Elsevier

Biochemical Pharmacology

Volume 151, May 2018, Pages 201-213
Biochemical Pharmacology

On the G protein-coupling selectivity of the native A2B adenosine receptor

https://doi.org/10.1016/j.bcp.2017.12.003Get rights and content

Abstract

A2B adenosine receptor (A2BAR) activation induces Gs-dependent cyclic AMP accumulation. However, A2BAR G protein-coupling to other signaling events, e.g. ERK1/2 and calcium, is not well documented. We explored Gi, Gq/11 and Gs coupling in 1321 N1 astrocytoma, HEK293, and T24 bladder cancer cells endogenously expressing human A2BAR, using NECA or nonnucleoside BAY60-6583 as agonist, selective Gi, Gs and Gq/11 blockers, and CRISPR/Cas9-based Gq- and Gs-null HEK293 cells. In HEK293 cells, A2BAR-mediated ERK1/2 activity occurred via both Gi and Gs, but not Gq/11. However, HEK293 cell calcium mobilization was completely blocked by Gq/11 inhibitor UBO-QIC and by Gq/11 knockout. In T24 cells, Gi was solely responsible for A2BAR-mediated ERK1/2 stimulation, and Gs suppressed ERK1/2 activity. A2BAR-mediated intracellular calcium mobilization in T24 cells was mainly via Gi, although Gs may also play a role, but Gq/11 is not involved. In 1321 N1 astrocytoma cells A2BAR activation suppressed rather than stimulated ERK1/2 activity. The ERK1/2 activity decrease was reversed by Gs downregulation using cholera toxin, but potentiated by Gi inhibitor pertussis toxin, and UBO-QIC had no effect. EPACs played an important role in A2BAR-mediated ERK1/2 signaling in all three cells. Thus, A2BAR may: couple to the same downstream pathway via different G proteins in different cell types; activate different downstream events via different G proteins in the same cell type; activate Gi and Gs, which have opposing or synergistic roles in different cell types/signaling pathways. The findings, relevant to drug discovery, address some reported controversial roles of A2BAR and could apply to signaling mechanisms in other GPCRs.

Introduction

The A2B adenosine receptor (A2BAR) is involved in many important physiological and pathophysiological functions, such as the hematopoietic stem cell emergence [1], inflammation [2], [3], [4], cell proliferation [5], [6], [7]; reactive oxygen species (ROS) production [8], erectile function [9], bone homeostasis [10], cardiac protection [11] and diabetes mellitus [12]. However, the signaling mechanisms related to those functions are not well understood. In particular, the mechanisms leading to ERK1/2 phosphorylation, one of those canonical signaling processes, have not been well explored and currently published data are inconsistent. For example, it has been suggested A2BAR-mediated ERK1/2 activity in HEK293 cells is via the Gq/11 protein [13]. However, Schulte and Fredholm [14] suggested that A2BAR-mediated ERK1/2 activity in CHO cells expressing the recombinant A2BAR is mediated via Gs rather than Gq/11. Epperson et al. [15] also found a lack of A2BAR-coupling to Gq protein in cardiac fibroblasts. PKC is a typical modulator of the A2BAR- and other GPCR-mediated ERK1/2 activity [16]. However, Fang and Olah [17] showed that EPAC1 but not PKC is involved in A2BAR-mediated ERK1/2 phosphorylation in human vascular endothelial cells (HUVEC). In rat skeletal muscle, A2BAR activation was found not to modulate ERK1/2, although it stimulated cAMP accumulation [18]. In trophoblast cells, it was found that A2BAR downregulates ERK1/2 activity [19]. ERK1/2 activity stimulated by adenosine-5′-N-ethyluronamide (NECA) in A2BAR-expressing CHO is via PI3K, but not PKA [14]. However, Sun et al. [20] showed that A2BAR activation stimulates ERK1/2 activity via PKA in sickle erythrocytes. Thus, it seems signal mechanisms diverge in various studies and in different cell types under different conditions. Indeed, A2BAR has been shown to cause both proliferation and antiproliferation in different cell types [5], [21], [22], [23], [24], [25], [26], [27]. A2BAR has been reported to involve both pro- and anti-inflammatory actions [2], [28], [29], [30], which might be related the different G protein-coupling in different types of immune cells. Thus, it is important to understand the types of G proteins or signaling pathways involved in A2BAR-mediated functions.

Recent studies show that various A2BAR agonists have different maximum effect (Emax) and signaling bias in different signaling pathways [31], [32]. Nonnucleoside agonist BAY60-6583 stimulates ERK1/2 activity in HEK293 cells but not in T24 bladder cancer cells, both endogenously expressing the A2BAR, although nucleosides NECA and adenosine-5′-N-cyclopropyluronamide (CPCA) stimulated ERK1/2 activity in both cell types [31]. It was demonstrated that BAY60-6583 behaves as an antagonist and induces insulin secretion in a mouse pancreatic cell line, MIN-6 [31]. Some controversies concerning the A2BAR might also be due to different agonists used in those earlier studies.

In the present study, we explore the coupling of the native human A2BAR to Gi, Gq/11 and Gs in three different cell types, 1321 N1 astrocytoma, HEK293, and T24 bladder cancer cells. We used NECA and BAY60-6583, selective blockers for Gi, Gs and Gq/11, as well as the clustered regularly interspaced short palindromic repeat-associated protein-9 nuclease (CRISPR/Cas9)-based Gq/11- and Gs-null HEK293 cells [33], [34]. As ERK1/2 activity and calcium mobilization are involved in many A2B-mediated functions and are the convergence of multiple G protein-signaling pathways directed under the A2BAR, we explored the contributions of various G proteins and downstream signaling molecules to ERK1/2 activity and calcium mobilization. These pathways were compared in three different cell types endogenously expressing almost solely the A2BAR with the other three AR subtypes being at very low levels. We found that the A2BAR may couple to different downstream signaling events via different G proteins in the same cell type, and the A2BAR may couple to the same downstream pathway via different G proteins in different cell types. Gi and Gs proteins activated by the A2BAR may play opposing or synergistic roles in different cell types and in different signaling pathways. Any one of the three G proteins, Gi, Gq/11, and Gs, may be solely responsible for a specific signaling event mediated via the A2BAR depending on cell type. The findings could be related to controversial roles of the A2BAR-mediated effects reported in literature, e.g. proliferation and antiproliferation, or proinflammatory vs. anti-inflammatory. These findings could be applicable to the study of signaling pathways of other GPCRs, and should be of considerable relevance to future drug discovery and development.

Section snippets

Materials

GO6983, CE3F4, ESI-09, HJC0350, H89, PSB603, NECA (adenosine-5′-N-ethyluronamide) and MRS2365 ([[(1R,2R,3S,4R,5S)-4-[6-amino-2-(methylthio)-9H-purin-9-yl]-2,3-dihydroxybicyclo[3.1.0]hex-1-yl]methyl] diphosphoric acid monoester trisodium salt) were from Tocris (Ellisville, MO). CGS21680, Carbachol, CTX and PTX were from Sigma (St. Louis, MO). UBO-QIC was purchased from University of Bonn (Bonn, Germany). BAY60-6583 (LUF6210, termed hereafter ‘BAY’) was synthesized at Leiden/Amsterdam Center for

Gene expression of four AR subtypes in three cell types

In order to study the coupling of the endogenous human A2BAR to different G proteins, we selected three cell types, HEK293, T24 and 1321 N1, which endogenously express the A2BAR highly in comparison to the other three AR subtypes. The reason for our use of AR agonist NECA is that it is still the only established full agonist for the A2BAR that is commercially available, although nonselective. The nonnucleoside agonist BAY60-6583 is a partial agonist, albeit selective. Previously, we have found

Discussion

By using three cell types, HEK293 (including wild-type, Gs- and Gq/11-null HEK293), 1321 N1 astrocytoma and T24 bladder cancer cells, it was demonstrated in the present study that the endogenous A2BAR may couple to the same downstream signaling pathway via different G proteins in different cell types, and it may couple to different signaling pathways via different G proteins in the same type of cells (Table 1). Furthermore, Gs and Gi may have both opposing effects and synergistic effects in

Acknowledgements

Supported the Intramural Research Program of the National Institute of Diabetes and Digestive and Kidney Diseases (ZIADK031117), National Institutes of Health, Bethesda, MD, USA. AI was funded by the PRIME from Japan Agency for Medical Research and Development (AMED) and Japan Society for the Promotion of Science KAKENHI Grant Number 17 K08264. The authors thank Jurgen Wess (NIDDK, NIH) for advice, Ad IJzerman (Leiden, The Netherlands) for providing BAY60-6583 (LUF6210), E. Kiselev (NIDDK, NIH)

Conflict of interest

The authors have no conflict of interest to declare.

References (67)

  • D. Courilleau et al.

    Identification of a tetrahydroquinoline analog as a pharmacological inhibitor of the cAMP-binding protein Epac

    J. Biol. Chem.

    (2012)
  • K.J. Soeder et al.

    The beta3-adrenergic receptor activates mitogen-activated protein kinase in adipocytes through a Gi-dependent mechanism

    J. Biol. Chem.

    (1999)
  • A. Herrlich et al.

    Involvement of Gs and Gi proteins in dual coupling of the luteinizing hormone receptor to adenylyl cyclase and phospholipase C

    J. Biol. Chem.

    (1996)
  • G. Wu et al.

    Interaction of the delta-opioid receptor with GM1 ganglioside: conversion from inhibitory to excitatory mode

    Brain Res. Mol. Brain. Res.

    (1997)
  • R.B. Raffa et al.

    Could dual G-protein coupling explain [D-Met2]FMRFamide's mixed action in vivo?

    Peptides

    (1996)
  • A.M. Zamah et al.

    Protein kinase A-mediated phosphorylation of the beta2-adrenergic receptor regulates its coupling to Gs and Gi. Demonstration in a reconstituted system

    J. Biol. Chem.

    (2002)
  • P. Crespo et al.

    Dual effect of beta-adrenergic receptors on mitogen-activated protein kinase. Evidence for a beta gamma-dependent activation and a G alpha s-cAMP-mediated inhibition

    J. Biol. Chem.

    (1995)
  • Z.G. Gao et al.

    On the selectivity of the Gαq inhibitor UBO-QIC: A comparison with the Gαi inhibitor pertussis toxin

    Biochem. Pharmacol.

    (2016)
  • B.G. Dekkers et al.

    Distinct PKA and Epac compartmentalization in airway function and plasticity

    Pharmacol. Ther.

    (2013)
  • O.H. Petersen

    Calcium signalling and secretory epithelia

    Cell Calcium

    (2014)
  • L. Jing et al.

    Adenosine signaling promotes hematopoietic stem and progenitor cell emergence

    J. Exp. Med.

    (2015)
  • H. Liu et al.

    Beneficial role of erythrocyte adenosine A2B receptor-mediated AMP-activated protein kinase activation in high-altitude hypoxia

    Circulation

    (2016)
  • A. Ohta et al.

    Extracellular adenosine-mediated modulation of regulatory T cells

    Front. Immunol.

    (2014)
  • Q. Wei et al.

    A2B adenosine receptor blockade inhibits growth of prostate cancer cells

    Purinergic Signal.

    (2013)
  • D. Mittal et al.

    Adenosine 2B receptor expression on cancer cells promotes metastasis

    Cancer Res.

    (2016)
  • E. Ntantie et al.

    An adenosine-mediated signaling pathway suppresses prenylation of the GTPase Rap1B and promotes cell scattering

    Sci. Signal.

    (2013)
  • X. Yang et al.

    A2B adenosine receptors inhibit superoxide production from mitochondrial complex I in rabbit cardiomyocytes via a mechanism sensitive to Pertussis toxin

    Br. J. Pharmacol.

    (2011)
  • C. Ning et al.

    Xia Y Excess adenosine A2B receptor signaling contributes to priapism through HIF-1α mediated reduction of PDE5 gene expression

    FASEB J.

    (2014)
  • C. Corciulo et al.

    Adenosine A2B receptors play an important role in bone homeostasis

    Purinergic Signal.

    (2016)
  • L. Antonioli et al.

    Adenosine signalling in diabetes mellitus–pathophysiology and therapeutic considerations

    Nat. Rev. Endocrinol.

    (2015)
  • S.A. Epperson et al.

    Adenosine receptors and second messenger signaling pathways in rat cardiac fibroblasts

    Am. J. Physiol. Cell. Physiol.

    (2009)
  • H. Wei et al.

    Independent beta-arrestin 2 and G protein-mediated pathways for angiotensin II activation of extracellular signal-regulated kinases 1 and 2

    Proc. Nat. Acad. Sci. U.S.A.

    (2003)
  • Y. Fang et al.

    Cyclic AMP-dependent, protein kinase A-independent activation of extracellular signal-regulated kinase 1/2 following adenosine receptor stimulation in human umbilical vein endothelial cells: role of exchange protein activated by cAMP 1 (Epac1)

    J. Pharmacol. Exp. Ther.

    (2007)
  • Cited by (35)

    • Biased agonism at adenosine receptors

      2021, Cellular Signalling
      Citation Excerpt :

      Accordingly, efforts to dissect agonist-mediated G protein-dependent and G protein-independent signalling is an area of intense research. Bioluminescence resonance energy transfer (BRET), Förster resonance energy transfer (FRET) and split-luciferase (NanoBiT) technologies have been used to quantify GPCR preferences for G protein [9,106–110] and arrestin coupling [111], including application to adenosine receptors [18,20,112]. Independent of biosensors, the recent utilization of CRISPR-Cas9 technology has enabled the selective deletion of certain Gα subtypes, facilitating the delineation of the specific GPCR-effector interactions involved in downstream signalling.

    • An experimental strategy to probe Gq contribution to signal transduction in living cells

      2021, Journal of Biological Chemistry
      Citation Excerpt :

      The central research question therefore is whether FR and YM are indeed not as selective as previously anticipated (24, 25, 27) or, whether inhibition by FR or YM of biological responses may, instead, indicate necessity of and/or specific contribution by Gq. The approach presented in this study attempts to fill this knowledge gap by providing a molecular toolbox for unambiguous discrimination between on-target Gq and off-target non-Gq action for the two widely applied natural product inhibitors (14, 29–55, 128). One of its core elements is GαqF75K I190W, a drug-resistant Gαq mutant, which is catalytically active, yet entirely refractory toward inhibitor action.

    • Heterotrimeric G<inf>q</inf> proteins as therapeutic targets?

      2020, Journal of Biological Chemistry
      Citation Excerpt :

      Selective inhibition by FR of Gq, G11, and G14 over all other mammalian G proteins, its molecular mechanism of GDI action, and the potential to probe the Gq contribution to complex biological processes in physiology and disease were not addressed until 2015, when a comprehensive study provided in vitro and ex vivo characterization at a level of detail sufficient to reinvigorate the field of Gq protein inhibitors (11) (Fig. 3). Indeed, this very study impacted G protein inhibitor research in manifold beneficial ways: it (i) created scientific community awareness for the existence of a most valuable signal transduction inhibitor, (ii) triggered independent confirmatory studies to re-examine FR's selectivity profile (80–82), (iii) helped fuel the competitive efforts to identify the best-suited synthetic methodology for preparing the complex molecule by chemical synthesis (83–86), (iv) sparked broad interest for the application of FR and YM to explore the biological consequences that arise from specific Gq inhibition (7, 87–107), and (v) provided experimental evidence that Gq inhibition may qualify as an effective postreceptor strategy to target oncogenic signaling in cancer cells with elevated Gq activity. The first signs for FR efficacy in cancer treatment were obtained when exposing a panel of skin melanoma cells to FR in cell culture (11).

    View all citing articles on Scopus
    View full text