biol Chem.
274
1999
13205
13210
135
Chaker
MB
Tharp
MD
Bergstresser
PR
Rodent epidermal Langerhans cells demonstrate greater histochemical specificity for ADP than for ATP and AMP.
J Invest Dermatol.
82
1984
496
500
136
Girolomoni
G
Santantonio
ML
Pastore
S
Bergstresser
PR
Giannetti
A
Cruz
PD
Jr
Epidermal Langerhans cells are resistant to the permeabilizing effects of extracellular ATP: in vitro evidence supporting a protective role of membrane ATPase.
J Invest Dermatol.
100
1993
282
287
137
Coutinho-Silva
R
Alves
LA
Campos-de-Carvalho
AC
Savino
W
Persechini
PM
Characterization of P2Z purinergic receptors on phagocytic cells of the thymic reticulum in culture.
Biochim Biophys Acta.
1280
1996
217
222
138
Alves
LA
Coutinho-Silva
R
Savino
W
Extracellular ATP: a further modulator in neuroendocrine control of the thymus.
Neuroimmunomodulation.
6
1999
81
89
139
Liu
Q
Bohlen
H
Titzer
S
et al
Expression and a role of functionally coupled P2Y receptors in human dendritic cells.
FEBS Lett.
445
1999
402
408
140
Berchtold
S
Ogilvie
AL
Bogdan
C
et al
Human monocyte derived dendritic cells express functional P2X and P2Y receptors as well as ecto-nucleotidases.
FEBS Lett.
458
1999
424
428
141
Mutini
C
Falzoni
S
Ferrari
D
et al
Mouse dendritic cells express the P2X7 purinergic receptor: characterization and possible participation in antigen presentation.
J Immunol.
163
1999
1958
1965
142
Ferrari D, La Sala A, Chiozzi P, et al. The P2 purinergic receptors of human dendritic cells: identification and coupling to cytokine release. FASEB J. In press.
143
Nihei
OK
de Carvalho
AC
Savino
W
Alves
LA
Pharmacological properties of P2Z/P2X7 receptor characterized in murine dendritic cells: role on the induction of apoptosis.
Blood.
96
2000
996
1005
144
Marriott
I
Inscho
EW
Bost
KL
Extracellular uridine nucleotides initiate cytokine production by murine dendritic cells.
Cell Immunol.
195
1999
147
156
145
Coutinho-Silva
R
Persechini
PM
Bisaggio
RD
et al
P2Z/P2X7 receptor-dependent apoptosis of dendritic cells.
Am J Physiol.
276
1999
C1139
C1147
146
Gregory
SH
Kern
M
Adenosine and adenine nucleotides are mitogenic for mouse thymocytes.
Biochem Biophys Res Commun.
83
1978
1111
1116
147
Fishman
RF
Rubin
AL
Novogrodsky
A
Stenzel
KH
Selective suppression of blastogenesis induced by different mitogens: effect of noncyclic adenosine-containing compounds.
Cell Immunol.
54
1980
129
139
148
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.
127
1981
1834
1838
149
Schmidt
A
Ortaldo
JR
Herberman
RB
Inhibition of human natural killer cell reactivity by exogenous adenosine 5′-triphosphate.
J Immunol.
132
1984
146
150
150
Lin
J
Krishnaraj
R
Kemp
RG
Exogenous ATP enhances calcium influx in intact thymocytes.
J Immunol.
135
1985
3403
3410
151
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.
927
1987
437
444
152
el-Moatassim
C
Maurice
T
Mani
JC
Dornand
J
The [Ca2+]i increase induced in murine thymocytes by extracellular ATP does not involve ATP hydrolysis and is not related to phosphoinositide metabolism.
FEBS Lett.
242
1989
391
396
153
el-Moatassim
C
Bernad
N
Mani
JC
Dornand
J
Extracellular ATP induces a non-specific permeability of thymocyte plasma membranes.
Biochem Cell Biol.
67
1989
495
502
154
Wiley
JS
Dubyak
GR
Extracellular adenosine triphosphate increases cation permeability of chronic lymphocytic leukemic lymphocytes.
Blood.
73
1989
1316
1323
155
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 effect of ATP.
J Immunol.
143
1989
1955
1960
156
Filippini
A
Taffs
RE
Agui
T
Sitkovsky
MV
Ecto-ATPase activity in cytolytic T lymphocytes. Protection from the cytolytic effects of extracellular ATP.
J Biol Chem.
265
1990
334
340
157
Di Virgilio
F
Pizzo
P
Zanovello
P
Bronte
V
Collavo
D
Extracellular ATP as a possible mediator of cell-mediated cytotoxicity.
Immunol Today.
11
1990
274
277
158
Padeh
S
Cohen
A
Roifman
CM
ATP-induced activation of human B lymphocytes via P2-purinoceptors.
J Immunol.
146
1991
1626
1632
159
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.
305
1993
54
60
160
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.
267
1994
C886
C892
161
Markwardt
F
Lohn
M
Bohm
T
Klapperstuck
M
Purinoceptor-operated cationic channels in human B-lymphocytes.
J Physiol.
498
1997
143
151
162
Wiley
JS
Gargett
CE
Zhang
W
Snook
MB
Jamieson
GP
Partial agonists and antagonists reveal a second permeability state of human lymphocyte P2Z/P2X7 channel.
Am J Physiol.
275
1998
C1224
C1231
163
Baricordi
OR
Ferrari
D
Melchiorri
L
et al
An ATP-activated channel is involved in mitogenic stimulation of human T lymphocytes.
Blood.
87
1996
682
690
164
Chvatchko
Y
Valera
S
Aubry
JP
Renno
T
Buell
G
Bonnefoy
JY
The involvement of an ATP-gated ion channel, P2X1, in thymocyte apoptosis.
Immunity.
5
1996
275
283
165
Apasov
SG
Koshiba
M
Chused
TM
Sitkovsky
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.
158
1997
5095
5105
166
Chused
TM
Apasov
S
Sitkovsky
M
Murine T lymphocytes modulate activity of an ATP-activated P2Z-type purinoceptor during differentiation.
J Immunol.
157
1996
1371
1380
167
Koshiba
M
Apasov
S
Sverdlov
V
et al
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.
94
1997
831
836
168
Ross
PE
Ehring
GR
Cahalan
MD
Dynamics of ATP-induced calcium signalling in single mouse thymocytes.
J Cell Biol.
138
1997
987
998
169
Zheng
LM
Zychlinsky
A
Liu
CC
Ojcius
DM
Young
JD
Extracellular ATP as a trigger for apoptosis or programmed cell death.
J Cell Biol.
112
1991
279
288
170
Steinberg
TH
Di Virgilio
F
Cell-mediated cytotoxicity: ATP as an effector and the role of target cells.
Curr Opin Immunol.
3
1991
71
75
171
Zambon
A
Bronte
V
Di Virgilio
F
et al
Role of extracellular ATP in cell-mediated cytotoxicity: a study with ATP-sensitive and ATP-resistant macrophages.
Cell Immunol.
156
1994
458
467
172
Baricordi
OR
Melchiorri
L
Adinolfi
E
et al
Increased proliferation rate of lymphoid cells transfected with the P2X7 ATP receptor.
J Biol Chem.
274
1999
33206
33208
173
Jamieson
GP
Snook
MB
Thurlow
PJ
Wiley
JS
Extracellular ATP causes loss of L-selectin from human lymphocytes via occupancy of P2Z purinoceptors.
J Cell Physiol.
166
1996
637
642
174
Gu
B
Bendall
LG
Wiley
JS
Adenosine triphosphate-induced shedding of CD23 and L-selectin (CD62L) from lymphocytes is mediated by the same receptor but different metalloproteases.
Blood.
92
1998
946
951
175
Tou
JS
Maier
C
Phospholipid metabolism and lysosomal enzyme secretion by leukocytes. Effects of dibutyryl cyclic adenosine 3′:5′-monophosphate and ATP.
Biochim Biophys Acta.
451
1976
353
362
176
LeRoy
EC
Ager
A
Gordon
JL
Effects of neutrophil elastase and other proteases on porcine aortic endothelial prostaglandin I2 production, adenine nucleotide release, and responses to vasoactive agents.
J Clin Invest.
74
1984
1003
1010
177
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 nucleotides.
Lab Invest.
58
1988
37
47
178
Ward
PA
Cunningham
TW
McCulloch
KK
Johnson
KJ
Regulatory effects of adenosine and adenine nucleotides on oxygen radical responses of neutrophils.
Lab Invest.
58
1988
438
447
179
Kuroki
M
Minakami
S
Extracellular ATP triggers superoxide production in human neutrophils.
Biochem Biophys Res Commun.
162
1989
377
380
180
Kuroki
M
Takeshige
K
Minakami
S
ATP-induced calcium mobilization in human neutrophils.
Biochim Biophys Acta.
1012
1989
103
106
181
Dubyak
GR
Cowen
DS
Activation of inositol phospholipid-specific phospholipase C by P2-purinergic receptors in human phagocytic leukocytes: role of pertussis toxin-sensitive G proteins.
Ann N Y Acad Sci.
603
1990
227
245
182
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.
245
1989
25
29
183
Balazovich
KJ
Boxer
LA
Extracellular adenosine nucleotides stimulate protein kinase C activity and human neutrophil activation.
J Immunol.
144
1990
631
637
184
Cowen
DS
Sanders
M
Dubyak
G
P2-purinergic receptors activate a guanine nucleotide-dependent phospholipase C in membranes from HL-60 cells.
Biochim Biophys Acta.
1053
1990
195
203
185
Freyer
DR
Boxer
LA
Axtell
RA
Todd
RF
Stimulation of human neutrophil adhesive properties by adenine nucleotides.
J Immunol.
141
1988
580
586
186
Robson
SC
Kaczmarek
E
Siegel
JB
et al
Loss of ATP diphosphohydrolase activity with endothelial cell activation.
J Exp Med.
185
1997
153
163
187
Dawicki
DD
McGowan-Jordan
J
Bullard
S
Pond
S
Round
S
Extracellular nucleotides stimulate leukocyte adherence to cultured pulmonary artery endothelial cells.
Am J Physiol.
268
1995
L666
L673
188
Rounds
S
Likar
LL
Harrington
EO
et al
Nucleotide-induced PMN adhesion to cultured epithelial cells: possible role of MUC1 mucin.
Am J Physiol.
277
1999
L874
L880
189
Cronstein
BN
Van de Stouwe
M
Druska
L
Levin
RI
Weismann
G
Nonsteroidal antiinflammatory agents inhibit stimulated neutrophil adhesion to endothelium: adenosine dependent and independent mechanisms.
Inflammation.
18
1994
323
325
190
Sud'ina
GF
Mirzoeva
OK
Galkina
SI
Pushkareva
MA
Ullrich
V
Involvement of ecto-ATPase and extracellular ATP in polymorphonuclear granulocyte-endithelial interactions.
FEBS Lett.
423
1998
243
248
191
Cowen
DS
Berger
M
Nuttle
L
Dubyak
GR
Chronic treatment with P2-purinergic receptor agonists induces phenotypic modulation of the HL-60 and U937 human myelogenous leukemia cell lines.
J Leukoc Biol.
50
1991
109
122
192
Clifford
EE
Martin
KA
Dalal
P
Thomas
R
Dubyak
GR
Stage-specific expression of P2Y receptors, ecto-apyrase and ecto-5′-nucleotidase in myeloid leukocytes.
Am J Physiol.
273
1997
C973
C987
193
Gasmi
L
McLennan
AG
Edwards
SW
Diadenosine polyphosphates induce intracellular Ca2+ mobilization in human neutrophils via a pertussis toxin sensitive G-protein.
Immunology.
90
1997
154
159
194
Saito
H
Ebisawa
M
Reason
DC
et al
Extracellular ATP stimulates interleukin-dependent cultured mast cell and eosinophils through calcium mobilization.
Int Arch Allergy Appl Immunol.
94
1991
68
70
195
Dichmann
S
Idzko
M
Zimpfer
U
et al
Adenosine triphosphate-induced oxygen radical production and CD11b up-regulation: Ca2+ mobilization and actin reorganization in human neutrophils.
Blood.
95
2000
973
978
196
Gaarder
A
Jonsen
A
Laland
S
Hellem
AJ
Owren
P
Adenosine diphosphate in red cells as a factor in the adhesiveness of human blood platelets.
Nature.
192
1961
531
532
197
Born
GV
Aggregation of blood platelets by adenosine diphosphate and its reversal.
Nature.
194
1962
927
928
198
Born
GV
Kratzer
MAA
Source and concentration of extracellular adenosine triphosphate during haemostasis in rats, rabbits and man.
J Physiol.
354
1984
419
429
199
Gachet
C
Hechler
B
Leon
C
et al
Activation of ADP receptors and platelet function.
Thromb Haemost.
78
1997
271
275
200
Hourani
S
Hall
DA
Receptors for ADP of human blood platelets.
Trends Pharmacol Sci.
15
1994
103
108
201
Mills
DC
ADP receptors on platelets.
Thromb Haemost.
76
1996
835
856
202
Packham
MA
Bryant
NL
Guccione
MA
Kinlough-Rathbone
RL
Mustard
JF
Effect of the concentration of Ca2+ in the suspending medium on the responses of human and rabbit platelets to aggregating agents.
Thromb Haemost.
62
1989
968
976
203
Pengo
V
Boschello
M
Marzari
A
Baca
M
Schivazappa
L
Dalla Volta
S
Adenosine diphosphate (ADP)-induced alpha granules release from platelets of native whole blood is reduced by ticlopidine but not by aspirin or dipyridamole.
Thromb Haemost.
56
1986
147
150
204
MacKenzie
AB
Mahaut-Smith
MP
Sage
SO
Activation of receptor-operated cation channels via P2X1 not P2T purinoceptors in human platelets.
J Biol Chem.
271
1996
2879
2881
205
Vial
C
Hechler
B
Leon
C
Cazenave
JP
Gachet
C
Presence of P2X1 purinoceptors in human platelets and megakarioblastic cell lines.
Thromb Haemost.
78
1997
1500
1504
206
Sun
B
Li
J
Okahara
K
Kambayashi
J
P2X1 purinoceptor in human platelets.
J Biol Chem.
273
1998
11544
11547
207
Clifford
EE
Parker
K
Humphreys
BD
Kertesy
SB
Dubyak
GR
The P2X1 receptor, an adenosine triphosphate-gated cation channel, is expressed in human platelets but not in human blood leukocytes.
Blood.
91
1998
3172
3181
208
Leon
C
Hechler
B
Vial
C
Leray
C
Cazenave
JP
Gachet
C
The P2Y1 receptor is an ADP receptor antagonized by ATP and expressed in platelets amd megakarioblastic cells.
FEBS Lett.
403
1997
26
30
209
Savi
P
Beauverger
P
Labouret
C
et al
Role of P2Y1 purinoceptor in ADP-induced platelet activation.
FEBS Lett.
422
1998
291
295
210
Daniel
JL
Dangelmaier
C
Jin
J
Ashby
B
Smith
JB
Kunapuli
SP
Molecular basis for ADP-induced platelet activation, I: evidence for three distinct ADP receptors on human platelets.
J Biol Chem.
273
1998
2024
2029
211
Park
HS
Hourani
SM
Differential effects of adenine nucleotide analogues on shape change and aggregation induced by adenosine 5-diphosphate (ADP) in human platelets.
Br J Pharmacol.
127
1999
1359
1366
212
Hourani
SMO
Pharmacology of the platelet ADP receptors: agonist and antagonist.
Haematologica.
85
2000
58
65
213
Fabre
JE
Nguyen
M
Latour
A
et al
Decreased platelet aggregation, increased bleeding time and resistance to thromboembolism in P2Y1-deficient mice.
Nat Med.
5
1999
1199
1202
214
Mulryan
K
Gitterman
DP
Lewis
CJ
et al
Reduced vas deferens contraction and male infertility in mice lacking P2X1 receptors.
Nature.
403
2000
86
89
215
Cattaneo
M
Gachet
C
ADP receptors and clinical bleeding disorders.
Arterioscler Thromb Vasc Biol.
19
1999
2281
2285
216
Leon
C
Vial
C
Gachet
C
et al
The P2Y1 receptor is normal in a patient presenting a severe deficiency of ADP-induced platelet aggregation.
Thromb Haemost.
81
1999
775
781
217
Cusack
NJ
Hourani
SMO
Platelet P2 receptors: from curiosity to clinical targets.
J Auton Nerv Syst.
81
2000
37
43
218
Lages
B
Weiss
HJ
Secreted dense granule adenine nucleotides promote calcium influx and the maintenance of elevated cytosolic calcium levels in stimulated human platelets.
Thromb Haemost.
81
1999
286
292
219
Vischer
UM
Wollheim
CB
Purine nucleotides induce regulated secretion of von Willebrand factor: involvement of cytosolic Ca2+ and cyclic adenosine monophosphate-dependent signaling in endothelial exocytosis.
Blood.
91
1998
118
127
220
Enjyoji
K
Sevigny
J
Lin
Y
et al
Targeted disruption of cd39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation.
Nat Med.
5
1999
1010
1017
221
Parker
JC
Snow
RL
Influence of external ATP on permeability and metabolism of dog red blood cells.
Am J Physiol.
223
1972
888
893
222
Trams
EG
Kaufman
H
Burnstock
G
A proposal for the role of ecto-enzymes and adenylates in traumatic shock.
J Theor Biol.
87
1980
609
621
223
Bergfeld
GR
Forrester
T
Release of ATP from human erythrocytes in response to a brief period of hypoxia and hypercapnia.
Cardiovasc Res.
26
1992
40
47
224
Berrie
CO
Hawkins
PT
Stephens
LR
Harden
TK
Downes
CP
Phosphatidylinositol 4,5-bisphosphate hydrolysis in turkey erythrocytes is regulated by P2Y purinoceptors.
Mol Pharmacol.
35
1989
526
532
225
Boyer
JL
Downes
CP
Harden
TK
Kinetics of activation of phospholipase C by P2 purinergic agonists and guanine nucleotides.
J Biol Chem.
264
1989
884
890
226
Boeynaems
J-M
Pearson
JD
P2 purinoceptors on vascular endothelial cells: physiological significance and transduction mechanisms.
Trends Pharmacol Sci.
11
1990
34
37
227
Boarder
MR
Hourani
SM
The regulation of vascular function by P2 receptors: multiple sites and multiple receptors.
Trends Pharmacol Sci.
19
1998
99
107
228
Modderman
WE
Vrijheid-Lammers
T
Lowik
CW
Nijweide
PJ
Removal of hematopoietic cells and macrophages from mouse bone marrow cultures: isolation of fibroblast-like stromal cells.
Exp Hematol.
22
1994
194
201
229
Nijweide
PJ
Modderman
WE
Hagenaars
CE
Extracellular adenosine triphosphate: a shock to hemopoietic cells.
Clin Orthop.
313
1995
92
102
230
Osipchuk
Y
Cahalan
M
Cell-to-cell calcium signals mediated by ATP receptors in mast cells.
Nature.
359
1992
241
244
231
Qian
YX
McCloskey
MA
Activation of mast cell K+ channels through multiple G protein-linked receptors.
Proc Natl Acad Sci U S A.
90
1993
7844
7848
232
Buell
G
Michel
AD
Lewis
C
Collo
G
Humphrey
PP
Surprenant
A
P2X1 receptor activation in HL-60 cells.
Blood.
87
1996
2659
2664
233
Perregaux
DG
McNiff
P
Laliberte
R
Conklyn
M
Gabel
G
ATP acts as an agonist to promote stimulus-induced secretion of IL-1β and IL-18 in human blood.
J Immunol.
165
2000
4615
4623
234
Mehta VB, Hart J, Wewers D. ATP stimulated release of IL-1β and IL-18 requires priming by LPS and is independent of caspase-1 cleavage. J Biol Chem. In press.
235
Schnurr
M
Then
F
Galambos
P
Scholz
C
Siegmund
B
Endres
S
Eigler
A
Extracellular ATP and TNF-α synergize in the activation of human dendritic cells.
J Immunol.
165
2000
4704
4709
236
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 T-helper 1 responses. J Immunol. In press.

Two recent papers suggest that circulating human monocytes express a functional P2X7 receptor coupled to IL-1β and IL-18 release.233,234 In addition, two papers show that exogeneous ATP can be a differentiation factor for human dendritic cells.235,236 

Author notes

Francesco Di Virgilio, Department of Experimental and Diagnostic Medicine, Section of General Pathology, University of Ferrara, Via Borsari, 46, I-44100 Ferrara, Italy; e-mail:fdv@dns.unife.it.

Sign in via your Institution