Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Adenosine receptors as drug targets — what are the challenges?

Key Points

  • It has been established for several decades that extracellular adenosine is an important modulator of physiological and pathological processes that can be safely targeted by adenosine receptor agonists or antagonists. Experimental studies have clarified the biological functions of four adenosine receptors, which represent pharmacological targets for the treatment of several human diseases including neurological, inflammatory or ischaemic conditions.

  • Despite several years of efforts, including several large Phase III clinical trials of adenosine receptor drugs, very few agents have actually made it to the clinic owing to insufficient efficacy and/or unacceptable side effects.

  • Based on this discrepancy, we discuss the therapeutic potential of adenosine receptor modulators, focusing on the key biological factors limiting their clinical development and the hurdles that could and should be overcome.

  • A major challenge in developing adenosine receptor ligands for specific clinical applications is that adenosine has so many roles. Thus, demonstrating the effects of drugs causing adenosine receptor activation or inactivation on specific systems under distinct experimental settings is not sufficient to prove that they can be delivered in a manner that is clinically effective and safe for treating human disease.

  • We discuss the complexity of adenosine signalling and drug effects over the continuum of specific disease courses, addressing the implications for the use of adenosine receptor-targeting agents.

  • The adenosine receptor antagonist caffeine, which is commonly ingested, complicates the interpretation of clinical trials, and a careful assessment of the caffeine intake of individual patients is vital in assessing a patient's response to adenosine receptor-targeting drugs.

Abstract

Adenosine signalling has long been a target for drug development, with adenosine itself or its derivatives being used clinically since the 1940s. In addition, methylxanthines such as caffeine have profound biological effects as antagonists at adenosine receptors. Moreover, drugs such as dipyridamole and methotrexate act by enhancing the activation of adenosine receptors. There is strong evidence that adenosine has a functional role in many diseases, and several pharmacological compounds specifically targeting individual adenosine receptors — either directly or indirectly — have now entered the clinic. However, only one adenosine receptor-specific agent — the adenosine A2A receptor agonist regadenoson (Lexiscan; Astellas Pharma) — has so far gained approval from the US Food and Drug Administration (FDA). Here, we focus on the biology of adenosine signalling to identify hurdles in the development of additional pharmacological compounds targeting adenosine receptors and discuss strategies to overcome these challenges.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Local amplification of adenosine signalling in response to insults or hypoxia.

Similar content being viewed by others

References

  1. Fredholm, B. B. et al. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol. Rev. 53, 527–552 (2001). This is an overview of the current knowledge of adenosine receptor biology, from gene structure to expression, distribution, biochemical properties, pharmacological profiles, signalling and behavioural responses, focusing on receptor nomenclature and classification.

    CAS  PubMed  Google Scholar 

  2. Fredholm, B. B. et al. International Union of Basic and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors — an update. Pharmacol. Rev. 63, 1–34 (2011). This is an update of reference 1, based on 10 more years of research.

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Eltzschig, H. K., Sitkovsky, M. V. & Robson, S. C. Purinergic signaling during inflammation. N. Engl. J. Med. 367, 2322–2333 (2012). This is a comprehensive review summarizing the biomedical implications of extracellular ATP, ADP and adenosine signalling in the context of a broad range of inflammatory diseases.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Eltzschig, H. K. Adenosine: an old drug newly discovered. Anesthesiology 111, 904–915 (2009).

    CAS  PubMed  Google Scholar 

  5. Johansson, S. M., Yang, J. N., Lindgren, E. & Fredholm, B. B. Eliminating the antilipolytic adenosine A1 receptor does not lead to compensatory changes in the antilipolytic actions of PGE2 and nicotinic acid. Acta Physiol. 190, 87–96 (2007).

    CAS  Google Scholar 

  6. Grenz, A. et al. Equilibrative nucleoside transporter 1 (ENT1) regulates postischemic blood flow during acute kidney injury in mice. J. Clin. Invest. 122, 693–710 (2012). This report shows that a crosstalk pathway — between epithelium-expressed adenosine transporters (such as ENT1) and endothelium-expressed A 2B receptors — dampens renal ischaemia and reperfusion injury by preventing a no-reflow phenomenon.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Sun, D. et al. Mediation of tubuloglomerular feedback by adenosine: evidence from mice lacking adenosine 1 receptors. Proc. Natl Acad. Sci. USA 98, 9983–9988 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Rosenberger, P. et al. Hypoxia-inducible factor-dependent induction of netrin-1 dampens inflammation caused by hypoxia. Nature Immunol. 10, 195–202 (2009). This study demonstrates that the neuronal guidance molecule netrin 1 has important functions outside brain development, showing that hypoxia-dependent induction of netrin 1 dampens mucosal inflammation by enhancing a purinergic signalling event, particularly through the A 2B receptor.

    CAS  Google Scholar 

  9. Huang, Z. L. et al. Adenosine A2A, but not A1, receptors mediate the arousal effect of caffeine. Nature Neurosci. 8, 858–859 (2005).

    CAS  PubMed  Google Scholar 

  10. Lazarus, M. et al. Arousal effect of caffeine depends on adenosine A2A receptors in the shell of the nucleus accumbens. J. Neurosci. 31, 10067–10075 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu, X. L. et al. Genetic inactivation of the adenosine A2A receptor attenuates pathologic but not developmental angiogenesis in the mouse retina. Invest. Ophthalmol. Vis. Sci. 51, 6625–6632 (2010).

    PubMed  PubMed Central  Google Scholar 

  12. Hasko, G., Linden, J., Cronstein, B. & Pacher, P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nature Rev. Drug Discov. 7, 759–770 (2008). This is an expert and focused review on the molecular and cellular biology of adenosine receptors and their pharmacological actions, with emphasis on immune and inflammatory responses.

    CAS  Google Scholar 

  13. Eltzschig, H. K. & Carmeliet, P. Hypoxia and inflammation. N. Engl. J. Med. 364, 656–665 (2011). This review describes the relationship between hypoxia and inflammation, and demonstrates the functional roles of their interdepencence during intestinal inflammation, acute lung injury, ischaemia–reperfusion injury and cancer.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Eltzschig, H. K. & Eckle, T. Ischemia and reperfusion —from mechanism to translation. Nature Med. 17, 1391–1401 (2011).

    CAS  PubMed  Google Scholar 

  15. Fredholm, B. B. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ. 14, 1315–1323 (2007).

    CAS  PubMed  Google Scholar 

  16. Jacobson, K. A. & Gao, Z. G. Adenosine receptors as therapeutic targets. Nature Rev. Drug Discov. 5, 247–264 (2006). This is an expert and comprehensive review on the medicinal chemistry of the first and second generations of adenosine receptor agonists and antagonists as well as their possible clinical uses.

    CAS  Google Scholar 

  17. Delacretaz, E. Clinical practice. Supraventricular tachycardia. N. Engl. J. Med. 354, 1039–1051 (2006).

    CAS  PubMed  Google Scholar 

  18. Muller, C. E. & Jacobson, K. A. Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochim. Biophys. Acta 1808, 1290–1308 (2011).

    PubMed  Google Scholar 

  19. Ghimire, G., Hage, F. G., Heo, J. & Iskandrian, A. E. Regadenoson: a focused update. J. Nucl. Cardiol. 11 Dec 2012 (doi:10.1007/s12350-012-9661-3).

    Google Scholar 

  20. Fredholm, B., Chen, J.-F., Masino, S. A. & Vaugeois, J.-M. Actions of adeosine at its receptors in the CNS: insights from knckouts and drugs. Annu. Rev. Pharmacol. Toxicol. 45, 385–412 (2005). This is a comprehensive review on the development and characterization of genetic knockout models for adenosine receptors (from 1997 to 2005), focusing on new insights into brain functions under physiological and pathological conditions.

    CAS  PubMed  Google Scholar 

  21. Massie, B. M. et al. Rolofylline, an adenosine A1-receptor antagonist, in acute heart failure. N. Engl. J. Med. 363, 1419–1428 (2010). This is a report on the clinical Phase III trial for the A 1 receptor antagonist rolofylline in 2,033 patients with acute heart failure; the drug had a disappointing clinical outcome.

    PubMed  Google Scholar 

  22. Teerlink, J. R. et al. The safety of an adenosine A1-receptor antagonist, rolofylline, in patients with acute heart failure and renal impairment: findings from PROTECT. Drug Saf. 35, 233–244 (2012).

    CAS  PubMed  Google Scholar 

  23. Fredholm, B. B. et al. Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch. Pharmacol. 362, 364–374 (2000).

    CAS  PubMed  Google Scholar 

  24. Daly, J. W. & Padgett, W. L. Agonist activity of 2- and 5′-substituted adenosine analogs and their N6-cycloalkyl derivatives at A1- and A2-adenosine receptors coupled to adenylate cyclase. Biochem. Pharmacol. 43, 1089–1093 (1992).

    CAS  PubMed  Google Scholar 

  25. Koeppen, M., Eckle, T. & Eltzschig, H. K. Selective deletion of the A1 adenosine receptor abolishes heart-rate slowing effects of intravascular adenosine in vivo. PLoS ONE 4, e6784 (2009).

    PubMed  PubMed Central  Google Scholar 

  26. Corvol, J. C., Studler, J. M., Schonn, J. S., Girault, J. A. & Herve, D. Gαolf is necessary for coupling D1 and A2a receptors to adenylyl cyclase in the striatum. J. Neurochem. 76, 1585–1588 (2001).

    CAS  PubMed  Google Scholar 

  27. Kull, B., Svenningsson, P. & Fredholm, B. B. Adenosine A2A receptors are colocalized with and activate Golf in rat striatum. Mol. Pharmacol. 58, 771–777 (2000).

    CAS  PubMed  Google Scholar 

  28. Schulte, G. & Fredholm, B. B. Signalling from adenosine receptors to mitogen-activated protein kinases. Cell. Signal. 15, 813–827 (2003).

    CAS  PubMed  Google Scholar 

  29. Eckle, T. et al. A2B adenosine receptor dampens hypoxia-induced vascular leak. Blood 111, 2024–2035 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Eckle, T. et al. Adora2b-elicited Per2 stabilization promotes a HIF-dependent metabolic switch crucial for myocardial adaptation to ischemia. Nature Med. 18, 774–782 (2012). This study provides a mechanism of how A 2B receptor signalling can provide cardioprotection against ischaemia–reperfusion injury. The findings indicate that myocardial A 2B signalling leads to the stabilization of the circadian rhythm protein PER2 (period circadian clock 2), which mediates a metabolic switch that enhances myocardial glycolytic capacity, thereby providing enhanced ischaemia tolerance.

    CAS  PubMed  Google Scholar 

  31. Hart, M. L. et al. Hypoxia-inducible factor-1α-dependent protection from intestinal ischemia/reperfusion injury involves ecto-5′-nucleotidase (CD73) and the A2B adenosine receptor. J. Immunol. 186, 4367–4374 (2011).

    CAS  PubMed  Google Scholar 

  32. Schingnitz, U. et al. Signaling through the A2B adenosine receptor dampens endotoxin-induced acute lung injury. J. Immunol. 184, 5271–5279 (2010).

    CAS  PubMed  Google Scholar 

  33. Frick, J. S. et al. Contribution of adenosine A2B receptors to inflammatory parameters of experimental colitis. J. Immunol. 182, 4957–4964 (2009).

    CAS  PubMed  Google Scholar 

  34. Zhong, H. et al. Activation of murine lung mast cells by the adenosine A3 receptor. J. Immunol. 171, 338–345 (2003).

    CAS  PubMed  Google Scholar 

  35. Ochaion, A. et al. The anti-inflammatory target A3 adenosine receptor is over-expressed in rheumatoid arthritis, psoriasis and Crohn's disease. Cell. Immunol. 258, 115–122 (2009).

    CAS  PubMed  Google Scholar 

  36. Gessi, S. et al. Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clin. Cancer Res. 10, 5895–5901 (2004).

    CAS  PubMed  Google Scholar 

  37. Fishman, P., Bar-Yehuda, S., Liang, B. T. & Jacobson, K. A. Pharmacological and therapeutic effects of A3 adenosine receptor agonists. Drug Discov. Today 17, 359–366 (2012).

    CAS  PubMed  Google Scholar 

  38. King, A. E., Ackley, M. A., Cass, C. E., Young, J. D. & Baldwin, S. A. Nucleoside transporters: from scavengers to novel therapeutic targets. Trends Pharmacol. Sci. 27, 416–425 (2006).

    CAS  PubMed  Google Scholar 

  39. MacDonald, P. E., Braun, M., Galvanovskis, J. & Rorsman, P. Release of small transmitters through kiss-and-run fusion pores in rat pancreatic β cells. Cell Metab. 4, 283–290 (2006).

    CAS  PubMed  Google Scholar 

  40. Zhang, Z. et al. Regulated ATP release from astrocytes through lysosome exocytosis. Nature Cell Biol. 9, 945–953 (2007).

    CAS  PubMed  Google Scholar 

  41. Chekeni, F. B. et al. Pannexin 1 channels mediate 'find-me' signal release and membrane permeability during apoptosis. Nature 467, 863–867 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Elliott, M. R. et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 461, 282–286 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Anselmi, F. et al. ATP release through connexin hemichannels and gap junction transfer of second messengers propagate Ca2+ signals across the inner ear. Proc. Natl. Acad. Sci. USA 105, 18770–18775 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kanneganti, T. D. et al. Pannexin-1-mediated recognition of bacterial molecules activates the cryopyrin inflammasome independent of Toll-like receptor signaling. Immunity 26, 433–443 (2007).

    CAS  PubMed  Google Scholar 

  45. Faigle, M., Seessle, J., Zug, S., El Kasmi, K. C. & Eltzschig, H. K. ATP release from vascular endothelia occurs across Cx43 hemichannels and is attenuated during hypoxia. PLoS ONE 3, e2801 (2008).

    PubMed  PubMed Central  Google Scholar 

  46. Ballarin, M., Fredholm, B. B., Ambrosio, S. & Mahy, N. Extracellular levels of adenosine and its metabolites in the striatum of awake rats: inhibition of uptake and metabolism. Acta Physiol. Scand. 142, 97–103 (1991).

    CAS  PubMed  Google Scholar 

  47. Wei, C. J., Li, W. & Chen, J. F. Normal and abnormal functions of adenosine receptors in the central nervous system revealed by genetic knockout studies. Biochim. Biophys. Acta 1808, 1358–1379 (2011).

    CAS  PubMed  Google Scholar 

  48. Fedele, D. E., Li, T., Lan, J. Q., Fredholm, B. B. & Boison, D. Adenosine A1 receptors are crucial in keeping an epileptic focus localized. Exp. Neurol. 200, 184–190 (2006).

    CAS  PubMed  Google Scholar 

  49. Gimenez-Llort, L. et al. Mice lacking the adenosine A1 receptor are anxious and aggressive, but are normal learners with reduced muscle strength and survival rate. Eur. J. Neurosci. 16, 547–550 (2002).

    PubMed  Google Scholar 

  50. Schwarzschild, M. A., Agnati, L., Fuxe, K., Chen, J. F. & Morelli, M. Targeting adenosine A2A receptors in Parkinson's disease. Trends Neurosci. 29, 647–654 (2006). This is an expert and focused review on the rationale as well as preclinical and clinical evidence for the efficacy of an A 2A receptor antagonist in Parkinson's disease.

    CAS  PubMed  Google Scholar 

  51. Ferre, S., Fredholm, B. B., Morelli, M., Popoli, P. & Fuxe, K. Adenosine–dopamine receptor–receptor interactions as an integrative mechanism in the basal ganglia. Trends Neurosci. 20, 482–487 (1997).

    CAS  PubMed  Google Scholar 

  52. Richardson, P. J., Kase, H. & Jenner, P. G. Adenosine A2A receptor antagonists as new agents for the treatment of Parkinson's disease. Trends Pharmacol. Sci. 18, 338–344 (1997).

    CAS  PubMed  Google Scholar 

  53. Wei, C. J. et al. Selective inactivation of adenosine A2A receptors in striatal neurons enhances working memory and reversal learning. Learn. Mem. 18, 459–474 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Zhou, S. J. et al. Preferential enhancement of working memory in mice lacking adenosine A2A receptors. Brain Res. 1303, 74–83 (2009).

    CAS  PubMed  Google Scholar 

  55. Yu, C., Gupta, J., Chen, J. F. & Yin, H. H. Genetic deletion of A2A adenosine receptors in the striatum selectively impairs habit formation. J. Neurosci. 29, 15100–15103 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ledent, C. et al. Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature 388, 674–678 (1997). This milestone study describes the generation and initial characterization of the first genetic knockout mice for adenosine receptors — namely the Adora2a -knockout mice.

    CAS  PubMed  Google Scholar 

  57. Chen, J. F. et al. A2A adenosine receptor deficiency attenuates brain injury induced by transient focal ischemia in mice. J. Neurosci. 19, 9192–9200 (1999). This study describes the generation and characterization of the second line of the Adora2a -knockout mice and provides compelling evidence that A 2A receptor inactivation confers neuroprotection against ischaemia.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Xiao, D. et al. Forebrain adenosine A2A receptors contribute to l-3,4-dihydroxyphenylalanine-induced dyskinesia in hemiparkinsonian mice. J. Neurosci. 26, 13548–13555 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ohta, A. & Sitkovsky, M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 414, 916–920 (2001). This landmark study demonstrates that endogenous adenosine via the activation of A 2A receptors functions to limit excessive inflammation and collateral tissue damage.

    CAS  PubMed  Google Scholar 

  60. Day, Y. J. et al. Renal protection from ischemia mediated by A2A adenosine receptors on bone marrow-derived cells. J. Clin. Invest. 112, 883–891 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Chen, J. F. et al. Adenosine A2A receptors and brain injury: broad spectrum of neuroprotection, multifaceted actions and “fine tuning” modulation. Prog. Neurobiol. 83, 310–331 (2007).

    CAS  PubMed  Google Scholar 

  62. Yu, L. et al. Selective inactivation or reconstitution of adenosine A2A receptors in bone marrow cells reveals their significant contribution to the development of ischemic brain injury. Nature Med. 10, 1081–1087 (2004). This study demonstrates a novel role of the A 2A receptor in bone marrow-derived cells: it protects against ischaemic brain injury.

    CAS  PubMed  Google Scholar 

  63. Huang, Z. L., Urade, Y. & Hayaishi, O. The role of adenosine in the regulation of sleep. Curr. Top. Med. Chem. 11, 1047–1057 (2011).

    CAS  PubMed  Google Scholar 

  64. Yang, D. et al. The A2B adenosine receptor protects against inflammation and excessive vascular adhesion. J. Clin. Invest. 116, 1913–1923 (2006). This is the first description of Adora2b -knockout mice, which were found to be prone to vascular inflammation — a finding that was later confirmed in many other models of vascular inflammation.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Eckle, T. et al. Cardioprotection by ecto-5′-nucleotidase (CD73) and A2B adenosine receptors. Circulation 115, 1581–1590 (2007).

    CAS  PubMed  Google Scholar 

  66. Schulte, G. & Fredholm, B. B. Human adenosine A1, A2A, A2B, and A3 receptors expressed in Chinese hamster ovary cells all mediate the phosphorylation of extracellular-regulated kinase 1/2. Mol. Pharmacol. 58, 477–482 (2000).

    CAS  PubMed  Google Scholar 

  67. Hart, M. L., Jacobi, B., Schittenhelm, J., Henn, M. & Eltzschig, H. K. Cutting Edge: A2B adenosine receptor signaling provides potent protection during intestinal ischemia/reperfusion injury. J. Immunol. 182, 3965–3968 (2009).

    CAS  PubMed  Google Scholar 

  68. Chouker, A. et al. In vivo hypoxic preconditioning protects from warm liver ischemia-reperfusion injury through the adenosine A2B receptor. Transplantation 94, 894–902 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Grenz, A. et al. The reno-vascular A2B adenosine receptor protects the kidney from ischemia. PLoS Med. 5, e137 (2008).

    PubMed  PubMed Central  Google Scholar 

  70. Eckle, T., Grenz, A., Laucher, S. & Eltzschig, H. K. A2B adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice. J. Clin. Invest. 118, 3301–3315 (2008). This head-to-head comparison of all four Adora -knockout mice revealed that Adora2b -knockout mice are particularly prone to ventilator-induced lung injury. The A 2B receptor agonist BAY 60-6583 enhanced alveolar fluid transport, thereby attenuating pulmonary oedema and concomittant lung inflammation during acute lung injury.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Takedachi, M. et al. CD73-generated adenosine promotes osteoblast differentiation. J. Cell. Physiol. 227, 2622–2631 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Carroll, S. H. et al. A2B adenosine receptor promotes mesenchymal stem cell differentiation to osteoblasts and bone formation in vivo. J. Biol. Chem. 287, 15718–15727 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Johnston-Cox, H. et al. The A2b adenosine receptor modulates glucose homeostasis and obesity. PLoS ONE 7, e40584 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Koupenova, M. et al. A2b adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation 125, 354–363 (2012).

    CAS  PubMed  Google Scholar 

  75. Cekic, C. et al. Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J. Immunol. 188, 198–205 (2012).

    CAS  PubMed  Google Scholar 

  76. Dai, Y. et al. A2B adenosine receptor-mediated induction of IL-6 promotes CKD. J. Am. Soc. Nephrol. 22, 890–901 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Wei, W. et al. Blocking A2B adenosine receptor alleviates pathogenesis of experimental autoimmune encephalomyelitis via inhibition of IL-6 production and Th17 differentiation. J. Immunol. 190, 138–146 (2013).

    CAS  PubMed  Google Scholar 

  78. Bjorklund, O. et al. Decreased behavioral activation following caffeine, amphetamine and darkness in A3 adenosine receptor knock-out mice. Physiol. Behav. 95, 668–676 (2008).

    PubMed  Google Scholar 

  79. Yang, J. N., Wang, Y., Garcia-Roves, P. M., Bjornholm, M. & Fredholm, B. B. Adenosine A3 receptors regulate heart rate, motor activity and body temperature. Acta Physiol. 199, 221–230 (2010).

    CAS  Google Scholar 

  80. 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. 198, 783–796 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Thompson, L. F. et al. Crucial role for ecto-5′-nucleotidase (CD73) in vascular leakage during hypoxia. J. Exp. Med. 200, 1395–1405 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Sitkovsky, M. V. et al. Physiological control of immune response and inflammatory tissue damage by hypoxia-inducible factors and adenosine A2A receptors. Annu. Rev. Immunol. 22, 657–682 (2004). This important review discusses the relationship between hypoxia and adenosine signalling through the A 2A receptor, and implicates A 2A signalling as an endogenous feedback signal to dampen collateral tissue injury.

    CAS  PubMed  Google Scholar 

  83. Gomes, C. V., Kaster, M. P., Tome, A. R., Agostinho, P. M. & Cunha, R. A. Adenosine receptors and brain diseases: neuroprotection and neurodegeneration. Biochim. Biophys. Acta 1808, 1380–1399 (2011).

    CAS  PubMed  Google Scholar 

  84. Boison, D. Adenosine dysfunction in epilepsy. Glia 60, 1234–1243 (2012).

    PubMed  Google Scholar 

  85. Linden, J. Regulation of leukocyte function by adenosine receptors. Adv. Pharmacol. 61, 95–114 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Synnestvedt, K. et al. Ecto-5′-nucleotidase (CD73) regulation by hypoxia-inducible factor-1 mediates permeability changes in intestinal epithelia. J. Clin. Invest. 110, 993–1002 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Zetterstrom, T. et al. Purine levels in the intact rat brain. Studies with an implanted perfused hollow fibre. Neurosci. Lett. 29, 111–115 (1982).

    CAS  PubMed  Google Scholar 

  88. Pedata, F., Corsi, C., Melani, A., Bordoni, F. & Latini, S. Adenosine extracellular brain concentrations and role of A2A receptors in ischemia. Ann. NY Acad. Sci. 939, 74–84 (2001).

    CAS  PubMed  Google Scholar 

  89. Andine, P., Rudolphi, K. A., Fredholm, B. B. & Hagberg, H. Effect of propentofylline (HWA 285) on extracellular purines and excitatory amino acids in CA1 of rat hippocampus during transient ischaemia. Br. J. Pharmacol. 100, 814–818 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Dux, E., Fastbom, J., Ungerstedt, U., Rudolphi, K. & Fredholm, B. B. Protective effect of adenosine and a novel xanthine derivative propentofylline on the cell damage after bilateral carotid occlusion in the gerbil hippocampus. Brain Res. 516, 248–256 (1990).

    CAS  PubMed  Google Scholar 

  91. Johansson, B. et al. Hyperalgesia, anxiety, and decreased hypoxic neuroprotection in mice lacking the adenosine A1 receptor. Proc. Natl Acad. Sci. USA 98, 9407–9412 (2001). This is the first report of the generation and charaterization of an Adora1 -knockout mouse with a focus on its effects in the central nervous system.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Eckle, T., Koeppen, M. & Eltzschig, H. K. Role of extracellular adenosine in acute lung injury. Physiology 24, 298–306 (2009).

    CAS  PubMed  Google Scholar 

  93. Ahmad, A. et al. Adenosine A2A receptor is a unique angiogenic target of HIF-2α in pulmonary endothelial cells. Proc. Natl Acad. Sci. USA 106, 10684–10689 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Eckle, T., Kohler, D., Lehmann, R., El Kasmi, K. C. & Eltzschig, H. K. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation 118, 166–175 (2008). These findings reveal a functional role of HIF in mediating cardioprotection during ischaemic preconditioning by enhancing extracelluluar adenosine production and A 2B receptor signalling.

    CAS  PubMed  Google Scholar 

  95. Kong, T., Westerman, K. A., Faigle, M., Eltzschig, H. K. & Colgan, S. P. HIF-dependent induction of adenosine A2B receptor in hypoxia. FASEB J. 20, 2242–2250 (2006).

    CAS  PubMed  Google Scholar 

  96. Eltzschig, H. K. et al. Central role of Sp1-regulated CD39 in hypoxia/ischemia protection. Blood 113, 224–232 (2009). In contrast to NT5E induction by hypoxia, which is mediated by HIF, this study identifies that ENTPD1 induction during conditions of limited oxygen availability is mediated by the transcription factor SP1.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Hart, M. L., Gorzolla, I. C., Schittenhelm, J., Robson, S. C. & Eltzschig, H. K. SP1-dependent induction of CD39 facilitates hepatic ischemic preconditioning. J. Immunol. 184, 4017–4024 (2010).

    CAS  PubMed  Google Scholar 

  98. Kohler, D. et al. CD39/ectonucleoside triphosphate diphosphohydrolase 1 provides myocardial protection during cardiac ischemia/reperfusion injury. Circulation 116, 1784–1794 (2007).

    PubMed  Google Scholar 

  99. Reutershan, J. et al. Adenosine and inflammation: CD39 and CD73 are critical mediators in LPS-induced PMN trafficking into the lungs. FASEB J. 23, 473–482 (2009).

    CAS  PubMed  Google Scholar 

  100. Hart, M. L. et al. Extracellular adenosine production by ecto-5′-nucleotidase protects during murine hepatic ischemic preconditioning. Gastroenterology 135, 1739–1750. e3 (2008).

    CAS  PubMed  Google Scholar 

  101. Csoka, B. et al. A2B adenosine receptors protect against sepsis-induced mortality by dampening excessive inflammation. J. Immunol. 185, 542–550 (2010).

    CAS  PubMed  Google Scholar 

  102. Lappas, C. M., Day, Y. J., Marshall, M. A., Engelhard, V. H. & Linden, J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J. Exp. Med. 203, 2639–2648 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Day, Y. J. et al. A2A adenosine receptors on bone marrow-derived cells protect liver from ischemia-reperfusion injury. J. Immunol. 174, 5040–5046 (2005).

    CAS  PubMed  Google Scholar 

  104. Enjyoji, K. et al. Targeted disruption of CD39/ATP diphosphohydrolase results in disordered hemostasis and thromboregulation. Nature Med. 5, 1010–1017 (1999).

    CAS  PubMed  Google Scholar 

  105. Marcus, A. J. et al. Metabolic control of excessive extracellular nucleotide accumulation by CD39/ecto-nucleotidase-1: implications for ischemic vascular diseases. J. Pharmacol. Exp. Ther. 305, 9–16 (2003).

    CAS  PubMed  Google Scholar 

  106. Eckle, T. et al. Identification of ectonucleotidases CD39 and CD73 in innate protection during acute lung injury. J. Immunol. 178, 8127–8137 (2007).

    CAS  PubMed  Google Scholar 

  107. Grenz, A. et al. Contribution of E-NTPDase1 (CD39) to renal protection from ischemia-reperfusion injury. FASEB J. 21, 2863–2873 (2007).

    CAS  PubMed  Google Scholar 

  108. Grenz, A. et al. Protective role of ecto-5′-nucleotidase (CD73) in renal ischemia. J. Am. Soc. Nephrol. 18, 833–845 (2007).

    CAS  PubMed  Google Scholar 

  109. Hasko, G. et al. Ecto-5′-nucleotidase (CD73) decreases mortality and organ injury in sepsis. J. Immunol. 187, 4256–4267 (2011).

    CAS  PubMed  Google Scholar 

  110. Castillo, C. A., Leon, D., Ruiz, M. A., Albasanz, J. L. & Martin, M. Modulation of adenosine A1 and A2A receptors in C6 glioma cells during hypoxia: involvement of endogenous adenosine. J. Neurochem. 105, 2315–2329 (2008).

    CAS  PubMed  Google Scholar 

  111. Angelatou, F. et al. Upregulation of A1 adenosine receptors in human temporal lobe epilepsy: a quantitative autoradiographic study. Neurosci. Lett. 163, 11–14 (1993).

    CAS  PubMed  Google Scholar 

  112. Biber, K. et al. Interleukin-6 upregulates neuronal adenosine A1 receptors: implications for neuromodulation and neuroprotection. Neuropsychopharmacology 33, 2237–2250 (2008).

    CAS  PubMed  Google Scholar 

  113. Jin, D. et al. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res. 70, 2245–2255 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Sun, X. et al. CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology 139, 1030–1040 (2010).

    CAS  PubMed  Google Scholar 

  115. Ohta, A. et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl Acad. Sci. USA 103, 13132–13137 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Popoli, P. et al. Blockade of striatal adenosine A2A receptor reduces, through a presynaptic mechanism, quinolinic acid-induced excitotoxicity: possible relevance to neuroprotective interventions in neurodegenerative diseases of the striatum. J. Neurosci. 22, 1967–1975 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Dai, S. S. et al. Local glutamate level dictates adenosine A2A receptor regulation of neuroinflammation and traumatic brain injury. J. Neurosci. 30, 5802–5810 (2010). This study demonstrates that a local increase in glutamate levels switches the anti-inflammatory effect mediated by A 2A receptor activation to a pro-inflammatory effect and exacerbates traumatic brain injury, providing a partial explanation for the opposing effects on tissue damage caused by A 2A receptor activation in the brain versus in peripheral tissues.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Li, W. et al. Genetic inactivation of adenosine A2A receptors attenuates acute traumatic brain injury in the mouse cortical impact model. Exp. Neurol. 215, 69–76 (2009).

    CAS  PubMed  Google Scholar 

  119. Chen, J. F. et al. Neuroprotection by caffeine and A2A adenosine receptor inactivation in a model of Parkinson's disease. J. Neurosci. 21, RC143 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Canas, P. M. et al. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by β-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 29, 14741–14751 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Blum, D. et al. A dual role of adenosine A2A receptors in 3-nitropropionic acid-induced striatal lesions: implications for the neuroprotective potential of A2A antagonists. J. Neurosci. 23, 5361–5369 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Chou, S. Y. et al. CGS21680 attenuates symptoms of Huntington's disease in a transgenic mouse model. J. Neurochem. 93, 310–320 (2005).

    CAS  PubMed  Google Scholar 

  123. Li, Y. et al. Mouse spinal cord compression injury is reduced by either activation of the adenosine A2A receptor on bone marrow-derived cells or deletion of the A2A receptor on non-bone marrow-derived cells. Neuroscience 141, 2029–2039 (2006).

    CAS  PubMed  Google Scholar 

  124. Bar-Yehuda, S. et al. The A3 adenosine receptor agonist CF102 induces apoptosis of hepatocellular carcinoma via de-regulation of the Wnt and NF-κB signal transduction pathways. Int. J. Oncol. 33, 287–295 (2008).

    CAS  PubMed  Google Scholar 

  125. Deaglio, S. et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J. Exp. Med. 204, 1257–1265 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Eltzschig, H. K. et al. HIF-1-dependent repression of equilibrative nucleoside transporter (ENT) in hypoxia. J. Exp. Med. 202, 1493–1505 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Loffler, M., Morote-Garcia, J. C., Eltzschig, S. A., Coe, I. R. & Eltzschig, H. K. Physiological roles of vascular nucleoside transporters. Arterioscler. Thromb. Vasc. Biol. 27, 1004–1013 (2007).

    PubMed  Google Scholar 

  128. Morote-Garcia, J. C., Rosenberger, P., Nivillac, N. M., Coe, I. R. & Eltzschig, H. K. Hypoxia-inducible factor-dependent repression of equilibrative nucleoside transporter 2 attenuates mucosal inflammation during intestinal hypoxia. Gastroenterology 136, 607–618 (2009).

    CAS  PubMed  Google Scholar 

  129. Sitkovsky, M. V. Damage control by hypoxia-inhibited AK. Blood 111, 5424–5425 (2008).

    CAS  PubMed  Google Scholar 

  130. Morote-Garcia, J. C., Rosenberger, P., Kuhlicke, J. & Eltzschig, H. K. HIF-1-dependent repression of adenosine kinase attenuates hypoxia-induced vascular leak. Blood 111, 5571–5580 (2008).

    CAS  PubMed  Google Scholar 

  131. Khoa, N. D. et al. Inflammatory cytokines regulate function and expression of adenosine A2A receptors in human monocytic THP-1 cells. J. Immunol. 167, 4026–4032 (2001).

    CAS  PubMed  Google Scholar 

  132. Haschemi, A. et al. Cross-regulation of carbon monoxide and the adenosine A2a receptor in macrophages. J. Immunol. 178, 5921–5929 (2007).

    CAS  PubMed  Google Scholar 

  133. Zhang, Y., Palmblad, J. & Fredholm, B. B. Biphasic effect of ATP on neutrophil functions mediated by P2U and adenosine A2A receptors. Biochem. Pharmacol. 51, 957–965 (1996).

    CAS  PubMed  Google Scholar 

  134. Colgan, S. P. & Eltzschig, H. K. Adenosine and hypoxia-inducible factor signaling in intestinal injury and recovery. Annu. Rev. Physiol. 74, 153–175 (2011).

    PubMed  Google Scholar 

  135. Fredholm, B. B. Adenosine receptors as drug targets. Exp. Cell Res. 316, 1284–1288 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Dhalla, A. K., Shryock, J. C., Shreeniwas, R. & Belardinelli, L. Pharmacology and therapeutic applications of A1 adenosine receptor ligands. Curr. Top. Med. Chem. 3, 369–385 (2003).

    CAS  PubMed  Google Scholar 

  137. Chen, J. F. The adenosine A2A receptor as an attractive target for Parkinson's disease treatment. Drug News Perspect. 16, 597–604 (2003).

    CAS  PubMed  Google Scholar 

  138. Jenner, P. et al. Adenosine, adenosine A2A antagonists, and Parkinson's disease. Parkinsonism Relat. Disord. 15, 406–413 (2009).

    CAS  PubMed  Google Scholar 

  139. Hauser, R. A. et al. Preladenant in patients with Parkinson's disease and motor fluctuations: a phase 2, double-blind, randomised trial. Lancet Neurol. 10, 221–229 (2011).

    CAS  PubMed  Google Scholar 

  140. Guttman, M. Efficacy of istradefylline in Parkinson's diease patients treated with levodopa with motor response complications: results of the KW-6002 US-018 study. Mov. Disord. 21 (Suppl. 15), 585 (2006).

    Google Scholar 

  141. Meissner, W. G. et al. Priorities in Parkinson's disease research. Nature Rev. Drug Discov. 10, 377–393 (2011).

    CAS  Google Scholar 

  142. Vallon, V., Muhlbauer, B. & Osswald, H. Adenosine and kidney function. Physiol. Rev. 86, 901–940 (2006).

    CAS  PubMed  Google Scholar 

  143. Cotter, G. et al. The PROTECT pilot study: a randomized, placebo-controlled, dose-finding study of the adenosine A1 receptor antagonist rolofylline in patients with acute heart failure and renal impairment. J. Card. Fail. 14, 631–640 (2008).

    CAS  PubMed  Google Scholar 

  144. Voors, A. A. et al. Effects of the adenosine A1 receptor antagonist rolofylline on renal function in patients with acute heart failure and renal dysfunction: results from PROTECT (placebo-controlled randomized study of the selective adenosine A1 receptor antagonist rolofylline for patients hospitalized with acute decompensated heart failure and volume overload to assess treatment effect on congestion and renal function). J. Am. Coll. Cardiol. 57, 1899–1907 (2011). This paper describes the design and clinical outcome of the largest clinical Phase III trial (PROTECT) of the A 1 receptor antagonist rolofylline in acute heart failure.

    CAS  PubMed  Google Scholar 

  145. Gessi, S. et al. Adenosine receptor targeting in health and disease. Expert Opin. Investig. Drugs 20, 1591–1609 (2011).

    CAS  PubMed  Google Scholar 

  146. Silverman, M. H. et al. Clinical evidence for utilization of the A3 adenosine receptor as a target to treat rheumatoid arthritis: data from a phase II clinical trial. J. Rheumatol. 35, 41–48 (2008).

    CAS  PubMed  Google Scholar 

  147. Avni, I. et al. Treatment of dry eye syndrome with orally administered CF101: data from a phase 2 clinical trial. Ophthalmology 117, 1287–1293 (2010).

    PubMed  Google Scholar 

  148. Montinaro, A. et al. Adoptive immunotherapy with Cl-IB-MECA-treated CD8+ T cells reduces melanoma growth in mice. PLoS ONE 7, e45401 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Chen, Z. et al. Controlling murine and rat chronic pain through A3 adenosine receptor activation. FASEB J. 26, 1855–1865 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Gessi, S. et al. A3 adenosine receptors in human neutrophils and promyelocytic HL60 cells: a pharmacological and biochemical study. Mol. Pharmacol. 61, 415–424 (2002).

    CAS  PubMed  Google Scholar 

  151. Baraldi, P. G., Preti, D., Borea, P. A. & Varani, K. Medicinal chemistry of A3 adenosine receptor modulators: pharmacological activities and therapeutic implications. J. Med. Chem. 55, 5676–5703 (2012).

    CAS  PubMed  Google Scholar 

  152. Wallace, K. L. & Linden, J. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood 116, 5010–5020 (2010). This paper shows that adenosine signalling, through the activation of A 2A receptors expressed on natural killer T cells, dampens ischaemia–reperfusion injury in models of sickle cell disease.

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Zhang, Y. et al. Detrimental effects of adenosine signaling in sickle cell disease. Nature Med. 17, 79–86 (2011).

    CAS  PubMed  Google Scholar 

  154. Field, J. J., Nathan, D. G. & Linden, J. Targeting iNKT cells for the treatment of sickle cell disease. Clin. Immunol. 140, 177–183 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Gladwin, M. T. Adenosine receptor crossroads in sickle cell disease. Nature Med. 17, 38–40 (2011).

    CAS  PubMed  Google Scholar 

  156. Schmitt, L. I., Sims, R. E., Dale, N. & Haydon, P. G. Wakefulness affects synaptic and network activity by increasing extracellular astrocyte-derived adenosine. J. Neurosci. 32, 4417–4425 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Lovatt, D. et al. Neuronal adenosine release, and not astrocytic ATP release, mediates feedback inhibition of excitatory activity. Proc. Natl Acad. Sci. USA 109, 5913–5914 (2012).

    Google Scholar 

  158. Ramlackhansingh, A. F. et al. Adenosine 2A receptor availability in dyskinetic and nondyskinetic patients with Parkinson disease. Neurology 76, 1811–1816 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Mishina, M. et al. Adenosine A2A receptors measured with CTMSX PET in the striata of Parkinson's disease patients. PLoS ONE 6, e17338 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Cha, J. H. et al. Altered brain neurotransmitter receptors in transgenic mice expressing a portion of an abnormal human huntington disease gene. Proc. Natl Acad. Sci. USA 95, 6480–6485 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Popoli, P. et al. Functions, dysfunctions and possible therapeutic relevance of adenosine A2A receptors in Huntington's disease. Prog. Neurobiol. 81, 331–348 (2007).

    CAS  PubMed  Google Scholar 

  162. Thiel, M. et al. Oxygenation inhibits the physiological tissue-protecting mechanism and thereby exacerbates acute inflammatory lung injury. PLoS Biol. 3, e174 (2005). This is a landmark paper showing that the therapeutic use of high inspired oxygen concentrations (mechanical ventilation with 100% oxygen) — as is frequently necessary for the treatment of patients suffering from acute lung injury to maintain adequate oxygen tissue levels — has detrimental side effects.

    PubMed  PubMed Central  Google Scholar 

  163. Eltzschig, H. K., Rivera-Nieves, J. & Colgan, S. P. Targeting the A2B adenosine receptor during gastrointestinal ischemia and inflammation. Expert Opin. Ther. Targets 13, 1267–1277 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Day, Y. J. et al. Protection from ischemic liver injury by activation of A2A adenosine receptors during reperfusion: inhibition of chemokine induction. Am. J. Physiol. Gastrointest. Liver Physiol. 286, G285–G293 (2004).

    CAS  PubMed  Google Scholar 

  165. Zhou, Y., Schneider, D. J. & Blackburn, M. R. Adenosine signaling and the regulation of chronic lung disease. Pharmacol. Ther. 123, 105–116 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Chunn, J. L. et al. Adenosine-dependent pulmonary fibrosis in adenosine deaminase-deficient mice. J. Immunol. 175, 1937–1946 (2005).

    CAS  PubMed  Google Scholar 

  167. Sun, C. X. et al. Role of A2B receptor signaling in adenosine-dependent pulmonary inflammation and injury. J. Clin. Invest. 116, 1–10 (2006).

    Google Scholar 

  168. Peng, Z. et al. Adenosine signaling contributes to ethanol-induced fatty liver in mice. J. Clin. Invest. 119, 582–594 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Chan, E. S. et al. Adenosine A2A receptors in diffuse dermal fibrosis: pathogenic role in human dermal fibroblasts and in a murine model of scleroderma. Arthritis Rheum. 54, 2632–2642 (2006).

    CAS  PubMed  Google Scholar 

  170. Wen, J. et al. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A2B adenosine receptor signaling. FASEB J. 24, 740–749 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Mi, T. et al. Excess adenosine in murine penile erectile tissues contributes to priapism via A2B adenosine receptor signaling. J. Clin. Invest. 118, 1491–1501 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Volmer, J. B., Thompson, L. F. & Blackburn, M. R. Ecto-5′-nucleotidase (CD73)-mediated adenosine production is tissue protective in a model of bleomycin-induced lung injury. J. Immunol. 176, 4449–4458 (2006).

    CAS  PubMed  Google Scholar 

  173. Zhou, Y. et al. Enhanced airway inflammation and remodeling in adenosine deaminase-deficient mice lacking the A2B adenosine receptor. J. Immunol. 182, 8037–8046 (2009).

    CAS  PubMed  Google Scholar 

  174. Sun, C. X. et al. Role of A2B adenosine receptor signaling in adenosine-dependent pulmonary inflammation and injury. J. Clin. Invest. 116, 2173–2182 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Zhou, Y. et al. Distinct roles for the A2B adenosine receptor in acute and chronic stages of bleomycin-induced lung injury. J. Immunol. 186, 1097–1106 (2011).

    CAS  PubMed  Google Scholar 

  176. Sullivan, G. W., Fang, G., Linden, J. & Scheld, W. M. A2A adenosine receptor activation improves survival in mouse models of endotoxemia and sepsis. J. Infect. Dis. 189, 1897–1904 (2004).

    CAS  PubMed  Google Scholar 

  177. Nemeth, Z. H. et al. Adenosine A2A receptor inactivation increases survival in polymicrobial sepsis. J. Immunol. 176, 5616–5626 (2006).

    CAS  PubMed  Google Scholar 

  178. Csoka, B. et al. A2A adenosine receptors and C/EBPβ are crucially required for IL-10 production by macrophages exposed to Escherichia coli. Blood 110, 2685–2695 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  179. Chan, E. S. et al. Adenosine A2A receptors play a role in the pathogenesis of hepatic cirrhosis. Br. J. Pharmacol. 148, 1144–1155 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  180. Martire, A. et al. Opposite effects of the A2A receptor agonist CGS21680 in the striatum of Huntington's disease versus wild-type mice. Neurosci. Lett. 417, 78–83 (2007).

    CAS  PubMed  Google Scholar 

  181. Obrenovitch, T. P. & Urenjak, J. Altered glutamatergic transmission in neurological disorders: from high extracellular glutamate to excessive synaptic efficacy. Prog. Neurobiol. 51, 39–87 (1997).

    CAS  PubMed  Google Scholar 

  182. Haydon, P. G. & Carmignoto, G. Astrocyte control of synaptic transmission and neurovascular coupling. Physiol. Rev. 86, 1009–1031 (2006).

    CAS  PubMed  Google Scholar 

  183. Jacobson, K. A., von Lubitz, D. K., Daly, J. W. & Fredholm, B. B. Adenosine receptor ligands: differences with acute versus chronic treatment. Trends Pharmacol. Sci. 17, 108–113 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Fredholm, B. B., Battig, K., Holmen, J., Nehlig, A. & Zvartau, E. E. Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol. Rev. 51, 83–133 (1999). This is an expert and comprehensive review on the complex pharmacology of the actions of caffeine, with an emphasis on the psychostimulatory effects of caffeine in the brain.

    CAS  PubMed  Google Scholar 

  185. de Mendonca, A., Sebastiao, A. M. & Ribeiro, J. A. Adenosine: does it have a neuroprotective role after all? Brain Res. Brain Res. Rev. 33, 258–274 (2000).

    CAS  PubMed  Google Scholar 

  186. Von Lubitz, D. K. et al. Chronic administration of selective adenosine A1 receptor agonist or antagonist in cerebral ischemia. Eur. J. Pharmacol. 256, 161–167 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Von Lubitz, D. K. et al. Reduction of postischemic brain damage and memory deficits following treatment with the selective adenosine A1 receptor agonist. Eur. J. Pharmacol. 302, 43–48 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. Burgueno, J. et al. The adenosine A2A receptor interacts with the actin-binding protein α-actinin. J. Biol. Chem. 278, 37545–37552 (2003).

    CAS  PubMed  Google Scholar 

  189. Chern, Y., Lai, H. L., Fong, J. C. & Liang, Y. Multiple mechanisms for desensitization of A2a adenosine receptor-mediated cAMP elevation in rat pheochromocytoma PC12 cells. Mol. Pharmacol. 44, 950–958 (1993).

    CAS  PubMed  Google Scholar 

  190. Halldner, L., Lozza, G., Lindstrom, K. & Fredholm, B. B. Lack of tolerance to motor stimulant effects of a selective adenosine A2A receptor antagonist. Eur. J. Pharmacol. 406, 345–354 (2000).

    CAS  PubMed  Google Scholar 

  191. Pinna, A., Fenu, S. & Morelli, M. Motor stimulant effects of the adenosine A2A receptor antagonist SCH 58261 do not develop tolerance after repeated treatments in 6-hydroxydopamine-lesioned rats. Synapse 39, 233–238 (2001).

    CAS  PubMed  Google Scholar 

  192. Bastia, E. et al. A crucial role for forebrain adenosine A2A receptors in amphetamine sensitization. Neuropsychopharmacology 30, 891–900 (2005).

    CAS  PubMed  Google Scholar 

  193. Yu, L. et al. Adenosine A2A receptor antagonists exert motor and neuroprotective effects by distinct cellular mechanisms. Ann. Neurol. 63, 338–346 (2008).

    CAS  PubMed  Google Scholar 

  194. Shen, H. Y. et al. A critical role of the adenosine A2A receptor in extrastriatal neurons in modulating psychomotor activity as revealed by opposite phenotypes of striatum and forebrain A2A receptor knock-outs. J. Neurosci. 28, 2970–2975 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Scammell, T. E. et al. Focal deletion of the adenosine A1 receptor in adult mice using an adeno-associated viral vector. J. Neurosci. 23, 5762–5770 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Adamantidis, A. R., Zhang, F., Aravanis, A. M., Deisseroth, K. & de Lecea, L. Neural substrates of awakening probed with optogenetic control of hypocretin neurons. Nature 450, 420–424 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Deisseroth, K. et al. Next-generation optical technologies for illuminating genetically targeted brain circuits. J. Neurosci. 26, 10380–10386 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  198. Lerchner, W. et al. Reversible silencing of neuronal excitability in behaving mice by a genetically targeted, ivermectin-gated Cl channel. Neuron 54, 35–49 (2007).

    CAS  PubMed  Google Scholar 

  199. Alexander, G. M. et al. Remote control of neuronal activity in transgenic mice expressing evolved G protein-coupled receptors. Neuron 63, 27–39 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  200. Nawaratne, V. et al. New insights into the function of M4 muscarinic acetylcholine receptors gained using a novel allosteric modulator and a DREADD (designer receptor exclusively activated by a designer drug). Mol. Pharmacol. 74, 1119–1131 (2008).

    CAS  PubMed  Google Scholar 

  201. Aden, U. et al. Aggravated brain damage after hypoxic ischemia in immature adenosine A2A knockout mice. Stroke 34, 739–744 (2003).

    PubMed  Google Scholar 

  202. Burnstock, G., Fredholm, B. B. & Verkhratsky, A. Adenosine and ATP receptors in the brain. Curr. Top. Med. Chem. 11, 973–1011 (2011).

    CAS  PubMed  Google Scholar 

  203. Boison, D., Chen, J. F. & Fredholm, B. B. Adenosine signaling and function in glial cells. Cell Death Differ. 17, 1071–1082 (2010).

    CAS  PubMed  Google Scholar 

  204. Turner, C. P. et al. A1 adenosine receptors mediate hypoxia-induced ventriculomegaly. Proc. Natl Acad. Sci. USA 100, 11718–11722 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  205. Chabre, M. & le Maire, M. Monomeric G-protein-coupled receptor as a functional unit. Biochemistry 44, 9395–9403 (2005).

    CAS  PubMed  Google Scholar 

  206. Whorton, M. R. et al. A monomeric G protein-coupled receptor isolated in a high-density lipoprotein particle efficiently activates its G protein. Proc. Natl Acad. Sci. USA 104, 7682–7687 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Ferre, S. et al. Building a new conceptual framework for receptor heteromers. Nature Chem. Biol. 5, 131–134 (2009).

    CAS  Google Scholar 

  208. Orru, M., Quiroz, C., Guitart, X. & Ferre, S. Pharmacological evidence for different populations of postsynaptic adenosine A2A receptors in the rat striatum. Neuropharmacology 61, 967–974 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  209. Armentero, M. T. et al. Past, present and future of A2A adenosine receptor antagonists in the therapy of Parkinson's disease. Pharmacol. Ther. 132, 280–299 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Moriyama, K. & Sitkovsky, M. V. Adenosine A2A receptor is involved in cell surface expression of A2B receptor. J. Biol. Chem. 285, 39271–39288 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Canals, M. et al. Adenosine A2A–dopamine D2 receptor–receptor heteromerization: qualitative and quantitative assessment by fluorescence and bioluminescence energy transfer. J. Biol. Chem. 278, 46741–46749 (2003).

    CAS  PubMed  Google Scholar 

  212. Ciruela, F. et al. Presynaptic control of striatal glutamatergic neurotransmission by adenosine A1–A2A receptor heteromers. J. Neurosci. 26, 2080–2087 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Ferre, S. et al. Synergistic interaction between adenosine A2A and glutamate mGlu5 receptors: implications for striatal neuronal function. Proc. Natl Acad. Sci. USA 99, 11940–11945 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  214. Pin, J. P. et al. International Union of Basic and Clinical Pharmacology. LXVII. Recommendations for the recognition and nomenclature of G protein-coupled receptor heteromultimers. Pharmacol. Rev. 59, 5–13 (2007).

    CAS  PubMed  Google Scholar 

  215. Yang, J. N. et al. Mice heterozygous for both A1 and A2A adenosine receptor genes show similarities to mice given long-term caffeine. J. Appl. Physiol. 106, 631–639 (2009).

    CAS  PubMed  Google Scholar 

  216. Ascherio, A. et al. Prospective study of caffeine consumption and risk of Parkinson's disease in men and women. Ann. Neurol. 50, 56–63 (2001).

    CAS  PubMed  Google Scholar 

  217. Ross, G. W. et al. Association of coffee and caffeine intake with the risk of Parkinson disease. JAMA 283, 2674–2679 (2000). References 216 and 217 provide strong epidemiological evidence — from the large, longitudinal study by the Honolulu Heart Program and the Health Professional Study — that caffeine consumption is inversely correlated with the risk of developing Parkinson's disease.

    CAS  PubMed  Google Scholar 

  218. Lindsay, J. et al. Risk factors for Alzheimer's disease: a prospective analysis from the Canadian Study of Health and Aging. Am. J. Epidemiol. 156, 445–453 (2002).

    PubMed  Google Scholar 

  219. van Boxtel, M. P., Schmitt, J. A., Bosma, H. & Jolles, J. The effects of habitual caffeine use on cognitive change: a longitudinal perspective. Pharmacol. Biochem. Behav. 75, 921–927 (2003).

    CAS  PubMed  Google Scholar 

  220. Ritchie, K. et al. The neuro-protective effects of caffeine: a prospective population study (the Three City Study). Neurology 69, 536–545 (2007).

    CAS  PubMed  Google Scholar 

  221. Higdon, J. V. & Frei, B. Coffee and health: a review of recent human research. Crit. Rev. Food Sci. Nutr. 46, 101–123 (2006).

    CAS  PubMed  Google Scholar 

  222. Salazar-Martinez, E. et al. Coffee consumption and risk for type 2 diabetes mellitus. Ann. Intern. Med. 140, 1–8 (2004).

    PubMed  Google Scholar 

  223. Schmidt, B. et al. Caffeine therapy for apnea of prematurity. N. Engl. J. Med. 354, 2112–2121 (2006).

    CAS  PubMed  Google Scholar 

  224. Schmidt, B. et al. Long-term effects of caffeine therapy for apnea of prematurity. N. Engl. J. Med. 357, 1893–1902 (2007).

    CAS  PubMed  Google Scholar 

  225. Saaksjarvi, K. et al. Prospective study of coffee consumption and risk of Parkinson's disease. Eur. J. Clin. Nutr. 62, 908–915 (2008).

    CAS  PubMed  Google Scholar 

  226. Powers, K. M. et al. Combined effects of smoking, coffee, and NSAIDs on Parkinson's disease risk. Mov. Disord. 23, 88–95 (2008).

    PubMed  Google Scholar 

  227. Fernandez, H. H. et al. Istradefylline as monotherapy for Parkinson disease: results of the 6002-US-051 trial. Parkinsonism Relat. Disord. 16, 16–20 (2010).

    CAS  PubMed  Google Scholar 

  228. Chen, J. F. et al. What knock-out animals tell us about the effects of caffeine. J. Alzheimers Dis. 20 (Suppl. 1), 17–24 (2010).

    Google Scholar 

  229. Tejani, F. H., Thompson, R. C., Iskandrian, A. E., McNutt, B. E. & Franks, B. Effect of caffeine on SPECT myocardial perfusion imaging during regadenoson pharmacologic stress: rationale and design of a prospective, randomized, multicenter study. J. Nucl. Cardiol. 18, 73–81 (2011).

    PubMed  Google Scholar 

  230. Cabalag, M. S. et al. Recent caffeine ingestion reduces adenosine efficacy in the treatment of paroxysmal supraventricular tachycardia. Acad. Emerg. Med. 17, 44–49 (2010).

    PubMed  Google Scholar 

  231. Pourcher, E. et al. Istradefylline for Parkinson's disease patients experiencing motor fluctuations: results of the KW-6002-US-018 study. Parkinsonism Relat. Disord. 18, 178–184 (2012).

    PubMed  Google Scholar 

  232. Factor, S. et al. A long-term study of istradefylline in subjects with fluctuating Parkinson's disease. Parkinsonism Relat. Disord. 16, 423–426 (2010).

    PubMed  Google Scholar 

  233. Hauser, R. A. et al. Study of istradefylline in patients with Parkinson's disease on levodopa with motor fluctuations. Mov. Disord. 23, 2177–2185 (2008).

    PubMed  Google Scholar 

  234. Rao, N. et al. A study of the pharmacokinetic interaction of istradefylline, a novel therapeutic for Parkinson's disease, and atorvastatin. J. Clin. Pharmacol. 48, 1092–1098 (2008).

    CAS  PubMed  Google Scholar 

  235. Stacy, M. et al. A 12-week, placebo-controlled study (6002-US-006) of istradefylline in Parkinson disease. Neurology 70, 2233–2240 (2008).

    CAS  PubMed  Google Scholar 

  236. LeWitt, P. A. et al. Adenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson's disease: a double-blind, randomized, multicenter clinical trial (6002-US-005). Ann. Neurol. 63, 295–302 (2008). This paper describes the design and clinical outcome of one of six of the randomized, placebo-controlled, Phase III trials of the A 2A receptor antagonist istradefylline in patients with advanced Parkinson's disease.

    CAS  PubMed  Google Scholar 

  237. Robak, T. & Robak, P. Purine nucleoside analogs in the treatment of rarer chronic lymphoid leukemias. Curr. Pharm. Des. 18, 3373–3388 (2012).

    CAS  PubMed  Google Scholar 

  238. Bruns, R. F. & Fergus, J. H. Allosteric enhancement of adenosine A1 receptor binding and function by 2-amino-3-benzoylthiophenes. Mol. Pharmacol. 38, 939–949 (1990).

    CAS  PubMed  Google Scholar 

  239. Katritch, V., Cherezov, V. & Stevens, R. C. Diversity and modularity of G protein-coupled receptor structures. Trends Pharmacol. Sci. 33, 17–27 (2012).

    CAS  PubMed  Google Scholar 

  240. Chen, J. F. et al. 8-(3-chlorostyryl)caffeine may attenuate MPTP neurotoxicity through dual actions of monoamine oxidase inhibition and A2A receptor antagonism. J. Biol. Chem. 277, 36040–36044 (2002).

    CAS  PubMed  Google Scholar 

  241. Huang, N. K. et al. A new drug design targeting the adenosinergic system for Huntington's disease. PLoS ONE 6, e20934 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. El-Tayeb, A. et al. Nucleoside-5′-monophosphates as prodrugs of adenosine A2A receptor agonists activated by ecto-5′-nucleotidase. J. Med. Chem. 52, 7669–7677 (2009).

    CAS  PubMed  Google Scholar 

  243. Flogel, U. et al. Selective activation of adenosine A2A receptors on immune cells by a CD73-dependent prodrug suppresses joint inflammation in experimental rheumatoid arthritis. Sci. Transl. Med. 4, 146ra108 (2012).

    PubMed  Google Scholar 

  244. Fredholm, B. B., Chern, Y., Franco, R. & Sitkovsky, M. Aspects of the general biology of adenosine A2A signaling. Prog. Neurobiol. 83, 263–276 (2007).

    CAS  PubMed  Google Scholar 

  245. Allen, J. A. et al. Discovery of β-arrestin-biased dopamine D2 ligands for probing signal transduction pathways essential for antipsychotic efficacy. Proc. Natl Acad. Sci. USA 108, 18488–18493 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  246. Langemeijer, E. V., Verzijl, D., Dekker, S. J. & Ijzerman, A. P. Functional selectivity of adenosine A1 receptor ligands? Purinergic Signal. 7, 171–192 (2012).

    Google Scholar 

  247. Lebon, G. et al. Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature 474, 521–525 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Xu, F. et al. Structure of an agonist-bound human A2A adenosine receptor. Science 332, 322–327 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  249. Jaakola, V. P. et al. The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science 322, 1211–1217 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Liu, W. et al. Structural basis for allosteric regulation of GPCRs by sodium ions. Science 337, 232–236 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  251. Kobilka, B. K. Structural insights into adrenergic receptor function and pharmacology. Trends Pharmacol. Sci. 32, 213–218 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  252. Hocher, B. Adenosine A1 receptor antagonists in clinical research and development. Kidney Int. 78, 438–445 (2010).

    CAS  PubMed  Google Scholar 

  253. Bennet, D. W. & Drury, A. N. Further observations relating to the physiological activity of adenine compounds. J. Physiol. 72, 288–320 (1931).

    CAS  PubMed  PubMed Central  Google Scholar 

  254. Drury, A. N. & Szent-Gyorgyi, A. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J. Physiol. 68, 213–237 (1929).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Stoner, H. B. & Green, H. N. Experimental limb ischaemia in man with especial reference to the role of adenosine triphosphate. Clin. Sci. 5, 159–175 (1945).

    CAS  PubMed  Google Scholar 

  256. Massion, W. H. Value of high energy compounds in the treatment of shock. Am. J. Surg. 110, 342–347 (1965).

    CAS  PubMed  Google Scholar 

  257. Yang, D. et al. A new role for the A2b adenosine receptor in regulating platelet function. J. Thromb. Haemost. 8, 817–827 (2010).

    PubMed  Google Scholar 

  258. Berne, R. M. Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. Am. J. Physiol. 204, 317–322 (1963).

    CAS  PubMed  Google Scholar 

  259. Gerlach, E., Deuticke, B. & Dreisbach, R. H. Der Nucleotid-Abbau im Herzmuskel bei Sauerstoffmangel und seine mögliche Bedeutung für die Coronardurchblutung. Naturwissenschaften 50, 228–229 (in German) (1963).

    CAS  Google Scholar 

  260. Mustafa, S. J., Berne, R. M. & Rubio, R. Adenosine metabolism in cultured chick-embryo heart cells. Am. J. Physiol. 228, 1474–1478 (1975).

    CAS  PubMed  Google Scholar 

  261. Duncker, D. J. & Merkus, D. Exercise hyperaemia in the heart: the search for the dilator mechanism. J. Physiol. 583, 847–854 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Lankford, A. R. et al. Effect of modulating cardiac A1 adenosine receptor expression on protection with ischemic preconditioning. Am. J. Physiol. Heart Circ. Physiol. 290, H1469–H1473 (2006).

    CAS  PubMed  Google Scholar 

  263. Yang, A., Palmer, A. A. & de Wit, H. Genetics of caffeine consumption and responses to caffeine. Psychopharmacology 211, 245–257 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Retey, J. V. et al. A functional genetic variation of adenosine deaminase affects the duration and intensity of deep sleep in humans. Proc. Natl Acad. Sci. USA 102, 15676–15681 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  265. Landolt, H. P. Sleep homeostasis: a role for adenosine in humans? Biochem. Pharmacol. 75, 2070–2079 (2008).

    CAS  PubMed  Google Scholar 

  266. Retey, J. V. et al. A genetic variation in the adenosine A2A receptor gene (ADORA2A) contributes to individual sensitivity to caffeine effects on sleep. Clin. Pharmacol. Ther. 81, 692–698 (2007).

    CAS  PubMed  Google Scholar 

  267. Childs, E. et al. Association between ADORA2A and DRD2 polymorphisms and caffeine-induced anxiety. Neuropsychopharmacology 33, 2791–2800 (2008).

    CAS  PubMed  Google Scholar 

  268. Domschke, K. et al. ADORA2A gene variation, caffeine, and emotional processing: a multi-level interaction on startle reflex. Neuropsychopharmacology 37, 759–769 (2012).

    CAS  PubMed  Google Scholar 

  269. Alsene, K., Deckert, J., Sand, P. & de Wit, H. Association between A2a receptor gene polymorphisms and caffeine-induced anxiety. Neuropsychopharmacology 28, 1694–1702 (2003).

    CAS  PubMed  Google Scholar 

  270. Taherzadeh-Fard, E., Saft, C., Wieczorek, S., Epplen, J. T. & Arning, L. Age at onset in Huntington's disease: replication study on the associations of ADORA2A, HAP1 and OGG1. Neurogenetics 11, 435–439 (2010).

    CAS  PubMed  Google Scholar 

  271. Popat, R. A. et al. Coffee, ADORA2A, and CYP1A2: the caffeine connection in Parkinson's disease. Eur. J. Neurol. 18, 756–765 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  272. Amin, N. et al. Genome-wide association analysis of coffee drinking suggests association with CYP1A1/CYP1A2 and NRCAM. Mol. Psychiatry 17, 1116–1129 (2011).

    PubMed  PubMed Central  Google Scholar 

  273. Hamza, T. H. et al. Genome-wide gene-environment study identifies glutamate receptor gene GRIN2A as a Parkinson's disease modifier gene via interaction with coffee. PLoS Genet. 7, e1002237 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Tantry, U. S. et al. First analysis of the relation between CYP2C19 genotype and pharmacodynamics in patients treated with ticagrelor versus clopidogrel: the ONSET/OFFSET and RESPOND genotype studies. Circ. Cardiovasc. Genet. 3, 556–566 (2010).

    CAS  PubMed  Google Scholar 

  275. Elzein, E. & Zablocki, J. A1 adenosine receptor agonists and their potential therapeutic applications. Expert Opin. Investig. Drugs 17, 1901–1910 (2008).

    CAS  PubMed  Google Scholar 

  276. Pinna, A. Novel investigational adenosine A2A receptor antagonists for Parkinson's disease. Expert Opin. Investig. Drugs 18, 1619–1631 (2009).

    CAS  PubMed  Google Scholar 

  277. Brown, R. et al. Abolished tubuloglomerular feedback and increased plasma renin in adenosine A1 receptor-deficient mice. Am. J. Physiol. Regul. Integr. Comp. Physiol. 281, R1362–R1367 (2001).

    CAS  PubMed  Google Scholar 

  278. Tawfik, H. E., Schnermann, J., Oldenburg, P. J. & Mustafa, S. J. Role of A1 adenosine receptors in regulation of vascular tone. Am. J. Physiol. Heart Circ. Physiol. 288, H1411–H1416 (2005).

    CAS  PubMed  Google Scholar 

  279. Wang, Y., Yang, J. N., Arner, A., Boels, P. J. & Fredholm, B. B. Adenosine A1 receptors and vascular reactivity. Acta Physiol. 199, 211–220 (2010).

    CAS  Google Scholar 

  280. Hua, X. et al. Involvement of A1 adenosine receptors and neural pathways in adenosine-induced bronchoconstriction in mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 293, L25–L32 (2007).

    CAS  PubMed  Google Scholar 

  281. Johansson, S. M. et al. A1 receptor deficiency causes increased insulin and glucagon secretion in mice. Biochem. Pharmacol. 74, 1628–1635 (2007).

    CAS  PubMed  Google Scholar 

  282. Yang, J. N. et al. Sex differences in mouse heart rate and body temperature and in their regulation by adenosine A1 receptors. Acta Physiol. 190, 63–75 (2007).

    CAS  Google Scholar 

  283. Kara, F. M. et al. Adenosine A1 receptors regulate bone resorption in mice: adenosine A1 receptor blockade or deletion increases bone density and prevents ovariectomy-induced bone loss in adenosine A1 receptor-knockout mice. Arthritis Rheum. 62, 534–541 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  284. Stenberg, D. et al. Sleep and its homeostatic regulation in mice lacking the adenosine A1 receptor. J. Sleep Res. 12, 283–290 (2003).

    PubMed  Google Scholar 

  285. Oishi, Y., Huang, Z. L., Fredholm, B. B., Urade, Y. & Hayaishi, O. Adenosine in the tuberomammillary nucleus inhibits the histaminergic system via A1 receptors and promotes non-rapid eye movement sleep. Proc. Natl Acad. Sci. USA 105, 19992–19997 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  286. Wu, W. P. et al. Increased nociceptive response in mice lacking the adenosine A1 receptor. Pain 113, 395–404 (2005).

    CAS  PubMed  Google Scholar 

  287. Schulte, G. et al. Adenosine A receptors are necessary for protection of the murine heart by remote, delayed adaptation to ischaemia. Acta Physiol. Scand. 182, 133–143 (2004).

    CAS  PubMed  Google Scholar 

  288. Lappas, C. M., Rieger, J. M. & Linden, J. A2A adenosine receptor induction inhibits IFN-γ production in murine CD4+ T cells. J. Immunol. 174, 1073–1080 (2005).

    CAS  PubMed  Google Scholar 

  289. Carman, A. J., Mills, J. H., Krenz, A., Kim, D. G. & Bynoe, M. S. Adenosine receptor signaling modulates permeability of the blood–brain barrier. J. Neurosci. 31, 13272–13280 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  290. Chen, X., Lan, X., Roche, I., Liu, R. & Geiger, J. D. Caffeine protects against MPTP-induced blood–brain barrier dysfunction in mouse striatum. J. Neurochem. 107, 1147–1157 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  291. Chen, X., Gawryluk, J. W., Wagener, J. F., Ghribi, O. & Geiger, J. D. Caffeine blocks disruption of blood brain barrier in a rabbit model of Alzheimer's disease. J. Neuroinflamm. 5, 12 (2008).

    Google Scholar 

  292. Morrison, R. R., Talukder, M. A., Ledent, C. & Mustafa, S. J. Cardiac effects of adenosine in A2A receptor knockout hearts: uncovering A2B receptors. Am. J. Physiol. Heart Circ. Physiol. 282, H437–H444 (2002).

    CAS  PubMed  Google Scholar 

  293. Montesinos, M. C. et al. Adenosine promotes wound healing and mediates angiogenesis in response to tissue injury via occupancy of A2A receptors. Am. J. Pathol. 160, 2009–2018 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  294. Tilley, S. L., Wagoner, V. A., Salvatore, C. A., Jacobson, M. A. & Koller, B. H. Adenosine and inosine increase cutaneous vasopermeability by activating A3 receptors on mast cells. J. Clin. Invest. 105, 361–367 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  295. Hua, X. et al. Adenosine induces airway hyperresponsiveness through activation of A3 receptors on mast cells. J. Allergy Clin. Immunol. 122, 107–113. e7 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  296. Li, L. et al. Dendritic cells tolerized with adenosine A2AR agonist attenuate acute kidney injury. J. Clin. Invest. 122, 3931–3942 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  297. Field, J. J. et al. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood 1 Feb 2013 (doi:10.1182/blood-2012-11-465963).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank E. Augusto and M. Marco for help with the figure. This research was supported by: a grant from the US National Institutes of Health (NIH/NS 041083–07) and the Cogan Foundation, and grants from the Jerry McDonald Huntington's Disease Research Fund to J.C.; grants from the US National Institutes of Health (NIH), the US National Heart, Lung, and Blood Institute (NHLBI) and the US National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (R01-HL0921, R01-DK083385 and R01-HL098294) and a grant from the Crohn's & Colitis Foundation of America (CCFA) to H.K.E.; and a grant from the Swedish Science Research Council (grant no. 2553) to B.B.F.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bertil B. Fredholm.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary information S1 (table)

17 clinical trials with istradeffyline, KW6002, in advanced Parkinson's disease patients (PDF 252 kb)

Related links

Related links

FURTHER INFORMATION

Jiang-Fan Chen's Laboratory

Holger K. Eltzschig's laboratory

Bertil B. Fredholm's group

ClinicalTrials.gov website

Glossary

Methylxanthines

Purine derivatives with a common xanthine core molecule and methyl group attached in various combinations to nitrogens. The most common methylxanthines include caffeine, theophylline, theobromine and paraxanthine.

G protein-coupled receptor

(GPCR). A cell membrane protein characterized by a seven-transmembrane structure, which is coupled to trimeric G proteins; GPCRs elicit diverse sets of signalling and biological functions.

Ectonucleoside triphosphate diphosphohydrolase 1

(ENTPD1; also known as CD39). A membrane-bound enzyme with enzymatic activity in the extracellular space; ENTPD1 catalyses the conversion of extracellular ATP and/or ADP to AMP — an important step in generating extracellular adenosine.

Ecto-5′-nucleotidase

(NT5E; also known as CD73). A membrane-bound enzyme with enzymatic activity in the extracellular space; NT5E catalyses the conversion of extracellular AMP to adenosine, thereby functioning as a pacemaker enzyme for generating extracellular ATP-derived adenosine.

Acute heart failure

A gradual or rapid change in the signs and symptoms of heart failure. Many pathological factors, including worsening renal function, persistent neurohormonal activation and progressive deterioration in myocardial function, all contribute to the development of acute heart failure.

Parkinson's disease

A neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra pars compacta of the midbrain and the accumulation of proteinaceous intracellular inclusions (Lewy bodies), leading to decreased dopamine levels in the striatum and cardinal motor symptoms.

Ischaemic preconditioning

A phenomenon in which repeated short periods of exposure to sublethal ischaemia induce tolerance and protection against subsequent lethal ischaemic injury.

Adenosine kinase

An enzyme that converts intracellular adenosine to AMP, which is critically important in setting the basal adenosine level. Hypoxia is associated with transcriptional repression of adenosine kinase, thereby resulting in increased intracellular adenosine levels and enhanced extracellular adenosine signalling.

Hypoxia-inducible factor

(HIF). Key transcription factor for hypoxia-induced responses that are critical in adapting hypoxic or ischaemic tissues to conditions of limited oxygen availability.

Bleomycin-induced lung injury

A widely used antitumour agent causing single- and double-stranded breaks in cellular DNA, leading to genomic instability of damaged cells. Bleomycin induces apoptosis and increases the production of reactive oxygen species, resulting in oxidative stress and pulmonary fibrosis.

Adenosine deaminase

An enzyme that converts adenosine to inosine (and deoxyadenosine to deoxyinosine). Lack of adenosine deaminase causes immune deficiency.

Oligomerization

G protein-coupled receptors (GPCRs) can exist in a monomeric state or form dimeric, multimeric or oligomeric structures. Hetero-oligomerization of GPCRs, involving several gene products, can potentially lead to an altered biological response repertoire.

Allosteric enhancers

Allosteric modulators do not have any activity by themselves, but as they bind to the allosteric site (which is distinct from the primary ligand binding orthosteric site) they can alter the receptor confirmation by an orthosteric ligand in such a way that the response to it is increased.

Equilibrative nucleoside transporter 1

(ENT1). A channel located in the cell membrane; along with ENT2, ENT1 speeds up the bi-directional transport of adenosine across the cell membrane along its gradient.

Functional selectivity

Also known as biased signalling; an emerging concept of G protein-coupled receptor (GPCR) function. For example, some GPCR ligands preferentially activate signals via β-arrestins, others via G proteins.

β-arrestin

An adaptor protein that was initially recognized as a negative regulator of G protein signalling but is now recognized to be a multifunctional adaptor that can not only mediate G protein-coupled receptor (GPCR) internalization and desensitization but also produce distinct intracellular signalling and hence functional consequences.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Chen, JF., Eltzschig, H. & Fredholm, B. Adenosine receptors as drug targets — what are the challenges?. Nat Rev Drug Discov 12, 265–286 (2013). https://doi.org/10.1038/nrd3955

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrd3955

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research