Elsevier

Biochemical Pharmacology

Volume 99, 1 January 2016, Pages 101-112
Biochemical Pharmacology

Quantification of adenosine A1 receptor biased agonism: Implications for drug discovery

https://doi.org/10.1016/j.bcp.2015.11.013Get rights and content

Abstract

Adenosine A1 receptor (A1AR) stimulation is a powerful protective mechanism in cerebral and cardiac ischemia⿿reperfusion injury. Despite this, therapeutic targeting of the A1AR for the treatment of ischemia⿿reperfusion injury has been largely unsuccessful, as high concentrations of prototypical A1AR agonists impart significant hemodynamic effects, particularly pronounced bradycardia, atrioventricular block and hypotension. Exploiting the phenomenon of biased agonism to develop ligands that promote A1AR cytoprotection in the absence of adverse hemodynamic effects remains a relatively unexplored, but exciting, approach to overcome current limitations. In native systems, the atypical A1AR agonists VCP746 and capadenoson retain cytoprotective signaling in the absence of bradycardia, a phenomenon suggestive of biased agonism. The current study used pharmacological inhibitors to investigate A1AR mediated cytoprotective signal transduction in a CHO FlpIn cell background, thus identifying candidate pathways for quantitative bias profiling, including cAMP, extracellular signal-regulated kinases 1 and 2 and Akt1/2/3. Subsequently, effects on cell survival and the bias profile of VCP746 and capadenoson were determined and compared to that of the prototypical A1AR agonists, NECA, R-PIA, MeCCPA and CPA. We found that prototypical agonists do not display significant bias for any of the pathways assessed. In contrast, VCP746 and capadenoson show significant bias away from calcium mobilization relative to all pathways tested. These studies demonstrate that quantitative ⿿fingerprinting⿿ of biased agonism within a model system can enable ligands to be clustered by their bias profile, which in turn may be predictive of preferential physiologically relevant in vivo pharmacology.

Introduction

Adenosine is an ubiquitous purine nucleoside that is involved in a range of physiological functions [1]. The nucleoside achieves this functionality by binding to one of the four adenosine receptors (ARs), namely, the A1AR, A2AAR, A2BAR, and A3AR, all of which are G protein-coupled receptors (GPCRs) [2]. The A1AR and A3AR preferentially couple to Gi/o proteins, whilst the A2AAR and A2BAR preferentially couple to Gs proteins [2], [3].

Under conditions of cellular stress, such as inflammation and hypoxia, adenosine can reach micromolar concentrations in the pericellular environment, acting via adenosine receptors to promote tissue protection and repair [4], [5]. This is particularly pertinent in ischemia⿿reperfusion injury (IRI), where lack of blood supply coupled with pro-inflammatory reperfusion, leads to serious organ and tissue damage [6], [7]. Activation of A1ARs can elicit powerful cytoprotection in myocardial or cerebral IRI [8], [9], [10], likely involving signaling downstream of the mitogen activated protein kinases (MAPK), the protein kinase B (Akt/PKB) family of serine/threonine protein kinases and a number of protein kinase C (PKC) isoforms [11], [12], [13], [14], [15], [16], [17], [18], [19].

Whilst A1AR agonism promotes profound cardioprotection, clinical trials of A1AR agonists have shown only limited success, as the trialed dosing was suboptimal due to undesirable hemodynamic side effects, including bradycardia [20]. Under these conditions, A1AR-mediated bradycardia largely occurs via an increase in the current of inward rectifying potassium channels in the pacemaker cells of the heart [21], [22], [23]. Additionally, in the presence of increased sympathetic tone, A1AR activation can reduce heart rate via inhibition of adenylate cyclase activation and subsequent effects on hyperpolarization-activated cyclic nucleotide-gated channels [22]. Consequently, the difficulty of A1AR-targeted medicines is that both the therapeutic effect, cytoprotection, and the clinically limiting effect, bradycardia, are mediated by the same target.

One approach that may allow for the separation of on-target beneficial effects from adverse effects is exploitation of biased agonism. This is a phenomenon whereby different ligands, acting at the same GPCR and in the same cellular background, can promote distinct receptor conformations that bias the signal either towards or away from a subset of the cellular pathways normally mediated by activation of the receptor [24]; in the case of the A1AR, this could represent signaling promoting cytoprotection in the absence of signaling producing on-target bradycardia. Importantly, we recently designed a novel bitopic (hybrid orthosteric/allosteric) A1AR ligand, 4-(5-amino-4-benzoyl-3-(3-(trifluoromethyl)phenyl)thiophen-2-yl)-N-(6-(9-((2R,3R,4S,5R)-3,4-dihydroxy-5 (hydroxymethyl)tetrahydrofuran-2-yl)-9H-purin-6-ylamino)hexyl)benzamide (VCP746), that displayed biased agonism in a heterologous expression system, as well as reducing isolated neonatal rat cardiomyocyte death due to simulated ischemia while having no effect on isolated rat atrial rate [25]. This finding provided the first proof of concept that potentially therapeutically useful biased agonism can be observed at the A1AR, but also raises a number of important questions. For instance, is the bias promoted by VCP746 due to its atypical extended structure, given that it is a hybrid molecule composed of orthosteric and allosteric pharmacophores? Can other atypical A1AR ligands promote similar or distinct patterns of bias? Is it possible to develop a ⿿signaling fingerprint⿿ in a recombinant model system that can be used to group A1AR ligands with regards to their propensity to engender physiologically relevant bias in a native cellular system?

The aim of the current study was to begin addressing these questions. We first extended our preliminary determination of VCP746 bias by identifying a broad range of signaling pathways linked to A1AR activation in a heterologous system, that are potentially relevant to cytoprotection, and can thus be used to generate a ⿿bias fingerprint⿿ as a surrogate predictor of physiologically relevant bias. We subsequently compared the bias profile of VCP746 to that of the prototypical A1AR agonists 5⿲-N-ethylcarboxamidoadenosine (NECA), (R-)-N6-phenylisopropyladenosine (R-PIA), 2-chloro-N-cyclopentyl-2⿲-methyl adenosine (MeCCPA) and N6-cyclopentyladenosine (CPA), as well as another atypical agonist, capadenoson (Fig. 1). The latter was chosen because it is also an extended non-nucleoside molecule that mediates cardioprotection in the absence of adverse hemodynamic effects [26], [27]. We find that both VCP746 and capadenoson display a distinct pattern of bias, whereas canonical A1AR ligands do not, providing further support for the role of quantitative bias profiling in model systems to better inform potential drug candidate selection.

Section snippets

Materials

Fluo4⿢, Dulbecco⿿s modified Eagle medium (DMEM) and penicillin⿿streptomycin were purchased from Invitrogen (Carlsbad, CA). Adenosine deaminase (ADA) and hygromycin-B were purchased from Roche (Basel, Switzerland). 2-[1-(3-Dimethylaminopropyl)indol-3-yl]-3-(indol-3-yl)maleimide (GF109203X) was purchased from Tocris Bioscience (Ellisville, MO). Fetal bovine serum (FBS) was purchased from ThermoTrace (Melbourne, Australia). AlphaScreen⿢ SureFire⿢ extracellular signal-regulated kinases 1 and 2 (ERK

A1AR activation promotes cell survival in A1-FlpIn-CHO cells

Exposure of A1-FlpIn-CHO cells to serum starvation conditions was used to promote a 40⿿50% reduction in cell survival after 24 h. The A1AR prototypical agonists, NECA, MeCCPA, R-PIA, and CPA, as well as the A1AR atypical agonists, VCP746 and capadenoson, mediated a concentration-dependent increase in A1-FlpIn-CHO cell survival, restoring total cell survival to approximately 90% (Fig. 2A and B).

A1AR-mediated cell survival is Gi/o dependent

Concomitant treatment of A1-FlpIn-CHO cells with A1AR agonists and pharmacological inhibitors was used

Discussion

This study illustrates the utility of developing quantitative biased agonist ⿿fingerprints⿿ in a heterologous system that can be used to cluster molecules in terms of their propensity to mediate distinct physiological effects. Specifically, the novel bitopic A1AR agonist, VCP746, and the non-nucleoside A1AR agonist, capadenoson, were significantly biased away from A1AR-mediated calcium mobilization relative to the reference agonist, NECA, whilst the prototypical agonists MeCCPA, R-PIA and CPA

Acknowledgements

The authors thank Drs. Michael Crouch and Ron Osmond for providing the ERK 1/2 phosphorylation assay kit. This work was funded by the National Health and Medical Research Council of Australia (NHMRC) [Program Grant APP1055134 & Project Grant APP1084487]. LTM is a recipient of an Australian Research Council (ARC) Discovery Early Career Researcher Award (DECRA), AC and PMS are Principal Research Fellows of the NHMRC, KJG is a NHMRC Overseas Biomedical Postdoctoral Training Fellow.

References (52)

  • J. Corbisier et al.

    Biased signaling at chemokine receptors

    J. Biol. Chem.

    (2015)
  • K.J. Gregory et al.

    Stimulus bias provides evidence for conformational constraints in the structure of a G protein-coupled receptor

    J. Biol. Chem.

    (2012)
  • K.A. Jacobson et al.

    Adenosine receptors as therapeutic targets

    Nat. Rev. Drug Discov.

    (2006)
  • B.B. Fredholm et al.

    International union of pharmacology. XXV. Nomenclature and classification of adenosine receptors

    Pharmacol. Rev.

    (2001)
  • B.B. Fredholm

    Adenosine, an endogenous distress signal, modulates tissue damage and repair

    Cell Death Differ.

    (2007)
  • G.J. Harrison et al.

    Extracellular adenosine levels and cellular energy metabolism in ischemically preconditioned rat heart

    Cardiovasc. Res.

    (1998)
  • S. Latini et al.

    Adenosine in the central nervous system: release mechanisms and extracellular concentrations

    J. Neurochem.

    (2001)
  • V.E. Laubach et al.

    Targeting of adenosine receptors in ischemia⿿reperfusion injury

    Expert Opin. Ther. Targets

    (2011)
  • R.A. Cunha

    Neuroprotection by adenosine in the brain: from A1 receptor activation to A2A receptor blockade

    Purinergic Signal.

    (2005)
  • R.D. Lasley et al.

    Delayed adenosine A1 receptor preconditioning in rat myocardium is MAPK dependent but iNOS independent

    Am. J. Physiol. Heart Circ. Physiol.

    (2005)
  • E.A. Reid et al.

    In vivo adenosine receptor preconditioning reduces myocardial infarct size via subcellular ERK signaling

    Am. J. Physiol. Heart Circ. Physiol.

    (2005)
  • A. Dana et al.

    Adenosine A1 receptor induced delayed preconditioning in rabbits: induction of p38 mitogen-activated protein kinase activation and Hsp27 phosphorylation via a tyrosine kinase- and protein kinase C-dependent mechanism

    Circ. Res.

    (2000)
  • N.V. Solenkova et al.

    Endogenous adenosine protects preconditioned heart during early minutes of reperfusion by activating Akt

    Am. J. Physiol. Heart Circ. Physiol.

    (2006)
  • G. Simonis et al.

    Dual mechanism of autoregulation of protein kinase C in myocardial ischemia

    Mol. Cell. Biochem.

    (2006)
  • K. Mubagwa et al.

    Adenosine, adenosine receptors and myocardial protection: an updated overview

    Cardiovasc. Res.

    (2001)
  • R. Ciccarelli et al.

    Molecular signalling mediating the protective effect of A1 adenosine and mGlu3 metabotropic glutamate receptor activation against apoptosis by oxygen/glucose deprivation in cultured astrocytes

    Mol. Pharmacol.

    (2007)
  • Cited by (52)

    • Biased agonism at adenosine receptors

      2021, Cellular Signalling
      Citation Excerpt :

      In light of this understanding, the bias profiles of capadenoson and neladenoson have been assessed in vitro. Remarkably, similar to VCP746, both capadenoson and neladenoson were biased away from calcium mobilisation relative to the reference agonist NECA, suggesting that these in vitro signalling fingerprints may indeed have the capacity to cluster compounds according to their effects on heart rate in vivo [83–86]. Interestingly, when compared to capadenoson and VCP746, neladenoson displayed additional, significant bias away from ERK1/2 and Akt phosphorylation [86].

    • Bifunctional Tools to Study Adenosine Receptors

      2023, Topics in Medicinal Chemistry
    View all citing articles on Scopus
    View full text