Skip to main content
The Journal of Clinical Investigation logoLink to The Journal of Clinical Investigation
. 2006 Jul 3;116(7):1835–1837. doi: 10.1172/JCI29125

New insights into the regulation of inflammation by adenosine

Joel Linden 1
PMCID: PMC1483168  PMID: 16823484

Abstract

Adenosine, long known as a regulator of cardiovascular function, has recently been identified as a significant paracrine inhibitor of inflammation that acts primarily by activation of A2A adenosine receptors (A2AARs) on lymphoid or myeloid cells. In this issue of the JCI, Yang et al. describe a proinflammatory phenotype resulting from deletion of the gene encoding the A2B adenosine receptor (A2BAR) in the mouse, suggesting that activation of the A2BAR can also have antiinflammatory effects (see the related article beginning on page 1913). Nevertheless, the role of the A2BAR remains enigmatic since its activation can either stimulate or inhibit the release of proinflammatory cytokines in different cells and tissues.


There is growing interest in elucidating the mechanisms by which adenosine inhibits the immune system, since these inhibitory adenosine receptors and their downstream signaling pathways are promising targets for new antiinflammatory therapies. By signaling through the A2A adenosine receptor (A2AAR), adenosine suppresses the immune system, primarily by inhibiting lymphoid or myeloid cells including neutrophils (1), macrophages (2), lymphocytes (3, 4), and platelets (5). These responses are amplified by rapid induction of A2AAR mRNA in macrophages and T lymphocytes in response to inflammatory or ischemic stimuli (2, 3, 6, 7). The A2B adenosine receptor (A2BAR) also appears to mediate antiinflammatory effects in macrophages by inhibiting the production of TNF-α and IL-1β, stimulating IL-10 and inhibiting macrophage proliferation (811) (Figure 1A). Macrophage A2BAR signaling increases during inflammation, as the macrophage-activating cytokine IFN-γ causes induction of macrophage A2BAR mRNA (12). However, the A2BAR is somewhat unusual in that it is dually coupled to the generally antiinflammatory G protein Gs and the generally proinflammatory G protein Gq (13); in addition, numerous proinflammatory effects mediated by activation of the A2BAR have also been described (Table 1).

Figure 1. Anti- and proinflammatory effects mediated by activation of the A2B AR.

Figure 1

(A) In macrophages, the release of TNF-α and other proinflammatory cytokines is inhibited as a result of activating either the A2AAR or the A2BAR. These effects are amplified if TNF-α production is stimulated by LPS, which signals through TLR4 and other Toll-like receptors. Activation of the A2BAR also stimulates production of the antiinflammatory cytokine IL-10. (B) In endothelial cells and other cells noted in Table 1, activation of the A2BAR stimulates IL-6 and other proinflammatory cytokines. Hypoxia increases the intracellular production of adenosine, which is transported outside the cell by nucleoside transport proteins. Hypoxia and possibly activation of the A2BAR stimulate the release of ATP and the production of the A2BAR and ectoenzymes (CD39 and CD73) that convert ATP to adenosine. Vasodilation in response to A2BAR activation may increase shear stress to stimulate ATP release.

Table 1 .

Proinflammatory effects of activating A2BR

graphic file with name JCI0629125.t1.jpg

Inflamed phenotype of the A2B AR knockout mouse

Given the opposing cellular effects mediated by A 2BAR activation, the phenotype that would result from deletion of the gene encoding A 2BAR was uncertain. In this issue of the JCI, Yang and coworkers use a receptor knockout/reporter gene knock-in approach to confirm the expected expression pattern of the A 2BAR transcript, noting high levels in monocytes/macrophages and the vasculature (14). Interestingly, they report a moderately inflamed phenotype in the A 2BAR knockout mouse at baseline, including elevated adhesion molecule expression on vascular endothelial cells and elevated cytokine production. The inflammatory phenotype is exaggerated in mice given endotoxin. The baseline response to A 2BAR knockout is surprising since the A 2BAR is recognized to have a low affinity for adenosine, yet the data imply that baseline adenosine levels are high enough to produce some activation of the A 2BAR. A well-recognized limitation of the global knockout approach is that the resulting phenotype may reflect a developmental adaptation to the gene knockout and not an acute direct consequence of deleting the gene product. In order to address this possibility, Yang et al. examined the effect of deleting the gene that codes for A 2BAR on mRNA levels of the other adenosine receptor subtypes and found no changes. However, other possible adaptations have yet to be examined. It has recently been demonstrated that hypoxia triggers coordinate upregulation on endothelial cells of mRNA for the A 2BAR and ectoenzymes involved in the conversion of extracellular adenine nucleotides to adenosine (15) (Figure 1B). In addition, the release of adenine nucleotides from various cells, including neutrophils and endothelial cells, is a regulated process (16). It will be of interest in future studies to determine whether deletion of the gene encoding A 2BAR influences either nucleotide release or extracellular nucleotide metabolism. Such effects could indirectly influence inflammation by altering the availability of adenosine to other adenosine receptor subtypes, particularly the A 2AAR. Deletion of either the ecto-apyrase CD39 (also known as NTPDase1; ref. 17) or the ecto–5′-nucleotidase (also known as CD73; refs. 18, 19) results in a proinflammatory phenotype.

Future questions

It will also be of interest in future studies to examine the response of the A 2BAR knockout mouse in the setting of hypoxia or ischemia, which elicit the accumulation of large levels of adenosine. A selective A 2BAR antagonist blocks myocardial preconditioning when applied to the isolated rabbit heart after ischemia (20). This could occur either because A 2BARs on the heart mediate cardioprotection or, as discussed above, because A 2BAR activation facilitates the release and/or metabolism of adenine nucleotides to indirectly enhance adenosine production. The latter scheme is consistent with the observation that myocardial preconditioning has a remote adenosine-mediated effect on platelet function (21).

In conclusion, the study by Yang et al. (14) bolsters the conclusion that activation of A2BARs on certain cells, particularly macrophages, inhibits inflammation. A proinflammatory phenotype noted at rest is somewhat unexpected, and the mechanism underlying this inflammation is not yet known. In view of a number of previous reports indicating that A2BAR activation can be proinflammatory (Table 1), it will be of interest to use the newly available A2BAR knockout mouse in order to determine whether A2BAR activation on different cells can elicit both pro- and antiinflammatory responses.

Footnotes

Nonstandard abbreviations used: A2AAR, A2A adenosine receptor; A2BAR, A2B adenosine receptor.

Conflict of interest: The author owns stock in Adenosine Therapeutics, LLC.

Citation for this article: J. Clin. Invest. 116:1835–1837 (2006). doi:10.1172/JCI29125.

See the related article beginning on page 1913.

References

  • 1.Fredholm B.B. Purines and neutrophil leukocytes. Gen. Pharmacol. 1997;28:345–350. doi: 10.1016/s0306-3623(96)00169-3. [DOI] [PubMed] [Google Scholar]
  • 2.Murphree L.J., Sullivan G.W., Marshall M.A., Linden J. Lipopolysaccharide rapidly modifies adenosine receptor transcripts in murine and human macrophages: role of NF-kappaB in A(2A) adenosine receptor induction. Biochem. J. 2005;391:575–580. doi: 10.1042/BJ20050888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lappas C.M., Rieger J.M., Linden J. A2A adenosine receptor induction inhibits IFN-gamma production in murine CD4+ T cells. J. Immunol. 2005;174:1073–1080. doi: 10.4049/jimmunol.174.2.1073. [DOI] [PubMed] [Google Scholar]
  • 4.Koshiba M., Rosin D.L., Hayashi N., Linden J., Sitkovsky M.V. Patterns of A2A extracellular adenosine receptor expression in different functional subsets of human peripheral T cells. Flow cytometry studies with anti-A2A receptor monoclonal antibodies. Mol. Pharmacol. 1999;55:614–624. [PubMed] [Google Scholar]
  • 5.Varani K., Gessi S., Dalpiaz A., Borea P.A. Pharmacological and biochemical characterization of purified A2a adenosine receptors in human platelet membranes by [3H]-CGS 21680 binding. Br. J. Pharmacol. . 1996;117:1693–1701. doi: 10.1111/j.1476-5381.1996.tb15341.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Toufektsian M.C., et al. Stimulation of A2A-adenosine receptors after myocardial infarction suppresses inflammatory activation and attenuates contractile dysfunction in the remote left ventricle. Am. J. Physiol. Heart Circ. Physiol. 2006;290:H1410–H1418. doi: 10.1152/ajpheart.00860.2005. [DOI] [PubMed] [Google Scholar]
  • 7.Napieralski R., Kempkes B., Gutensohn W. Evidence for coordinated induction and repression of ecto-5′-nucleotidase (CD73) and the A2a adenosine receptor in a human B cell line. Biol. Chem. 2003;384:483–487. doi: 10.1515/BC.2003.054. [DOI] [PubMed] [Google Scholar]
  • 8.Nemeth Z.H., 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]
  • 9.Xaus J., et al. Adenosine inhibits macrophage colony-stimulating factor-dependent proliferation of macrophages through the induction of p27kip-1 expression. J. Immunol. 1999;163:4140–4149. [PubMed] [Google Scholar]
  • 10.Kreckler L.M., Wan T.C., Ge Z.D., Auchampach J.A. 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]
  • 11.Sipka S., et al. Adenosine inhibits the release of interleukin-1beta in activated human peripheral mononuclear cells. Cytokine. 2005;31:258–263. doi: 10.1016/j.cyto.2005.05.002. [DOI] [PubMed] [Google Scholar]
  • 12.Xaus J., et al. IFN-gamma up-regulates the A2B adenosine receptor expression in macrophages: a mechanism of macrophage deactivation. J. Immunol. 1999;162:3607–3614. [PubMed] [Google Scholar]
  • 13.Linden J., Auchampach J.A., Jin X., Figler R.A. The structure and function of A1 and A2B adenosine receptors. Life Sci. 1998;62:1519–1524. doi: 10.1016/s0024-3205(98)00100-3. [DOI] [PubMed] [Google Scholar]
  • 14.Yang D., et al. The A2B adensoine 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]
  • 15.Eltzschig H.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]
  • 16.Burnstock G. Purinergic signalling. Br. J. Pharmacol. 2006;147(Suppl. 1):S172–S181. doi: 10.1038/sj.bjp.0706429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dwyer K.M., et al. Thromboregulatory manifestations in human CD39 transgenic mice and the implications for thrombotic disease and transplantation. J. Clin. Invest. 2004;113:1440–1446. doi: 10.1172/JCI200419560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Koszalka P., et al. Targeted disruption of cd73/ecto-5′-nucleotidase alters thromboregulation and augments vascular inflammatory response. Circ. Res. 2004;95:814–821. doi: 10.1161/01.RES.0000144796.82787.6f. [DOI] [PubMed] [Google Scholar]
  • 19.Zernecke A., et al. CD73/ecto-5′-nucleotidase protects against vascular inflammation and neointima formation. Circulation. 2006;113:2120–2127. doi: 10.1161/CIRCULATIONAHA.105.595249. [DOI] [PubMed] [Google Scholar]
  • 20.Solenkova N.V., Solodushko V., Cohen M.V., Downey J.M. Endogenous adenosine protects preconditioned heart during early minutes of reperfusion by activating Akt. Am. J. Physiol. Heart Circ. Physiol. 2006;290:H441–H449. doi: 10.1152/ajpheart.00589.2005. [DOI] [PubMed] [Google Scholar]
  • 21.Hata K., Whittaker P., Kloner R.A., Przyklenk K. Brief myocardial ischemia attenuates platelet thrombosis in remote, damaged, and stenotic carotid arteries. Circulation. 1999;100:843–848. doi: 10.1161/01.cir.100.8.843. [DOI] [PubMed] [Google Scholar]
  • 22.Ryzhov S., et al. 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]
  • 23.Feoktistov I., Polosa R., Holgate S.T., Biaggioni I. Adenosine A2B receptors: a novel therapeutic target in asthma? Trends Pharmacol. Sci. 1998;19:148–153. doi: 10.1016/s0165-6147(98)01179-1. [DOI] [PubMed] [Google Scholar]
  • 24.Auchampach J.A., Jin X., Wan T.C., Caughey G.H., Linden J. Canine mast cell adenosine receptors: cloning and expression of the A3 receptor and evidence that degranulation is mediated by the A2B receptor. Mol. Pharmacol. 1997;52:846–860. doi: 10.1124/mol.52.5.846. [DOI] [PubMed] [Google Scholar]
  • 25.Zhong H., et al. A(2B) adenosine receptors increase cytokine release by bronchial smooth muscle cells. Am. J. Respir. Cell Mol. Biol. 2004;30:118–125. doi: 10.1165/rcmb.2003-0118OC. [DOI] [PubMed] [Google Scholar]
  • 26.Sitaraman S.V., 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]
  • 27.Fiebich B.L., et al. Adenosine A2b receptors mediate an increase in interleukin (IL)-6 mRNA and IL-6 protein synthesis in human astroglioma cells. . J. Neurochem. 1996;66:1426–1431. doi: 10.1046/j.1471-4159.1996.66041426.x. [DOI] [PubMed] [Google Scholar]
  • 28.Fiebich B.L., et al. IL-6 expression induced by adenosine A2b receptor stimulation in U373 MG cells depends on p38 mitogen activated kinase and protein kinase C. Neurochem. Int. 2005;46:501–512. doi: 10.1016/j.neuint.2004.11.009. [DOI] [PubMed] [Google Scholar]
  • 29.Schwaninger M., Neher M., Viegas E., Schneider A., Spranger M. Stimulation of interleukin-6 secretion and gene transcription in primary astrocytes by adenosine. J. Neurochem. 1997;69:1145–1150. doi: 10.1046/j.1471-4159.1997.69031145.x. [DOI] [PubMed] [Google Scholar]
  • 30.Evans B.A., et al. Human osteoblast precursors produce extracellular adenosine, which modulates their secretion of IL-6 and osteoprotegerin. . J. Bone Miner. Res. 2006;21:228–236. doi: 10.1359/JBMR.051021. [DOI] [PubMed] [Google Scholar]
  • 31.Zhong H., Belardinelli L., Maa T., Zeng D. Synergy between A2B adenosine receptors and hypoxia in activating human lung fibroblasts. Am. J. Respir. Cell Mol. Biol. 2005;32:2–8. doi: 10.1165/rcmb.2004-0103OC. [DOI] [PubMed] [Google Scholar]
  • 32.Rees D.A., et al. Adenosine-induced IL-6 expression in pituitary folliculostellate cells is mediated via A2b adenosine receptors coupled to PKC and p38 MAPK. Br. J. Pharmacol. 2003;140:764–772. doi: 10.1038/sj.bjp.0705488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Li Y., Wang W., Parker W., Clancy J.P. Adenosine regulation of cystic fibrosis transmembrane conductance regulator through prostenoids in Airway epithelia. Am. J. Respir. Cell Mol. Biol. 2006;34:600–608. doi: 10.1165/rcmb.2005-0421OC. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical Investigation are provided here courtesy of American Society for Clinical Investigation

RESOURCES