Abstract
The directional movement of cells can be regulated by ATP, certain other nucleotides (e.g., ADP, UTP), and adenosine. Such regulation occurs for cells that are “professional phagocytes” (e.g., neutrophils, macrophages, certain lymphocytes, and microglia) and that undergo directional migration and subsequent phagocytosis. Numerous other cell types (e.g., fibroblasts, endothelial cells, neurons, and keratinocytes) also change motility and migration in response to ATP, other nucleotides, and adenosine. In this article, we review how nucleotides and adenosine modulate chemotaxis and motility and highlight the importance of nucleotide- and adenosine-regulated cell migration in several cell types: neutrophils, microglia, endothelial cells, and cancer cells. We also discuss difficulties in conducting experiments and drawing conclusions regarding the ability of nucleotides and adenosine to modulate the migration of professional and non-professional phagocytes.
Keywords: Regulation, Chemotaxis, Nucleotides, Adenosine, Receptors
Introduction
Motility and migration of cells are fundamental biological properties that contribute to normal development and differentiation. These processes are particularly important in the function of “professional phagocytes,” which include neutrophils, macrophages, and microglia [1–3]. Such cells have essential roles in migrating toward and engulfing microorganisms and apoptotic cells, events that depend on the detection of chemoattractants or paracrine “find-me/eat-me” signals, respectively [4, 5]. Directed migration is also important for other cell types and contributes to normal cell function (e.g., development and angiogenesis) and pathophysiological states (e.g., cancer metastasis). Evidence in recent years has indicated the importance of extracellular nucleotides and adenosine in the regulation of homeostatic mechanisms and migration of professional phagocytes and other cell types (e.g., fibroblasts, endothelial cells, keratinocytes, and neurons), including stem cells and malignant cells [6, 7].
Chemotaxis, the directed migration of cells in response to chemical cues, is characterized by three distinct processes: motility, cell polarization, and gradient sensing ([8]; Fig. 1). Cell motility, which can occur in the absence of extracellular cues [8], is driven by an actin-mediated extension of self-regulating pseudopod protrusions that facilitate random and directed cell migration. Such migration can occur in the absence of external cues as a “correlated random walk” [9], movement of cells by their sequential formation of pseudopods [10]. Such pseudopod formation is biased (i.e., occurs at one end of the cell) even in the absence of an external signal. Cell polarization involves the detection of extracellular cues and progression through a series of cytoskeletal rearrangements that promote the protrusion of a leading edge of a cell and the release of integrin-based contacts with the substratum and contraction at the rear of the cell (i.e., the uropod; [11]). Cell polarization is central to the migration of many cell types.
Fig. 1.
Cellular events involved in chemotaxis. Cell motility is driven by the biased, actin-mediated extension of pseudopod-like extensions at one end of the cell. Some cells are capable of coordinating intracellular and extracellular signaling to promote cell polarization (middle panel) and establish persistent leading and trailing edges. Furthermore, even while immobile, some cells can detect and amplify external chemoattractant gradients, a process termed gradient sensing (right panel)
Different cell types have varying levels of intrinsic polarity. Certain cell types exposed to a uniform concentration of chemoattractant do not polarize, but instead undergo chemokinesis (i.e., random cell migration; [11]). However, some types of cells (e.g., neutrophils and the slime mold Dictyostelium discoidium) can polarize under such conditions; such cell types can detect and amplify spatial gradients even while immobile, a process called gradient sensing [8].
The release of ATP and other nucleotides (e.g., UTP; or adenosine 3′,5′ cyclic AMP [cAMP] by D. discoidium) in response to extracellular stimuli is critical for different aspects of the chemotactic response and involves mechanisms that are discussed elsewhere in this issue by Lazarowski [12]. Extracellular ATP promotes cell migration through both autocrine and paracrine pathways. For example, professional phagocytes (e.g., neutrophils; Fig. 2) release ATP in response to stimulation with some chemoattractants. This autocrine release of ATP is critical for the chemotactic response to the chemoattractants and is discussed below. ATP released from apoptotic cells may act as a paracrine “find-me” signal that promotes phagocytic clearance [5]. There is debate as to whether ATP is a chemoattractant (see [13]) or if it promotes random cell migration (i.e., chemokinesis), acting as a local “touch-me” signal rather than a long-range “find-me” signal [14]. It is tempting to postulate that similar mechanisms may also recruit immune cells to cancerous tissues.
Fig. 2.
Autocrine/paracrine stimulation of chemotaxis. Extracellular nucleotides promote cell migration through autocrine and paracrine pathways. In the case of neutrophils, stimulation with some chemoattractants, such as IL-8 from host cells (e.g., macrophages) or formyl peptides from damaged cells or bacteria, results in a release of ATP. Through various autocrine signaling pathways, released nucleotides promote cell migration. Alternatively, the direct release of ATP from apoptotic cells has been proposed to act as a paracrine “find me” signal to promote phagocytic clearance. Elevated levels of extracellular ATP in tumor microenvironments may similarly mediate neutrophil migration
Released ATP can undergo numerous fates. Cells utilize multiple mechanisms to generate and respond to extracellular ATP and adenosine (Fig. 3). These include an interaction of nucleotides with one or more P2X (ion channel; [15]) and P2Y (G-protein-coupled; [16]) receptors and the enzymatic hydrolysis of ATP (e.g., via the sequential actions of NTPDases and CD73/ecto-5′-nucleotidase; [17]) to generate adenosine. Extracellular adenosine, in turn, can interact with one or more G-protein-coupled P1 receptors [18] or can be taken back up into cells via ENT (equilibrative nucleoside transporter [19]) to undergo metabolic fates that include the synthesis of ATP. One consequence of the activation of P1 (and certain P2Y) receptors is a change in cellular levels of cAMP, which can exit cells by certain multidrug resistant proteins (MRPs 4 and 5; [20]). The conversion of extracellular cAMP to AMP by D. discoidium occurs via extracellular phosphodiesterases [21]. Although similar mechanisms are not as clearly defined in mammalian tissues, metabolism of cAMP to AMP has been demonstrated in several systems (e.g., [22], [23]). In such tissues, this conversion likely occurs via ectonucleotide pyrophosphates (i.e., NPP1-3) that have phosphodiesterase activity [17].
Fig. 3.
P1/P2 receptor signaling pathways. A diverse array of intracellular and extracellular receptors and enzymes facilitate P1/P2 receptor signaling pathways. ATP released into the extracellular space can activate G protein-coupled P2Y receptors or P2X ion channels. Ectonucleotidases (e.g., NTPDases/CD39 and ecto-5′-nucleotidases/CD73) hydrolyze released ATP, generating extracellular ADP, AMP, and adenosine (ado). Adenosine, which can also be released from or internalized by cells through equilibrative nucleoside transporters (ENTs), can activate G-protein-coupled P1 receptors. It has been proposed that cell-surface F1F0 synthase perpetuates ATP signals by facilitating the conversion of ADP to ATP. Adenylyl cyclase, the activity of which is controlled by GPCRs, generates the second messenger cAMP, which cells can release via multidrug resistance proteins (MRPs). Extracellular nucleotide pyrophosphatase/phosphodiesterases (ENPPs) facilitate the conversion of released cAMP to adenosine. In the intracellular environment, adenosine kinase (AK), adenylate kinase (ADK), and phosphodiesterases (PDE) contribute to ATP generation, while 5′NT generates adenosine. The conversion of ADP to ATP is primarily driven by mitochondrial pathways that are upregulated as intracellular AMP concentrations are increased
Although extracellular ATP can promote the release of chemottractant cytokines from a variety of cells (e.g., through P2X7-mediated activation of the NALP inflammasomes; [24, 25]), ATP, and its metabolites, acting by the pathways described above, can also amplify chemoattractant signals and directly stimulate cell migration. Below, we review the modulation of chemotaxis and cell motility by nucleotides and adenosine and emphasize similarities and differences between professional and non-professional phagocytes. We focus on articles related to these topics that have appeared in the past 5 years (2006–2011). Table 1 provides an overview of the cell types in which P1/P2 receptors and certain effectors regulate cell migration and chemotaxis. In the subsequent text, we highlight findings for neutrophils, microglia, endothelial cells, and cancer cells.
Table 1.
Cell types in which P1/P2 receptors and effectors regulate cell migration and chemotaxis
| Cell type | Receptor/effector | Stimulation of response (unless noted otherwise; reference number) |
|---|---|---|
| Astrocyte progenitor cells | P2Y1 | Migration [113] |
| Breast cancer cells | P2X7 | Migration [89] |
| CD73 | Migration [92, 93] | |
| A2b | Migration [93] | |
| Cardiac fibroblasts | P2Y2 | Migration [114] |
| Corneal epithelial cells | P2X7 | Migration [115] |
| P2Y2 | Migration [116, 117] | |
| Dendritic cells | P2Y2 | Chemotaxis [83] |
| Eosinophils chemotaxis | P2Y2 | VCAM-1 regulation [118] and [83, 118]; monosodium urate-stimulated |
| Cytokine release and migration [119] | ||
| Endothelial cells | ATP | PI3K/Rho/ROCK- [77] and PI3K/Akt/mTOR-modulated [73] migration |
| ADP | Rac1-modulated migration [120] | |
| P2 | RhoA/FAK-modulated migration [75] | |
| P2Y1 | MAPK-modulated migration [71, 72] | |
| P2Y13 | Migration [74] | |
| P2X4 | Migration [81] | |
| F1/F0 synthase | Migration [79, 81] | |
| MRP4 | ↓ Migration [121] | |
| Glioma cells | P2X7 | Migration [88] |
| Hematopoietic stem sells | P2Y/UTP | Migration [122] |
| Hepatic stellate cells | A2a | ↓ PDGF-stimulated chemotaxis [123] |
| CD73 | ↓ Migration [124] | |
| Keratinocytes | P2/ATP | HB-EGF synthesis and release, ↑ migration [125] |
| P2Y2 | Inhibition of spreading, ↓ IGF-1/PI3K dependent migration [126, 127] | |
| Macrophages | P2/ATP | Facilitates C5a-mediated chemotaxis [32]; ↓ ‘basal’ |
| migration/LPS-induced IL-6/TNFa release [55]; | ||
| P2X7 | CD44 (adhesion molecule) shedding, in P388D1 cells [128], | |
| In vivo bladder macrophage migration [129] | ||
| A2a | ↓ Chemokine secretion [130], ↓ CCR7 protein expression, | |
| ↓ CCR7-stimulated migration [131] | ||
| Mesenchymal stromal cells | CD73 | Migration [132] |
| Microglia | ATP | Chemotaxis [51–56] |
| P2Y12 | PI3K-dependent [133] chemotaxis [57] | |
| P2X4 | PI3K-dependent [133] chemotaxis [57] | |
| CD39 | Chemotaxis [68] | |
| A2b | ↓ Chemotaxis [64] | |
| Monocytes | P2Y2/P2Y6 | Regulation of IL-8 secretion/migration [134] |
| Natural killer cells | ATP/P2Y11 | Inhibition of CX3CL1- and enhancement of CXCL12-induced chemotaxis [135] |
| Neural progenitors | P2Y1 | Migration [136] |
| Neural stem cells | ATP/UTP | Migration [137] |
| Neutrophils | P2 | IL-8-induced chemotaxis [37], LPS-induced chemotaxis |
| [138] | ||
| P2Y2 | Chemotaxis, orientation in chemoattractant gradients [29, 35] | |
| P2X1 | Chemotaxis [30] | |
| CD39 | Negative regulation of ATP-induced neutrophil trafficking [139] | |
| Chemotaxis [47] | ||
| A3 | Chemoattractant-induced migration [29] | |
| ↓ Chemotaxis [44] | ||
| A1 | ↓ Transmigration [43] | |
| A2a | ↓ Chemotaxis [40, 140] | |
| NG108-15 hybridoma cells | P2Y2 | Migration [141] |
| Oligodendrocyte progenitors | P2Y1 | Migration [142] |
| Pre-adipocyte fibroblasts | P2Y | Chemokinesis/Migration [143] |
| Renal mesangial cells | ATP/UTP | Migration [144] |
| Smooth muscle cells | ATPgS | Chemotaxis [145] |
| P2Y2 | Migration [146] | |
| P2Y12 | Migration of smooth muscle-like cells [147] | |
| CD39 | Facilitation of ATP/UTP/PDGF-induced migration [148] | |
| Spermatazoa | A3 | Motility [149] |
| ATP/P2 | Motility [150] |
Neutrophils
Neutrophils represent 50–75 % of circulating leukocytes [26] and are the most abundant of the body’s professional phagocytes. As part of the first line of defense of the innate immune system, neutrophils detect and direct their movement in response to chemoattractant signals released by pathogens and damaged tissues. The net forward movement of neutrophils results from PI3K-mediated F-actin polymerization at the front (i.e., the leading edge or pseudopod) and the Rho GTPase RhoA-mediated contraction of actin-myosin II complexes at the back of the cell (the trailing edge, i.e., uropod) [27]. The ability of neutrophils to orient themselves in chemoattractant gradients as shallow as 1 % implies that the cells amplify external gradients [28], although the signaling pathways involved in this amplification are not fully understood.
Extracellular ATP and autocrine/paracrine P1/P2 receptor signaling pathways are critical mediators of neutrophil chemotaxis. Upon stimulation with chemoattractants, such as the bacteria-derived formyl peptide receptor agonist fMet-Leu-Phe (fMLP), human neutrophils release ATP in a polarized fashion at their leading edge [29]. ATP does not act as a chemoattractant but promotes chemokinesis; the chemotactic response to fMLP is inhibited if neutrophils are incubated with the ATP scavenger apyrase, indicating that this response requires extracellular ATP [29]. Apyrase also inhibits chemotaxis of mouse neutrophils in response to W-peptide (Trp-Lys-Tyr-Met-Val-d-Met; [30]), a potent agonist of the mouse formyl peptide receptors [31]. In addition, apyrase inhibits chemotaxis of macrophages promoted by the complement agonist C5a [32]. Intraperitoneal injection of apyrase attenuates neutrophil accumulation in the lungs of mice following lipopolysaccharide (LPS)-induced injury [33] and reduces the number of adherent neutrophils in the microvasculature at sites of focal hepatic injury in mice [13]. Although in vivo imaging experiments in the latter study suggested that apyrase did not affect the percentage or speed of neutrophils undergoing chemotaxis and the authors attributed the effect of apyrase to suppression of P2X7-mediated activation of the macrophage Nlrp3 inflammasome, the local concentration of apyrase may have been too low to overcome the high extracellular concentration of ATP, which can reach 80 μM [34].
Nucleotide-activated P2 receptors are key participants in the ATP-promoted chemotactic pathway of neutrophils. Exposure of neutrophils to the non-hydrolyzable ATP analog ATPγS, which overwhelms the polarized endogenous extracellular ATP signals, impedes the ability of the cells to accurately migrate in response to point sources of fMLP, thus implicating P2 receptors in the polarization of neutrophils in chemoattractant gradients [29]. Experiments with P2Y2-knockout animals reveal reduced sequestration of neutrophils in the lungs in a mouse model of sepsis [29, 35]. Studies with neutrophil-like HL-60 cells suggest that P2Y2 receptors may co-localize with fMLP-activated formyl-peptide receptors at the cell surface [36]. In addition, ATP and P2 signaling are required for IL-8-induced neutrophil chemotaxis in vitro [37]. P2X receptors may also contribute to the regulation of neutrophil chemotaxis: A P2X1 receptor-selective agonist can induce chemokinesis and enhance IL-8-induced chemotaxis of human neutrophils by activating RhoA, which, as noted above, promotes the contraction of actin/myosin complexes at the trailing edge of migrating cells [30].
Adenosine-activated P1 receptors modulate neutrophil chemotaxis, though their precise role is unclear. Adenosine receptors couple to Gi (A1, A3) or Gs (A2a, A2b), which can exert distinct physiological responses [38]. A2a receptors inhibit neutrophil chemotaxis, as shown by studies that used selective A2a agonists [39] and A2a-knockout mice [40]. The role of Gi-coupled A1 and A3 receptors is more controversial. Early studies showed that A1 receptors exert pro-inflammatory effects by promoting neutrophil adhesion and chemotaxis [41, 42], but more recent data obtained with knockout mice suggest that A1 receptors inhibit neutrophil microvascular permeability [43]. Other evidence suggests that A3 receptors localize at the leading edge of human neutrophils during chemotaxis: Inhibition of A3 receptor signaling blunts migration in gradients of fMLP or IL-8 [29]. However, a study that used the A3-selective agonist CP-532-903 suggested that A3 receptor activation inhibits the migration of murine neutrophils [44]. In addition, A3 knockout mice have reduced accumulation of neutrophils in the lungs in a model of inflammation [45]. The authors of the study that used CP-532-903 reported that this inhibition is not due to cAMP production, alterations in Ca2+ signaling, or desensitization of fMLP receptors, and speculated that activation of A3 receptors may sequester Gi-linked signaling components that are necessary for neutrophil migration [46].
Although results of studies related to the effects of adenosine are inconsistent, together they indicate a role for A3 receptors in the chemotaxis of neutrophils and suggest that spatiotemporal aspects of signaling contribute to this action. This idea is supported by evidence that CD39 (NTPDase1), which initiates the hydrolysis of extracellular ATP to adenosine, localizes at the leading edge of migrating human neutrophils and is required for efficient chemotaxis in response to fMLP, IL-8, and C5a [47].
Microglia
Microglia, the resident macrophages and primary professional phagocytes of the CNS, exist in two states: a resting, surveillance state and an activated state. The conversion to the activated state depends on extracellular signals [48]. Early studies revealed that extracellular ATP promotes the formation of microglial processes that are characteristic of the surveillance state [49]. Subsequent work has shown that microglia undergo ATP- and ADP-promoted chemotaxis, which is inhibited by AR-C69931MX, an antagonist of the ADP-activated P2Y12 receptor [50]. Additional evidence indicates that ATP/ADP promotes this chemotactic response [51–56] and that P2Y12 receptors have a critical role in the response [57]. In addition, both P2Y1 [58] and P2X4 [59] receptors have been implicated in microglial chemotactic responses. The chemotactic response of microglia to ATP appears to occur via a mechanism that is distinct from chemotaxis promoted by bradykinin [60] or C5a [53, 61]; moreover, the response to ATP may be driven by both chemotaxis and chemokinesis [53, 61]. Thus, the chemotactic response of microglia to extracellular ATP has several similarities with that of neutrophils.
ATP induces the formation of processes in resting microglia [49], but recent evidence shows that it can promote the retraction of such processes and induce chemorepulsion in activated microglia [62]. The latter response has been attributed to upregulation of A2a receptors and concomitant downregulation of P2Y12 [62] and A3 [63] receptors. Other studies implicate an inhibitory role of A2B [64] and A3 [65] receptors in microglial migration. Microglia (akin to neutrophils) express CD39 on their cell surface, thereby enabling the clearance of extracellular ATP and facilitating the generation of adenosine [66, 67]. CD39-mediated hydrolysis of extracellular ATP is critical for this chemotaxis of microglia: Microglia isolated from CD39-knockout mice do not migrate in response to ATP or adenosine [68], but this response is restored in microglia treated with ATP and apyrase. Thus, the chemotactic response of microglia is akin to that of neutrophils and involves both P1 and P2 receptors. Altered expression of the key receptors in this process during states of microglial activity (e.g., upregulation of inhibitory receptors and downregulation of stimulatory receptors upon microglial activation) may help fine-tune potentially catastrophic inflammatory responses in the CNS. Indeed, enhancement in P2-mediated signalling and migration responses has been observed in microglia in a mouse model of status epilepticus [69].
Endothelial cells
Although endothelial cells are not professional phagocytes, their migration is an essential contributor to angiogenesis, which is important for tissue healing, development, and cancer progression [70]. Many different types of endothelial cells exhibit a chemotactic response to extracellular nucleotides; the P2Y1 receptor has been implicated as a mediator of this process [71, 72]. Activation of P2Y1 receptors by ADP promotes human umbilical vein endothelial cell (HUVEC) migration via mitogen-activated protein kinase pathways (including ERK1/2, JNK, and p38; [71]). Similarly, in vasa vasorum endothelial cells (VVECs), but not pulmonary artery or aortic endothelial cells, ATP promotes cell migration and potentiates the DNA synthesis-stimulating activity of VEGF and bFGF [73]. The nucleotide-induced migration of VVECs occurs via a pathway that is similar to, but distinct from, platelet-mediated pathways [74] and likely occurs by P2Y1-mediated activation of ERK1/2, PI3K, and mTOR [73].
The chemotactic response of endothelial cells to extracellular nucleotides may also involve other signaling pathways. Exposure to hypergravity induces the release of ATP and migration of bovine aortic endothelial cells in a RhoA activation/FAK phosphorylation-dependent manner [75]. Studies of HUVECs reveal that extracellular ATP and UTP both induce FAK phosphorylation and promote endothelial migration through the activation of P2Y2 and P2Y4 receptors [76]. In addition, extracellular ATP, via P2Y receptors, activates PI3K and Rho/ROCK pathways in VVECs, which in turn promote the release of ATP [77]. Such studies suggest that nucleotide receptor-mediated positive feedback loops may promote endothelial cell migration.
Enzymes that control the availability of extracellular nucleotides play an important role in angiogenesis. Although there has been debate regarding its contribution to extracellular ATP synthesis [78], F1F0 synthase appears to be an important positive regulator of endothelial cell migration [79–81]. Interestingly, F1F0 synthase is also the major endothelial binding site for angiostatin, a potent inhibitor of angiogenesis and endothelial cell migration [82, 83].
Cancer cells
Cancer cells can use extracellular nucleotide-mediated migration pathways to facilitate their proliferation and migration and facilitate tumor angiogenesis. Various cancer cells over-express P2X7 receptors [84–87] that regulate their motility (e.g., C6 glioma cells [88] and breast cancer cells [89]). ATP enhances the migration of TGFβ1-treated A549 lung cancer cells, although the precise receptor responsible for this effect has not yet been identified [90].
Enzymes that control the availability of extracellular ATP can play an important role in tumorigenesis. Tumor cells express F1F0 synthase on their extracellular surface; some evidence suggests that this enzyme is the target of angiostatin [91], a protein that is cytotoxic to tumors by inhibiting angiogenesis. Human breast cancer cells release an adenosine 5′-diphosphate transphosphorylase (sNDPK) that promotes the extracellular synthesis of ATP, which is proposed to induce P2Y1-dependent endothelial cell migration and VEGFR-2 activation [72].
The P1 receptor pathway has also been implicated in promoting cancer cell migration; both CD73 [92, 93] and A2b activity [93] can contribute to this response. Furthermore, adenosine upregulates the expression of the chemokine receptor CXCR4 in cancer cells, an effect that is proposed to occur through the activation of A2a/A2b receptors [94]. The extracellular pyrophosphatase autotaxin (NPP2) stimulates motility of cancer cells [95] and migration of endothelial cells [96]. Studies of autotaxin in cancer progression have focused on its lysophosphatidic acid (LPA)-producing activity [97]. However, because LPA can stimulate ATP release from human endothelial cells [98], it is tempting to speculate that, akin to what occurs in neutrophils, LPA-induced angiogenesis depends upon P2-receptor signaling.
It is not known if the elevated extracellular ATP levels in tumor microenvironments help recruit immune cells. Such recruitment contributes to the pathogenesis of cancer [99] but can also potentially be exploited to promote the destruction of cancerous tissue [100, 101]; thus, ATP signaling pathways represent potential therapeutic targets in the treatment of cancer.
Perspectives and conclusions
Many different cell types migrate in response to extracellular cues; cell migration plays critical roles in a diverse array of physiological processes, including inflammation [102], angiogenesis [103], wound healing [104], and development [105]. Cell migration can also contribute to pathology, helping to mediate autoimmune responses, developmental disorders, tumor progression, and metastasis. Thus, cell migration pathways are potential targets for the treatment of a diverse array of diseases. As noted above, autocrine/paracrine P1/P2 signaling pathways influence cell migration and as such, may be therapeutic targets for settings in which such migration is defective or excessive.
Studies of P1/P2-receptor-mediated cell migration and its possible therapeutic targeting have a unique set of challenges. The complex, multi-component nature of these signaling mechanisms makes it difficult to identify specific receptors for individual pathways, a problem that is exacerbated by the limited availability of selective P2-receptor agonists and antagonists. Certain approaches can help circumvent such difficulties. For example, the use of enzymes, such as apyrase and adenosine deaminase, to hydrolyze nucleotides and adenosine, respectively, can aid in defining their contribution to cellular events. Non-hydrolyzable analogs, such as ATPγS, can also help distinguish P1- and P2-mediated effects. Caution is required in using agents such as suramin, a P2 receptor inhibitor that also interacts with other signaling components (e.g., G-proteins [106] and protein arginine methyltransferase 1 [107]). Furthermore, because cells often express multiple P1/P2 receptor subtypes, one must employ drug concentrations that are appropriate for the receptors of interest. The use of knockout mice and siRNA/shRNA approaches are alternatives to the administration of pharmacological agents for assessing and defining the role of particular P1 and P2 receptors.
Studies of cell migration have a unique set of challenges. Transwell assays, which are often used to assess cell migration, facilitate the rapid acquisition of large amounts of data. However, the chemoattractant gradients created in transwell assays can be atypically steep [108], and the information obtained from such assays is limited, for example, with respect to the directionality of cell migration and in differentiating chemotaxis from random movement (chemokinesis). Because of the polarized nature of signaling in cell migration, pharmacological agents can have different effects if added to the upper or lower wells of such assays, which may explain inconsistent findings (e.g., for A3 adenosine receptors in neutrophil migration). Microscope-based 2D chemotaxis methods can yield detailed data regarding the speed and trajectory of migrating cells but are technically challenging and require specialized equipment. The latter include cell migration slides, which, although expensive, may provide useful information (e.g., [108]). Given the limitations of individual methods, investigators should seek to use multiple, complementary approaches when studying cell migration. Comprehensive overviews of currently available techniques and the advantages and disadvantages of each can be found in the reviews of Zigmond et al. [109] and Roussos et al. [99].
Considerable evidence implicates P1/P2-mediated cell migration pathways in pathophysiology [69]. For example, recent studies show that pathogens such as HIV [110] and malaria [111] promote infection by “hijacking” P1/P2 signaling pathways. These pathways may provide therapeutic targets in such settings. Interest in the possible use of inhibitors of cell migration has increased in recent years. In addition, there is an increased prevalence of autoimmune disorders, such as type I diabetes mellitus, and a growing recognition of a need for therapies that downregulate immune cell activity without abolishing the ability to remove pathogens [112]. P1/P2-mediated migration thus may offer therapeutic opportunities, in particular, if one can identify unique pathways and exploit information regarding receptor/ecto-nucleotidase stoichiometry so as to define “signaling bottlenecks” as novel therapeutic targets. Efforts directed at altering P1/P2-mediated cell migration pathways may thus prove to be therapeutically useful.
Footnotes
Work in the authors’ laboratories is supported by grants from the NIH and the MRC.
References
- 1.Flannagan RS, Cosio G, Grinstein S. Antimicrobial mechanisms of phagocytes and bacterial evasion strategies. Nat Rev Microbiol. 2009;7(5):355–366. doi: 10.1038/nrmicro2128. [DOI] [PubMed] [Google Scholar]
- 2.Krause KH. Professional phagocytes: predators and prey of microorganisms. Schweiz Med Wochenschr. 2000;130(4):97–100. [PubMed] [Google Scholar]
- 3.Brown EJ. Phagocytosis. Bioessays. 1995;17(2):109–117. doi: 10.1002/bies.950170206. [DOI] [PubMed] [Google Scholar]
- 4.Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, Park D, Woodson RI, Ostankovich M, Sharma P, Lysiak JJ, Harden TK, Leitinger N, Ravichandran KS. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature. 2009;461(7261):282–286. doi: 10.1038/nature08296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ravichandran KS. Find-me and eat-me signals in apoptotic cell clearance: progress and conundrums. J Exp Med. 2010;207(9):1807–1817. doi: 10.1084/jem.20101157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Junger WG. Immune cell regulation by autocrine purinergic signalling. Nat Rev Immunol. 2011;11(3):201–212. doi: 10.1038/nri2938. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Linden J. Regulation of leukocyte function by adenosine receptors. Adv Pharmacol. 2011;61:95–114. doi: 10.1016/B978-0-12-385526-8.00004-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Iglesias PA, Devreotes PN. Navigating through models of chemotaxis. Curr Opin Cell Biol. 2008;20(1):35–40. doi: 10.1016/j.ceb.2007.11.011. [DOI] [PubMed] [Google Scholar]
- 9.Patlak C. Random walk with persistence and external bias. Bull Math Biol. 1953;15(3):311–338. [Google Scholar]
- 10.Bosgraaf L, Haastert PJM. The ordered extension of pseudopodia by amoeboid cells in the absence of external cues. PLoS One. 2009;4(4):e5253. doi: 10.1371/journal.pone.0005253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Petrie RJ, Doyle AD, Yamada KM. Random versus directionally persistent cell migration. Nat Rev Mol Cell Biol. 2009;10(8):538–549. doi: 10.1038/nrm2729. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lazarowski E (2012) Vesicular and conductive mechanisms of nucleotide release. Purinergic Signalling. doi:10.1007/s11302-012-9304-9 [DOI] [PMC free article] [PubMed]
- 13.McDonald B, Pittman K, Menezes GB, Hirota SA, Slaba I, Waterhouse CCM, Beck PL, Muruve DA, Kubes P (2010) Intravascular danger signals guide neutrophils to sites of sterile inflammation. Science 330 (6002):362–366. doi:10.1126/science.1195491 [DOI] [PubMed]
- 14.Isfort K, Ebert F, Bornhorst J, Sargin S, Kardakaris R, Pasparakis M, Baehler M, Schwerdtle T, Schwab A, Hanley PJ (2011) Real-time imaging reveals that P2Y2 and P2Y12 receptor agonists are not chemoattractants and macrophage chemotaxis to C5a is PI3K- and p38 MAPK-independent. J Biol Chem. doi:10.1074/jbc.M111.289793 [DOI] [PMC free article] [PubMed]
- 15.Surprenant A, North RA. Signaling at purinergic P2X receptors. Annu Rev Physiol. 2009;71(1):333–359. doi: 10.1146/annurev.physiol.70.113006.100630. [DOI] [PubMed] [Google Scholar]
- 16.Kugelgen I, Harden TK. Molecular pharmacology, physiology, and structure of the P2Y receptors. Adv Pharmacol. 2011;61:373–415. doi: 10.1016/B978-0-12-385526-8.00012-6. [DOI] [PubMed] [Google Scholar]
- 17.Zimmermann H. Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedebergs Arch Pharmacol. 2000;362(4–5):299–309. doi: 10.1007/s002100000309. [DOI] [PubMed] [Google Scholar]
- 18.Fredholm BB, Ijzerman AP, Jacobson KA, Linden J, Muller CE. International union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors—an update. Pharmacol Rev. 2011;63(1):1–34. doi: 10.1124/pr.110.003285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Young JD, Yao SY, Sun L, Cass CE, Baldwin SA. Human equilibrative nucleoside transporter (ENT) family of nucleoside and nucleobase transporter proteins. Xenobiotica. 2008;38(7–8):995–1021. doi: 10.1080/00498250801927427. [DOI] [PubMed] [Google Scholar]
- 20.Sager G, Ravna AW. Cellular efflux of cAMP and cGMP—a question about selectivity. Mini Rev Med Chem. 2009;9(8):1009–1013. doi: 10.2174/138955709788681654. [DOI] [PubMed] [Google Scholar]
- 21.Bader S, Kortholt A, Van Haastert PJM. Seven dictyostelium discoideum phosphodiesterases degrade three pools of cAMP and cGMP. Biochem J. 2007;402(1):153–161. doi: 10.1042/BJ20061153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Rosenberg PA, Dichter MA. Extracellular cAMP accumulation and degradation in rat cerebral cortex in dissociated cell culture. J Neurosci. 1989;9(8):2654–2663. doi: 10.1523/JNEUROSCI.09-08-02654.1989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Jackson EK, Mi Z, Dubey RK. The extracellular cAMP-adenosine pathway significantly contributes to the in vivo production of adenosine. J Pharmacol Exp Ther. 2007;320(1):117–123. doi: 10.1124/jpet.106.112748. [DOI] [PubMed] [Google Scholar]
- 24.Virgilio F. Liaisons dangereuses: P2X(7) and the inflammasome. Trends Pharmacol Sci. 2007;28(9):465–472. doi: 10.1016/j.tips.2007.07.002. [DOI] [PubMed] [Google Scholar]
- 25.Martinon F, Gaide O, Pétrilli V, Mayor A, Tschopp J. NALP inflammasomes: a central role in innate immunity. Semin Immunopathol. 2007;29(3):213–229. doi: 10.1007/s00281-007-0079-y. [DOI] [PubMed] [Google Scholar]
- 26.Jandl JH (1996) Blood: textbook of hematology. Boston, Little Brown and Co.
- 27.Bagorda A, Parent CA. Eukaryotic chemotaxis at a glance. J Cell Sci. 2008;121(16):2621–2624. doi: 10.1242/jcs.018077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Zigmond SH. Ability of polymorphonuclear leukocytes to orient in gradients of chemotactic factors. J Cell Biol. 1977;75(2 Pt 1):606–616. doi: 10.1083/jcb.75.2.606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A, Nizet V, Insel PA, Junger WG. ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors. Science. 2006;314(5806):1792–1795. doi: 10.1126/science.1132559. [DOI] [PubMed] [Google Scholar]
- 30.Lecut C, Frederix K, Johnson DM, Deroanne C, Thiry M, Cl F, Maree Rl, Evans RJ, Volders PGA, Bours V, Oury Cc. P2X1 ion channels promote neutrophil chemotaxis through rho kinase activation. J Immunol. 2009;183(4):2801–2809. doi: 10.4049/jimmunol.0804007. [DOI] [PubMed] [Google Scholar]
- 31.He R, Tan L, Browning DD, Wang JM, Ye RD. The synthetic peptide Trp-Lys-Tyr-Met-Val-D-Met is a potent chemotactic agonist for mouse formyl peptide receptor. J Immunol. 2000;165(8):4598–4605. doi: 10.4049/jimmunol.165.8.4598. [DOI] [PubMed] [Google Scholar]
- 32.Kronlage M, Song J, Sorokin L, Isfort K, Schwerdtle T, Leipziger J, Robaye B, Conley PB, Kim H-C, Sargin S, Schon P, Schwab A, Hanley PJ. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci Signal. 2010;3(132):ra55. doi: 10.1126/scisignal.2000588. [DOI] [PubMed] [Google Scholar]
- 33.Jr R, Vollmer I, Stark S, Wagner R, Ngamsri K-C, Eltzschig HK. Adenosine and inflammation: CD39 and CD73 are critical mediators in LPS-induced PMN trafficking into the lungs. FASEB J. 2009;23(2):473–482. doi: 10.1096/fj.08-119701. [DOI] [PubMed] [Google Scholar]
- 34.Corriden R, Insel PA, Junger WG. A novel method using fluorescence microscopy for real-time assessment of ATP release from individual cells. Am J Physiol Cell Physiol. 2007;293(4):C1420–C1425. doi: 10.1152/ajpcell.00271.2007. [DOI] [PubMed] [Google Scholar]
- 35.Inoue Y, Chen Y, Hirsh MI, Yip L, Junger WG. A3 and P2Y2 receptors control the recruitment of neutrophils to the lungs in a mouse model of sepsis. Shock. 2008;30(2):173–177. doi: 10.1097/shk.0b013e318160dad4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Chen Y, Yao Y, Sumi Y, Li A, To UK, Elkhal A, Inoue Y, Woehrle T, Zhang Q, Hauser C, Junger WG (2010) Purinergic signaling: a fundamental mechanism in neutrophil activation. Sci Signal 3 (125):ra45-. doi:10.1126/scisignal.2000549 [DOI] [PMC free article] [PubMed]
- 37.Kukulski F, Ben Yebdri F, Lecka J, Kauffenstein G, Levesque SA, Martín-Satué M, Sévigny J. Extracellular ATP and P2 receptors are required for IL-8 to induce neutrophil migration. Cytokine. 2009;46(2):166–170. doi: 10.1016/j.cyto.2009.02.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Fredholm BB, IJzerman AP, Jacobson KA, Klotz KN, Linden J. International union of pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001;53(4):527–552. [PMC free article] [PubMed] [Google Scholar]
- 39.Koizumi S, Odashima M, Otaka M, Jin M, Linden J, Watanabe S, Ohnishi H. Attenuation of gastric mucosal inflammation induced by indomethacin through activation of the A2A adenosine receptor in rats. J Gastroenterol. 2009;44(5):419–425. doi: 10.1007/s00535-009-0028-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang H, Zhang W, Tang R, Zhu C, Bucher C, Blazar BR, Geng JG, Zhang C, Linden J, Wu C, Huo Y. Adenosine receptor A2A deficiency in leukocytes increases arterial neointima formation in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2010;30(5):915–922. doi: 10.1161/ATVBAHA.109.202572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Neely CF, Jin J, Keith IM. A1-adenosine receptor antagonists block endotoxin-induced lung injury. Am J Physiol. 1997;272(2 Pt 1):L353–L361. doi: 10.1152/ajplung.1997.272.2.L353. [DOI] [PubMed] [Google Scholar]
- 42.Cronstein BN, Levin RI, Philips M, Hirschhorn R, Abramson SB, Weissmann G. Neutrophil adherence to endothelium is enhanced via adenosine A1 receptors and inhibited via adenosine A2 receptors. J Immunol. 1992;148(7):2201–2206. [PubMed] [Google Scholar]
- 43.Ngamsri KC, Wagner R, Vollmer I, Stark S, Reutershan J. Adenosine receptor A1 regulates polymorphonuclear cell trafficking and microvascular permeability in lipopolysaccharide-induced lung injury. J Immunol. 2010;185(7):4374–4384. doi: 10.4049/jimmunol.1000433. [DOI] [PubMed] [Google Scholar]
- 44.Hoeven D, Wan TC, Auchampach JA. Activation of the A(3) adenosine receptor suppresses superoxide production and chemotaxis of mouse bone marrow neutrophils. Mol Pharmacol. 2008;74(3):685–696. doi: 10.1124/mol.108.048066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wagner R, Ngamsri K-C, Stark S, Vollmer I, Jr R. Adenosine receptor A3 is a critical mediator in LPS-induced pulmonary inflammation. Am J Physiol Lung Cell Mol Physiol. 2010;299(4):L502–L512. doi: 10.1152/ajplung.00083.2010. [DOI] [PubMed] [Google Scholar]
- 46.Hoeven D, Gizewski ET, Auchampach JA. Activation of the A(3) adenosine receptor inhibits fMLP-induced Rac activation in mouse bone marrow neutrophils. Biochem Pharmacol. 2010;79(11):1667–1673. doi: 10.1016/j.bcp.2010.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Corriden R, Chen Y, Inoue Y, Beldi G, Robson SC, Insel PA, Junger WG. Ecto-nucleoside triphosphate diphosphohydrolase 1 (E-NTPDase1/CD39) regulates neutrophil chemotaxis by hydrolyzing released ATP to adenosine. J Biol Chem. 2008;283(42):28480–28486. doi: 10.1074/jbc.M800039200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev. 2011;91(2):461–553. doi: 10.1152/physrev.00011.2010. [DOI] [PubMed] [Google Scholar]
- 49.Neary JT, Baker L, Jorgensen SL, Norenberg MD. Extracellular ATP induces stellation and increases glial fibrillary acidic protein content and DNA synthesis in primary astrocyte cultures. Acta Neuropathol. 1994;87(1):8–13. doi: 10.1007/BF00386249. [DOI] [PubMed] [Google Scholar]
- 50.Honda S, Sasaki Y, Ohsawa K, Imai Y, Nakamura Y, Inoue K, Kohsaka S. Extracellular ATP or ADP induce chemotaxis of cultured microglia through Gi/o-coupled P2Y receptors. J Neurosci. 2001;21(6):1975–1982. doi: 10.1523/JNEUROSCI.21-06-01975.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Kurpius D, Nolley EP, Dailey ME. Purines induce directed migration and rapid homing of microglia to injured pyramidal neurons in developing hippocampus. Glia. 2007;55(8):873–884. doi: 10.1002/glia.20509. [DOI] [PubMed] [Google Scholar]
- 52.Duan Y, Sahley CL, Muller KJ. ATP and NO dually control migration of microglia to nerve lesions. Dev Neurobiol. 2009;69(1):60–72. doi: 10.1002/dneu.20689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Miller AM, Stella N. Microglial cell migration stimulated by ATP and C5a involve distinct molecular mechanisms: quantification of migration by a novel near-infrared method. Glia. 2009;57(8):875–883. doi: 10.1002/glia.20813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Choi M, Cho K, Shin S, Ko H, Kwon K, Shin C, Ko K. ATP induced microglial cell migration through non-transcriptional activation of matrix metalloproteinase-9. Arch Pharm Res. 2010;33(2):257–265. doi: 10.1007/s12272-010-0211-8. [DOI] [PubMed] [Google Scholar]
- 55.Lambert C, Ase AR, Seguela P, Antel JP. Distinct migratory and cytokine responses of human microglia and macrophages to ATP. Brain Behav Immun. 2010;24(8):1241–1248. doi: 10.1016/j.bbi.2010.02.010. [DOI] [PubMed] [Google Scholar]
- 56.Samuels SE, Lipitz JB, Dahl G, Muller KJ. Neuroglial ATP release through innexin channels controls microglial cell movement to a nerve injury. J Gen Physiol. 2010;136(4):425–442. doi: 10.1085/jgp.201010476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci. 2006;9(12):1512–1519. doi: 10.1038/nn1805. [DOI] [PubMed] [Google Scholar]
- 58.Simone R, Niturad CE, Nuccio C, Ajmone-Cat MA, Visentin S, Minghetti L. TGF-β and LPS modulate ADP-induced migration of microglial cells through P2Y1 and P2Y12 receptor expression. J Neurochem. 2010;115(2):450–459. doi: 10.1111/j.1471-4159.2010.06937.x. [DOI] [PubMed] [Google Scholar]
- 59.Horvath RJ, DeLeo JA. Morphine enhances microglial migration through modulation of P2X4 receptor signaling. J Neurosci. 2009;29(4):998–1005. doi: 10.1523/JNEUROSCI.4595-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Ifuku M, Farber K, Okuno Y, Yamakawa Y, Miyamoto T, Nolte C, Merrino VF, Kita S, Iwamoto T, Komuro I, Wang B, Cheung G, Ishikawa E, Ooboshi H, Bader M, Wada K, Kettenmann H, Noda M. Bradykinin-induced microglial migration mediated by B1-bradykinin receptors depends on Ca2+ influx via reverse-mode activity of the Na+/Ca2+ exchanger. J Neurosci. 2007;27(48):13065–13073. doi: 10.1523/JNEUROSCI.3467-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cudaback E, Li X, Montine KS, Montine TJ, Keene CD. Apolipoprotein E isoform-dependent microglia migration. FASEB J. 2011;25(6):2082–2091. doi: 10.1096/fj.10-176891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Orr AG, Orr AL, Li XJ, Gross RE, Traynelis SF. Adenosine A(2A) receptor mediates microglial process retraction. Nat Neurosci. 2009;12(7):872–878. doi: 10.1038/nn.2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Putten C, Zuiderwijk-Sick EA, Straalen L, Geus ED, Boven LA, Kondova I, Ijzerman AP, Bajramovic JJ. Differential expression of adenosine A3 receptors controls adenosine A2A receptor-mediated inhibition of TLR responses in microglia. J Immunol. 2009;182(12):7603–7612. doi: 10.4049/jimmunol.0803383. [DOI] [PubMed] [Google Scholar]
- 64.Bernascone S, Erriquez J, Ferraro M, Genazzani AA, Distasi C (2010) Novel adenosine and cAMP signalling pathways in migrating glial cells. Cell Calcium 48 (1):83–90 [DOI] [PubMed]
- 65.Choi I-Y, Lee J-C, Ju C, Hwang S, Cho G-S, Lee HW, Choi WJ, Jeong LS, Kim W-K (2011) A3 adenosine receptor agonist reduces brain ischemic injury and inhibits inflammatory cell migration in rats. Am J Pathol 179 (4):2042–2052 [DOI] [PMC free article] [PubMed]
- 66.Braun N, Sevigny J, Robson SC, Enjyoji K, Guckelberger O, Hammer K, Virgilio F, Zimmermann H. Assignment of ecto-nucleoside triphosphate diphosphohydrolase-1/cd39 expression to microglia and vasculature of the brain. Eur J Neurosci. 2000;12(12):4357–4366. [PubMed] [Google Scholar]
- 67.Nedeljkovic N, Bjelobaba I, Subasic S, Lavrnja I, Pekovic S, Stojkov D, Vjestica A, Rakic L, Stojiljkovic M. Up-regulation of ectonucleotidase activity after cortical stab injury in rats. Cell Biol Int. 2006;30(6):541–546. doi: 10.1016/j.cellbi.2006.03.001. [DOI] [PubMed] [Google Scholar]
- 68.Farber K, Markworth S, Pannasch U, Nolte C, Prinz V, Kronenberg G, Gertz K, Endres M, Bechmann I, Enjyoji K, Robson SC, Kettenmann H. The ectonucleotidase cd39/ENTPDase1 modulates purinergic-mediated microglial migration. Glia. 2008;56(3):331–341. doi: 10.1002/glia.20606. [DOI] [PubMed] [Google Scholar]
- 69.Avignone E, Ulmann L, Levavasseur F, Rassendren F, Audinat E. Status epilepticus induces a particular microglial activation state characterized by enhanced purinergic signaling. J Neurosci. 2008;28(37):9133–9144. doi: 10.1523/JNEUROSCI.1820-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Folkman J. Angiogenesis. Annu Rev Med. 2006;57(1):1–18. doi: 10.1146/annurev.med.57.121304.131306. [DOI] [PubMed] [Google Scholar]
- 71.Shen J, DiCorleto PE. ADP stimulates human endothelial cell migration via P2Y1 nucleotide receptor-mediated mitogen-activated protein kinase pathways. Circ Res. 2008;102(4):448–456. doi: 10.1161/CIRCRESAHA.107.165795. [DOI] [PubMed] [Google Scholar]
- 72.Yokdang N, Tellez JD, Tian H, Norvell J, Barsky SH, Valencik M, Buxton IL. A role for nucleotides in support of breast cancer angiogenesis: heterologous receptor signalling. Br J Cancer. 2011;104(10):1628–1640. doi: 10.1038/bjc.2011.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Gerasimovskaya E, Woodward H, Tucker D, Stenmark K. Extracellular ATP is a pro-angiogenic factor for pulmonary artery vasa vasorum endothelial cells. Angiogenesis. 2008;11(2):169–182. doi: 10.1007/s10456-007-9087-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Roedersheimer M, Nijmeh H, Burns N, Sidiakova AA, Stenmark KR, Gerasimovskaya EV. Complementary effects of extracellular nucleotides and platelet-derived extracts on angiogenesis of vasa vasorum endothelial cells in vitro and subcutaneous Matrigel plugs in vivo. Vasc Cell. 2011;3(1):4. doi: 10.1186/2045-824X-3-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Koyama T, Kimura C, Hayashi M, Watanabe M, Karashima Y, Oike M. Hypergravity induces ATP release and actin reorganization via tyrosine phosphorylation and RhoA activation in bovine endothelial cells. Pflugers Arch. 2009;457(4):711–719. doi: 10.1007/s00424-008-0544-z. [DOI] [PubMed] [Google Scholar]
- 76.Kaczmarek E, Erb L, Koziak K, Jarzyna R, Wink MR, Guckelberger O, Blusztajn JK, Trinkaus-Randall V, Weisman GA, Robson SC. Modulation of endothelial cell migration by extracellular nucleotides: involvement of focal adhesion kinase and phosphatidylinositol 3-kinase-mediated pathways. Thromb Haemost. 2005;93(4):735–742. doi: 10.1267/THRO05040735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Woodward HN, Anwar A, Riddle S, Taraseviciene-Stewart L, Fragoso M, Stenmark KR, Gerasimovskaya EV. PI3K, Rho, and ROCK play a key role in hypoxia-induced ATP release and ATP-stimulated angiogenic responses in pulmonary artery vasa vasorum endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2009;297(5):L954–L964. doi: 10.1152/ajplung.00038.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Quillen EE, Haslam GC, Samra HS, Amani-Taleshi D, Knight JA, Wyatt DE, Bishop SC, Colvert KK, Richter ML, Kitos PA. Ectoadenylate kinase and plasma membrane ATP synthase activities of human vascular endothelial cells. J Biol Chem. 2006;281(30):20728–20737. doi: 10.1074/jbc.M513042200. [DOI] [PubMed] [Google Scholar]
- 79.Zhang X, Gao F, L-l Yu, Peng Y, H-h L, J-y L, Yin M, Ni J. Dual functions of a monoclonal antibody against cell surface F1F0 ATP synthase on both HUVEC and tumor cells1. Acta Pharmacol Sin. 2008;29(8):942–950. doi: 10.1111/j.1745-7254.2008.00830.x. [DOI] [PubMed] [Google Scholar]
- 80.Notari L, Arakaki N, Mueller D, Meier S, Amaral J, Becerra SP. Pigment epithelium-derived factor binds to cell-surface F(1)-ATP synthase. FEBS J. 2010;277(9):2192–2205. doi: 10.1111/j.1742-4658.2010.07641.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Freeman KW, Bowman BR, Zetter BR. Regenerative protein thymosin Œ ≤ −4 is a novel regulator of purinergic signaling. FASEB J. 2011;25(3):907–915. doi: 10.1096/fj.10-169417. [DOI] [PubMed] [Google Scholar]
- 82.Moser TL, Stack MS, Asplin I, Enghild JJ, Hojrup P, Everitt L, Hubchak S, Schnaper HW, Pizzo SV. Angiostatin binds ATP synthase on the surface of human endothelial cells. Proc Natl Acad Sci USA. 1999;96(6):2811–2816. doi: 10.1073/pnas.96.6.2811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Muller T, Robaye B, Vieira RP, Ferrari D, Grimm M, Jakob T, Martin SF, Virgilio F, Boeynaems JM, Virchow JC, Idzko M. The purinergic receptor P2Y2 receptor mediates chemotaxis of dendritic cells and eosinophils in allergic lung inflammation. Allergy. 2010;65(12):1545–1553. doi: 10.1111/j.1398-9995.2010.02426.x. [DOI] [PubMed] [Google Scholar]
- 84.Slater M, Danieletto S, Pooley M, Cheng Teh L, Gidley-Baird A, Barden JA. Differentiation between cancerous and normal hyperplastic lobules in breast lesions. Breast Cancer Res Treat. 2004;83(1):1–10. doi: 10.1023/B:BREA.0000010670.85915.0f. [DOI] [PubMed] [Google Scholar]
- 85.Slater M, Danieletto S, Gidley-Baird A, Teh LC, Barden JA. Early prostate cancer detected using expression of non-functional cytolytic P2X7 receptors. Histopathology. 2004;44(3):206–215. doi: 10.1111/j.0309-0167.2004.01798.x. [DOI] [PubMed] [Google Scholar]
- 86.Adinolfi E, Melchiorri L, Falzoni S, Chiozzi P, Morelli A, Tieghi A, Cuneo A, Castoldi G, Virgilio F, Baricordi OR. P2X7 receptor expression in evolutive and indolent forms of chronic B lymphocytic leukemia. Blood. 2002;99(2):706–708. doi: 10.1182/blood.v99.2.706. [DOI] [PubMed] [Google Scholar]
- 87.Solini A, Cuccato S, Ferrari D, Santini E, Gulinelli S, Callegari MG, Dardano A, Faviana P, Madec S, Virgilio F, Monzani F. Increased P2X7 receptor expression and function in thyroid papillary cancer: a new potential marker of the disease? Endocrinology. 2008;149(1):389–396. doi: 10.1210/en.2007-1223. [DOI] [PubMed] [Google Scholar]
- 88.Wei W, Ryu JK, Choi HB, McLarnon JG. Expression and function of the P2X(7) receptor in rat C6 glioma cells. Cancer Lett. 2008;260(1–2):79–87. doi: 10.1016/j.canlet.2007.10.025. [DOI] [PubMed] [Google Scholar]
- 89.Jelassi B, Chantome A, Alcaraz-Perez F, Baroja-Mazo A, Cayuela ML, Pelegrin P, Surprenant A, Roger S. P2X(7) receptor activation enhances SK3 channels- and cystein cathepsin-dependent cancer cells invasiveness. Oncogene. 2011;30(18):2108–2122. doi: 10.1038/onc.2010.593. [DOI] [PubMed] [Google Scholar]
- 90.Miki K, Tanaka H, Nagai Y, Kimura C, Oike M (2010) Transforming growth factor β1 alters calcium mobilizing properties and endogenous ATP release in A549 cells: possible implications for cell migration. J Pharmacol Sci: 1007220420. doi:10.1254/jphs.10124FP [DOI] [PubMed]
- 91.Chi SL, Wahl ML, Mowery YM, Shan S, Mukhopadhyay S, Hilderbrand SC, Kenan DJ, Lipes BD, Johnson CE, Marusich MF, Capaldi RA, Dewhirst MW, Pizzo SV. Angiostatin-like activity of a monoclonal antibody to the catalytic subunit of F1F0 ATP synthase. Cancer Res. 2007;67(10):4716–4724. doi: 10.1158/0008-5472.CAN-06-1094. [DOI] [PubMed] [Google Scholar]
- 92.Wang L, Zhou X, Zhou T, Ma D, Chen S, Zhi X, Yin L, Shao Z, Ou Z, Zhou P. Ecto-5′-nucleotidase promotes invasion, migration and adhesion of human breast cancer cells. J Cancer Res Clin Oncol. 2008;134(3):365–372. doi: 10.1007/s00432-007-0292-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, Dwyer KM, Smyth MJ. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci U S A. 2010;107(4):1547–1552. doi: 10.1073/pnas.0908801107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Richard CL, Tan EY, Blay J. Adenosine upregulates CXCR4 and enhances the proliferative and migratory responses of human carcinoma cells to CXCL12/SDF-1α. Int J Cancer. 2006;119(9):2044–2053. doi: 10.1002/ijc.22084. [DOI] [PubMed] [Google Scholar]
- 95.Stracke ML, Krutzsch HC, Unsworth EJ, Arestad A, Cioce V, Schiffmann E, Liotta LA. Identification, purification, and partial sequence analysis of autotaxin, a novel motility-stimulating protein. J Biol Chem. 1992;267(4):2524–2529. [PubMed] [Google Scholar]
- 96.Ptaszynska MM, Pendrak ML, Stracke ML, Roberts DD. Autotaxin signaling via lysophosphatidic acid receptors contributes to vascular endothelial growth factor-induced endothelial cell migration. Mol Cancer Res. 2010;8(3):309–321. doi: 10.1158/1541-7786.MCR-09-0288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Panupinthu N, Lee HY, Mills GB. Lysophosphatidic acid production and action: critical new players in breast cancer initiation and progression. Br J Cancer. 2010;102(6):941–946. doi: 10.1038/sj.bjc.6605588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hirakawa M, Oike M, Karashima Y, Ito Y. Sequential activation of RhoA and FAK/paxillin leads to ATP release and actin reorganization in human endothelium. J Physiol. 2004;558(Pt 2):479–488. doi: 10.1113/jphysiol.2004.065334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Roussos ET, Condeelis JS, Patsialou A (2011) Chemotaxis in cancer. Nat Rev Cancer 11 (8):573–587 [DOI] [PMC free article] [PubMed]
- 100.Quezada SA, Peggs KS, Simpson TR, Allison JP (2011) Shifting the equilibrium in cancer immunoediting: from tumor tolerance to eradication. Immunol Rev 241 (1):104–118. doi:10.1111/j.1600-065X.2011.01007.x [DOI] [PMC free article] [PubMed]
- 101.Schuster M, Nechansky A, Kircheis R. Cancer immunotherapy. Biotechnol J. 2006;1(2):138–147. doi: 10.1002/biot.200500044. [DOI] [PubMed] [Google Scholar]
- 102.Barreiro O, Martin P, Gonzalez-Amaro R, Sanchez-Madrid F. Molecular cues guiding inflammatory responses. Cardiovasc Res. 2010;86(2):174–182. doi: 10.1093/cvr/cvq001. [DOI] [PubMed] [Google Scholar]
- 103.Birmingham JM, Busik JV, Hansen-Smith FM, Fenton JI. Novel mechanism for obesity-induced colon cancer progression. Carcinogenesis. 2009;30(4):690–697. doi: 10.1093/carcin/bgp041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Velnar T, Bailey T, Smrkolj V. The wound healing process: an overview of the cellular and molecular mechanisms. J Int Med Res. 2009;37(5):1528–1542. doi: 10.1177/147323000903700531. [DOI] [PubMed] [Google Scholar]
- 105.Aman A, Piotrowski T. Cell migration during morphogenesis. Dev Biol. 2010;341(1):20–33. doi: 10.1016/j.ydbio.2009.11.014. [DOI] [PubMed] [Google Scholar]
- 106.Chung WC, Kermode JC. Suramin disrupts receptor-G protein coupling by blocking association of G protein alpha and betagamma subunits. J Pharmacol Exp Ther. 2005;313(1):191–198. doi: 10.1124/jpet.104.078311. [DOI] [PubMed] [Google Scholar]
- 107.Feng Y, Li M, Wang B, Zheng YG. Discovery and mechanistic study of a class of protein arginine methylation inhibitors. J Med Chem. 2010;53(16):6028–6039. doi: 10.1021/jm100416n. [DOI] [PubMed] [Google Scholar]
- 108.Zengel P, Nguyen-Hoang A, Schildhammer C, Zantl R, Kahl V, Horn E. mu-Slide chemotaxis: a new chamber for long-term chemotaxis studies. BMC Cell Biol. 2011;12(1):21. doi: 10.1186/1471-2121-12-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Zigmond SH, Foxman EF, Segall JE (2001) Chemotaxis assays for eukaryotic cells. In: Current protocols in cell biology. John Wiley & Sons, Inc. doi:10.1002/0471143030.cb1201s00 [DOI] [PubMed]
- 110.Seror C, Melki MT, Subra F, Raza SQ, Bras M, Saidi H, Nardacci R, Voisin L, Paoletti A, Law F, Martins I, Amendola A, Abdul-Sater AA, Ciccosanti F, Delelis O, Niedergang F, Thierry S, Said-Sadier N, Lamaze C, Metivier D, Estaquier J, Fimia GM, Falasca L, Casetti R, Modjtahedi N, Kanellopoulos J, Mouscadet JF, Ojcius DM, Piacentini M, Gougeon ML, Kroemer G, Perfettini JL. Extracellular ATP acts on P2Y2 purinergic receptors to facilitate HIV-1 infection. J Exp Med. 2011;208(9):1823–1834. doi: 10.1084/jem.20101805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Tanneur V, Duranton C, Brand VB, Sandu CD, Akkaya C, Kasinathan RS, Gachet C, Sluyter R, Barden JA, Wiley JS, Lang F, Huber SM. Purinoceptors are involved in the induction of an osmolyte permeability in malaria-infected and oxidized human erythrocytes. FASEB J. 2006;20(1):133–135. doi: 10.1096/fj.04-3371fje. [DOI] [PubMed] [Google Scholar]
- 112.Mackay CR. Moving targets: cell migration inhibitors as new anti-inflammatory therapies. Nat Immunol. 2008;9(9):988–998. doi: 10.1038/ni.f.210. [DOI] [PubMed] [Google Scholar]
- 113.Striedinger K, Meda P, Scemes E. Exocytosis of ATP from astrocyte progenitors modulates spontaneous Ca2+ oscillations and cell migration. Glia. 2007;55(6):652–662. doi: 10.1002/glia.20494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Braun OO, Lu D, Aroonsakool N, Insel PA. Uridine triphosphate (UTP) induces profibrotic responses in cardiac fibroblasts by activation of P2Y2 receptors. J Mol Cell Cardiol. 2010;49(3):362–369. doi: 10.1016/j.yjmcc.2010.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Mayo C, Ren R, Rich C, Stepp MA, Trinkaus-Randall V. Regulation by P2X7: epithelial migration and stromal organization in the cornea. Invest Ophthalmol Vis Sci. 2008;49(10):4384–4391. doi: 10.1167/iovs.08-1688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Crooke A, Mediero A, Guzman-Aranguez A, Pintor J. Silencing of P2Y2 receptor delays Ap4A-corneal re-epithelialization process. Mol Vis. 2009;15:1169–1178. [PMC free article] [PubMed] [Google Scholar]
- 117.Boucher I, Rich C, Lee A, Marcincin M, Trinkaus-Randall V. The P2Y2 receptor mediates the epithelial injury response and cell migration. Am J Physiol Cell Physiol. 2010;299(2):C411–C421. doi: 10.1152/ajpcell.00100.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Vanderstocken G, Bondue B, Horckmans M, Pietrantonio L, Robaye B, Boeynaems JM, Communi D. P2Y2 receptor regulates VCAM-1 membrane and soluble forms and eosinophil accumulation during lung inflammation. J Immunol. 2010;185(6):3702–3707. doi: 10.4049/jimmunol.0903908. [DOI] [PubMed] [Google Scholar]
- 119.Kobayashi T, Kouzaki H, Kita H. Human eosinophils recognize endogenous danger signal crystalline uric acid and produce proinflammatory cytokines mediated by autocrine ATP. J Immunol. 2010;184(11):6350–6358. doi: 10.4049/jimmunol.0902673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hess CN, Kou R, Johnson RP, Li GK, Michel T. ADP signaling in vascular endothelial cells. J Biol Chem. 2009;284(47):32209–32224. doi: 10.1074/jbc.M109.032656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Tagami M, Kusuhara S, Imai H, Uemura A, Honda S, Tsukahara Y, Negi A. MRP4 knockdown enhances migration, suppresses apoptosis, and produces aggregated morphology in human retinal vascular endothelial cells. Biochem Biophys Res Commun. 2010;400(4):593–598. doi: 10.1016/j.bbrc.2010.08.109. [DOI] [PubMed] [Google Scholar]
- 122.Rossi L, Manfredini R, Bertolini F, Ferrari D, Fogli M, Zini R, Salati S, Salvestrini V, Gulinelli S, Adinolfi E, Ferrari S, Virgilio F, Baccarani M, Lemoli RM. The extracellular nucleotide UTP is a potent inducer of hematopoietic stem cell migration. Blood. 2007;109(2):533–542. doi: 10.1182/blood-2006-01-035634. [DOI] [PubMed] [Google Scholar]
- 123.Hashmi AZ, Hakim W, Kruglov EA, Watanabe A, Watkins W, Dranoff JA, Mehal WZ. Adenosine inhibits cytosolic calcium signals and chemotaxis in hepatic stellate cells. Am J Physiol Gastrointest Liver Physiol. 2007;292(1):G395–G401. doi: 10.1152/ajpgi.00208.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Andrade CM, Lopez PL, Noronha BT, Wink MR, Borojevic R, Margis R, Lenz G, Battastini AM, Guma FC. Ecto-5′-nucleotidase/CD73 knockdown increases cell migration and mRNA level of collagen I in a hepatic stellate cell line. Cell Tissue Res. 2011;344(2):279–286. doi: 10.1007/s00441-011-1140-7. [DOI] [PubMed] [Google Scholar]
- 125.Giltaire S, Lambert S, Poumay Y. HB-EGF synthesis and release induced by cholesterol depletion of human epidermal keratinocytes is controlled by extracellular atp and involves both p38 and ERK1/2 signaling pathways. J Cell Physiol. 2011;226(6):1651–1659. doi: 10.1002/jcp.22496. [DOI] [PubMed] [Google Scholar]
- 126.Taboubi S, Milanini J, Delamarre E, Parat F, Garrouste F, Pommier G, Takasaki J, Hubaud JC, Kovacic H, Lehmann M. G alpha(q/11)-coupled P2Y2 nucleotide receptor inhibits human keratinocyte spreading and migration. FASEB J. 2007;21(14):4047–4058. doi: 10.1096/fj.06-7476com. [DOI] [PubMed] [Google Scholar]
- 127.Taboubi S, Garrouste F, Parat F, Pommier G, Faure E, Monferran S, Kovacic H, Lehmann M. Gq-coupled purinergic receptors inhibit insulin-like growth factor-I/phosphoinositide 3-kinase pathway-dependent keratinocyte migration. Mol Biol Cell. 2010;21(6):946–955. doi: 10.1091/mbc.E09-06-0497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lin C, Ren S, Zhang L, Jin H, Sun J, Zuo Y (2011) Extracellular ATP induces CD44 shedding from macrophage-like P388D1 cells via the P2X7 receptor. Hematol Oncol. doi:10.1002/hon.1008 [DOI] [PubMed]
- 129.Martins JP, Silva RB, Coutinho-Silva R, Takiya CM, Battastini AM, Morrone FB, Campos MM (2011) P2X7 purinergic receptor and its role in inflammatory and nociceptive alterations associated to cyclophosphamide-induced hemorrhagic cystitis in mice. Br J Pharmacol. doi:10.1111/j.1476-5381.2011.01535.x [DOI] [PMC free article] [PubMed]
- 130.Koroskenyi K, Duro E, Pallai A, Sarang Z, Kloor D, Ucker DS, Beceiro S, Castrillo A, Chawla A, Ledent CA, Fesus L, Szondy Z. Involvement of adenosine A2A receptors in engulfment-dependent apoptotic cell suppression of inflammation. J Immunol. 2011;186(12):7144–7155. doi: 10.4049/jimmunol.1002284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Williams AJ, Cronstein BN (2011) The Effect of A(2A) adenosine receptor activation on C-C chemokine receptor 7 expression in human THP1 macrophages during inflammation. Inflammation. doi:10.1007/s10753-011-9353-1 [DOI] [PMC free article] [PubMed]
- 132.Ode A, Kopf J, Kurtz A, Schmidt-Bleek K, Schrade P, Kolar P, Buttgereit F, Lehmann K, Hutmacher DW, Duda GN, Kasper G. CD73 and CD29 concurrently mediate the mechanically induced decrease of migratory capacity of mesenchymal stromal cells. Eur Cell Mater. 2011;22:26–42. doi: 10.22203/ecm.v022a03. [DOI] [PubMed] [Google Scholar]
- 133.Ohsawa K, Irino Y, Nakamura Y, Akazawa C, Inoue K, Kohsaka S. Involvement of P2X4 and P2Y12 receptors in ATP-induced microglial chemotaxis. Glia. 2007;55(6):604–616. doi: 10.1002/glia.20489. [DOI] [PubMed] [Google Scholar]
- 134.Ben Yebdri F, Kukulski F, Tremblay A, Sévigny J. Concomitant activation of P2Y2 and P2Y6 receptors on monocytes is required for TLR1/2-induced neutrophil migration by regulating IL-8 secretion. Eur J Immunol. 2009;39(10):2885–2894. doi: 10.1002/eji.200939347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Gorini S, Callegari G, Romagnoli G, Mammi C, Mavilio D, Rosano G, Fini M, Virgilio F, Gulinelli S, Falzoni S, Cavani A, Ferrari D, Sala A. ATP secreted by endothelial cells blocks CXCL 1-elicited natural killer cell chemotaxis and cytotoxicity via P2Y receptor activation. Blood. 2010;116(22):4492–4500. doi: 10.1182/blood-2009-12-260828. [DOI] [PubMed] [Google Scholar]
- 136.Liu X, Hashimoto-Torii K, Torii M, Haydar TF, Rakic P. The role of ATP signaling in the migration of intermediate neuronal progenitors to the neocortical subventricular zone. Proc Natl Acad Sci USA. 2008;105(33):11802–11807. doi: 10.1073/pnas.0805180105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Grimm I, Ullsperger SN, Zimmermann H. Nucleotides and epidermal growth factor induce parallel cytoskeletal rearrangements and migration in cultured adult murine neural stem cells. Acta Physiol (Oxf) 2010;199(2):181–189. doi: 10.1111/j.1748-1716.2010.02092.x. [DOI] [PubMed] [Google Scholar]
- 138.Kukulski F, Ben Yebdri F, Bahrami F, Levesque SA, Martin-Satue M, Sevigny J. The P2 receptor antagonist PPADS abrogates LPS-induced neutrophil migration in the murine air pouch via inhibition of MIP-2 and KC production. Mol Immunol. 2010;47(4):833–839. doi: 10.1016/j.molimm.2009.09.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Hyman MC, Petrovic-Djergovic D, Visovatti SH, Liao H, Yanamadala S, Bouis D, Su EJ, Lawrence DA, Broekman MJ, Marcus AJ, Pinsky DJ. Self-regulation of inflammatory cell trafficking in mice by the leukocyte surface apyrase CD39. J Clin Invest. 2009;119(5):1136–1149. doi: 10.1172/JCI36433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Reutershan J, Cagnina RE, Chang D, Linden J, Ley K. Therapeutic anti-inflammatory effects of myeloid cell adenosine receptor A2a stimulation in lipopolysaccharide-induced lung injury. J Immunol. 2007;179(2):1254–1263. doi: 10.4049/jimmunol.179.2.1254. [DOI] [PubMed] [Google Scholar]
- 141.Ando K, Obara Y, Sugama J, Kotani A, Koike N, Ohkubo S, Nakahata N (2010) P2Y2 receptor-Gq/11 signaling at lipid rafts is required for UTP-induced cell migration in NG 108–15 cells. J Pharmacol Exp Ther. doi:10.1124/jpet.110.167528 [DOI] [PubMed]
- 142.Agresti C, Meomartini ME, Amadio S, Ambrosini E, Serafini B, Franchini L, Volonte C, Aloisi F, Visentin S. Metabotropic P2 receptor activation regulates oligodendrocyte progenitor migration and development. Glia. 2005;50(2):132–144. doi: 10.1002/glia.20160. [DOI] [PubMed] [Google Scholar]
- 143.Omatsu-Kanbe M, Inoue K, Fujii Y, Yamamoto T, Isono T, Fujita N, Matsuura H. Effect of ATP on preadipocyte migration and adipocyte differentiation by activating P2Y receptors in 3T3-L1 cells. Biochem J. 2006;393(Pt 1):171–180. doi: 10.1042/BJ20051037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Klawitter S, Hofmann LP, Pfeilschifter J, Huwiler A. Extracellular nucleotides induce migration of renal mesangial cells by upregulating sphingosine kinase-1 expression and activity. Br J Pharmacol. 2007;150(3):271–280. doi: 10.1038/sj.bjp.0706983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Lin CC, Lin WN, Cheng SE, Tung WH, Wang HH, Yang CM (2011) Transactivation of EGFR/PI3K/Akt involved in ATP-induced inflammatory protein expression and cell motility. J Cell Physiol. doi:10.1002/jcp.22880 [DOI] [PubMed]
- 146.Yu N, Erb L, Shivaji R, Weisman GA, Seye CI. Binding of the P2Y2 nucleotide receptor to filamin A regulates migration of vascular smooth muscle cells. Circ Res. 2008;102(5):581–588. doi: 10.1161/CIRCRESAHA.107.162271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Harada K, Matsumoto Y, Umemura K. Adenosine diphosphate receptor P2Y12-mediated migration of host smooth muscle-like cells and leukocytes in the development of transplant arteriosclerosis. Transplantation. 2011;92(2):148–154. doi: 10.1097/TP.0b013e318221d407. [DOI] [PubMed] [Google Scholar]
- 148.Behdad A, Sun X, Khalpey Z, Enjyoji K, Wink M, Wu Y, Usheva A, Robson S. Vascular smooth muscle cell expression of ectonucleotidase CD39 (ENTPD1) is required for neointimal formation in mice. Purinergic Signalling. 2009;5(3):335–342. doi: 10.1007/s11302-009-9158-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Burnett LA, Blais EM, Unadkat JD, Hille B, Tilley SL, Babcock DF. Testicular expression of Adora3i2 in Adora3 knockout mice reveals a role of mouse A3Ri2 and human A3Ri3 adenosine receptors in sperm. J Biol Chem. 2010;285(44):33662–33670. doi: 10.1074/jbc.M110.156075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Rodriguez-Miranda E, Buffone MG, Edwards SE, Ord TS, Lin K, Sammel MD, Gerton GL, Moss SB, Williams CJ. Extracellular adenosine 5′-triphosphate alters motility and improves the fertilizing capability of mouse sperm. Biol Reprod. 2008;79(1):164–171. doi: 10.1095/biolreprod.107.065565. [DOI] [PMC free article] [PubMed] [Google Scholar]



