<<

Oncogene (2010) 29, 5346–5358 & 2010 Macmillan Publishers Limited All rights reserved 0950-9232/10 www.nature.com/onc REVIEW Extracellular triphosphate and adenosine in

J Stagg and MJ Smyth

Cancer Program, Sir Donald and Lady Trescowthick Laboratories, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia

Adenosine triphosphate (ATP) is actively released in the mulated the hypothesis of purinergic extracellular environment in response to tissue damage (Burnstock, 1972). Burnstock’s hypothesis that ATP and cellular stress. Through the activation of P2X and could be released by cells to perform intercellular P2Y receptors, extracellular ATP enhances tissue repair, signaling was initially met with skepticism, as it seemed promotes the recruitment of immune and unlikely that a that acts as an intracellular dendritic cells, and acts as a co-activator of NLR family, source of energy would also as an extracellular pyrin domain-containing 3 (NLRP3) inflammasomes. messenger. Nevertheless, Burnstock pursued his work The conversion of extracellular ATP to adenosine, in and, together with Che Su and John Bevan, reported contrast, essentially through the enzymatic activity of the that ATP was also released from sympathetic nerves ecto-nucleotidases CD39 and CD73, acts as a negative- during stimulation (Su et al., 1971). Three decades later, mechanism to prevent excessive immune responses. following the cloning and characterization of ATP and Here we review the effects of extracellular ATP and adenosine adenosine surface receptors, purinergic signaling is a on tumorigenesis. First, we summarize the functions of well-established concept and constitutes an expanding extracellular ATP and adenosine in the context of tumor field of research in health and disease, including cancer . Second, we present an overview of the immuno- (Burnstock, 2007). Although early studies focused on suppressive and pro-angiogenic effects of extracellular the role of purinergic receptors in neurotransmission, it adenosine. Third, we present experimental evidence that soon became obvious that extracellular ATP and extracellular ATP and adenosine receptors are expressed adenosine have important roles in another system by tumor cells and enhance tumor growth. Finally, we discuss requiring efficient cell-to-cell : the recent studies, including our own work, which suggest that . Over the last decade, there has been therapeutic approaches that promote ATP-mediated activa- increasing interest not only in the effects of extracellular tion of inflammasomes, or inhibit the accumulation of tumor- and on inflammatory responses, derived extracellular adenosine, may constitute effective new but also on general biological pathways, such as cell means to induce anticancer activity. survival, proliferation, differentiation and motility. Oncogene (2010) 29, 5346–5358; doi:10.1038/onc.2010.292; It is becoming increasingly clear that the release of published online 26 July 2010 extracellular purines and pyrimidines represents a ubiquitous means of intercellular communication Keywords: immunosuppression; adenosine; used by different cell types, involved in various ecto-nucleotidases; inflammasome; ATP biological processes and conserved throughout evolu- tion, as evidenced by the discovery of ATP receptors in invertebrates (Fountain et al., 2007) and plants (Kim et al., 2006). Introduction Extracellular ATP in immunity For many scientists in the 1960s, cells could not possibly release a molecule as fundamental as adenosine tripho- ATP receptors sphate (ATP). Owing to its established role in the Krebs In 1978, Burnstock proposed two types of purinergic cycle, there was considerable skepticism to the notion receptors: P1 receptors selective for adenosine and that ATP—and purines in general—could exert extra- P2 receptors selective for ATP and ADP. Some P2 cellular function. In 1970, Burnstock et al. described the receptors additionally bind UTP or UDP (Burnstock, release of extracellular ATP as a transmitter substance by 2006). In 1985, a pharmacological approach was non-adrenergic inhibitory nerves, and later in 1972, for- proposed to distinguish between two types of P2 receptors: ionotropic P2X and metabotropic P2Y Correspondence: Dr J Stagg or Professor MJ Smyth, Cancer receptors. Currently, four subtypes of P1 receptors Immunology Program, Sir Donald and Lady Trescowthick Labora- (A1, A2A, A2B and A3), seven subtypes of P2X tories, Peter MacCallum Cancer Centre, Locked Bag 1, A’Beckett receptors and eight subtypes of P2Y receptors have Street, East Melbourne, Victoria 8006, Australia. E-mails: [email protected] or [email protected] been identified (Figure 1). P2Y receptors are subdivided Received 12 May 2010; revised 11 June 2010; accepted 13 June 2010; into five Gq/G11-coupled subtypes (P2Y1, P2Y2, P2Y4, published online 26 July 2010 P2Y6 and P2Y11) and three Gi/o-coupled subtypes Extracellular and adenosine J Stagg and MJ Smyth 5347

Figure 1 Adenosine triphosphate (ATP) and adenosine signaling. Extracellular ATP binds G--coupled P2Y and trimeric channel P2X receptors. P2Y receptors are subdivided into Gq/G11-coupled subtypes that activate the C and triphosphate pathways, and Gi/o-coupled subtypes that inhibit . P2X7 is an atypical ATP receptor, forming ion channels at low concentrations of ATP, activating NLRP3 inflammasomes by K þ efflux and the recruitment of the -1 hemichannel, and inducing cell death at high concentrations of ATP. Extracellular adenosine binds Gi/o-coupled A1 and A3 receptors and Gs-coupled A2A and A2B receptors. In contrast to A1 and A3 receptors, A2A and A2B receptors increase intracellular cyclic AMP levels.

(, P2Y13 and P2Y14). Gq/G11-coupled P2Y recep- 2006) and the processing and of the tors generally activate the and inositol -1b (IL-1b) and IL-18, important activators triphosphate pathways, whereas Gi/o-coupled P2Y of innate and adaptive immune responses. The NLRP3 receptors generally inhibit adenylyl cyclase and mod- inflammasome is the best-characterized inflammasome ulate ion channels (Abbracchio et al., 2009). and belongs to the intracellular NOD-like receptor In contrast to G-protein-coupled P2Y receptors, P2X (NLR) family. Together with Toll-like receptors (TLRs) receptors are trimeric cationic channels permeable to and C-type lectins, NLRs scan the extracellular and Na þ ,Kþ and Ca2 þ upon activation. Six homomeric intracellular environment for pathogen-associated mo- (P2X1À5 and P2X7) and six heteromeric (P2X1/2, P2X1/4, lecular patterns and host-derived danger-associated P2X1/5, P2X2/3, P2X2/6 and P2X4/6) receptors have been molecular patterns to alert the immune system (Schro- described. The homomeric P2X7 receptor is an atypical der and Tschopp, 2010). The NLRP3 inflammasome is with a longer carboxy-terminal activated in response to various danger , such as tail and a number of polymorphisms or spliced variants co-activation of P2X7 and TLRs, increased cytosolic (Gunosewoyo et al., 2007; Wu et al., 2009). The DNA levels, monosodium urate crystals, fibrillar activation of P2X7 receptors is highly regulated by amyloid-b and high extracellular glucose levels extracellular levels of ATP. Whereas low ATP concen- (Schroder and Tschopp, 2010). The NLRP3 inflamma- trations activate P2X7 ion channels to become perme- some can also be activated independently of TLR able to small , high ATP concentrations result in signaling (Kanneganti et al., 2007). Schroder and pore formation permeable to as large as Tschopp (2010) recently proposed that a unifying 900 kDa, which ultimately causes cell death (Khakh and element in NLRP3 activation might be the generation North, 2006). of . The activation of P2X7 receptors and the recruitment of the pannexin-1 membrane pore may also allow NLRP3 to Extracellular ATP and the NLR family, pyrin enter the cells and directly activate the inflammasome domain-containing 3 inflammasome (Kanneganti et al., 2007). In the context of inflammation, the release of ATP by activated monocytes, dying, injured or stressed cells, degranulating platelets or secreted by them- Extracellular ATP and T helper type 17 cell immune selves, acts as a co-activator of the NLR family, pyrin responses domain-containing 3 (NLRP3) (cryopirin/NALP3) T-helper type 17 cell immune responses are required for inflammasome (Piccini et al., 2008; Netea et al., 2009). the control of infectious agents (Cho et al., 2010) and are The NLRP3 inflammasome is a multiprotein complex involved in the pathogenesis of various autoimmune that triggers caspase-1 activation (Mariathasan et al., diseases, such as multiple sclerosis (Axtell et al., 2010)

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5348 and inflammatory bowel disease (Cho and Weaver, responses. Extracellular ATP, by activation of P2Y 2007). The generation of Th17 cells is controlled by the receptors, has been identified as a potent chemotactic cytokines IL-6, tumor (TGF)-b and IL-23 for immature DCs (Idzko et al., 2002). In (Bettelli et al., 2007). ‘Naturally occurring’ IL-17- contrast, mature DCs exposed to ATP have decreased producing CD4 þ T cells are also present in the gut. migratory capacity (Schnurr et al., 2003). Thus, the Atarashi et al. (2008) recently investigated the contribu- accumulation of ATP at sites of infection may allow the tion of commensal bacteria in the development of Th17 recruitment of immature DCs. The transient inhibition cells. Surprisingly, the study showed that extracellular of DCs’ migration after maturation might then prolong ATP is constitutively released from commensal bacteria antigen encounter. However, extensive exposure to and drives the differentiation of Th17 CD4 þ cells. This extracellular ATP can also inhibit DC functions. ATP- Th17-skewing effect of extracellular ATP has also been stimulated DCs produce less pro-inflammatory shown in monocyte-derived dendritic cells (DCs) cytokines, more IL-10 and synergize with (Schnurr et al., 2005). Thus, excessive accumulation of (IFN)-g in upregulating indoleamine 2,3-dioxygenase extracellular ATP may have an important role in the levels (Marteau et al., 2005). pathogenesis of autoimmune diseases. In support of this hypothesis, mice deficient for the ecto- triphos- phate diphosphohydrolase, CD39, which hydrolyzes ATP, develop exacerbated experimental colitis (Fried- Extracellular adenosine in immunity man et al., 2009). Moreover, patients with low levels of Adenosine receptors CD39 activity have an increased susceptibility for The conversion of extracellular ATP to adenosine, developing Crohn’s disease (Friedman et al., 2009). essentially through the enzymatic activity of the Taken together, these studies are consistent with a membrane-bound nucleotidases CD39 and CD73, acts role for extracellular ATP in driving Th17 immune responses. as a negative-feedback mechanism that prevents ex- cessive immune reactions. The immunosuppressive effects of adenosine are summarized in Figure 2. Extracellular ATP and Extracellular adenosine activates four distinct G-pro- Activation of the ATP receptor P2Y2 has been recently tein-coupled receptors: Gi/o-coupled A1 and A3 recep- implicated in the directed migration (that is, chemotaxis) tors that decrease intracellular cyclic AMP (cAMP) of immune phagocytes, such as macrophages and levels, and Gs-coupled A2A and A2B receptors that neutrophils. However, the means by which P2Y2 increase cAMP levels through the activation of adenylyl regulate the chemotaxis of macrophages and neutrophils cyclase (Hasko and Cronstein, 2004). In contrast to the are markedly different. In monocytes/macrophages, other adenosine receptors, which are all activated extracellular ATP released from an extrinsic source, at physiological concentrations of adenosine (that is, such as apoptotic cells, directly acts as a ‘find-me’ 30–300 nM in interstitial fluid), the A2B receptor requires (Elliott et al., 2009). In contrast, migrating neutrophils high levels of adenosine generated in response to do not migrate toward gradients of ATP, but release pathological conditions (Fredholm et al., 2001). ATP at their leading edge in response to a chemo- attractant, which, through the feedback activation of signaling P2Y and A3 receptors, provides chemotaxis amplifica- 2 A1 and A3 adenosine receptors inhibit adenylyl cyclase tion (Chen et al., 2006). Thus, whereas both macro- and protein A activity, thus decreasing intracel- phages and neutrophils exploit P2Y signaling, only in 2 lular cAMP levels, with studies revealing an important macrophages does it stimulate a non-redundant chemo- function in cardioprotection (Liang and Jacobson, tactic signal. Extracellular ‘find-me’ signals, such as 1998; Reichelt et al., 2005; Hochhauser et al., 2007), ATP, act in concert with ‘keep-out’ signals to induce (Clark et al., 2007) and protection macrophage recruitment. For instance, the release of against septic shock (Gallos et al., 2005). A1 and A3 lactoferrin selectively inhibits the migration of granulo- adenosine receptors have also been linked to phospha- cytes, but not macrophages (Bournazou et al., 2009). tidylinositol 3-kinase, mitogen-activated These recent studies offer a new level of complexity to and protein kinase C pathways (Hasko and Cronstein, the mechanisms governing cellular chemotaxis. The 2004; Gessi et al., 2008). In contrast to other P1 generally conserved functions of purinergic receptors receptors, A3 adenosine receptor presents multiple sites and their ubiquitous expression in different tissues of that are important for receptor suggest that ATP and adenosine receptors may be desensitization after activation (Palmer and Stiles, implicated in the motility of other cell types, including 2000). tumor cells. A2A and A2B adenosine receptors activate adenylyl cyclase and protein kinase A, thereby increasing Extracellular ATP and dendritic cells intracellular cAMP levels (Fredholm et al., 2007). A2A Although monocytes and macrophages are generally and A2B receptors can further activate mitogen- responsible for the non-immunogenic removal of activated protein kinase and protein kinase C. Owing apoptotic cells, DCs are specialized antigen-presenting to its high affinity for adenosine, the A2A adenosine cells whose role is to initiate or regulate immune receptor has a non-redundant immunosuppressive role

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5349

Figure 2 Immunosuppressive effects of extracellular adenosine. The phosphohydrolysis of extracellular adenosine triphosphate (ATP) to adenosine, essentially through the activity of the ecto-nucleotidases CD39 and CD73, acts as a potent negative-feedback mechanism to suppress the functions of innate and adaptive immune cells.

(Ohta and Sitkovsky, 2001; Odashima et al., 2005). The (Cronstein et al., 1990; Firestein et al., 1995; Bouma A2A adenosine receptor is expressed on monocytes/ et al., 1997). macrophages, mast cells, granulocytes, lymphocytes, DCs, natural killer (NK) cells, natural killer T (NKT) Effect of adenosine on NK cells. In contrast to cells, endothelial cells and airway epithelial cells macrophages and neutrophils, the effect of extracellular (Fredholm et al., 2007). Importantly, the regulation of adenosine on NK cells is not as clearly defined. The A2A receptor expression is driven by at least four activation of A3 adenosine receptors have been shown independent promoters (Yu et al., 2004), which most to enhance NK cell functions, although this could be the likely accounts for tissue-specific functions. A2A recep- result of chronic exposure to A3 agonists that induce tor expression is induced by hypoxia, as well as by receptor desensitization (Priebe et al., 1990; Harish (TNF)-a and IL-1a by nuclear et al., 2003). In contrast, activation of A2A adenosine factor-kB (Khoa et al., 2001). The A2B adenosine receptor has been shown to inhibit IFN-g production receptor differs from the other adenosine receptors by its (Lappas et al., 2005). The inhibitory effect of the A2A relative low affinity for adenosine (Feoktistov and receptor on NK cells most likely reflects its ability to Biaggioni, 1997). A2B is transcriptionally induced by increase intracellular cAMP levels, which has been hypoxia-inducible factor-1a activation (Eltzschig et al., shown to suppress NK cell functions (Goto et al., 1983). 2003; Feoktistov et al., 2004), TNF-a,IL-1b, IFN-g and extracellular adenosine (Xaus et al., 1999; Kolachala et al., 2005; St Hilaire et al., 2008). Effect of adenosine on NKT cells. Natural killer T cells are a unique population of T cells involved in the regulation of , infection, immune tole- Adenosine-mediated immunosuppression rance and tumor immunosurveillance (Godfrey and Effect of adenosine on immune phagocytes. The accu- Kronenberg, 2004; Swann et al., 2009). NKT cells mulation of extracellular adenosine profoundly inhibits recognize glycolipids presented in the context of the the function of phagocytes. In macrophages, adenosine major histocompatibility complex class I-like molecule inhibits phagocytosis, production of superoxide and CD1d and produce large amounts of cytokines upon (Eppell et al., 1989) (Hasko et al., 1996), activation. NKT cells deficient in A2A adenosine TNF-a release, major histocompatibility complex class receptor were found to produce significantly less IL-4, II expression and IL-12 production (Hasko et al., 2000). IL-10 and TGF-b, but more IFN-g upon stimulation In neutrophils, adenosine inhibits degranulation, adhe- with alpha-galactosylceramide (Nowak et al., 2010). sion to endothelial cells and superoxide production Consistent with a role for adenosine in regulating NKT

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5350 cell function, CD39-deficient NKT cells have been receptor , T cells fail to proliferate and to shown to produce less IL-4 upon activation (Beldi produce IFN-g upon reactivation, thus essentially et al., 2008). promoting T-cell anergy (Zarek et al., 2008). In vivo activation of A2A adenosine receptors drives CD4 þ þ Effect of adenosine on DCs. Extracellular adenosine T-cell differentiation toward Foxp3 regulatory T cells increases the chemotaxis of immature monocyte-derived (Tregs), most likely owing to a concomitant increase in DCs, but inhibits IL-12 production following activation TGF-b and decrease in IL-6 production following A2A (Panther et al., 2001, 2003). Similarly, immature receptor activation (Zarek et al., 2008). In addition, plasmacytoid DCs also migrate toward a gradient of activation of A2A receptors was found to promote the þ adenosine (Schnurr et al., 2004). Upon maturation, DCs generation of LAG-3 Tregs. alter the expression profile of adenosine receptors, switching from Gi- to Gs-coupled receptors, resulting Adenosine and Tregs. Foxp3 þ Tregs are crucial for the in a loss of chemotactic response and an inhibition of control of autoimmune responses and tumor immunity -producing function (Schnurr et al., 2004). and rely on multiple mechanisms for their immunosup- Recently, Novitskiy et al. (2008) investigated the effect pressive function. Recent studies have shown that the of extracellular adenosine on the differentiation of ecto- CD39 and CD73 are important contribu- human DCs from blood monocytes. The activation of tors to the regulatory function of Foxp3 þ Tregs A2B adenosine receptors on monocytes was shown to (Deaglio et al., 2007). In mice, Foxp3 þ Tregs express divert DC differentiation toward a population that high levels of both CD39 and CD73, upregulate CD73 produced high levels of vascular endothelial growth upon activation, and partly rely on ecto-nucleotidase factor, IL-8, IL-6, IL-10, -2, TGF-b and activity and adenosine for immunosuppression (Ring indoleamine 2, 3-dioxygenase. Remarkably, when adop- et al., 2010). Human Foxp3 þ Tregs also express CD39, tively transferred to tumor-bearing mice, adenosine- which is further upregulated upon Treg activation differentiated DCs significantly enhanced tumor growth. (Miyara et al., 2009), whereas CD73 expression remains unclear. The analysis of CD73 expression may be Effect of adenosine on B cells. The activation of A2A affected by -mediated shedding (Airas et al., adenosine receptors, by increased protein kinase A 1997). Nevertheless, inhibition of CD73 has been shown activity, has been shown to inhibit B-cell receptor- to reduce human Treg-mediated suppression in vitro induced NF-kB activation (Minguet et al., 2005). (Mandapathil et al., 2010). A recent study has further Consistent with a role for adenosine in regulating identified extracellular ATP as an activating molecule of B-cell functions, isotype-switched B cells, and more Tregs (Ring et al., 2010). Thus, extracellular release of specifically a subpopulation of memory B cells, upregu- ATP and its hydrolysis to adenosine by CD39 and CD73 late the expression of CD73, an essential ecto-nucleo- have a crucial role in Treg function. tidase for the generation of extracellular adenosine (Anderson et al., 2007). Further in support of this Production of extracellular adenosine. The extracellular model, a recent study revealed that memory B cells have concentration of adenosine is generally constant in most increased expression of involved in adenosine tissues, but can rapidly increase 100-fold in hypoxic signaling, including A2A adenosine receptor, compared tissue and in response to inflammation (Schulte and with naive B cells (Tomayko et al., 2008). Taken Fredholm, 2003). The dominant pathway leading to together, these studies strongly suggest that isotype- high extracellular adenosine levels is the extracellular switched B cells, especially memory B cells, use phosphohydrolysis of ATP by ecto-nucleotidases. adenosine receptors to control antigenic re-stimulation. These include ecto-nucleoside triphosphate diphospho- hydrolases, ecto- pyrophosphatase and Effect of adenosine on T cells. Extracellular adenosine phosphodiesterases, alkaline and CD73 is also an important inhibitor of T-cell functions. Similar (ecto-50-nucleotidase). Ecto-nucleoside triphosphate to its effect on B cells, adenosine inhibits T-cell receptor- diphosphohydrolases (predominantly CD39) and ecto- induced NF-kB activation (Majumdar and Aggarwal, nucleotide pyrophosphatase and phosphodiesterases 2003). The A2A receptor dominantly regulates T-cell hydrolyze ATP and ADP to AMP, which is further functions and is significantly upregulated upon hydrolyzed to adenosine by CD73. Thus, the membrane- T-cell activation. Activation of A2A receptors during bound nucleotidases CD39 and CD73 are key regulators T-cell activation significantly inhibits cytotoxicity and of the phosphohydrolysis of ATP into adenosine. cytokine production (Ohta et al., 2009) and has been CD39 (NTPDase1) is expressed on endothelial cells reported to inhibit T- (Zhang et al., and subsets of leukocytes, where it hydrolyzes ATP and 2004; Deaglio et al., 2007). T cells activated in the ADP released by activated platelets, thereby inhibiting presence of an A2A receptor agonist maintain a platelet recruitment and thrombus formation (Robson suppressed phenotype after removal of the agonist et al., 2006). CD39 expression on leukocytes also (Ohta et al., 2009). A2A receptor activation in T cells regulates leukocyte migration across the suppresses the development of both Th1 and Th2 into ischemic tissue (Hyman et al., 2009). CD39 also has immune responses, in vitro and in vivo (Csoka et al., important immunosuppressive functions through the 2008). When activated in the presence of an A2A phosphohydrolysis of pro-inflammatory extracellular

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5351 ATP and the generation of extracellular adenosine when et al., 2008), glioma (Bavaresco et al., 2008), glioblas- co-expressed together with CD73 (Dwyer et al., 2007). toma (Ludwig et al., 1999), melanoma (Sadej et al., 2006), ovarian cancer (Jin et al., 2010), cancer CD73 (ecto-50-nucleotidase) (Kondo et al., 2006), esophageal cancer (Fukuda et al., The membrane-bound CD73 (or ecto-50-nucleotidase) is 2004), gastric cancer (Durak et al., 1994), colon cancer considered as the rate-limiting in the generation (Eroglu et al., 2000), prostate cancer and breast cancer of extracellular adenosine (Resta et al., 1998). CD73 (Spychala et al., 2004). Notably, CD73 expression has catalyzes the of and pyrimi- been associated with a pro-metastatic phenotype in dine ribo- and deoxyribonucleoside monophosphates to melanoma and breast cancer (Lee et al., 2003; Leth- the corresponding nucleoside. CD73 expression is Larsen et al., 2009). In breast cancer cells, CD73 upregulated by hypoxia-inducible factor-1a activation expression was regulated by receptor-a, where- and exposure to type I IFNs, and has been associated by loss of -a significantly enhanced with Wnt signaling (Synnestvedt et al., 2002; Niemela CD73 expression (Spychala et al., 2004). In addition, et al., 2004; Spychala and Kitajewski, 2004) and protein CD73 expression was increased in breast tumor cells kinase C activation (Kitakaze et al., 1996). In addition, resistant to doxorubicin (Ujhazy et al., 1996). CD73 also IL-1b,TNF-a and E2 have been shown to increased the resistance of Jurkat leukemic cells to enhance CD73 activity, whereas IFN-g and IL-4 have tumor necrosis factor-related -inducing - been shown to downregulate CD73 (Savic et al., 1990; mediated apoptosis (Mikhailov et al., 2008). Christensen et al., 1992). The structure of CD73 has been reviewed elsewhere (Strater, 2006). Purinergic signaling in cancer CD73 functions. CD73 is expressed on various cell types, including subsets of lymphocytes, endothelial cells Tumor-suppressive effects of extracellular ATP and epithelial cells (Colgan et al., 2006). The expression Owing to its pro-inflammatory role and its direct of CD73 is critical for the regulation of hypoxia-induced cytotoxic function, the ATP receptor P2X7 might act vascular leakage and for the control of inflammatory as a tumor suppressor. Consistent with this hypothesis, responses (Thompson et al., 2004). CD73 upregulation P2X7 levels are significantly lower in various types of on endothelial cells decreases endothelial vascular cell cancer cells compared with adjacent normal tissues of adhesion molecule-1 expression and prevents leukocyte the same origin (Li et al., 2006). In addition, P2X7 attachment. CD73 expression on high endothelial receptors have been found to be cytolytically non- venules also negatively regulates the migration of functional in tumor cells (Slater et al., 2004). Several T and B cells from the blood into draining lymph nodes mechanisms regulate P2X7-induced cell death, including in response to TLR4 activation (Takedachi et al., 2008). glycosylation (Feng et al., 2005), trafficking (Guerra In contrast to its role in lymph nodes, CD73 promotes et al., 2003), oligomerization and internalization (Feng T-cell extravasation in peripheral tissue. Accordingly, et al., 2006). In support of a selective advantage for decreased extravasation of pathogenic T cells was tumor cells with decreased P2X7 levels, a recent study recently suggested as the cause of resistance of CD73- identified putative target sites for micro-RNA-186 and deficient mice to experimental autoimmune encephalo- micro-RNA À150 within the P2X7 , and showed myelitis (Mills et al., 2008). Through the production of that micro-RNA À186 and micro-RNA À150 levels are adenosine and the activation of adenosine receptors on significantly higher in cancer cells than in normal cells endothelial cells, it was suggested that CD73 triggers (Zhou et al., 2008). More evidence that P2X7 may act as adhesion molecules, such as intercellular adhesion a tumor suppressor comes from the analysis of breast molecule 1 on endothelial cells. cancer patients with a genetically dysfunctional P2X7. In addition to its enzymatic function, CD73 has been Breast cancer patients harboring a P2X7 gene associated suggested to have a role in T- (Resta et al., with decreased P2X7 activity have a significantly greater 1998) and . Human T cells treated with a risk of progression to metastatic disease (Ghiringhelli plate-bound anti-CD73 monoclonal antibody and sub- et al., 2009). Taken together, these studies support a mitogenic concentrations of anti-CD3 monoclonal anti- tumor-suppressive role for P2X7. body proliferate more readily and produce more IL-2 (Massaia et al., 1990). CD73 has been shown to regulate Pro-tumorigenic effects of extracellular ATP T-cell adhesion in a mechanism dependent on lympho- Other studies, in contrast, suggest that extracellular cyte function-associated antigen-1 (Airas et al., 2000). ATP can promote tumor growth, either directly by CD73 has also been shown to interact directly with activation of P2 receptors on tumor cells (including , such as , fibronec- P2X7) or indirectly via non-transformed cells. During tin and tenascin C (Dieckhoff et al., 1986; Sadej et al., skin wound healing, the release of extracellular ATP has 2006). been shown to be critical for efficient epithelial repair, revealing the role of purinergic signaling in general cell CD73 on cancer cells. CD73 has been found to be functions. Indeed, activation of ATP receptors stimu- overexpressed in several types of cancer, including lates the proliferation of epidermal keratinocytes (Greig bladder cancer (Stella et al., 2010), leukemia (Mikhailov et al., 2003) and cells (Wilden et al.,

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5352 1998), and initiates migration of epithelial cells (Boucher against trastuzumab (Herceptin) therapy, which relies et al., 2007). Likewise, extracellular ATP released from on p27 upregulation (Lane et al., 2001). A1 adenosine hepatocytes and Kupffer cells immediately after hepa- receptor has also recently been shown to act as a target tectomy has been shown to promote liver regeneration and regulator of estrogen receptor-a in breast cancer (Gonzales et al., 2010). One mechanism by which cells (Lin et al., 2010). extracellular ATP enhances cell function is through the transactivation of receptor A3 receptors in cancer. A3 receptor is also upregulated (EGFR). G-protein-coupled receptors are important in transformed versus adjacent non-transformed cells transactivators of EGFR, thereby promoting cell (Madi et al., 2004). In the case of A3 adenosine survival and proliferation. A recent study has identified receptors, however, synthetic agonists have been shown a P2Y-dependent pathway (most likely P2Y2) that leads to directly suppress tumor cell survival and/or prolifera- to the release of the EGFR ligand TGF-a and tion (Fishman et al., 2004). The antiproliferative effects transactivation of EGFR, through a mechanism invol- of A3 agonists most likely rely on the downregulation of ving mitochondrial production of reactive oxygen A3 adenosine receptors following chronic activation. A3 species (Myers et al., 2009). Interestingly, other groups agonists have been shown to decrease the activity of have also reported the ability of P2Y2 to induce reactive protein kinase A and Akt, resulting in a suppression of oxygen species, including in prostate cancer cells, cyclin D1, c- and cell proliferation (Fishman et al., resulting in increased tumor growth (Sauer et al., 2004). A3 receptor agonists have also been shown to 2001). Although transactivation of EGFR can trigger enhance the antitumor activity of NK cells by IL-12 kinase-dependent pathways, it may further promote cell upregulation (Harish et al., 2003). Finally, in some survival independently of its kinase activity. Indeed, circumstances, the pro-apoptotic effects of A3 receptor recent studies have shown that EGFR maintains basal agonists can be independent of A3 receptor activation intracellular glucose levels in a kinase-independent (Morello et al., 2008). manner (Weihua et al., 2008). Whether extracellular ATP can maintain intracellular glucose levels by EGFR transactivation remains unknown. However, it has been A2A receptors in cancer. The activation of A2A observed that activation of purinergic receptors in- adenosine receptors on endothelial cells significantly creases intracellular ATP content (Di Virgilio et al., enhances tumor angiogenesis (Ahmad et al., 2009). This is consistent with the fact that endothelial cells 2009). The activation of P2X7 has also been shown to increase tumor cell survival. Accordingly, overexpres- associated with tumors have significantly higher levels sion of P2X increased cell survival in serum-free of A2A adenosine receptors. Analysis of A2A receptor 7 expression in samples and matched normal conditions, whereas blocking P2X7 suppressed proli- feration of lymphocytic leukemia cells (Adinolfi et al., lung tissue revealed that there was a significant increased 2002). Taken together, these studies suggest that expression of A2A in early-stage cancer samples accumulation of extracellular ATP can induce impor- (Ahmad et al., 2009). Other studies have shown that tant pro-tumorigenic effects. activation of A2A adenosine receptors directly promotes In addition to its direct effect on tumor cells, tumor cell survival. Indeed, activation of A2A adenosine extracellular ATP may support tumor growth and receptors was found to significantly enhance the metastasis by indirect means by activation of purinergic proliferation of the -dependent breast cancer receptors on non-transformed cells. Chronic exposure of cell line MCF-7 (Etique et al., 2009). C6 glioma cells to ATP, for instance, enhances the In addition to its pro-angiogenic and proliferative release of the pro-inflammatory factors MCP-1, IL-8 effects, the A2A adenosine receptor is a potent and vascular endothelial growth factor (Jantaratnotai suppressor of endogenous tumor immunosurveillance. Accordingly, Ohta et al. (2006) showed that A2A et al., 2009). In thyroid papillary carcinoma cells, P2X7 activation induces IL-6 production (Solini et al., 2008). receptor-deficient mice mount spontaneous antitumor T-cell responses able to induce T-cell-dependent P2X7 has also been shown to trigger the release of the growth factor substance P from neuroblastoma cells tumor rejection. This study suggested that blocking A2A receptors enhanced existing antitumor immune (Raffaghello et al., 2006). Finally, activation of P2X7 in C6 glioma cells has been shown to increase tumor cell responses and increased IFN-g production from anti- þ migration (Wei et al., 2008). tumor CD8 T cells was suggested as the main mechanism of action.

Adenosine receptors in cancer A2B receptors in cancer. A2B adenosine receptors also A1 receptors in cancer. A1 adenosine receptors have have an important role in angiogenesis (Feoktistov been found to be overexpressed in primary breast tumor et al., 2002). The pro-angiogenic effect of A2B receptors tissues and to enhance breast cancer cell proliferation is due to its expression on both endothelial and hemato- (Mirza et al., 2005). RNA interference knockdown poietic cells. Accordingly, studies in A2B-deficient studies suggested that activation of A1 adenosine mice have shown that tumor-infiltrating CD45 þ cells, receptors decreases p27 expression, thereby upregulating most likely CD11b þ myeloid cells, produce up to CDK4 activity (Mirza et al., 2005). The activation of A1 fivefold more vascular endothelial growth factor when adenosine receptors may thus contribute to resistance proficient in A2B adenosine receptors (Ryzhov et al.,

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5353 2008). The expression of A2B adenosine receptors on et al., 2007). However, the addition of recombinant mast cells may also significantly enhance tumor angio- CRT and high-mobility group box 1 to live tumor cells genesis. The stimulation of A2B adenosine receptors in is not sufficient to induce antitumor immune responses, mast cells has been shown to induce the production of implying another level of regulation. As activation of IL-4, IL-13, IL-8 and vascular endothelial growth factor inflammasomes is central to the instigation of immune (Ryzhov et al., 2004). responses, Zitvogel and colleagues hypothesized that the NLRP3 inflammasome may constitute a third level of regulation of immunogenic chemotherapy (Ghiringhelli et al., 2009). Zitvogel and colleagues recently showed Modulating purinergic signaling for cancer therapy that activation of the NLRP3 inflammasome in DCs is indeed a critical event in the induction of antitumor ATP release and immunogenic chemotherapy immune responses following treatment with doxorubicin, Specific classes of chemotherapeutic , such as mitoxanthrone and oxaliplatin (Ghiringhelli et al., 2009). anthracyclines, have recently been shown to be potent Ghiringhelli et al. (2009) further tested whether the release of inducers of immunogenic cancer cell death, thereby ATP from dying tumor cells was involved in the activation triggering antitumor immune responses (Ghiringhelli of the NLRP3 inflammasome. Numerous chemotherapeu- et al., 2009) (Figure 3). Most remarkably, the antitumor tic drugs—that is, cadmium, etoposide, mitomycin C, immune responses induced by these specific chemother- oxaliplatin, cisplatin, staurosporine, thapsigargin, mi- apeutic drugs were shown to be essential for therapeutic toxanthrone and doxorubicin—could indeed trigger activity in mice. Intriguingly, drugs such as anthra- the release of extracellular ATP from tumor cells before cyclines and oxaliplatin induce potent immunogenic cell and during apoptosis (Martins et al., 2009). Remark- death, whereas other drugs, such as etoposide and ably, mice deficient in the ATP receptor P2X7 bearing mitomycin C, fail to do so. In an attempt to elucidate EL4 or EG7 tumors failed to respond to oxaliplatin the mechanism of immunogenic chemotherapy, Obeid treatment and failed to mount tumor-specific CD8 þ et al. (2007) showed that pro-inflammatory apoptosis of T-cell responses, a phenotype that could be rescued by tumor cells was directly linked to the rapid translocation the adoptive transfer of wild-type DCs. Taken together, of (CRT) to the cell surface upon exposure these studies strongly suggest that anthracyclines and to the . Dying tumor cells must thus translocate the oxaliplatin induce immunogenic cell death by the endoplasmic reticulum-resident CRT and disulfide iso- translocation of CRT on tumor cells and the activation, merase ERp57 to the plasma membrane to be efficiently on DCs, of TLR4 by high-mobility group box 1 and phagocytosed by DCs. In addition to CRT transloca- P2X7 by ATP release. These events in turn activate the tion, the release of chromatin-binding high-mobility NLRP3 inflammasome in DCs, stimulate antigen group box 1 protein by dying tumor cells and its presentation and IL-1b release, thereby generating activation of TLR4 on DCs are also required (Apetoh IFN-g-producing tumor-specific CD8 þ T cells.

Figure 3 Modulating purinergic signaling for cancer therapy. Altering the balance between extracellular adenosine triphosphate (ATP) and adenosine can induce anticancer activity. Anthracyclines and oxaliplatin trigger ATP release, calreticulin (CRT) translocation and high-mobility group box 1 (HMGB1) release from tumor cells, thereby activating NLRP3 inflammasome and inducing antitumor immune responses that are essential for therapeutic activity (Ghiringhelli et al., 2009). However, tumor cells often overexpress ecto-nucleotidases, such as CD39 and CD73, which hydrolyze extracellular ATP to adenosine. Two recent studies, including our own work, have identified tumor-derived CD73 as an important mechanism of tumor immune escape (Stagg et al., 2010). Antibody-based therapy against CD73 can significantly inhibit tumor growth, chemotaxis and metastasis, suggesting that CD73 may constitute a potential cancer target.

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5354 Blocking A2A adenosine receptors involving co-activation of the dependence receptor As discussed above, Ohta et al. (2006) showed that mice DCC (Rodrigues et al., 2007). genetically deficient in A2A receptors are significantly protected against CL8-1 and RMA tumor challenges, most likely by enhanced antitumor CD8 þ T-cell Conclusion response. The authors of that study also tested whether pharmacological inhibition of A2A receptors could One of the most important consequences of tumorigen- induce antitumor activity in wild-type mice. Indeed, esis is the disruption of tissue architecture. Among other injection of A2A antagonists significantly enhanced the pathways, tissue damage triggers the release of extra- therapeutic activity of adoptively transferred tumor- cellular ATP, which activates P2X and P2Y receptors to specific CD8 þ T cells. No therapeutic effect was promote tissue repair. The activation of ATP receptors observed against non-immunogenic B16 tumors, or in enhances cell survival, proliferation and migration, and nude mice with CL8-1 tumors, suggesting a T-cell- acts as a chemotactic molecule for the recruitment of dependent mechanism. Interestingly, pharmacological immune phagocytes and DCs. In the presence of danger- inhibition of A2A receptors was associated with the associated molecules, extracellular ATP further development of self-resolving autoimmunity as evi- activates NLRP3 inflammasomes, thereby triggering a denced by transient hair loss. As pointed out by the pro-inflammatory cascade leading to the activation of authors, ‘it is important to carefully consider the known innate and adaptive immune responses. Concurrently, cardiovascular and neurological effects of A2A and A2B regulatory mechanisms are in place to prevent excessive antagonists’. Potential side effects of A2A receptor tissue damage that might be caused by unresolved antagonists might include increased blood and inflammation. Tissue hypoxia and the accumulation of inflammation. However, based on the results of recent specific cytokines, such as type I IFNs and TNF-a, are clinical trials with A2A receptor antagonists against important activators of these regulatory mechanisms. Parkinson’s disease, A2A antagonists seem to be well- One of the most important immunosuppressive regula- tolerated (LeWitt et al., 2008). tory pathways is the phosphohydrolysis of extracellular ATP to adenosine. In the context of cancer, tumor cells can directly Targeting CD73 exploit ATP and adenosine receptors for survival, Although pharmacological antagonists to adenosine proliferation and motility. In addition, tumor cells have receptors may hold therapeutic potential, an alternative been found to express high levels of ecto-nucleotidases, is to target the ecto-nucleotidase CD73. The observation such as CD73, that catabolize the formation of that CD73 is expressed at high levels in various types of extracellular adenosine. Extracellular adenosine gener- cancer supports this approach. Using RNA-interference ated from the activity of CD73 promotes tumor knockdown of CD73, we have recently shown that angiogenesis, immune escape and metastasis. The CD73 expression by breast tumor cells significantly accumulation of extracellular adenosine thus constitutes impairs adaptive antitumor immune responses (Stagg an important regulatory mechanism in the generation of et al., 2010). Another independent group recently antitumor immunity. Our data and that of others corroborated our findings, revealing that tumor-derived strongly suggest that the ecto-nucleotidase CD73 may CD73 inhibited T-cell immunosurveillance of murine constitute a previously unrecognized target for cancer ovarian tumors (Jin et al., 2010). The latter study therapy. showed that CD73 on tumor cells inhibited T-cell It has become increasingly evident that antitumor function and induced T-cell apoptosis. immune responses are essential not only for the We have investigated whether CD73 could be a potential endogenous control of tumorigenesis, but also for the target for antibody-based therapy (clone TY/23). Tumors therapeutic activity of standard treatment, such as of treated mice were significantly reduced in size chemotherapy. However, several regulatory mechanisms, compared with control-treated mice (Stagg et al., or ‘immune-checkpoints’, limit the generation of anti- 2010). As 4T1.2 tumors can spontaneously metastasize cancer immune responses. Various strategies targeting after injection in the mammary fat pad, we also specific immune-checkpoints can be deployed to enhance observed that anti-CD73 monoclonal antibody treat- antitumor immunity. Understanding the relative impor- ment significantly reduced the number of spontaneous tance of each of these immune-checkpoints in specific lung metastases. Our data also revealed that CD73- cancer types in the context of patients’ individual derived adenosine enhanced tumor cell chemotaxis. polymorphism will be of primary importance. Thorough Others had previously shown that CD73 expression in translational studies will thus be essential for the breast cancer cells was associated with increased development of safe and effective new cancer treatments migration (Zhi et al., 2007). Our data further suggested aiming to harness antitumor immune responses. a role for A2B adenosine receptors in tumor cell metastasis. A2B receptors could potentially enhance receptor expression (Richard et al., 2006) or act as an important co-receptor. Accordingly, A2B Conflict of interest adenosine receptors have been shown to promote human colon cancer cell motility by a mechanism The authors declare no conflict of interest.

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5355 Acknowledgements Breast Cancer Foundation, the Victorian Breast Consortium, the Victorian Cancer Agency, and by JS is supported by a Canadian Institutes of Health Research a National Health and Medical Research Council of Australia (CIHR) Fellowship. MJS is supported by the Susan G Komen (NH&MRC) Australia Fellowship and Program Grant.

References

Abbracchio MP, Burnstock G, Verkhratsky A, Zimmermann H. Chen Y, Corriden R, Inoue Y, Yip L, Hashiguchi N, Zinkernagel A (2009). in the : an overview. et al. (2006). ATP release guides neutrophil chemotaxis via P2Y2 Trends Neurosci 32: 19–29. and A3 receptors. Science 314: 1792–1795. Adinolfi E, Melchiorri L, Falzoni S, Chiozzi P, Morelli A, Tieghi A Cho JH, Weaver CT. (2007). The of inflammatory bowel et al. (2002). P2X7 receptor expression in evolutive and indolent disease. Gastroenterology 133: 1327–1339. forms of chronic B lymphocytic leukemia. Blood 99: 706–708. Cho JS, Pietras EM, Garcia NC, Ramos RI, Farzam DM, Monroe Ahmad A, Ahmad S, Glover L, Miller SM, Shannon JM, Guo X et al. HR et al. (2010). IL-17 is essential for host defense against (2009). is a unique angiogenic target of cutaneous Staphylococcus aureus infection in mice. J Clin Invest HIF-2alpha in pulmonary endothelial cells. Proc Natl Acad Sci USA 120: 1762–1773. 106: 10684–10689. Christensen LD, Andersen V, Nygaard P, Bendtzen K. (1992). Effects Airas L, Niemela J, Jalkanen S. (2000). CD73 engagement promotes of immunomodulators on ecto-50-nucleotidase activity on blood lymphocyte binding to endothelial cells via a lymphocyte function- mononuclear cells in vitro. Scand J Immunol 35: 407–413. associated antigen-1-dependent mechanism. J Immunol 165: Clark AN, Youkey R, Liu X, Jia L, Blatt R, Day YJ et al. (2007). A1 5411–5417. adenosine receptor activation promotes angiogenesis and release of Airas L, Niemela J, Salmi M, Puurunen T, Smith DJ, Jalkanen S. VEGF from monocytes. Circ Res 101: 1130–1138. (1997). Differential regulation and function of CD73, a glycosyl- Colgan SP, Eltzschig HK, Eckle T, Thompson LF. (2006). -linked 70-kD adhesion molecule, on lympho- Physiological roles for ecto-50-nucleotidase (CD73). Purinergic cytes and endothelial cells. J Cell Biol 136: 421–431. Signal 2: 351–360. Anderson SM, Tomayko MM, Ahuja A, Haberman AM, Shlomchik Cronstein BN, Daguma L, Nichols D, Hutchison AJ, Williams M. MJ. (2007). New markers for murine memory B cells that define (1990). The adenosine/neutrophil paradox resolved: human neu- mutated and unmutated subsets. J Exp Med 204: 2103–2114. trophils possess both A1 and A2 receptors that promote chemotaxis Apetoh L, Ghiringhelli F, Tesniere A, Obeid M, Ortiz C, Criollo A and inhibit O2 generation, respectively. J Clin Invest 85: 1150–1157. et al. (2007). Toll-like receptor 4-dependent contribution of the Csoka B, Himer L, Selmeczy Z, Vizi ES, Pacher P, Ledent C et al. immune system to anticancer chemotherapy and radiotherapy. Nat (2008). Adenosine A2A receptor activation inhibits T helper 1 and Med 13: 1050–1059. T helper 2 cell development and function. FASEB J 22: Atarashi K, Nishimura J, Shima T, Umesaki Y, Yamamoto M, Onoue 3491–3499. M et al. (2008). ATP drives lamina propria T(H)17 cell differentia- Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A et al. tion. Nature 455: 808–812. (2007). Adenosine generation catalyzed by CD39 and CD73 Axtell RC, de Jong BA, Boniface K, van der Voort LF, Bhat R, De expressed on regulatory T cells mediates immune suppression. Sarno P et al. (2010). T helper type 1 and 17 cells determine efficacy J Exp Med 204: 1257–1265. of interferon-beta in multiple sclerosis and experimental encephalo- Di Virgilio F, Ferrari D, Adinolfi E. (2009). P2X(7): a growth- myelitis. Nat Med 16: 406–412. promoting receptor—implications for cancer. Purinergic Signal 5: Bavaresco L, Bernardi A, Braganhol E, Cappellari AR, Rockenbach 251–256. L, Farias PF et al. (2008). The role of ecto-50-nucleotidase/CD73 in Dieckhoff J, Mollenhauer J, Kuhl U, Niggemeyer B, von der Mark K, glioma cell line proliferation. Mol Cell Biochem 319: 61–68. Mannherz HG. (1986). The extracellular matrix proteins laminin Beldi G, Wu Y, Banz Y, Nowak M, Miller L, Enjyoji K et al. (2008). and fibronectin modify the AMPase activity of 50-nucleotidase from Natural killer T cell dysfunction in CD39-null mice protects against chicken gizzard smooth muscle. FEBS Lett 195: 82–86. concanavalin A-induced hepatitis. Hepatology 48: 841–852. Durak I, Cetin R, Canbolat O, Cetin D, Yurtarslani Z, Unal A. (1994). Bettelli E, Oukka M, Kuchroo VK. (2007). T(H)-17 cells in the circle Adenosine deaminase, 50-nucleotidase, guanase and deami- of immunity and autoimmunity. Nat Immunol 8: 345–350. nase activities in gastric tissues from patients with gastric cancer. Boucher I, Yang L, Mayo C, Klepeis V, Trinkaus-Randall V. (2007). Cancer Lett 84: 199–202. Injury and induce phosphorylation of epidermal growth Dwyer KM, Deaglio S, Gao W, Friedman D, Strom TB, Robson SC. factor receptor: MMP and HB-EGF dependent pathway. Exp Eye (2007). CD39 and control of cellular immune responses. Purinergic Res 85: 130–141. Signal 3: 171–180. Bouma MG, Jeunhomme TM, Boyle DL, Dentener MA, Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Voitenok NN, van den Wildenberg FA et al. (1997). Adenosine Walk SF et al. (2009). Nucleotides released by apoptotic cells inhibits neutrophil degranulation in activated human whole blood: act as a find-me signal to promote phagocytic clearance. Nature 461: involvement of adenosine A2 and A3 receptors. J Immunol 158: 282–286. 5400–5408. Eltzschig HK, Ibla JC, Furuta GT, Leonard MO, Jacobson KA, Bournazou I, Pound JD, Duffin R, Bournazos S, Melville LA, Brown Enjyoji K et al. (2003). Coordinated adenine nucleotide phospho- SB et al. (2009). Apoptotic human cells inhibit migration of hydrolysis and nucleoside signaling in posthypoxic endothelium: granulocytes via release of lactoferrin. J Clin Invest 119: 20–32. role of and adenosine A2B receptors. J Exp Med Burnstock G. (1972). Purinergic nerves. Pharmacol Rev 24: 509–581. 198: 783–796. Burnstock G. (2006). Historical review: ATP as a . Eppell BA, Newell AM, Brown EJ. (1989). Adenosine receptors are Trends Pharmacol Sci 27: 166–176. expressed during differentiation of monocytes to macrophages Burnstock G. (2007). and pathophysiology of purinergic in vitro. Implications for regulation of phagocytosis. J Immunol 143: neurotransmission. Physiol Rev 87: 659–797. 4141–4145. Burnstock G, Campbell G, Satchell D, Smythe A. (1970). Evidence Eroglu A, Canbolat O, Demirci S, Kocaoglu H, Eryavuz Y, Akgul H. that adenosine triphosphate or a related nucleotide is the transmitter (2000). Activities of adenosine deaminase and 50-nucleotidase in substance released by non-adrenergic inhibitory nerves in the gut. cancerous and noncancerous human colorectal tissues. Med Oncol Br J Pharmacol 40: 668–688. 17: 319–324.

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5356 Etique N, Grillier-Vuissoz I, Lecomte J, Flament S. (2009). proliferation, differentiation, and apoptosis in human fetal epider- between adenosine receptor (A2A isoform) and ERalpha mis. J Invest Dermatol 121: 1145–1149. mediates ethanol action in MCF-7 breast cancer cells. Oncol Rep 21: Guerra AN, Fisette PL, Pfeiffer ZA, Quinchia-Rios BH, Prabhu U, 977–981. Aga M et al. (2003). Purinergic receptor regulation of LPS-induced Feng YH, Li X, Wang L, Zhou L, Gorodeski GI. (2006). A truncated signaling and pathophysiology. J Endotoxin Res 9: 256–263. P2X7 receptor variant (P2X7-j) endogenously expressed in cervical Gunosewoyo H, Coster MJ, Kassiou M. (2007). Molecular probes for cancer cells antagonizes the full-length P2X7 receptor through P2X7 receptor studies. Curr Med Chem 14: 1505–1523. hetero-oligomerization. J Biol Chem 281: 17228–17237. Harish A, Hohana G, Fishman P, Arnon O, Bar-Yehuda S. (2003). Feng YH, Wang L, Wang Q, Li X, Zeng R, Gorodeski GI. (2005). A3 adenosine receptor agonist potentiates natural killer cell activity. ATP stimulates GRK-3 phosphorylation and beta-arrestin-2- Int J Oncol 23: 1245–1249. dependent internalization of P2X7 receptor. Am J Physiol Cell Hasko G, Cronstein BN. (2004). Adenosine: an endogenous regulator Physiol 288: C1342–C1356. of innate immunity. Trends Immunol 25: 33–39. Feoktistov I, Biaggioni I. (1997). Adenosine A2B receptors. Pharmacol Hasko G, Kuhel DG, Salzman AL, Szabo C. (2000). ATP suppression Rev 49: 381–402. of interleukin-12 and tumour necrosis factor-alpha release from Feoktistov I, Goldstein AE, Ryzhov S, Zeng D, Belardinelli L, Voyno- macrophages. Br J Pharmacol 129: 909–914. Yasenetskaya T et al. (2002). Differential expression of adenosine Hasko G, Szabo C, Nemeth ZH, Kvetan V, Pastores SM, Vizi ES. receptors in human endothelial cells: role of A2B receptors in (1996). Adenosine receptor agonists differentially regulate IL-10, angiogenic factor regulation. Circ Res 90: 531–538. TNF-alpha, and nitric oxide production in RAW 264.7 macro- Feoktistov I, Ryzhov S, Zhong H, Goldstein AE, Matafonov A, Zeng phages and in endotoxemic mice. J Immunol 157: 4634–4640. D et al. (2004). Hypoxia modulates adenosine receptors in human Hochhauser E, Leshem D, Kaminski O, Cheporko Y, Vidne BA, endothelial and smooth muscle cells toward an A2B angiogenic Shainberg A. (2007). The protective effect of prior ischemia phenotype. Hypertension 44: 649–654. reperfusion adenosine A1 or A3 receptor activation in the Firestein GS, Bullough DA, Erion MD, Jimenez R, Ramirez-Wein- normal and hypertrophied heart. Interact Cardiovasc Thorac Surg house M, Barankiewicz J et al. (1995). Inhibition of neutrophil 6: 363–368. adhesion by adenosine and an adenosine kinase inhibitor. The role Hyman MC, Petrovic-Djergovic D, Visovatti SH, Liao H, Yanama- of selectins. J Immunol 154: 326–334. dala S, Bouis D et al. (2009). Self-regulation of inflammatory cell Fishman P, Bar-Yehuda S, Ohana G, Barer F, Ochaion A, Erlanger A trafficking in mice by the leukocyte surface apyrase CD39. J Clin et al. (2004). An agonist to the A3 adenosine receptor inhibits colon Invest 119: 1136–1149. carcinoma growth in mice via modulation of GSK-3 beta and NF- Idzko M, Dichmann S, Ferrari D, Di Virgilio F, la Sala A, Girolomoni kappa B. Oncogene 23: 2465–2471. G et al. (2002). Nucleotides induce chemotaxis and polymer- Fountain SJ, Parkinson K, Young MT, Cao L, Thompson CR, North ization in immature but not mature human dendritic cells RA. (2007). An intracellular P2X receptor required for osmoregula- via activation of pertussis toxin-sensitive P2y receptors. Blood 100: tion in Dictyostelium discoideum. Nature 448: 200–203. 925–932. Fredholm BB, AP IJ, Jacobson KA, Klotz KN, Linden J. (2001). Jantaratnotai N, Choi HB, McLarnon JG. (2009). ATP stimulates International Union of . XXV. Nomenclature and chemokine production via a store-operated calcium entry pathway classification of adenosine receptors. Pharmacol Rev 53: 527–552. in C6 glioma cells. BMC Cancer 9: 442. Fredholm BB, Chern Y, Franco R, Sitkovsky M. (2007). Aspects of Jin D, Fan J, Wang L, Thompson LF, Liu A, Daniel BJ et al. (2010). the general of adenosine A2A signaling. Prog Neurobiol 83: CD73 on tumor cells impairs antitumor T-cell responses: a novel 263–276. mechanism of tumor-induced immune suppression. Cancer Res 70: Friedman DJ, Kunzli BM, YI AR, Sevigny J, Berberat PO, Enjyoji K 2245–2255. et al. (2009). From the Cover: CD39 deletion exacerbates Kanneganti TD, Lamkanfi M, Kim YG, Chen G, Park JH, Franchi L experimental murine colitis and human polymorphisms increase et al. (2007). Pannexin-1-mediated recognition of bacterial mole- susceptibility to inflammatory bowel disease. Proc Natl Acad Sci cules activates the cryopyrin inflammasome independent of Toll-like USA 106: 16788–16793. receptor signaling. Immunity 26: 433–443. Fukuda K, Sakakura C, Miyagawa K, Kuriu Y, Kin S, Nakase Y et al. Khakh BS, North RA. (2006). P2X receptors as cell-surface ATP (2004). Differential gene expression profiles of radioresistant sensors in health and disease. Nature 442: 527–532. oesophageal cancer cell lines established by continuous fractionated Khoa ND, Montesinos MC, Reiss AB, Delano D, Awadallah N, irradiation. Br J Cancer 91: 1543–1550. Cronstein BN. (2001). Inflammatory cytokines regulate function Gallos G, Ruyle TD, Emala CW, Lee HT. (2005). A1 adenosine and expression of adenosine A(2A) receptors in human monocytic receptor knockout mice exhibit increased mortality, renal dysfunc- THP-1 cells. J Immunol 167: 4026–4032. tion, and hepatic injury in murine septic peritonitis. Am J Physiol Kim SY, Sivaguru M, Stacey G. (2006). Extracellular ATP in plants. Renal Physiol 289: F369–F376. Visualization, localization, and analysis of physiological significance Gessi S, Merighi S, Varani K, Leung E, Mac Lennan S, Borea PA. in growth and signaling. Plant Physiol 142: 984–992. (2008). The A3 adenosine receptor: an enigmatic player in cell Kitakaze M, Node K, Minamino T, Komamura K, Funaya H, biology. Pharmacol Ther 117: 123–140. Shinozaki Y et al. (1996). Role of activation of protein kinase C in Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C et al. the infarct size-limiting effect of ischemic preconditioning through (2009). Activation of the NLRP3 inflammasome in dendritic cells activation of ecto-50-nucleotidase. Circulation 93: 781–791. induces IL-1beta-dependent adaptive immunity against tumors. Nat Kolachala V, Asamoah V, Wang L, Obertone TS, Ziegler TR, Merlin Med 15: 1170–1178. D et al. (2005). TNF-alpha upregulates adenosine 2b (A2b) receptor Godfrey DI, Kronenberg M. (2004). Going both ways: immune expression and signaling in intestinal epithelial cells: a basis for regulation via CD1d-dependent NKT cells. J Clin Invest 114: A2bR overexpression in colitis. Cell Mol Sci 62: 2647–2657. 1379–1388. Kondo T, Nakazawa T, Murata SI, Katoh R. (2006). Expression of Gonzales E, Julien B, Serriere-Lanneau V, Nicou A, Doignon I, CD73 and its ecto-50-nucleotidase activity are elevated in papillary Lagoudakis L et al. (2010). ATP release after partial hepatectomy thyroid carcinomas. Histopathology 48: 612–614. regulates liver regeneration in the rat. J Hepatol 52: 54–62. Lane HA, Motoyama AB, Beuvink I, Hynes NE. (2001). Modulation Goto T, Herberman RB, Maluish A, Strong DM. (1983). Cyclic AMP of p27/Cdk2 complex formation through 4D5-mediated inhibition as a mediator of prostaglandin E-induced suppression of human of HER2 receptor signaling. Ann Oncol 12(Suppl 1): S21–S22. natural killer cell activity. J Immunol 130: 1350–1355. Lappas CM, Rieger JM, Linden J. (2005). A2A adenosine receptor Greig AV, Linge C, Cambrey A, Burnstock G. (2003). Purinergic induction inhibits IFN-gamma production in murine CD4 þ T cells. receptors are part of a signaling system for keratinocyte J Immunol 174: 1073–1080.

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5357 Lee H, Lin EC, Liu L, Smith JW. (2003). Gene expression profiling of human CD4 þ T cells expressing the FoxP3 . tumor xenografts: in vivo analysis of -specific metastasis. Int J Immunity 30: 899–911. Cancer 107: 528–534. Morello S, Petrella A, Festa M, Popolo A, Monaco M, Vuttariello E Leth-Larsen R, Lund R, Hansen HV, Laenkholm AV, Tarin D, Jensen et al. (2008). Cl-IB-MECA inhibits human thyroid cancer cell ON et al. (2009). Metastasis-related plasma membrane proteins of proliferation independently of A3 adenosine receptor activation. human breast cancer cells identified by comparative quantitative Cancer Biol Ther 7: 278–284. mass spectrometry. Mol Cell 8: 1436–1449. Myers TJ, Brennaman LH, Stevenson M, Higashiyama S, Russell WE, LeWitt PA, Guttman M, Tetrud JW, Tuite PJ, Mori A, Chaikin P Lee DC et al. (2009). Mitochondrial reactive oxygen species mediate et al. (2008). Adenosine A2A GPCR-induced TACE/ADAM17-dependent transforming growth (KW-6002) reduces ‘off’ time in Parkinson’s disease: a double-blind, factor-alpha shedding. Mol Biol Cell 20: 5236–5249. randomized, multicenter clinical trial (6002-US-005). Ann Neurol 63: Netea MG, Nold-Petry CA, Nold MF, Joosten LA, Opitz B, van der 295–302. Meer JH et al. (2009). Differential requirement for the activation of Li X, Zhou L, Feng YH, Abdul-Karim FW, Gorodeski GI. (2006). the inflammasome for processing and release of IL-1beta in The P2X7 receptor: a novel biomarker of uterine epithelial . monocytes and macrophages. Blood 113: 2324–2335. Cancer Epidemiol Biomarkers Prev 15: 1906–1913. Niemela J, Henttinen T, Yegutkin GG, Airas L, Kujari AM, Rajala P Liang BT, Jacobson KA. (1998). A physiological role of the adenosine et al. (2004). IFN-alpha induced adenosine production on the A3 receptor: sustained cardioprotection. Proc Natl Acad Sci USA endothelium: a mechanism mediated by CD73 (ecto-50-nucleotidase) 95: 6995–6999. up-regulation. J Immunol 172: 1646–1653. Lin Z, Yin P, Reierstad S, O’Halloran M, Coon VJ, Pearson EK et al. Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, (2010). , a target and regulator of estrogen Tikhomirov OY et al. (2008). Adenosine receptors in regulation of receptoralpha action, mediates the proliferative effects of estradiol dendritic cell differentiation and function. Blood 112: 1822–1831. in breast cancer. Oncogene 29: 1114–1122. Nowak M, Lynch L, Yue S, Ohta A, Sitkovsky M, Balk SP et al. Ludwig HC, Rausch S, Schallock K, Markakis E. (1999). Expression (2010). The A2aR adenosine receptor controls cytokine production of CD 73 (ecto-50-nucleotidase) in 165 glioblastomas by immuno- in iNKT cells. Eur J Immunol 40: 682–687. histochemistry and electronmicroscopic histochemistry. Anticancer Obeid M, Tesniere A, Ghiringhelli F, Fimia GM, Apetoh L, Perfettini Res 19: 1747–1752. JL et al. (2007). Calreticulin exposure dictates the immunogenicity Madi L, Ochaion A, Rath-Wolfson L, Bar-Yehuda S, Erlanger A, of cancer cell death. Nat Med 13: 54–61. Ohana G et al. (2004). The A3 adenosine receptor is highly Odashima M, Bamias G, Rivera-Nieves J, Linden J, Nast CC, expressed in tumor versus normal cells: potential target for tumor Moskaluk CA et al. (2005). Activation of A2A adenosine receptor growth inhibition. Clin Cancer Res 10: 4472–4479. attenuates intestinal inflammation in animal models of inflamma- Majumdar S, Aggarwal BB. (2003). Adenosine suppresses activation of tory bowel disease. Gastroenterology 129: 26–33. nuclear factor-kappaB selectively induced by tumor necrosis factor Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MK in different cell types. Oncogene 22: 1206–1218. et al. (2006). A2A adenosine receptor protects tumors from Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, antitumor T cells. Proc Natl Acad Sci USA 103: 13132–13137. Szajnik M, Ren J et al. (2010). Generation and accumulation of Ohta A, Madasu M, Kini R, Subramanian M, Goel N, Sitkovsky M. immunosuppressive adenosine by human CD4 þ CD25highFOXP3 þ (2009). A2A adenosine receptor may allow expansion of T cells regulatory T cells. JBiolChem285: 7176–7186. lacking effector functions in extracellular adenosine-rich micro- Mariathasan S, Weiss DS, Newton K, McBride J, O’Rourke K, environments. J Immunol 183: 5487–5493. Roose-Girma M et al. (2006). Cryopyrin activates the inflamma- Ohta A, Sitkovsky M. (2001). Role of G-protein-coupled adenosine some in response to toxins and ATP. Nature 440: 228–232. receptors in downregulation of inflammation and protection from Marteau F, Gonzalez NS, Communi D, Goldman M, Boeynaems JM. tissue damage. Nature 414: 916–920. (2005). Thrombospondin-1 and indoleamine 2, 3-dioxygenase are Palmer TM, Stiles GL. (2000). Identification of threonine residues major targets of extracellular ATP in human dendritic cells. Blood controlling the agonist-dependent phosphorylation and desensitiza- 106: 3860–3866. tion of the rat A(3) adenosine receptor. Mol Pharmacol 57: 539–545. Martins I, Tesniere A, Kepp O, Michaud M, Schlemmer F, Senovilla L Panther E, Corinti S, Idzko M, Herouy Y, Napp M, la Sala A et al. et al. (2009). Chemotherapy induces ATP release from tumor cells. (2003). Adenosine affects expression of membrane molecules, 8: 3723–3728. cytokine and chemokine release, and the T-cell stimulatory capacity Massaia M, Perrin L, Bianchi A, Ruedi J, Attisano C, Altieri D et al. of human dendritic cells. Blood 101: 3985–3990. (1990). Human T cell activation. Synergy between CD73 (ecto-50- Panther E, Idzko M, Herouy Y, Rheinen H, Gebicke-Haerter PJ, nucleotidase) and signals delivered through CD3 and CD2 Mrowietz U et al. (2001). Expression and function of adenosine molecules. J Immunol 145: 1664–1674. receptors in human dendritic cells. FASEB J 15: 1963–1970. Mikhailov A, Sokolovskaya A, Yegutkin GG, Amdahl H, West A, Piccini A, Carta S, Tassi S, Lasiglie D, Fossati G, Rubartelli A. (2008). Yagita H et al. (2008). CD73 participates in cellular multiresistance ATP is released by monocytes stimulated with pathogen-sensing program and protects against TRAIL-induced apoptosis. J Immunol receptor and induces IL-1beta and IL-18 secretion in an 181: 464–475. autocrine way. Proc Natl Acad Sci USA 105: 8067–8072. Mills JH, Thompson LF, Mueller C, Waickman AT, Jalkanen S, Priebe T, Ruiz L, Nelson JA. (1990). Role of natural killer cells in the Niemela J et al. (2008). CD73 is required for efficient entry of modulation of primary antibody production by purine lymphocytes into the central nervous system during experimental and their analogs. Cell Immunol 130: 513–519. autoimmune encephalomyelitis. Proc Natl Acad Sci USA 105: Raffaghello L, Chiozzi P, Falzoni S, Di Virgilio F, Pistoia V. (2006). 9325–9330. The P2X7 receptor sustains the growth of human neuroblastoma Minguet S, Huber M, Rosenkranz L, Schamel WW, Reth M, cells through a substance P-dependent mechanism. Cancer Res 66: Brummer T. (2005). Adenosine and cAMP are potent inhibitors 907–914. of the NF-kappa B pathway downstream of immunoreceptors. Eur Reichelt ME, Willems L, Molina JG, Sun CX, Noble JC, Ashton KJ J Immunol 35: 31–41. et al. (2005). Genetic deletion of the A1 adenosine receptor limits Mirza A, Basso A, Black S, Malkowski M, Kwee L, Pachter JA et al. myocardial ischemic tolerance. Circ Res 96: 363–367. (2005). RNA interference targeting of A1 receptor-overexpressing Resta R, Yamashita Y, Thompson LF. (1998). Ecto-enzyme and breast carcinoma cells leads to diminished rates of cell proliferation signaling functions of lymphocyte CD73. Immunol Rev 161: 95–109. and induction of apoptosis. Cancer Biol Ther 4: 1355–1360. Richard CL, Tan EY, Blay J. (2006). Adenosine upregulates CXCR4 and Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A et al. enhances the proliferative and migratory responses of human (2009). Functional delineation and differentiation dynamics of carcinoma cells to CXCL12/SDF-1alpha. Int J Cancer 119: 2044–2053.

Oncogene Extracellular adenosine triphosphate and adenosine J Stagg and MJ Smyth 5358 Ring S, Enk AH, Mahnke K. (2010). ATP activates regulatory T Cells Strater N. (2006). Ecto-50-nucleotidase: structure function relation- in vivo during contact reactions. J Immunol 184: ships. Purinergic Signal 2: 343–350. 3408–3416. Su C, Bevan JA, Burnstock G. (1971). [3H]adenosine triphosphate: Robson SC, Sevigny J, Zimmermann H. (2006). The E-NTPDase release during stimulation of enteric nerves. Science 173: family of ectonucleotidases: Structure function relationships and 336–338. pathophysiological significance. Purinergic Signal 2: 409–430. Swann JB, Uldrich AP, van Dommelen S, Sharkey J, Murray WK, Rodrigues S, De Wever O, Bruyneel E, Rooney RJ, Gespach C. (2007). Godfrey DI et al. (2009). Type I natural killer T cells suppress Opposing roles of netrin-1 and the dependence receptor DCC in tumors caused by p53 loss in mice. Blood 113: 6382–6385. cancer cell invasion, tumor growth and metastasis. Oncogene 26: Synnestvedt K, Furuta GT, Comerford KM, Louis N, Karhausen J, 5615–5625. Eltzschig HK et al. (2002). Ecto-50-nucleotidase (CD73) regulation Ryzhov S, Goldstein AE, Matafonov A, Zeng D, Biaggioni I, by hypoxia-inducible factor-1 mediates permeability changes in Feoktistov I. (2004). Adenosine-activated mast cells induce IgE intestinal epithelia. J Clin Invest 110: 993–1002. synthesis by B lymphocytes: an A2B-mediated process involving Takedachi M, Qu D, Ebisuno Y, Oohara H, Joachims ML, McGee ST Th2 cytokines IL-4 and IL-13 with implications for . et al. (2008). CD73-generated adenosine restricts lymphocyte J Immunol 172: 7726–7733. migration into draining lymph nodes. J Immunol 180: 6288–6296. Ryzhov S, Novitskiy SV, Zaynagetdinov R, Goldstein AE, Carbone Thompson LF, Eltzschig HK, Ibla JC, Van De Wiele CJ, Resta R, DP, Biaggioni I et al. (2008). Host A(2B) adenosine receptors Morote-Garcia JC et al. (2004). Crucial role for ecto-50-nucleotidase promote carcinoma growth. Neoplasia 10: 987–995. (CD73) in vascular leakage during hypoxia. J Exp Med 200: Sadej R, Spychala J, Skladanowski AC. (2006). Expression of ecto-50- 1395–1405. nucleotidase (eN, CD73) in cell lines from various stages of human Tomayko MM, Anderson SM, Brayton CE, Sadanand S, Steinel NC, melanoma. Melanoma Res 16: 213–222. Behrens TW et al. (2008). Systematic comparison of gene expression Sauer H, Klimm B, Hescheler J, Wartenberg M. (2001). Activation of between murine memory and naive B cells demonstrates that p90RSK and growth stimulation of multicellular tumor spheroids memory B cells have unique signaling capabilities. J Immunol 181: are dependent on reactive oxygen species generated after purinergic 27–38. receptor stimulation by ATP. FASEB J 15: 2539–2541. Ujhazy P, Berleth ES, Pietkiewicz JM, Kitano H, Skaar JR, Ehrke MJ Savic V, Stefanovic V, Ardaillou N, Ardaillou R. (1990). Induction of et al. (1996). Evidence for the involvement of ecto-50-nucleotidase ecto-50-nucleotidase of rat cultured mesangial cells by interleukin-1 (CD73) in drug resistance. Int J Cancer 68: 493–500. beta and tumour necrosis factor-alpha. Immunology 70: 321–326. Wei W, Ryu JK, Choi HB, McLarnon JG. (2008). Expression and Schnurr M, Toy T, Shin A, Hartmann G, Rothenfusser S, Soellner J function of the P2X(7) receptor in rat C6 glioma cells. Cancer Lett et al. (2004). Role of adenosine receptors in regulating chemotaxis 260: 79–87. and cytokine production of plasmacytoid dendritic cells. Blood 103: Weihua Z, Tsan R, Huang WC, Wu Q, Chiu CH, Fidler IJ et al. 1391–1397. (2008). Survival of cancer cells is maintained by EGFR independent Schnurr M, Toy T, Shin A, Wagner M, Cebon J, Maraskovsky E. of its kinase activity. Cancer Cell 13: 385–393. (2005). Extracellular nucleotide signaling by P2 receptors inhibits Wilden PA, Agazie YM, Kaufman R, Halenda SP. (1998). IL-12 and enhances IL-23 expression in human dendritic cells: a ATP-stimulated smooth muscle cell proliferation requires novel role for the cAMP pathway. Blood 105: 1582–1589. independent ERK and PI3 K signaling pathways. Am J Physiol Schnurr M, Toy T, Stoitzner P, Cameron P, Shin A, Beecroft T et al. 275: H1209–H1215. (2003). ATP gradients inhibit the migratory capacity of specific Wu PY, Lin YC, Chang CL, Lu HT, Chin CH, Hsu TT et al. (2009). human dendritic cell types: implications for P2Y11 receptor Functional decreases in P2X7 receptors are associated with retinoic signaling. Blood 102: 613–620. acid-induced neuronal differentiation of Neuro-2a neuroblastoma Schroder K, Tschopp J. (2010). The inflammasomes. Cell 140: cells. Cell Signal 21: 881–891. 821–832. Xaus J, Mirabet M, Lloberas J, Soler C, Lluis C, Franco R et al. Schulte G, Fredholm BB. (2003). Signalling from adenosine receptors (1999). IFN-gamma up-regulates the A2B adenosine receptor to mitogen-activated protein . Cell Signal 15: 813–827. expression in macrophages: a mechanism of macrophage deactiva- Slater M, Danieletto S, Gidley-Baird A, Teh LC, Barden JA. (2004). tion. J Immunol 162: 3607–3614. Early prostate cancer detected using expression of non-functional Yu L, Frith MC, Suzuki Y, Peterfreund RA, Gearan T, Sugano S et al. cytolytic P2X7 receptors. Histopathology 44: 206–215. (2004). Characterization of genomic organization of the adenosine Solini A, Cuccato S, Ferrari D, Santini E, Gulinelli S, Callegari MG A2A receptor gene by molecular and analyses. Brain et al. (2008). Increased P2X7 receptor expression and function in Res 1000: 156–173. thyroid papillary cancer: a new potential marker of the disease? Zarek PE, Huang CT, Lutz ER, Kowalski J, Horton MR, Linden J 149: 389–396. et al. (2008). A2A receptor signaling promotes peripheral tolerance Spychala J, Kitajewski J. (2004). Wnt and beta-catenin signaling target by inducing T-cell anergy and the generation of adaptive regulatory the expression of ecto-50-nucleotidase and increase extracellular T cells. Blood 111: 251–259. adenosine generation. Exp Cell Res 296: 99–108. Zhang H, Conrad DM, Butler JJ, Zhao C, Blay J, Hoskin DW. (2004). Spychala J, Lazarowski E, Ostapkowicz A, Ayscue LH, Jin A, Mitchell Adenosine acts through A2 receptors to inhibit IL-2-induced BS. (2004). Role of estrogen receptor in the regulation of ecto-50- tyrosine phosphorylation of STAT5 in T lymphocytes: role of nucleotidase and adenosine in breast cancer. Clin Cancer Res 10: cyclic adenosine 30,50-monophosphate and phosphatases. 708–717. J Immunol 173: 932–944. St Hilaire C, Koupenova M, Carroll SH, Smith BD, Ravid K. (2008). Zhi X, Chen S, Zhou P, Shao Z, Wang L, Ou Z et al. (2007). RNA TNF-alpha upregulates the A2B adenosine receptor gene: the role of interference of ecto-50-nucleotidase (CD73) inhibits human breast NAD(P)H oxidase 4. Biochem Biophys Res Commun 375: 292–296. cancer cell growth and invasion. Clin Exp Metast 24: 439–448. Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer Zhou L, Qi X, Potashkin JA, Abdul-Karim FW, Gorodeski GI. D et al. (2010). Anti-CD73 antibody therapy inhibits breast tumor (2008). miR-186 and miR-150 down-regulate expres- growth and metastasis. Proc Natl Acad Sci USA 107: 1547–1552. sion of the pro-apoptotic purinergic P2X7 receptor by activation of Stella J, Bavaresco L, Braganhol E, Rockenbach L, Farias PF, Wink instability sites at the 30-untranslated region of the gene that MR et al. (2010). Differential expression in human decrease steady-state levels of the transcript. J Biol Chem 283: bladder cancer cell lines. Urol Oncol 28: 260–267. 28274–28286.

Oncogene