Abstract
Recent progress in our understanding of the unique role of A2B receptors in the regulation of inflammation, immunity and tissue repair was considerably facilitated with the introduction of new pharmacological and genetic tools. However, it also led to seemingly conflicting conclusions on the role of A2B adenosine receptors in inflammation with some publications indicating pro-inflammatory effects and others suggesting the opposite. This chapter reviews the functions of A2B receptors in various cell types related to inflammation and integrated effects of A2B receptor modulation in several animal models of inflammation. It is argued that translation of current findings into novel therapies would require a better understanding of A2B receptors functions in diverse types of inflammatory responses in various tissues and at different points of their progression.
Keywords: Adenosine, Inflammation, Neovascularization, Receptors Purinergic P1, Tissue Remodeling
I. Introduction
The extracellular accumulation of adenosine contributes to the regulation of inflammation, immunity and tissue repair. Adenosine exerts its action by interacting with four adenosine receptor subtypes, A1, A2A, A2B and A3 that belong to the family of seven transmembrane G protein-coupled receptors (Fredholm et al., 2001a).
Among adenosine receptor subtypes the A2B receptor has the lowest affinity to adenosine requiring micromolar concentrations to become functional, whereas the affinities of other adenosine receptor subtypes are significantly higher, rendering them active well below micromolar concentrations of adenosine (Fredholm et al., 2001b). Therefore, it is likely that A2B receptors remain silent under the resting conditions when extracellular adenosine concentrations are low, estimated between ten and few hundred nM range (Fredholm, 2007), but their role becomes more important in pathophysiological conditions when adenosine concentrations are the highest.
Because both A2A and A2B receptor subtypes stimulate adenylate cyclase, A2B receptors were often viewed as being a redundant low affinity version of A2A receptors. However, there is also strong evidence that A2B receptors play a non-redundant role distinct from, and often opposite to that of A2A receptors.
II. Tools to study A2B adenosine receptor function
Recent progress in our understanding of the unique role of A2B receptors was considerably facilitated with the introduction of new pharmacological and genetic tools. Results from using these complementary approaches, however, have not always agreed, with some publications indicating pro-inflammatory effects and others suggesting the opposite. We will address these issues later in this chapter, but first will discuss potential pitfalls in using genetic and pharmacological tools that might have contributed to inadvertent misinterpretation of results.
Generation of mice deficient in A2B adenosine receptor provided a powerful tool for investigating its function in vivo and in vitro. Several studies have found significant differences between A2BKO and WT mouse phenotypes at rest. These differences include increases in basal TNF-α secretion, leukocyte adhesion and vascular permeability as well as changes in expression of E-selectin, P-selectin, ICAM-1, IκB and P2Y1 receptors (Eckle et al., 2008a;Yang et al., 2010;Yang et al., 2006). This is surprising because basal levels of adenosine are thought to be too low to activate A2B receptors. As with many gene knockouts there is always concern that potential compensatory changes in the expression of other proteins may confound the specific role of A2B receptors in any given process. Therefore, it is important to keep in mind limitations of gene knockouts so that appropriate controls can be included and correct conclusions can be made.
A number of potent and selective A2B receptor antagonists have been synthesized over the last decade (for review see (Kalla and Zablocki, 2009)). They were used together with antagonists of other adenosine receptor subtypes to elucidate the role of A2B receptors in inflammatory models. When interpreting these studies, however, attention should be paid to the use of antagonists at concentrations that remain selective for the receptor to be targeted. For example, DPCPX and ZM241385 are often used as selective A1 and A2A antagonists, respectively, but the fact that they can also bind to human and mouse A2B receptors at low nanomolar concentrations (Kreckler et al., 2006;Linden et al., 1999) is often overlooked. Table in this chapter summarizes binding affinities of adenosine antagonists at human and mouse adenosine receptor subtypes commonly used to study the role of A2B receptors.
Table.
Affinity of commonly used antagonists to human (h) and mouse (m) adenosine receptor subtypes determined in radioligand binding assays (inhibition constants in μM).
| Compounds | Subtypes
|
|||
|---|---|---|---|---|
| A1 | A2A | A2B | A3 | |
| ALT-801 | h 5.0a | h 0.7a | h 0.02a | h 6.3a |
| m 5.2a | m 3.5a | m 0.2a | m >10a | |
| CVT 6883 | h 1.9b | h 3.3b | h 0.02b | h 1.0b |
| MRS 1706 | h 0.2c | h 0.1c | h 0.001c | h 0.2c |
| MRS 1754 | h 0.4c | h 0.5c | h 0.002c | h 0.6c |
| m 0.009d | m × 10d | m 0.003d | m > 10d | |
| PSB1115 | h > 10b | h > 10b | h 0.05b | h > 10b |
| DPCPX | h 0.004e | h 0.1e | h 0.05f | h 4.0e |
| m 0.002d | m 0.6d | m 0.09d | m > 10d | |
| ZM 241385 | h 0.3g | h 0.0008g | h 0.03f | h > 10g |
| m 0.2d | m 0.0007d | m 0.03d | m > 100d | |
The goal of attaining selectivity for A2B agonists has been more elusive than for antagonists but several potent A2B agonists have been developed recently (for review see (Baraldi et al., 2009)). BAY 60-6583 has been described as a high-affinity (EC50 of 3 nM) and specific A2B agonist based on its effects on the activity of reporters co-expressed together with human adenosine receptors (Eckle et al., 2007). However, the reported affinity of BAY 60-6583 to A2B receptors determined by radiolgand binding were approximately two order magnitude lower (Ki of 330 – 750 nM) (Auchampach et al., 2009). It is likely, therefore, that the affinity of BAY 60-6583 to A2B receptors was overestimated in a functional reporter assay, possibly due to the significant receptor reserve in cells overexpressing A2B receptors (Linden et al., 1999). Comparative binding studies at all four adenosine receptor subtypes would be necessary to validate the selectivity of this compound toward A2B receptors.
In summary, the combination of novel genetic and pharmacological approaches provides powerful tools to dissect the role of adenosine signaling through A2B receptors in physiological and pathological processes. As important as these advances are, we wanted to alert the reader to the importance of taking into account the pitfalls and limitations of these approaches for the correct interpretation of results of studies on the role of A2B receptors in adenosine-dependent regulation of inflammatory responses.
III. A2B receptors on immune cells
A. Neutrophils, lymphocytes, platelets
A2B receptors are ubiquitously expressed and, therefore, it is not surprising that they are present on various cells of hematopoietic origin. In most cases, A2B receptors are co-expressed with A2A receptors. A2B receptor transcripts are found in neutrophils (Fredholm et al., 1996) lymphocytes (Gessi et al., 2005) and even platelets (Amisten et al., 2008). Little is known about their specific functions in these cells and A2A receptors appear to predominate (Csoka et al., 2008;Fredholm et al., 1996;Yang et al., 2010).
Recently, the neuronal guidance molecule netrin-1 was proposed to inhibit neutrophil migration through activation of A2B receptors located on these cells (Rosenberger et al., 2009). Although intriguing, the issue of interactions between netrin-1 and A2B receptors remains controversial. It has been previously hypothesized that A2B receptors can also serve as receptors for netrin-1 (Corset et al., 2000). However, there is evidence against a direct effect of netrin-1 on A2B receptors. First, binding assays in COS cells overexpressing A2B receptors failed to demonstrate netrin-1 binding to these receptors (McKenna et al., 2008). Second, the lack of effects of 100 μM DPCPX and ZM241385, known to inhibit human A2B receptors (see Table), on the netrin-1-dependent inhibition of neutrophil migration (Rosenberger et al., 2009) argues against this hypothesis.
B. Mast cells
1. Role of A2B receptors in mast cell degranulation
Adenosine has distinct effects on mast cell degranulation of pre-formed mediators and on release of newly generated cytokine/growth factors. Adenosine is known to potentiate antigen-induced degranulation of mast cells. This effect of adenosine is mediated by A3 adenosine receptors in rodent (Ramkumar et al., 1993;Salvatore et al., 2000), but perhaps not in human (Walker et al., 1997) or canine (Auchampach et al., 1997) mast cells.
The role of A2B receptors in mast cell degranulation is less clear. A2B, but not A3 receptors both stimulated directly and potentiated the effects of the calcium ionophore A23187 on degranulation of canine BR mastocytoma cells (Auchampach et al., 1997). Conversely, A2B receptors were proposed to play an inhibitory role in degranulation of mouse bone marrow derived mast cells (BMMCs), based on the finding that A2BKO mice show an exaggerated antigen-induced mast cell degranulation (Hua et al., 2007). Acknowledging the implausibility of explaining this phenomenon by tonic stimulation of the low-affinity A2B receptor by endogenous adenosine, Hua et al proposed the alternative explanation that A2B receptors may be constitutively active in wild type BMMCs even in the absence of an agonist. However, we determined that A2B receptors expressed in wild type BMMCs display no constitutive activity. Furthermore, our work demonstrated that A2B receptors do not inhibit the A3 receptor-mediated potentiation of antigen-induced degranulation of BMMCs (Ryzhov et al., 2008d). It is likely therefore that exaggerated antigen-induced degranulation in BMMCs deficient of A2B receptors is unrelated to the loss of adenosine signaling function of A2B receptors. Compensatory developmental changes in mice or rearrangement of proteins normally coupled to the A2B receptor as a result of the A2B knockout may have contributed in this phenomenon.
In contrast, pharmacological studies suggested that A2A but not A2B receptors can attenuate antigen-induced degranulation in human cord blood-derived mast cells (Suzuki et al., 1998) and primary human lung mast cells (Duffy et al., 2007). This effect was attributed to the ability of A2A receptors to close K+ channel KCa3.1 by a cAMP-independent mechanism (Duffy et al., 2007).
2. Role of A2B receptors in cytokine/growth factor secretion from mast cells
The role of A2B receptors in regulation of cytokine/growth factor secretion from mast cells is better understood than its effects on degranulation. Studies in HMC-1 cells showed that only A2B, but not A2A or A3 receptors stimulate secretion of angiogenic factors IL-8 and VEGF, and the Th2 cytokines IL-13 and IL-4 (Feoktistov and Biaggioni, 1995;Feoktistov et al., 2003;Ryzhov et al., 2006;Ryzhov et al., 2004). Conditioned media from A2B receptor-activated mast cells stimulated human umbilical vein endothelial cell (HUVEC) proliferation and migration, and induced capillary tube formation. These pro-angiogenic effects of A2B receptor-stimulated mast cells were attributed primarily to VEGF release, because they were blocked by anti-VEGF antibody (Feoktistov et al., 2003). Co-culturing B lymphocytes with A2B receptor-stimulated mast cells induced IgE production by B lymphocytes, an effect that appeared to be secondary to increased secretion of Th2 cytokines IL-4 and IL-13 by the mast cells (Ryzhov et al., 2004).
Like human HMC-1 cells, mouse BMMCs express A3, A2A and A2B, but not A1 receptors (Feoktistov et al., 2003;Meade et al., 2002;Ryzhov et al., 2008d). Activation of adenosine receptors stimulated IL-13 and VEGF secretion only in wild type (WT) but not in A2BKO BMMCs. Contrary to adenosine action on degranulation, which is only apparent in antigen-stimulated BMMCs, these effects do not require activation of FcεRI receptors, since they are evident even in the absence of antigen (Ryzhov et al., 2008d).
The notion that only A2B, but not A2A receptors co-expressed in the same cells, were able to stimulate cytokine/growth factor secretion could seemed paradoxical because both receptor subtypes were thought to act by stimulation of adenylate cyclase through Gs proteins. However, studies in HMC-1 revealed that in contrast to A2A receptors, A2B receptors are also coupled to phospholipase C (PLC), as evidenced by increase in inositol phosphate production with consequent mobilization of intracellular calcium. These A2B receptor-dependent pathways are stimulated through a cholera toxin- and pertussis toxin-insensitive G-protein, presumably of the Gq family (Feoktistov and Biaggioni, 1995). In addition, stimulation of A2B receptors activates the small GTP-binding protein p21ras. This event triggers ERK signaling pathway with sequential stimulation of Raf, MEK1/2 and ERK1/2 protein kinase activities (Feoktistov et al., 1999). We have also demonstrated the coupling of adenosine receptors to JNK and p38 MAPK signaling pathways (Feoktistov et al., 1999). The fact that A2B receptors are coupled to multiple intracellular signaling pathways in mast cells, explains their ability to regulate the generation and secretion of diverse cytokines and growth factors.
A2B receptors stimulate release of VEGF, IL-13 and IL-8 from mast cells by mechanisms that involve activation of ERK and p38 MAPK (Ryzhov et al., 2006;Ryzhov et al., 2008d). Stimulation of the receptor tyrosine kinase c-kit with stem cell factor (Meade et al., 2002) or the receptor complex ST2/IL1RAP with IL-33 (Silver et al., 2010) synergized with A2B receptors in the upregulation of IL-8 from HMC-1 cells. Functional analysis of cells transfected with full-length and truncated receptor constructs revealed that the A2B receptor C-terminus is important for coupling to Gs and Gq proteins. However, the A2B receptor C-terminus is not essential for upregulation of IL-8. Instead, integrity of the third intracellular loop of the A2B receptor was crucial for IL-8 stimulation (Ryzhov et al., 2009).
Whereas A2B receptor-mediated stimulation of IL-8 and IL-13 is cAMP-independent, stimulation of adenylate cyclase was required (but not sufficient) for upregulation of VEGF and IL-4 (Ryzhov et al., 2006;Ryzhov et al., 2008d). The dual coupling of A2B receptors to Gs/Gq proteins with concurrent stimulation of diverse intracellular pathways is necessary for adenosine-dependent regulation of IL-4 production in HMC-1. A2B adenosine receptors induce IL-4 generation via Gq-mediated stimulation of PLCβ, inositol trisphosphate-mediated mobilization of intracellular Ca2+ and activation of NFAT by calcineurin. This process is potentiated via Gs-mediated stimulation of adenylate cyclase and activation of protein kinase A (PKA), and may involve the increase in protein levels of NFATc1. Thus, the existence of cross-talk between Gq- PLCβ and Gs-adenylate cyclase signaling pathways in regulation of IL-4 secretion enables A2B receptors, coupled to both Gq and Gs, to effectively stimulate IL-4 production in mast cells (Ryzhov et al., 2006).
C. Dendritic cells
1. Dendritic cell functions
Dendritic cells play an important role in bridging innate and adaptive immunity. It is generally accepted that conventional dendritic cells arise from bone-marrow hematopoietic progenitors or peripheral blood monocytes that migrate into peripheral tissues and differentiate into immature dendritic cells. Immature dendritic cells in tissues are constantly sampling their microenvironment for the presence of antigens. Upon activation by pathogens and other inflammatory stimuli, dendritic cells undergo phenotypical maturation and migrate toward the secondary lymphoid organs. On reaching these organs, dendritic cells develop into mature cells capable to present antigens to naïve T lymphocytes, thus initiating the development of adaptive immune responses (Dominguez and Ardavin, 2010). The presence of A2B receptors on monocytes and dendritic cells (Novitskiy et al., 2008) suggests that their activation may influence both differentiation and maturation of dendritic cells.
2. Role of A2B receptors in dendritic cell differentiation
Using a combination of genetic and pharmacological approaches, we have recently shown that stimulation of A2B receptors in vitro and in vivo induces generation of a phenotypically and functionally distinct subset of dendritic cells. These “adenosine-differentiated” cells are impaired in their ability to induce T-cell proliferation and IFN-γ production. These cells also produce high levels of immunomodulatory cytokines IL-6, IL-10 and TGF-β. It is possible that by up-regulating IL-10 and TGF-β, adenosine-differentiated dentritic cells could affect Th1-mediated immune reactions, induce the generation of regulatory T cells, and polarize the immune response toward a Th2 type. Because these cells also express high levels of the tolerance-inducing enzymes indoleamine 2,3-dioxygenase (IDO) and arginase, they can impair T-cell signal transduction and function. Adenosine-differentiated dendritic cells also secrete high levels of angiogenic factors VEGF and IL-8. Both immunosuppressive and pro-angiogenic properties of these cells could be beneficial for tumor growth. Indeed, our studies in vivo demonstrated that the presence of “adenosine-differentiated” dendritic cells significantly promoted tumor growth in a mouse Lewis lung carcinoma model (Novitskiy et al., 2008).
3. Role of A2B receptors in dendritic cell maturation
In addition to modulation of cell differentiation, A2B receptors were shown also to affect maturation of dendritic cells. A2B receptors inhibited Th1 immune response-promoting cytokines IL-12 p70 and IL-23 but enhanced IL-10 secretion by TLR-activated bone-marrow-derived dendritic cells. Moreover, stimulation of A2B receptors during dendritic cell maturation increased expression and enzymatic activity of IDO and arginase in LPS-activated dendritic cells (Ben et al., 2008). A2B receptors were shown to be responsible for formation of a dendritic cell fraction expressing low levels of MHC-II and co-stimulatory molecule CD86. These cells were characterized by an increased expression of A2B receptor transcripts compared to the rest of dendritic cell population. Only this subset of “adenosine-matured” dendritic cells expressed lower levels of IL-12p40 but higher levels of IL-10 and had poor capacity to stimulate CD4+ T cells, compared to cells matured in the absence of adenosine stimulation (Wilson et al., 2009). Thus, stimulation of A2B receptors, not only during dendritic cell differentiation but also during maturation, may lead to the formation of a cell population capable of impairing Th1 differentiation of CD4+ T cells and promoting immune tolerance. The role of A2B receptors in adenosine actions on both differentiation and maturation of dendritic cells appears to be non-redundant because specific stimulation of other adenosine receptor subtypes did not produce similar effects. Furthermore, these effects were observed only in WT but not A2BKO cells and were inhibited by selective A2B antagonists (Ben et al., 2008;Novitskiy et al., 2008;Wilson et al., 2009).
D. Monocytes/Macropages
Adenosine has been recognized as an important regulator of monocyte/macrophage functions. Some responses to adenosine are mediated by cAMP-dependent mechanisms. These responses include the inhibition of LPS-induced TNF-α production (Kreckler et al., 2009), the potentiation of IL-10 production (Nemeth et al., 2005), and the inhibition of proliferation induced by monocyte colony stimulating factor (M-CSF) (Xaus et al., 1999b). The role of A2B receptors in adenosine-dependent regulation of these monocyte/macrophage functions is often masked by A2A receptors co-expressed in the same cells. A2B receptors can control cAMP-dependent functions only in those cell models where the expression of dominant A2A receptors is negligible or absent. For example, macrophages generated from mouse bone marrow in vitro expressed predominantly A2B adenosine receptors and only negligible levels of A2A receptors. Stimulation of A2B, but not A2A receptors increased cAMP levels in this cell preparation and inhibited M-CSF-induced macrophage proliferation by upregulating the cyclin-dependent kinase inhibitor p27Kip-1 via activation of cAMP-PKA pathway (Xaus et al., 1999b). IFN-γ further upregulated the expression of A2B receptors on bone marrow-derived macrophages resulting in an increased cAMP production in response to stimulation with NECA, which in turn down-regulated both MHC-II and iNOS expression (Xaus et al., 1999a). Due to negligible expression of A2A receptors on RAW264.7 cells, A2B receptors were capable of inhibiting TNF-α production and augmenting IL-10 production in this macrophage-like cell line following activation with LPS (Nemeth et al., 2005).
Similarly, A2B receptors inhibited LPS-induced TNF-α release in peritoneal macrophages isolated from mice lacking A2A receptor. However, selective inhibition of A2B receptors had no effect on the adenosine-dependent inhibition of LPS-induced TNF-α secretion from WT mouse peritoneal macrophages due to the dominant role of A2A receptors in regulation of this cAMP-dependent event (Kreckler et al., 2006). Indeed, NECA-induced cAMP accumulation was similar in peritoneal macrophages obtained from WT and A2BKO animals, indicating the dominant role of A2A receptors in this process. Furthermore, the absence of A2B adenosine receptors did not affect adenosine receptor-dependent suppression of LPS-activated TNF-α release from peritoneal macrophages (Ryzhov et al., 2008c). Comprehensive pharmacological analysis of adenosine-dependent inhibition of LPS-induced TNF-α release from human primary monocytes (Zhang et al., 2005) and alveolar macrophages (Buenestado et al., 2010) revealed that this effect was exclusively mediated by A2A receptors, corroborating the findings in mouse peritoneal macrophages.
However, A2B receptors may have distinctive functions in macrophages even in the presence of otherwise dominant A2A receptors. Pre-treatment of mouse primary alveolar macrophages with the selective A2B antagonist MRS 1706 or genetic ablation of A2B receptors resulted in a loss of NECA-stimulated increases in osteopontin expression (Schneider et al., 2010). Likewise, pharmacological inhibition with selective A2B antagonists or genetic ablation of A2B receptors completely abrogated NECA-induced increase in IL-6 release from peritoneal macrophages (Ryzhov et al., 2008c). Of interest, elevation of cAMP in murine macrophages attenuates LPS-induced TNF-α secretion (Kreckler et al., 2009), but has no effect on basal IL-6 release (Tang et al., 1998). It is possible that the differential regulation of TNF-α and IL-6 secretion by A2A and A2B receptors in mouse peritoneal macrophages can be explained by coupling of these receptors to distinct intracellular pathways. Further studies are needed to delineate the signaling pathways linking activation of A2B receptors to cytokine production in macrophages.
IV. A2B receptors on endothelial cells
A. Endothelial cells in inflammation
Vascular endothelium lines all vessels in the body and serves as a dynamic and selective barrier regulating the flow of nutrients, biologically active molecules and cells across blood vessel walls. Endothelial cells, in close cooperation with other cell types, play an important role in inflammation and subsequent tissue remodeling. Activation of endothelial cells by inflammatory stimuli increases vascular permeability and the expression of adhesion molecules on the endothelial surface, which in turn promote edema, and leukocyte attachment and extravasation leading to initiation of an inflammatory cascade. Later in inflammation, endothelial cells play a central role in the expansion, regression and remodeling of pre-existing blood vessels, a process commonly known as angiogenesis. Adenosine has been implicated in modulation of all these events (Sands and Palmer, 2005).
Differential expression of cell surface adenosine receptors is part of the phenotypic heterogeneity of endothelial cells, and endothelial responses to adenosine can differ depending on the relative expression of adenosine receptor subtypes. For example, HUVECs express predominantly A2A adenosine receptors, whereas the human microvascular endothelial cells HMEC-1 express predominantly A2B receptors (Feoktistov et al., 2002). Predominant expression of A2B receptors has been demonstrated also in cardiac microvascular endothelial cells (Ryzhov et al., 2008b) and endothelial cells of high endothelial venules (Takedachi et al., 2008). Furthermore, there are multiple factors that can modify an endothelial phenotype, including mechanical forces, biologically active compounds, the composition of extracellular matrix, and contact with circulating and tissue-based cells. Conditions present in inflammatory processes provide powerful stimuli for such phenotypic changes. The Th1 cytokines IL-1 and TNF-α increase expression of both A2A and A2B adenosine receptors in human dermal microvascular endothelial cells. IFN-γ treatment increases the expression of A2B receptors, but decreases the expression of A2A receptors (Khoa et al., 2003). Hypoxia, a condition often present in inflamed tissues, also selectively increases A2B expression in endothelial cells (Eltzschig et al., 2003;Feoktistov et al., 2004) by a mechanism that involves the oxygen-sensitive HIF-1α-dependent transactivation of the A2B receptor promoter (Kong et al., 2006). An increase in the A2B receptor expression may not only increase the effects of adenosine but also affect their outcomes. For example, HUVECs express predominantly A2A adenosine receptors, and do not produce VEGF in response to adenosine (Feoktistov et al., 2002). Hypoxia decreased A2A and increased A2B receptor expression in these cells. Consistent with these changes in receptor expression, adenosine stimulated VEGF release under hypoxic but not normoxic conditions, indicating that hypoxia increased the expression of A2B receptors that were functionally coupled to upregulation of VEGF (Feoktistov et al., 2004).
B. Role of A2B receptors in the expression of endothelial adhesion molecules
Adenosine has been shown to inhibit the expression of adhesion molecules and leukocyte recruitment by activated endothelial cells. Whereas A2A receptors have been implicated in these effects both in vitro and in vivo (Palmer and Trevethick, 2008), the role of A2B receptors is less understood.
Genetic ablation of A2B receptor in mice produced a phenotype that was interpreted as supporting an inhibitory role of A2B receptors in the expression of endothelial adhesion molecules and leukocyte recruitment. Intravital examination of mesenteric venules revealed an increased number of leukocytes rolling or adhered to the vascular wall of A2BKO mice as compared to WT mice. Based on these findings and also on the increased levels of the adhesion molecules ICAM-1, P-selectin and E-selectin in protein extracts isolated from mesenteric arteries of A2BKO mice, it has been suggested that A2B receptors tonically downregulate the expression of endothelial adhesion molecules and leukocyte recruitment (Yang et al., 2006). However, these effects of A2B receptor ablation in vivo are likely to be secondary to the increased basal plasma levels of TNF-α, a potent activator of endothelial adhesion molecules (Mackay et al., 1993). In contrast, our studies showed that stimulation of A2B receptors in cardiac microvascular endothelial cells induced rapid cell surface expression of P-selectin by a mechanism likely involving exocytosis of the content of Weibel-Palade bodies (Ryzhov et al., 2008b). Further studies are needed to elucidate the role of endothelial A2B receptor activation on the expression of adhesion molecules involved in the recruitment of inflammatory cells by activated endothelium.
C. Role of A2B receptors in regulation of endothelial barrier function
Numerous studies in vitro have shown that adenosine acting on A2A or/and A2B receptors decreases endothelial permeability (for review see (Biaggioni and Feoktistov, 2005)). In vivo, mice lacking either apyrase (CD39) or ecto-5′-nucleotidase (CD73), enzymes involved in the generation of extracellular adenosine, had a higher leakage of albumin through endothelium in various tissues, as measured by the Evans Blue technique (Eltzschig et al., 2003;Thompson et al., 2004). Remarkably, a similar phenotype was found in mice lacking A2B receptors but not other adenosine receptor subtypes (Eckle et al., 2008a). The increased basal vascular permeability was even further increased in A2BKO mice subjected to ambient hypoxia (Eckle et al., 2008a) supporting the previous evidence obtained in vitro on the role of A2B receptors in regulation of endothelial barrier function. Studies in bone marrow chimeric mice suggested a predominant role of vascular A2B receptors but not those located on bone marrow-derived cells in this response. Surprisingly, A2AKO mice did not demonstrate loss of barrier function in vivo (Eckle et al., 2008a) as it would be expected from in vitro studies that implicated A2A receptors in adenosine-dependent regulation of permeability of several endothelial cell types (Umapathy et al., 2010;Wang and Huxley, 2006). Although some of the differences in vascular permeability between A2BKO and A2AKO mice used in these studies may be attributed to their different genetic background (C57Bl6 and CD1, respectively), these results can be explained by the predominant role of A2B receptors in hypoxic endothelial cells due to hypoxia-inducible factor-1α (HIF-1α)-dependent increase in A2B receptor expression (Kong et al., 2006). HIF-1α was also shown to transactivate the promoter of CD73 (Synnestvedt et al., 2002) and repress the promoter of the equilibrative nucleoside transporter (Eltzschig et al., 2005). Taken together, these effects of hypoxia would upregulate A2B receptors and produce high levels of adenosine at the endothelial surface, thus promoting A2B receptor signaling. However the reason for the augmented vascular permeability in A2BKO mice at rest, when extracellular adenosine levels are low, is less clear. Explanation of this phenomenon will require further investigation.
Reduction of endothelial permeability by adenosine seems to be mediated by the cAMP-PKA-dependent pathway, because this effect was mimicked by reagents elevating cAMP or stimulating PKA. These effects were associated with a rearrangement of the F-actin component of the cytoskeleton, enhanced cell-surface expression of cell–cell junctional protein VE-cadherin and an involvement of myosin-light-chain phosphatase (Umapathy et al., 2010). However, the exact mechanism of adenosine-induced endothelial barrier enhancement remains largely unknown. It has been proposed that it can be explained in part by relaxation of actin cytoskeletal tension, as a result of phosphorylation by PKA of actin-associated, vasodilator-stimulated phosphoprotein (Comerford et al., 2002). Inhibition of RhoA-dependent pathway has been implicated in adenosine- and cAMP-dependent regulation of endothelial barrier function (Harrington et al., 2004;Waschke et al., 2004). In addition, stimulation of ERK via A2B receptors was also proposed to promote barrier function through dephosphorylation of the myosin II regulatory light chains (Srinivas et al., 2004).
D. Role of A2B receptors in regulation of endothelial cell proliferation
Depending on the endothelial cell studied, either A2A or A2B receptors have been implicated in stimulation of endothelial cell proliferation. Adenosine A2B receptors have been shown to mediate the proliferative actions of adenosine in human retinal microvascular endothelial cells (Afzal et al., 2003;Grant et al., 2001;Grant et al., 1999;Mino et al., 2001), in porcine coronary artery and rat aortic endothelial cells (Dubey et al., 2002). The proliferative effects of adenosine on endothelial cells are mediated at least partly by stimulating the production of growth factors that facilitate new blood vessel formation. Adenosine increased VEGF production in pig cerebral microvascular endothelial cells (Fischer et al., 1995), but the adenosine receptor subtype involved in VEGF upregulation in these cells remains uncertain because of non-specific concentrations of antagonists used in that study. Takagi and associates showed that adenosine upregulates VEGF mRNA in bovine retinal microvascular cells via A2A receptors (Takagi et al., 1996).
However, adenosine upregulated VEGF mRNA expression and protein secretion via A2B receptors in human retinal endothelial cells (Grant et al., 1999). VEGF is not the only angiogenic factor modulated by adenosine in endothelial cells. In human retinal microvascular endothelial cells, A2B receptor activation upregulated also basic fibroblast growth factor (bFGF) and insulin-like factor-1 (Grant et al., 1999). In immortalized human dermal microvascular endothelial cells HMEC-1, stimulation of A2B adenosine receptors upregulated bFGF and IL-8 in addition to VEGF (Feoktistov et al., 2002). The intracellular mechanisms mediating these effects are not clear. Although some data suggest that cAMP may play a role in the effects of adenosine on VEGF secretion (Takagi et al., 1996), other studies found that cAMP-PKA-independent stimulation of MAPK pathways was primarily responsible for pro-angiogenic effects of A2B receptors on microvascular endothelial cells (Feoktistov et al., 2002;Grant et al., 2001;Grant et al., 1999).
It is commonly accepted that VEGF production is regulated by HIF-1α. However, A2B receptors can upregulate VEGF production in microvascular endothelial cells by a HIF-1α-independent mechanism (Feoktistov et al., 2004). Because adenosine levels are increased in hypoxia, stimulation of A2B receptors on endothelial cells can complement HIF-1α-dependent actions of hypoxia in the regulation of angiogenesis. Stimulation of A2B receptors results in secretion of additional angiogenic factors (IL-8) not induced by hypoxia per se (when adenosine is scavenged by adenosine deaminase) and in greater VEGF production from endothelial cells (Ryzhov et al., 2007). Thus, A2B receptor-dependent release of angiogenic factors can contribute to the overall effect of hypoxia and provide an autocrine pathway regulating endothelial cell growth during chronic inflammation or in the resolution phase of acute inflammation.
V. A2B receptors on epithelial cells
A. Intestinal epithelial cells
Epithelial cells participate in inflammatory processes by maintaining mucosal integrity, producing biologically active mediators and modulating local immune responses. Functional adenosine A2B receptors are expressed on intestinal epithelial cells. Epithelial A2B receptors were shown to upregulate chloride secretion through the activation of apical cystic fibrosis conductance regulator (CFTR) (Strohmeier et al., 1995). The effect of A2B receptors on epithelial secretion has received particular attention because of its potential relevance to intestinal inflammation. As part of the pathophysiology of these disorders, neutrophils are recruited into intestinal crypts, where they release AMP, which is then converted to adenosine at the epithelial cell surface by CD73. It is adenosine that then acts on epithelial A2B receptors to stimulate chloride secretion (Madara et al., 1993). This pathway normally serves to hydrate the mucosal surface, thereby protecting the intestine by preventing the translocation of bacteria, bacterial products and antigens to lamina propria. When stimulated during inflammation, chloride secretion facilitates fluid movement to lumen in order to flush pathogens from mucosal surface, thus contributing to inflammation-associated diarrhea (Kolachala et al., 2008b).
Epithelial A2B receptors were also shown to modulate the release of proteins involved in intestinal inflammation. Stimulation of A2B receptors on colonic epithelial cells induced an increase in IL-6 secretion into the luminal compartment at levels sufficient to activate neutrophils (Sitaraman et al., 2001). Stimulation of A2B receptors also induced fibronectin synthesis and secretion from the apical surface of intestinal epithelial cells (Walia et al., 2004). Apical fibronectin significantly enhanced the adherence and invasion of Salmonella typhimurium to cultured epithelial cells as well as consequent IL-8 secretion (Walia et al., 2004). Intracellular mechanisms mediating these effects were suggested to involve stimulation of IL-6 and fibronectin transcription via cAMP/PKA-mediated activation of nuclear cAMP-responsive element-binding protein (CREB) (Sitaraman et al., 2001;Walia et al., 2004).
A2B receptor functions on intestinal epithelium can be modulated by INF-γ and TNF-α, factors elevated at various stages of inflammation. TNF-α reportedly increase A2B receptor expression in colonic epithelial cells by a post-transcriptional mechanism possibly involving downregulation of A2B receptor-specific micro RNA miR27b and miR128a (Kolachala et al., 2010), whereas INF-γ affects A2B receptor signaling through the direct inhibition of adenylate cyclase expression (Kolachala et al., 2005).
B. Pulmonary epithelial cells
A2B receptors have been suggested to play an important role in regulation of ion and water transport in airway epithelium. Airborne particles, including pathogens, are absorbed by the mucus layer lining the airways, where they are inactivated by the innate mucosal defense system and removed via mucociliary and cough clearance. This process is largely depends on active transepithelial salt transport involving the CFTR and amiloride-sensitive epithelial Na+ channel (ENaC). The role of A2B receptors in the regulation of CFTR was demonstrated in the human airway epithelial Calu-3 cell line and primary bronchial epithelial cells (Huang et al., 2001;Lazarowski et al., 2004). Like in intestinal epithelium, A2B receptors stimulate CFTR by a mechanism involving cAMP/PKA-dependent pathway (Lazarowski et al., 2004). In bronchial ciliated epithelium, CFTR can inhibit ENaC, and activation of CFTR in the presence of ENaC inhibition generates Cl- secretion and liquid transport to maintain airway surface liquid volume (Donaldson and Boucher, 2007). In distal airspaces of the lung, sodium movement through ENaC and chloride movement through CFTR may be coupled (Reddy et al., 1999), and activation of CFTR-mediated chloride transport can result in fluid absorption (Fang et al., 2006). Therefore, adenosine-dependent, cAMP-mediated pathways could promote alveolar ion absorption and fluid clearance. Indeed, adenosine receptors were shown to regulate alveolar liquid clearance in rat type II pneumocytes (Factor et al., 2007), and high levels of A2B receptor expression were described in murine type II alveolar epithelial cells (Cagnina et al., 2009).
In addition to stimulation of cAMP-dependent pathway, apical adenosine A2B receptors in Calu-3 cells can regulate anion secretion through stimulation of basolateral KCa channels via PLC/Ca2+ signaling. This pathway synergizes with cAMP-dependent modulation of apical CFTR channels for transepithelial anion secretion and a mixed secretion of chloride and bicarbonate (Wang et al., 2008). The mechanism of A2B receptor-mediated tranepithelial liquid transport appears even more complicated in view of the recent report that A2B receptor signaling regulates gap junctional intercellular communication between epithelial cells by the release of PGE2 and subsequent activation of basolateral EP4 receptors. It has been suggested that this mechanism may contribute to the spread of ions, second messenger and/or co-factor exchange between cells to fully activate CFTR and ensure efficient Cl- secretion (Scheckenbach et al., 2010).
Physical stimulation of airway surfaces was suggested to evoke liquid secretion by producing ATP that then locally converted to adenosine, and sensed by A2B adenosine receptors (Huang et al., 2001). In nasal epithelial cells A2B receptor agonists elicited sustained responses in ciliary beat frequences (Morse et al., 2001). Thus, A2B receptors were proposed to stimulate mucociliary clearance in response to injurious stimuli to remove them from airway surfaces. Whether tonic stimulation of A2B receptors is required at rest in order to maintain the mucus clearance is less clear given the low affinity of this receptor subtype. A recent report by Rollins et al appeared to support this hypothesis by demonstrating that the selective A2B antagonist ATL801 inhibited autoregulation of airway surface liquid height in human bronchial epithelial cells (Rollins et al., 2008). However, the lack of effects of micromolar concentrations of DPCPX and ZM241385 (Rollins et al., 2008) known to inhibit human A2B receptors (see Table) adds uncertainty to the authors’ conclusion.
A2B receptors can also regulate cytokine secretion from pulmonary epithelium. Stimulation of A2B receptors on human primary bronchial epithelial cells (HBECs) upregulated the expression of several cytokines including CXCL2, CXCL3 and IL-19 (Zhong et al., 2005). A2B receptor-stimulated IL-19 released from HBECs was able to activate the monocytic cell line THP-1 and induce TNF-α secretion. In turn, TNF-α released from THP-1 upregulated the expression of A2B receptors in HBECs, thus providing a positive feedback loop to facilitate the effects of adenosine (Zhong et al., 2005).
VI. Role of A2B receptors on fibroblasts
The human fibroblasts cell line VA13 was the first cell type where a low affinity adenosine receptor was originally described back in 1980 (Bruns, 1980), and later designated as A2B (Bruns et al., 1986). Fibroblasts represent a heterogeneous population of cells, which may differ in phenotype and function not only between anatomical sites but even within a single tissue where immature cells (often called mesenchymal fibroblasts) exist with fibroblasts of various degree of differentiation. Fibroblasts play an important role in the progression of inflammation by secreting various factors that define the tissue microenvironment and modulate immune cell functions. They also contribute to tissue remodeling by increased proliferation, differentiation and generation of various components of the extracellular matrix (for review see (Flavell et al., 2008)).
Stimulation of A2B receptors on mouse cardiac fibroblasts was shown to promote IL-6 release. This effect was independent of the Gs–cAMP–PKA pathway, but required protein kinase C δ and p38 MAPK activation (Feng et al., 2010). In contrast, A2B receptor-mediated increase of IL-6 release from human gingival fibroblasts was attributed, at least in part, to the activation of Gs–cAMP–PKA pathway (Murakami et al., 2000). Stimulation of these cells with high concentrations (10–50 μM) of adenosine agonists potentiated IL-6 and IL-8 release induced by IL-1β and upregulated the expression of hyaluronate synthase mRNA (Murakami et al., 2001). In contrast, stimulation of A2B receptors on synovial fibroblasts counteracted the effects of IL-1β by decreasing MMP1 mRNA expression (Boyle et al., 1996).
The role of A2B receptors in regulation of IL-6 secretion under normoxic and hypoxic conditions was studied in human pulmonary fibroblasts. Stimulation of A2B receptors under normoxic conditions increased the release of IL-6 and promoted the differentiation of human lung fibroblasts to myofibroblasts. Hypoxia amplified these effects by upregulating the expression of A2B adenosine receptors. The findings that adenosine increases the release of IL-6, and this cytokine in turn induces differentiation of fibroblasts into myofibroblasts suggest a mechanism whereby adenosine could participate in the remodeling process of chronic inflammatory diseases (Zhong et al., 2005).
Studies in corpus cavernosal fibroblast cells isolated from WT and A2BKO mice suggested that A2B receptors upregulate the expression of TGF-β1 and promote fibrosis. Pharmacological analysis in WT cells showed that NECA increased TGF-β1 mRNA expression and procollagen I mRNA levels, which was completely abolished by the A2B receptor specific antagonist, MRS1754. Furthermore, this effect was not seen in the A2BKO cells (Wen et al., 2010).
In contrast, A2B receptors diminished collagen production and proliferation of rat cardiac fibroblasts (Dubey et al., 2001;Epperson et al., 2009). The diverse effects of A2B receptors on fibroblasts of different origins can be attributed to the known phenotypical and functional heterogeneity of these cells.
VII. Role of A2B receptors in animal models of inflammation
The recent generation of A2BKO mice and the development of selective A2B antagonists have made it possible for the testing of an integrated role of A2B receptors in complex animal models of acute and chronic inflammation. Results have been interpreted as evidence for either “pro-inflammatory” or “anti-inflammatory” role of A2B receptors, and results from genetic and pharmacological approach have not always been in agreement.
A2BKO mice appear to have a pro-inflammatory phenotype compared to WT controls, characterized by elevated TNF-α plasma concentrations (Yang et al., 2006) and increased vascular permeability for albumin in the colon, kidney and lung (Eckle et al., 2008a). This is surprising because this is seen even at rest, when the low extracellular adenosine levels are not expected to stimulate A2B receptors. Short-term exposure of mice to ambient hypoxia induced significantly higher vascular leak in all organs of A2BKO mice compared to WT control (Eckle et al., 2008a). A2BKO mice had also increased pulmonary albumin leakage after acute lung injury produced by either mechanical ventilation (Eckle et al., 2008b) or LPS inhalation (Eckle et al., 2008b;Schingnitz et al., 2010), compared to WT controls. In parallel to pulmonary edema, an increase in neutrophil infiltration and tissue levels of IL-1β, IL-6 and TNF-α was also higher in A2BKO mice. Studies using bone marrow chimeric mice showed that A2B receptors located on stromal cells rather than those on bone marrow-derived cells contribute to these differences in lung injury between A2BKO and WT animals (Eckle et al., 2008b;Schingnitz et al., 2010).
Furthermore, it has been reported that genetic deficiency of A2B receptors increased the mortality of mice suffering from cecal ligation and puncture-induced sepsis. The increased mortality of A2B knockout mice was associated with increased inflammatory indices measured in the spleen, heart, and plasma in comparison with WT animals. Again, experiments using bone-marrow chimeras revealed that it is the lack of A2B receptors on nonhematopoietic cells that is primarily responsible for the increased inflammation of septic A2B receptor-deficient mice (Csoka et al., 2010). Another study employing bone-marrow chimeras suggested that vascular A2B receptors also play an important role in protective effects of ischemic preconditioning in a mouse model of acute renal failure from ischemia (Grenz et al., 2008). Thus, it is possible that an increase in vascular permeability in mice lacking A2B receptors may explain, at least in part, the observed exacerbation of acute tissue injury seen in these different disease models.
This evidence obtained in models of acute tissue injury seems to support an “anti-inflammatory” role of A2B receptors. However, even similar models of acute injury may produce opposite results in different tissues. For example, A2B receptors were proposed to play a protective role in gastro-intestinal model of ischemia-reperfusion injury based on enhanced intestinal injury observed in A2BKO mice (Hart et al., 2009). In contrast, the lungs of A2BKO mice were significantly protected in a pulmonary ischemia–reperfusion model, as evidenced by reduced pulmonary artery pressure, increased lung compliance, decreased myeloperoxidase, and reduced levels TNF-α, IL-6, CXCL-1, CCL2 and CCL5 (Anvari et al., 2010). These results would suggest a “pro-inflammatory” role of A2B receptors in the lung, in contrast to their “anti-inflammatory” actions in the intestine. Experiments using bone-marrow chimeras also suggested that these effects were due to A2B receptor activation primarily on resident pulmonary cells and not bone marrow-derived cells (Anvari et al., 2010).
It becomes increasingly clear that the final outcome of genetic ablation of A2B receptors may be dependent not only on the tissue but also on the model of inflammation studied. Although genetic ablation of adenosine A2B receptors in mice has been shown to facilitate acute inflammatory responses to antigen challenges in passively sensitized mice (Hua et al., 2007), this is not the case in chronic inflammation, a process dependent on the complex interplay between multiple cells and inflammatory factors. We studied the effects of A2B receptor gene ablation in the context of ovalbumin-induced chronic pulmonary inflammation. We found that repetitive airway allergen challenge induced a significant increase in adenosine levels in fluid recovered by bronchoalveolar lavage (BAL). Genetic ablation of A2B receptors significantly attenuated allergen-induced chronic pulmonary inflammation as evidenced by a reduction in the number of BAL eosinophils and in peribronchial eosinophilic infiltration. The most striking difference in the pulmonary inflammation induced in A2BKO and WT mice was the lack of allergen-induced IL-4 release in the airways of A2BKO animals, in line with a significant reduction in IL-4 protein and mRNA levels in lung tissue. In addition, attenuation of TGF-β1 release in airways of A2BKO mice correlated with reduced airway smooth muscle and goblet cell hyperplasia/hypertrophy. It was concluded, therefore, that genetic removal of A2B adenosine receptors in mice leads to inhibition of allergen-induced chronic pulmonary inflammation and airway remodeling (Zaynagetdinov et al., 2010). This conclusion was also supported by pharmacological evidence; antagonism of adenosine A2B receptors by the selective A2B antagonist CVT-6883 resulted in inhibition of airway inflammation induced by chronic exposure to allergen (Mustafa et al., 2007).
Inhibition of adenosine A2B receptors by the selective A2B antagonist CVT-6883 also reduced pulmonary inflammation and airway remodeling in adenosine deaminase (ADA)-deficient mice (Sun et al., 2006). These mice are characterized by elevated lung tissue levels of adenosine and exhibit a lung phenotype with features of lung inflammation, bronchial hyperresponsiveness, enhanced mucus secretion, increased IgE synthesis, and elevated levels of pro-inflammatory cytokines and angiogenic factors that could be reversed by lowering adenosine levels with exogenous ADA (Blackburn et al., 2000). Paradoxically, genetic removal of the A2B receptors from ADA-deficient mice led to enhanced pulmonary inflammation and airway destruction. The authors suggested that loss of pulmonary barrier function in A2BKO mice and excessive airway neutrophilia contributed to the enhanced tissue damage observed in this model (Zhou et al., 2009). In addition, TNF-α levels, known to be elevated in A2BKO mice at rest (Ryzhov et al., 2008c;Yang et al., 2006), were also markedly increased in the lungs of ADA/A2B double-knockout mice, which could activate pathways that influence the trafficking of neutrophils in the lung (Zhou et al., 2009). To explain the opposite effects of A2B receptor antagonism and genetic ablation, the authors emphasized that pharmacological inhibition of A2B receptors was introduced later in the disease process, 10 days after triggering pulmonary disease by withdrawing mice from ADA replacement therapy (Zhou et al., 2009). Thus, the differential effects of pharmacological antagonism and genetic deletion of A2B receptors in ADA-deficient mice may indicate the importance of timing when elimination of A2B receptor signaling could either promote or attenuate the development of pulmonary disease in ADA-deficient mouse model. The tonic increase in TNFα in A2BKO but not in WT mice may also contribute to this phenomenon.
The use of A2BKO mice has also resulted in different outcomes of bleomycin-induced pulmonary fibrosis depending on the research model used. Bleomycin instilled directly into the respiratory tract produced the acute damage resulting in extensive apoptosis of airway epithelial cells, followed by infiltration of granulocytes early after the challenge. These mice developed severe fibrosis due to failed wound healing. In contrast, in the intraperitoneal bleomycin model, lung injury and inflammation are not induced acutely, but mice do develop chronic pulmonary fibrosis. The fibrotic process was significantly reduced in A2BKO mice in the intraperitoneal bleomycin model, but not in the intratracheal model, compared to corresponding WT controls (Zhou, 2010). In contrast, pharmacological antagonism of A2B receptors with CVT-6883 attenuated pulmonary inflammation and fibrosis in WT mice subjected to lung injury induced by intratracheal instillation of bleomycin (Sun et al., 2006). These differences in the results obtained from genetic and pharmacological targeting of A2B receptors could be explained by the loss of pulmonary vascular barrier seen in A2BKO mice even at rest (Eckle et al., 2008a). This phenotype would be expected to amplify any model of acute pulmonary injury that involves an acute phase of inflammation with extensive edema and neutrophil infiltration.
The role of A2B receptors in promoting intestinal inflammation was demonstrated in both acute and chronic mouse models of colitis. Genetic removal or pharmacological antagonism of A2B receptors with ATL-801 attenuated clinical and histological features of intestinal inflammation in parallel with reduction of IL-6 and CXCL-1 secretion in several mouse models of experimental colitis (Kolachala et al., 2008a;Kolachala et al., 2008c). In contrast, both genetic ablation and pharmacological antagonism of A2B with PSB1115 promoted intestinal inflammation in a mouse model of acute colitis as reflected by increased weight loss, colonic shortening, and disease activity indices (Frick et al., 2009). The reason for disparate results of these studies is unclear but may reflect the complexity of events orchestrated in these models when small differences in protocols or in composition of intestinal flora present in animals may greatly affect outcomes.
Adding more to controversy on the role of A2B receptors in inflammatory responses, A2B receptors were proposed to play a role in protective effects of ischemic preconditioning in models of myocardial ischemia-reperfusion injury (Eckle et al., 2007;Kuno et al., 2007). Interestingly, in situ ischemic preconditioning conferred cardioprotection in A1KO, A2AKO, or A3KO mice but not in A2BKO mice (Eckle et al., 2007). On the other hand, a recent comprehensive study also employing A2BKO mice, and using the selective A2B antagonist ATL-801 both in mouse and rat models of myocardial ischemia-reperfusion injury argued against contribution of A2B receptors at least in the acute phase of ischemic preconditioning (Maas et al., 2010).
Stimulation of angiogenesis and modulation of immune response by A2B receptors may play an important role in promotion of cancer growth, which can be considered in broad terms to share characteristics with chronic inflammatory processes. Indeed, Lewis lung carcinoma tumors grown in host animals lacking A2B adenosine receptors contained significantly lower levels of VEGF and displayed lower intra-tumor vascular density compared to tumors grown in WT animals. This difference in neovascularization and tumor tissue VEGF levels was due to A2B receptor-dependent VEGF production by host tumor-associated cells. Furthermore, analysis of host immune cells in tumors suggested that A2B receptor signaling may favor the expansion of myeloid-derived suppressor cells. These observations raise the interesting possibility that host A2B receptors on immune cells not only stimulate tumor angiogenesis, but also suppress immune surveillance, thus engaging two distinct mechanisms to promote tumor survival and growth (Ryzhov et al., 2008a). It is also possible that not only tumors, but also infectious agents may exploit host A2B receptors for their advantage. Stimulation of host A2B receptors was implicated in persistence of chlamidal infection (Pettengill et al., 2009) and establishing of leishmania infection (de Almeida Marques-da-Silva et al., 2008). However, the role of other adenosine receptor subtypes in these infections is not known and the mechanisms responsible for these effects need to be defined in detail.
VIII. Conclusion
To be beneficial, the inflammatory reaction must be acute, destroying an injurious agent within a short period of time and in a localized area, while inducing an immune response. This is achieved through a complex series of events involving multiple cell types and secreted factors. Given the actions of adenosine to return the host to a homeostatic state, it is not surprising that A2B receptors may operate to control the extent of acute inflammatory responses. On the other hand, premature suppression of acute inflammation by adenosine may promote its progression to a chronic status to the detriment of the host.
We believe that labeling A2B receptors as “anti-inflammatory” or “pro-inflammatory” may be overly simplistic and misleading because it fails to appreciate just how complex each of the different types of inflammation is and conveys little about precise mechanisms. For example, the inflammation in a model of ventilation-induced lung injury is clearly different from the inflammation in a model of allergen-induced chronic lung disease. Therefore, it is not surprising that inhibition of A2B receptors produced opposite effects in these models. As we illustrated in this chapter, A2B receptors may play different roles even in similar types of inflammation but occurring in different tissues. Furthermore, A2B receptors can play different roles at different points in the progression of inflammation. In this respect, A2B receptor functions may be reflective of pleiotropic effects of the secreted factors. As an example, IL-6 was shown to promote inflammation in models of chronic inflammatory diseases, whereas in models of acute inflammation it can suppress local and systemic acute inflammatory responses (Gabay, 2006). Moreover, various combinations of cytokines released from A2B receptor-activated cells can produce different outcomes. TGF-β1 produced by “adenosine-differentiated” dendritic cells (Novitskiy et al., 2008) favors the emergence of adaptive Tregs, but in combination with IL-4 or IL-6, it may stimulate Th9 and Th17 cells, respectively (Ernst et al., 2010).
Targeting the low affinity A2B receptor, as opposed to other adenosine receptor subtypes, in the development of novel therapeutic approaches for treatment of inflammatory diseases is especially appealing because these receptors are likely activated only in the pathophysiological environment, while remaining silent in normal tissues. This characteristic could provide specificity in therapy of certain immune-related disorders, while decreasing likelihood of side effects. However, translation of current findings into novel therapies would require a better understanding of A2B receptors functions in diverse types of inflammatory responses in various tissues and at different points of their progression.
Acknowledgments
This work was supported in part by National Institutes of Health grants R01HL095787 and R01CA138923
Abbreviations
- ADA
adenosine deaminase
- BAL
bronchoalveolar lavage
- bFGF
basic fibroblast growth factor
- BMMC
bone marrow-derived mast cell
- CFTR
cystic fibrosis transmembrane conductance regulator
- ENaC
epithelial sodium channel
- ERK
extracellular signal-regulated kinase
- HIF-1α
hypoxia-inducible factor 1-alpha
- HBEC
human bronchial epithelial cell
- HMC-1
human mast cell-1
- HMEC-1
human microvascular endothelial cell-1
- HUVEC
human umbilical vein endothelial cell
- ICAM
inter-cellular adhesion molecule
- IDO
indoleamine 2,3-dioxygenase
- IL1RAP
interleukin-1 receptor accessory protein
- INF-γ
interferon-gamma
- JNK
c-Jun N-terminal kinase
- KO
knockout
- LPS
lipopolysaccharide
- MAPK
mitogen-activated protein kinase
- M-CSF
macrophage colony-stimulating factor
- MHC-II
major histocompatibility complex class II
- MMP-1
matrix metalloproteinase-1
- NFAT
nuclear factor of activated T-cells, PKA, protein kinase A
- PLC
phospholipase C
- TGF-β1
transforming growth factor beta one
- TLR
toll-like receptors
- TNF-α
tumor necrosis factor alpha
- VEGF
vascular endothelial growth factor
- WT
wild type
Footnotes
Conflict of interest: The authors have been recipients of research grants from Gilead Palo Alto, Inc.
References
- Afzal A, Shaw LC, Caballero S, Spoerri PE, Lewin AS, Zeng D, et al. Reduction in preretinal neovascularization by ribozymes that cleave the A2B adenosine receptor mRNA. Circ Res. 2003;93:500–506. doi: 10.1161/01.RES.0000091260.78959.BC. [DOI] [PubMed] [Google Scholar]
- Amisten S, Braun OO, Bengtsson A, Erlinge D. Gene expression profiling for the identification of G-protein coupled receptors in human platelets. Thromb Res. 2008;122:47–57. doi: 10.1016/j.thromres.2007.08.014. [DOI] [PubMed] [Google Scholar]
- Anvari F, Sharma AK, Fernandez LG, Hranjec T, Ravid K, Kron IL, et al. Tissue-derived proinflammatory effect of adenosine A2B receptor in lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg. 2010;140:871–877. doi: 10.1016/j.jtcvs.2010.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Auchampach JA, Jin X, Wan TC, Caughey GH, Linden J. Canine mast cell adenosine receptors: Cloning and expression of the A3 receptors and evidence that degranulation is mediated by the A2B receptor. Mol Pharmacol. 1997;52:846–860. doi: 10.1124/mol.52.5.846. [DOI] [PubMed] [Google Scholar]
- Auchampach JA, Kreckler LM, Wan TC, Maas JE, van der Hoeven D, Gizewski E, et al. Characterization of the A2B adenosine receptor from mouse, rabbit, and dog. J Pharmacol Exp Ther. 2009;329:2–13. doi: 10.1124/jpet.108.148270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Baraldi PG, Tabrizi MA, Fruttarolo F, Romagnoli R, Preti D. Recent improvements in the development of A2B adenosine receptor agonists. Purinergic Signal. 2009;5:3–19. doi: 10.1007/s11302-009-9140-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ben AA, Lefort A, Hua X, Libert F, Communi D, Ledent C, et al. Modulation of murine dendritic cell function by adenine nucleotides and adenosine: involvement of the A2B receptor. Eur J Immunol. 2008;38:1610–1620. doi: 10.1002/eji.200737781. [DOI] [PubMed] [Google Scholar]
- Biaggioni I, Feoktistov I. Adenosine and endothelial cell function. In: Hasko G, Cronstein BN, Szabo C, editors. Adenosine Receptors: Therapeutic Aspects for Inflammatory and Immune Deaseases. CRC Press; UK, USA: 2005. pp. 109–131. [Google Scholar]
- Blackburn MR, Volmer JB, Thrasher JL, Zhong H, Crosby JR, Lee JJ, et al. Metabolic consequences of adenosine deaminase deficiency in mice are associated with defects in alveogenesis, pulmonary inflammation, and airway obstruction. J Exp Med. 2000;192:159–170. doi: 10.1084/jem.192.2.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyle DL, Sajjadi FG, Firestein GS. Inhibition of synoviocyte collagenase gene expression by adenosine receptor stimulation. Arthr Rheum. 1996;39:923–930. doi: 10.1002/art.1780390608. [DOI] [PubMed] [Google Scholar]
- Bruns RF. Adenosine receptor activation in human fibroblasts: nucleoside agonists and antagonists. Can J Physiol Pharmacol. 1980;58:673–691. doi: 10.1139/y80-110. [DOI] [PubMed] [Google Scholar]
- Bruns RF, Lu GH, Pugsley TA. Characterization of the A2 adenosine receptor labeled by [3H]NECA in rat striatal membranes. Mol Pharmacol. 1986;29:331–346. [PubMed] [Google Scholar]
- Buenestado A, Grassin DS, Arnould I, Besnard F, Naline E, Blouquit-Laye S, et al. The role of adenosine receptors in regulating production of tumour necrosis factor-alpha and chemokines by human lung macrophages. Br J Pharmacol. 2010;159:1304–1311. doi: 10.1111/j.1476-5381.2009.00614.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cagnina RE, Ramos SI, Marshall MA, Wang G, Frazier CR, Linden J. Adenosine A2B receptors are highly expressed on murine type II alveolar epithelial cells. Am J Physiol. 2009;297:L467–L474. doi: 10.1152/ajplung.90553.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Comerford KM, Lawrence DW, Synnestvedt K, Levi BP, Colgan SP. Role of vasodilator-stimulated phosphoprotein in PKA-induced changes in endothelial junctional permeability. FASEB J. 2002;16:583–585. doi: 10.1096/fj.01-0739fje. [DOI] [PubMed] [Google Scholar]
- Corset V, Nguyen-Ba-Charvet KT, Forcet C, Moyse E, dotal A, Mehlen P. Netrin-1-mediated axon outgrowth and cAMP production requires interaction with adenosine A2b receptor. Nature. 2000;407:747–750. doi: 10.1038/35037600. [DOI] [PubMed] [Google Scholar]
- Csoka B, Himer L, Selmeczy Z, Vizi ES, Pacher P, Ledent C, et al. Adenosine A2A receptor activation inhibits T helper 1 and T helper 2 cell development and effector function. FASEB J. 2008;22:3491–3499. doi: 10.1096/fj.08-107458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Csoka B, Nemeth ZH, Rosenberger P, Eltzschig HK, Spolarics Z, Pacher P, et al. A2B adenosine receptors protect against sepsis-induced mortality by dampening excessive inflammation. J Immunol. 2010;185:542–550. doi: 10.4049/jimmunol.0901295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Almeida Marques-da-Silva, de Oliveira JC, Figueiredo AB, de Souza Lima JD, Carneiro CM, Rangel Fietto JL, et al. Extracellular nucleotide metabolism in Leishmania: influence of adenosine in the establishment of infection. Microbes Infect. 2008;10:850–857. doi: 10.1016/j.micinf.2008.04.016. [DOI] [PubMed] [Google Scholar]
- Dominguez PM, Ardavin C. Differentiation and function of mouse monocyte-derived dendritic cells in steady state and inflammation. Immunol Rev. 2010;234:90–104. doi: 10.1111/j.0105-2896.2009.00876.x. [DOI] [PubMed] [Google Scholar]
- Donaldson SH, Boucher RC. Sodium channels and cystic fibrosis. Chest. 2007;132:1631–1636. doi: 10.1378/chest.07-0288. [DOI] [PubMed] [Google Scholar]
- Dubey RK, Gillespie DG, Jackson EK. A2B adenosine receptors stimulate growth of porcine and rat arterial endothelial cells. Hypertension. 2002;39:530–535. doi: 10.1161/hy0202.103075. [DOI] [PubMed] [Google Scholar]
- Dubey RK, Gillespie DG, Zacharia LC, Mi Z, Jackson EK. A2b receptors mediate the antimitogenic effects of adenosine in cardiac fibroblasts. Hypertension. 2001;37:716–721. doi: 10.1161/01.hyp.37.2.716. [DOI] [PubMed] [Google Scholar]
- Duffy SM, Cruse G, Brightling CE, Bradding P. Adenosine closes the K+ channel KCa3.1 in human lung mast cells and inhibits their migration via the adenosine A2A receptor. Eur J Immunol. 2007;37:1653–1662. doi: 10.1002/eji.200637024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckle T, Faigle M, Grenz A, Laucher S, Thompson LF, Eltzschig HK. A2B adenosine receptor dampens hypoxia-induced vascular leak. Blood. 2008a;111:2024–2035. doi: 10.1182/blood-2007-10-117044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckle T, Grenz A, Laucher S, Eltzschig HK. A2B adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice. J Clin Invest. 2008b;118:3301–3315. doi: 10.1172/JCI34203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckle T, Krahn T, Grenz A, Kohler D, Mittelbronn M, Ledent C, et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation. 2007;115:1581–1590. doi: 10.1161/CIRCULATIONAHA.106.669697. [DOI] [PubMed] [Google Scholar]
- Eltzschig HK, Abdulla P, Hoffman E, Hamilton KE, Daniels D, Schonfeld C, et al. HIF-1-dependent repression of equilibrative nucleoside transporter (ENT) in hypoxia. J Exp Med. 2005;202:1493–1505. doi: 10.1084/jem.20050177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eltzschig HK, Ibla JC, Furuta GT, Leonard MO, Jacobson KA, Enjyoji K, et al. Coordinated adenine nucleotide phosphohydrolysis and nucleoside signaling in posthypoxic endothelium: role of ectonucleotidases and adenosine A2B receptors. J Exp Med. 2003;198:783–796. doi: 10.1084/jem.20030891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Epperson SA, Brunton LL, Ramirez-Sanchez I, Villarreal F. Adenosine receptors and second messenger signaling pathways in rat cardiac fibroblasts. Am J Physiol. 2009;296:C1171–C1177. doi: 10.1152/ajpcell.00290.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ernst PB, Garrison JC, Thompson LF. Much ado about adenosine: adenosine synthesis and function in regulatory T cell biology. J Immunol. 2010;185:1993–1998. doi: 10.4049/jimmunol.1000108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Factor P, Mutlu GM, Chen L, Mohameed J, Akhmedov AT, Meng FJ, et al. Adenosine regulation of alveolar fluid clearance. Proc Natl Acad Sci U S A. 2007;104:4083–4088. doi: 10.1073/pnas.0601117104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fang X, Song Y, Hirsch J, Galietta LJ, Pedemonte N, Zemans RL, et al. Contribution of CFTR to apical-basolateral fluid transport in cultured human alveolar epithelial type II cells. Am J Physiol. 2006;290:L242–L249. doi: 10.1152/ajplung.00178.2005. [DOI] [PubMed] [Google Scholar]
- Feng W, Song Y, Chen C, Lu ZZ, Zhang Y. Stimulation of adenosine A2B receptors induces interleukin-6 secretion in cardiac fibroblasts via the PKC-delta-P38 signalling pathway. Br J Pharmacol. 2010;159:1598–1607. doi: 10.1111/j.1476-5381.2009.00558.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feoktistov I, Biaggioni I. Adenosine A2b receptors evoke interleukin-8 secretion in human mast cells. An enprofylline-sensitive mechanism with implications for asthma. J Clin Invest. 1995;96:1979–1986. doi: 10.1172/JCI118245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feoktistov I, Goldstein AE, Biaggioni I. Role of p38 mitogen-activated protein kinase and extracellular signal-regulated protein kinase kinase in adenosine A2B receptor- mediated interleukin-8 production in human mast cells. Mol Pharmacol. 1999;55:726–735. [PubMed] [Google Scholar]
- Feoktistov I, Goldstein AE, Ryzhov S, Zeng D, Belardinelli L, Voyno-Yasenetskaya T, et al. Differential expression of adenosine receptors in human endothelial cells: role of A2B receptors in angiogenic factor regulation. Circ Res. 2002;90:531–538. doi: 10.1161/01.res.0000012203.21416.14. [DOI] [PubMed] [Google Scholar]
- Feoktistov I, Ryzhov S, Goldstein AE, Biaggioni I. Mast cell-mediated stimulation of angiogenesis: cooperative interaction between A2B and A3 adenosine receptors. Circ Res. 2003;92:485–492. doi: 10.1161/01.RES.0000061572.10929.2D. [DOI] [PubMed] [Google Scholar]
- Feoktistov I, Ryzhov S, Zhong H, Goldstein AE, Matafonov A, Zeng D, et al. Hypoxia modulates adenosine receptors in human endothelial and smooth muscle cells toward an A2B angiogenic phenotype. Hypertension. 2004;44:649–654. doi: 10.1161/01.HYP.0000144800.21037.a5. [DOI] [PubMed] [Google Scholar]
- Fischer S, Sharma HS, Karliczek GF, Schaper W. Expression of vascular permeability factor/vascular endothelial growth factor in pig cerebral microvascular endothelial cells and its upregulation by adenosine. Brain Res. 1995;28:141–148. doi: 10.1016/0169-328x(94)00193-i. [DOI] [PubMed] [Google Scholar]
- Flavell SJ, Hou TZ, Lax S, Filer AD, Salmon M, Buckley CD. Fibroblasts as novel therapeutic targets in chronic inflammation. Br J Pharmacol. 2008;153:S241–S246. doi: 10.1038/sj.bjp.0707487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 2007;14:1315–1323. doi: 10.1038/sj.cdd.4402132. [DOI] [PubMed] [Google Scholar]
- Fredholm BB, Ijzerman AP, Jacobson KA, Klotz KN, Linden J. International Union of Pharmacology. XXV Nomenclature and classification of adenosine receptors. Pharmacol Rev. 2001a;53:527–552. [PMC free article] [PubMed] [Google Scholar]
- Fredholm BB, Irenius E, Kull B, Schulte G. Comparison of the potency of adenosine as an agonist at human adenosine receptors expressed in Chinese hamster ovary cells. Biochem Pharmacol. 2001b;61:443–448. doi: 10.1016/s0006-2952(00)00570-0. [DOI] [PubMed] [Google Scholar]
- Fredholm BB, Zhang Y, van der Ploeg I. Adenosine A2a receptors mediate the inhibitory effect of adenosine on formyl-met-leu-phe-stimulated respiratory burst in neutrophil leucocytes. Naunyn-Schmiedeberg’s Arch Pharmacol. 1996;354:262–267. doi: 10.1007/BF00171056. [DOI] [PubMed] [Google Scholar]
- Frick JS, Macmanus CF, Scully M, Glover LE, Eltzschig HK, Colgan SP. Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. J Immunol. 2009;182:4957–4964. doi: 10.4049/jimmunol.0801324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gabay C. Interleukin-6 and chronic inflammation. Arthritis Res Ther. 2006;8:S3. doi: 10.1186/ar1917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gessi S, Varani K, Merighi S, Cattabriga E, Pancaldi C, Szabadkai Y, et al. Expression, pharmacological profile, and functional coupling of A2B receptors in a recombinant system and in peripheral blood cells using a novel selective antagonist radioligand, [3H]MRE 2029-F20. Mol Pharmacol. 2005;67:2137–2147. doi: 10.1124/mol.104.009225. [DOI] [PubMed] [Google Scholar]
- Grant MB, Davis MI, Caballero S, Feoktistov I, Biaggioni I, Belardinelli L. Proliferation, migration, and ERK activation in human retinal endothelial cells through A2B adenosine receptor stimulation. Invest Ophthalmol Visual Sci. 2001;42:2068–2073. [PubMed] [Google Scholar]
- Grant MB, Tarnuzzer RW, Caballero S, Ozeck MJ, Davis MI, Spoerri PE, et al. Adenosine receptor activation induces vascular endothelial growth factor in human retinal endothelial cells. Circ Res. 1999;85:699–706. doi: 10.1161/01.res.85.8.699. [DOI] [PubMed] [Google Scholar]
- Grenz A, Osswald H, Eckle T, Yang D, Zhang H, Tran ZV, et al. The reno-vascular A2B adenosine receptor protects the kidney from ischemia. PLoS Med. 2008;5:e137. doi: 10.1371/journal.pmed.0050137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harrington EO, Newton J, Morin N, Rounds S. Barrier dysfunction and RhoA activation are blunted by homocysteine and adenosine in pulmonary endothelium. Am J Physiol. 2004;287:L1091–L1097. doi: 10.1152/ajplung.00421.2003. [DOI] [PubMed] [Google Scholar]
- Hart ML, Jacobi B, Schittenhelm J, Henn M, Eltzschig HK. Cutting Edge: A2B Adenosine receptor signaling provides potent protection during intestinal ischemia/reperfusion injury. J Immunol. 2009;182:3965–3968. doi: 10.4049/jimmunol.0802193. [DOI] [PubMed] [Google Scholar]
- Hua X, Kovarova M, Chason KD, Nguyen M, Koller BH, Tilley SL. Enhanced mast cell activation in mice deficient in the A2b adenosine receptor. J Exp Med. 2007;204:117–128. doi: 10.1084/jem.20061372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang P, Lazarowski ER, Tarran R, Milgram SL, Boucher RC, Stutts MJ. Compartmentalized autocrine signaling to cystic fibrosis transmembrane conductance regulator at the apical membrane of airway epithelial cells. Proc Natl Acad Sci U S A. 2001;98:14120–14125. doi: 10.1073/pnas.241318498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalla RV, Zablocki J. Progress in the discovery of selective, high affinity A2B adenosine receptor antagonists as clinical candidates. Purinergic Signal. 2009;5:21–29. doi: 10.1007/s11302-008-9119-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Khoa ND, Montesinos MC, Williams AJ, Kelly M, Cronstein BN. Th1 cytokines regulate adenosine receptors and their downstream signaling elements in human microvascular endothelial cells. J Immunol. 2003;171:3991–3998. doi: 10.4049/jimmunol.171.8.3991. [DOI] [PubMed] [Google Scholar]
- Kim YC, Ji X, Melman N, Linden J, Jacobson KA. Anilide derivatives of an 8-phenylxanthine carboxylic congener are highly potent and selective antagonists at human A2B adenosine receptors. J Med Chem. 2000;43:1165–1172. doi: 10.1021/jm990421v. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klotz KN, Hessling J, Hegler J, Owman C, Kull B, Fredholm BB, et al. Comparative pharmacology of human adenosine receptor subtypes - characterization of stably transfected receptors in CHO cells. Naunyn-Schmiedeberg’s Arch Pharmacol. 1998;357:1–9. doi: 10.1007/pl00005131. [DOI] [PubMed] [Google Scholar]
- Kolachala V, Asamoah V, Wang L, Srinivasan S, Merlin D, Sitaraman SV. Interferon-gamma down-regulates adenosine 2b receptor-mediated signaling and short circuit current in the intestinal epithelia by inhibiting the expression of adenylate cyclase. J Biol Chem. 2005;280:4048–4057. doi: 10.1074/jbc.M409577200. [DOI] [PubMed] [Google Scholar]
- Kolachala V, Ruble B, Vijay-Kumar M, Wang L, Mwangi S, Figler H, et al. Blockade of adenosine A2B receptors ameliorates murine colitis. Br J Pharmacol. 2008a;155:127–137. doi: 10.1038/bjp.2008.227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolachala VL, Bajaj R, Chalasani M, Sitaraman SV. Purinergic receptors in gastrointestinal inflammation. Am J Physiol. 2008b;294:G401–G410. doi: 10.1152/ajpgi.00454.2007. [DOI] [PubMed] [Google Scholar]
- Kolachala VL, Vijay-Kumar M, Dalmasso G, Yang D, Linden J, Wang L, et al. A2B adenosine receptor gene deletion attenuates murine colitis. Gastroenterology. 2008c;135:861–870. doi: 10.1053/j.gastro.2008.05.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kolachala VL, Wang L, Obertone TS, Prasad M, Yan Y, Dalmasso G, et al. Adenosine 2B receptor expression is post-transcriptionally regulated by microRNA. J Biol Chem. 2010;285:18184–18190. doi: 10.1074/jbc.M109.066555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kong T, Westerman KA, Faigle M, Eltzschig HK, Colgan SP. HIF-dependent induction of adenosine A2B receptor in hypoxia. FASEB J. 2006;20:2242–2250. doi: 10.1096/fj.06-6419com. [DOI] [PubMed] [Google Scholar]
- Kreckler LM, Gizewski E, Wan TC, Auchampach JA. Adenosine suppresses lipopolysaccharide-induced tumor necrosis factor-alpha production by murine macrophages through a protein kinase A- and exchange protein activated by cAMP-independent signaling pathway. J Pharmacol Exp Ther. 2009;331:1051–1061. doi: 10.1124/jpet.109.157651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kreckler LM, Wan TC, Ge ZD, Auchampach JA. Adenosine inhibits tumor necrosis factor-alpha release from mouse peritoneal macrophages via A2A and A2B but not the A3 adenosine receptor. J Pharmacol Exp Ther. 2006;317:172–180. doi: 10.1124/jpet.105.096016. [DOI] [PubMed] [Google Scholar]
- Kuno A, Critz SD, Cui L, Solodushko V, Yang XM, Krahn T, et al. Protein kinase C protects preconditioned rabbit hearts by increasing sensitivity of adenosine A2b-dependent signaling during early reperfusion. J Mol Cell Cardiol. 2007;43:262–271. doi: 10.1016/j.yjmcc.2007.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lazarowski ER, Tarran R, Grubb BR, van Heusden CA, Okada S, Boucher RC. Nucleotide release provides a mechanism for airway surface liquid homeostasis. J Biol Chem. 2004;279:36855–36864. doi: 10.1074/jbc.M405367200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linden J, Thai T, Figler H, Jin X, Robeva AS. Characterization of human A2B adenosine receptors: radioligand binding, western blotting, and coupling to Gq in human embryonic kidney 293 cells and HMC-1 mast cells. Mol Pharmacol. 1999;56:705–713. [PubMed] [Google Scholar]
- Maas JE, Wan TC, Figler RA, Gross GJ, Auchampach JA. Evidence that the acute phase of ischemic preconditioning does not require signaling by the A2B adenosine receptor. J Mol Cell Cardiol. 2010;49:886–893. doi: 10.1016/j.yjmcc.2010.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mackay F, Loetscher H, Stueber D, Gehr G, Lesslauer W. Tumor necrosis factor alpha (TNF-α)-induced cell adhesion to human endothelial cells is under dominant control of one TNF receptor type, TNF-R55. J Exp Med. 1993;177:1277–1286. doi: 10.1084/jem.177.5.1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Madara JL, Patapoff TW, Gillece-Castro B, Colgan SP, Parkos CA, Delp C, et al. 5′-adenosine monophosphate is the neutrophil-derived paracrine factor that elicits chloride secretion from T84 intestinal epithelial cell monolayers. J Clin Invest. 1993;91:2320–2325. doi: 10.1172/JCI116462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKenna WL, Wong-Staal C, Kim GC, Macias H, Hinck L, Bartoe JL. Netrin-1-independent adenosine A2b receptor activation regulates the response of axons to netrin-1 by controlling cell surface levels of UNC5A receptors. J Neurochem. 2008;104:1081–1090. doi: 10.1111/j.1471-4159.2007.05040.x. [DOI] [PubMed] [Google Scholar]
- Meade CJ, Worrall L, Hayes D, Protin U. Induction of interleukin 8 release from the HMC-1 mast cell line: synergy between stem cell factor and activators of the adenosine A2b receptor. Biochem Pharmacol. 2002;64:317–325. doi: 10.1016/s0006-2952(02)01058-4. [DOI] [PubMed] [Google Scholar]
- Mino RP, Spoerri PE, Caballero S, Player D, Belardinelli L, Biaggioni I, et al. Adenosine receptor antagonists and retinal neovascularization in vivo. Invest Ophthalmol Visual Sci. 2001;42:3320–3324. [PubMed] [Google Scholar]
- Morse DM, Smullen JL, Davis CW. Differential effects of UTP, ATP, and adenosine on ciliary activity of human nasal epithelial cells. Am J Physiol. 2001;280:C1485–C1497. doi: 10.1152/ajpcell.2001.280.6.C1485. [DOI] [PubMed] [Google Scholar]
- Murakami S, Hashikawa T, Saho T, Takedachi M, Nozaki T, Shimabukuro Y, et al. Adenosine regulates the IL-1 beta-induced cellular functions of human gingival fibroblasts. Int Immunol. 2001;13:1533–1540. doi: 10.1093/intimm/13.12.1533. [DOI] [PubMed] [Google Scholar]
- Murakami S, Terakura M, Kamatani T, Hashikawa T, Saho T, Shimabukuro Y, et al. Adenosine regulates the production of interleukin-6 by human gingival fibroblasts via cyclic AMP/protein kinase A pathway. J Periodontal Res. 2000;35:93–101. doi: 10.1034/j.1600-0765.2000.035002093.x. [DOI] [PubMed] [Google Scholar]
- Mustafa SJ, Nadeem A, Fan M, Zhong H, Belardinelli L, Zeng D. Effect of a specific and selective A2B adenosine receptor antagonist on adenosine agonist AMP and allergen-induced airway responsiveness and cellular influx in a mouse model of asthma. J Pharmacol Exp Ther. 2007;320:1246–1251. doi: 10.1124/jpet.106.112250. [DOI] [PubMed] [Google Scholar]
- Nemeth ZH, Lutz CS, Csoka B, Deitch EA, Leibovich SJ, Gause WC, et al. Adenosine augments IL-10 production by macrophages through an A2B receptor-mediated posttranscriptional mechanism. J Immunol. 2005;175:8260–8270. doi: 10.4049/jimmunol.175.12.8260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY, et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood. 2008;112:1822–1831. doi: 10.1182/blood-2008-02-136325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ongini E, Dionisotti S, Gessi S, Irenius E, Fredholm BB. Comparison of CGS 15943, ZM 241385 and SCH 58261 as antagonists at human adenosine receptors. Naunyn-Schmiedebergs Arch Pharmacol. 1999;359:7–10. doi: 10.1007/pl00005326. [DOI] [PubMed] [Google Scholar]
- Palmer TM, Trevethick MA. Suppression of inflammatory and immune responses by the A2A adenosine receptor: an introduction. Br J Pharmacol. 2008;153:S27–S34. doi: 10.1038/sj.bjp.0707524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pettengill MA, Lam VW, Ojcius DM. The danger signal adenosine induces persistence of chlamydial infection through stimulation of A2b receptors. PLoS One. 2009;4:e8299. doi: 10.1371/journal.pone.0008299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramkumar V, Stiles GL, Beaven MA, Ali H. The A3 adenosine receptor is the unique adenosine receptor which facilitates release of allergic mediators in mast cells. J Biol Chem. 1993;268:16887–16890. [PubMed] [Google Scholar]
- Reddy MM, Light MJ, Quinton PM. Activation of the epithelial Na+ channel (ENaC) requires CFTR Cl- channel function. Nature. 1999;402:301–304. doi: 10.1038/46297. [DOI] [PubMed] [Google Scholar]
- Rollins BM, Burn M, Coakley RD, Chambers LA, Hirsh AJ, Clunes MT, et al. A2B adenosine receptors regulate the mucus clearance component of the lung’s innate defense system. Am J Respir Cell Mol Biol. 2008;39:190–197. doi: 10.1165/rcmb.2007-0450OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenberger P, Schwab JM, Mirakaj V, Masekowsky E, Mager A, Morote-Garcia JC, et al. Hypoxia-inducible factor-dependent induction of netrin-1 dampens inflammation caused by hypoxia. Nat Immunol. 2009;10:195–202. doi: 10.1038/ni.1683. [DOI] [PubMed] [Google Scholar]
- Ryzhov S, Goldstein AE, Biaggioni I, Feoktistov I. Cross-talk between Gs- and Gq-coupled pathways in regulation of interleukin-4 by A2B adenosine receptors in human mast cells. Mol Pharmacol. 2006;70:727–735. doi: 10.1124/mol.106.022780. [DOI] [PubMed] [Google Scholar]
- Ryzhov S, Goldstein AE, Matafonov A, Zeng D, Biaggioni I, Feoktistov I. Adenosine-activated mast cells induce IgE synthesis by B lymphocytes: an A2B-mediated process involving Th2 cytokines IL-4 and IL-13 with implications for asthma. J Immunol. 2004;172:7726–7733. doi: 10.4049/jimmunol.172.12.7726. [DOI] [PubMed] [Google Scholar]
- Ryzhov S, McCaleb JL, Goldstein AE, Biaggioni I, Feoktistov I. Role of adenosine receptors in the regulation of angiogenic factors and neovascularization in hypoxia. J Pharmacol Exp Ther. 2007;382:565–572. doi: 10.1124/jpet.106.114850. [DOI] [PubMed] [Google Scholar]
- Ryzhov S, Novitskiy SV, Zaynagetdinov R, Goldstein AE, Carbone DP, Biaggioni I, et al. Host A2B adenosine receptors promote carcinoma growth. Neoplasia (New York) 2008a;10:987–995. doi: 10.1593/neo.08478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryzhov S, Solenkova NV, Goldstein AE, Lamparter M, Fleenor T, Young PP, et al. Adenosine receptor-mediated adhesion of endothelial progenitors to cardiac microvascular endothelial cells. Circ Res. 2008b;102:356–363. doi: 10.1161/CIRCRESAHA.107.158147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryzhov S, Zaynagetdinov R, Goldstein AE, Matafonov A, Biaggioni I, Feoktistov I. Differential role of the carboxy-terminus of the A2B adenosine receptor in stimulation of adenylate cyclase, phospholipase Cβ, and interleukin-8. Purinergic Signal. 2009;5:289–298. doi: 10.1007/s11302-008-9129-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV, Blackburn MR, Biaggioni I, et al. Effect of A2B adenosine receptor gene ablation on adenosine-dependent regulation of proinflammatory cytokines. J Pharmacol Exp Ther. 2008c;324:694–700. doi: 10.1124/jpet.107.131540. [DOI] [PubMed] [Google Scholar]
- Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV, Dikov MM, Blackburn MR, et al. Effect of A2B adenosine receptor gene ablation on proinflammatory adenosine signaling in mast cells. J Immunol. 2008d;180:7212–7220. doi: 10.4049/jimmunol.180.11.7212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salvatore CA, Tilley SL, Latour AM, Fletcher DS, Koller BH, Jacobson MA. Disruption of the A3 adenosine receptor gene in mice and its effect on stimulated inflammatory cells. J Biol Chem. 2000;275:4429–4434. doi: 10.1074/jbc.275.6.4429. [DOI] [PubMed] [Google Scholar]
- Sands WA, Palmer TM. Adenosine receptors and the control of endothelial cell function in inflammatory disease. Immunol Lett. 2005;101:1–11. doi: 10.1016/j.imlet.2005.04.005. [DOI] [PubMed] [Google Scholar]
- Scheckenbach KL, Losa D, Dudez T, Bacchetta M, O’Grady S, Crespin S, et al. PGE2 regulation of CFTR activity and air surface liquid volume requires gap junctional communication. Am J Respir Cell Mol Biol. 2010 doi: 10.1165/rcmb.2009-0361OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schingnitz U, Hartmann K, Macmanus CF, Eckle T, Zug S, Colgan SP, et al. Signaling through the A2B adenosine receptor dampens endotoxin-induced acute lung injury. J Immunol. 2010;184:5271–5279. doi: 10.4049/jimmunol.0903035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider DJ, Lindsay JC, Zhou Y, Molina JG, Blackburn MR. Adenosine and osteopontin contribute to the development of chronic obstructive pulmonary disease. FASEB J. 2010;24:70–80. doi: 10.1096/fj.09-140772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silver MR, Margulis A, Wood N, Goldman SJ, Kasaian M, Chaudhary D. IL-33 synergizes with IgE-dependent and IgE-independent agents to promote mast cell and basophil activation. Inflamm Res. 2010;59:207–218. doi: 10.1007/s00011-009-0088-5. [DOI] [PubMed] [Google Scholar]
- Sitaraman SV, Merlin D, Wang L, Wong M, Gewirtz AT, Si-Tahar M, et al. Neutrophil-epithelial crosstalk at the intestinal lumenal surface mediated by reciprocal secretion of adenosine and IL-6. J Clin Invest. 2001;107:861–869. doi: 10.1172/JCI11783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srinivas SP, Satpathy M, Gallagher P, Lariviere E, Van Driessche W. Adenosine induces dephosphorylation of myosin II regulatory light chain in cultured bovine corneal endothelial cells. Exp Eye Res. 2004;79:543–551. doi: 10.1016/j.exer.2004.06.027. [DOI] [PubMed] [Google Scholar]
- Strohmeier GR, Reppert SM, Lencer WI, Madara JL. The A2b adenosine receptor mediates cAMP responses to adenosine receptor agonists in human intestinal epithelia. J Biol Chem. 1995;270:2387–2394. doi: 10.1074/jbc.270.5.2387. [DOI] [PubMed] [Google Scholar]
- Sun CX, Zhong H, Mohsenin A, Morschl E, Chunn JL, Molina JG, et al. Role of A2B adenosine receptor signaling in adenosine-dependent pulmonary inflammation and injury. J Clin Invest. 2006;116:2173–2182. doi: 10.1172/JCI27303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suzuki H, Takei M, Nakahata T, Fukamachi H. Inhibitory effect of adenosine on degranulation of human cultured mast cells upon cross-linking of Fc epsilon RI. Biochem Biophys Res Commun. 1998;242:697–702. doi: 10.1006/bbrc.1997.8040. [DOI] [PubMed] [Google Scholar]
- Synnestvedt K, Furuta GT, Comerford KM, Louis N, Karhausen J, Eltzschig HK, et al. Ecto-5′-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia. J Clin Invest. 2002;110:993–1002. doi: 10.1172/JCI15337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takagi H, King GL, Ferrara N, Aiello LP. Hypoxia regulates vascular endothelial growth factor receptor KDR/Flk gene expression through adenosine A2 receptors in retinal capillary endothelial cells. Invest Ophthalmol Visual Sci. 1996;37:1311–1321. [PubMed] [Google Scholar]
- Takedachi M, Qu D, Ebisuno Y, Oohara H, Joachims ML, McGee ST, et al. CD73-generated adenosine restricts lymphocyte migration into draining lymph nodes. J Immunol. 2008;180:6288–6296. doi: 10.4049/jimmunol.180.9.6288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tang Y, Feng Y, Wang X. Calcitonin gene-related peptide potentiates LPS-induced IL-6 release from mouse peritoneal macrophages. J Neuroimmunol. 1998;84:207–212. doi: 10.1016/s0165-5728(97)00257-9. [DOI] [PubMed] [Google Scholar]
- Thompson LF, Eltzschig HK, Ibla JC, Van De Wiele CJ, Resta R, Morote-Garcia JC, et al. Crucial role for ecto-5′-nucleotidase (CD73) in vascular leakage during hypoxia. J Exp Med. 2004;200:1395–1405. doi: 10.1084/jem.20040915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Umapathy NS, Fan Z, Zemskov EA, Alieva IB, Black SM, Verin AD. Molecular mechanisms involved in adenosine-induced endothelial cell barrier enhancement. Vascul Pharmacol. 2010;52:199–206. doi: 10.1016/j.vph.2009.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walia B, Castaneda FE, Wang L, Kolachala VL, Bajaj R, Roman J, et al. Polarized fibronectin secretion induced by adenosine regulates bacterial-epithelial interaction in human intestinal epithelial cells. Biochem J. 2004;382:589–596. doi: 10.1042/BJ20040021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walker BA, Jacobson MA, Knight DA, Salvatore CA, Weir T, Zhou D, et al. Adenosine A3 receptor expression and function in eosinophils. Am J Resp Cell Mol Biol. 1997;16:531–537. doi: 10.1165/ajrcmb.16.5.9160835. [DOI] [PubMed] [Google Scholar]
- Wang D, Sun Y, Zhang W, Huang P. Apical adenosine regulates basolateral Ca2+-activated potassium channels in human airway Calu-3 epithelial cells. Am J Physiol. 2008;294:C1443–C1453. doi: 10.1152/ajpcell.00556.2007. [DOI] [PubMed] [Google Scholar]
- Wang J, Huxley VH. Adenosine A2A receptor modulation of juvenile female rat skeletal muscle microvessel permeability. Am J Physiol Heart Circ Physiol. 2006;291:H3094–H3105. doi: 10.1152/ajpheart.00526.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waschke J, Drenckhahn D, Adamson RH, Barth H, Curry FE. cAMP protects endothelial barrier functions by preventing Rac-1 inhibition. Am J Physiol. 2004;287:H2427–H2433. doi: 10.1152/ajpheart.00556.2004. [DOI] [PubMed] [Google Scholar]
- Wen J, Jiang X, Dai Y, Zhang Y, Tang Y, Sun H, et al. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A2B adenosine receptor signaling. FASEB J. 2010;24:740–749. doi: 10.1096/fj.09-144147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilson JM, Ross WG, Agbai ON, Frazier R, Figler RA, Rieger J, et al. The A2B adenosine receptor impairs the maturation and immunogenicity of dendritic cells. J Immunol. 2009;182:4616–4623. doi: 10.4049/jimmunol.0801279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xaus J, Mirabet M, Lloberas J, Soler C, Lluis C, Franco R, et al. IFN-gamma up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J Immunol. 1999a;162:3607–3614. [PubMed] [Google Scholar]
- Xaus J, Valledor AF, Cardo M, Marques L, Beleta J, Palacios JM, et al. Adenosine inhibits macrophage colony-stimulating factor-dependent proliferation of macrophages through the induction of p27kip-1 expression. J Immunol. 1999b;163:4140–4149. [PubMed] [Google Scholar]
- Yang D, Chen H, Koupenova M, Carroll SH, Eliades A, Freedman JE, et al. A new role for the A2b adenosine receptor in regulating platelet function. J Thromb Haemost. 2010;8:817–827. doi: 10.1111/j.1538-7836.2010.03769.x. [DOI] [PubMed] [Google Scholar]
- Yang D, Zhang Y, Nguyen HG, Koupenova M, Chauhan AK, Makitalo M, et al. The A2B adenosine receptor protects against inflammation and excessive vascular adhesion. J Clin Invest. 2006;116:1913–1923. doi: 10.1172/JCI27933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zaynagetdinov R, Ryzhov S, Goldstein AE, Yin H, Novitskiy SV, Goleniewska K, et al. Attenuation of chronic pulmonary inflammation in A2B adenosine receptor knockout mice. Am J Respir Cell Mol Biol. 2010;42:564–571. doi: 10.1165/rcmb.2008-0391OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang JG, Hepburn L, Cruz G, Borman RA, Clark KL. The role of adenosine A2A and A2B receptors in the regulation of TNF-alpha production by human monocytes. Biochem Pharmacol. 2005;69:883–889. doi: 10.1016/j.bcp.2004.12.008. [DOI] [PubMed] [Google Scholar]
- Zhong H, Belardinelli L, Maa T, Zeng D. Synergy between A2B adenosine receptors and hypoxia in activating human lung fibroblasts. Am J Resp Cell Mol Biol. 2005;32:2–8. doi: 10.1165/rcmb.2004-0103OC. [DOI] [PubMed] [Google Scholar]
- Zhou Y. The role of A2B adenosine receptor signaling in adenosine dependent lung desease. UT Graduate School of Biomedical Sciences at Houston: Dissertations and Theses. 2010:Paper 13. http://digitalcommons.library.tmc.edu/utgsbs_dissertations/13.
- Zhou Y, Mohsenin A, Morschl E, Young HW, Molina JG, Ma W, et al. Enhanced airway inflammation and remodeling in adenosine deaminase-deficient mice lacking the A2B adenosine receptor. J Immunol. 2009;182:8037–8046. doi: 10.4049/jimmunol.0900515. [DOI] [PMC free article] [PubMed] [Google Scholar]
