Hostname: page-component-76fb5796d-dfsvx Total loading time: 0 Render date: 2024-04-28T00:37:50.540Z Has data issue: false hasContentIssue false

Cannabidiol negatively modulates adenosine A2A receptor functioning in living cells

Published online by Cambridge University Press:  22 August 2023

Nuria Sánchez-Fernández
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Laura Gómez-Acero
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Laura I. Sarasola
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Josep Argerich
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Andy Chevigné
Affiliation:
Immuno-Pharmacology and Interactomics, Department of Infection and Immunity, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
Kenneth A. Jacobson
Affiliation:
Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
Francisco Ciruela
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Víctor Fernández-Dueñas*
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
Ester Aso*
Affiliation:
Pharmacology Unit, Department of Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L’Hospitalet de Llobregat, Spain Neuropharmacology and Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, L’Hospitalet de Llobregat, Spain
*
Corresponding authors: Ester Aso; Email: ester.aso@ub.edu; Víctor Fernández-Dueñas; Email: vfernandez@ub.edu
Corresponding authors: Ester Aso; Email: ester.aso@ub.edu; Víctor Fernández-Dueñas; Email: vfernandez@ub.edu
Rights & Permissions [Opens in a new window]

Abstract

Objectives:

Cannabidiol (CBD) is a phytocannabinoid with great potential in clinical applications. The mechanism(s) of action of CBD require further investigation. Previous studies suggested that adenosine A2A receptors (A2ARs) could play a role in CBD-induced effects. Here, we evaluated the ability of CBD to modify the function of A2AR.

Methods:

We used HEK-293T cells transfected with the cDNA encoding the human A2AR and Gαs protein, both modified to perform bioluminescence-based assays. We first assessed the effect of CBD on A2AR ligand binding using an A2AR NanoLuciferase sensor. Next, we evaluated whether CBD modified A2AR coupling to mini-Gαs proteins using the NanoBiT™ assay. Finally, we further assessed CBD effects on A2AR intrinsic activity by recording agonist-induced cAMP accumulation.

Results:

CBD did not bind orthosterically to A2AR but reduced the coupling of A2AR to Gαs protein and the subsequent generation of cAMP.

Conclusion:

CBD negatively modulates A2AR functioning.

Type
Short Communication
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of Scandinavian College of Neuropsychopharmacology

Significant outcomes

  • Cannabidiol does not bind orthosterically to A2AR.

  • Cannabidiol reduces the functionality of A2AR

Limitations

  • This is an in vitro study, and results cannot be directly extrapolated to in vivo conditions.

  • Putative allosteric binding of CBD to A2AR cannot be confirmed or ruled out with the luminescence-based techniques employed in this study.

Introduction

Cannabidiol (CBD) is a phytocannabinoid isolated from Cannabis sativa without psychoactive properties, but with potential benefits against multiple pathological conditions (ElSohly et al., Reference ElSohly, Radwan, Gul, Chandra, Galal, Kinghorn, Falk, Gibbons and Kobayashi)2017). Several preclinical reports demonstrated protective and anti-inflammatory effects of CBD in a wide spectrum of neurodegenerative diseases, neuroinflammatory processes, stroke, colitis, liver, kidney injury, cardiovascular disease, arthritis, sepsis, diabetes, cancer, and epilepsy models (Pacher et al., Reference Pacher, Kogan and Mechoulam2020). Furthermore, CBD exerted positive effects in experimental models of other neuropsychiatric disorders such as epilepsy, anxiety, schizophrenia, dementia, addiction, and neonatal hypoxic-ischemic encephalopathy (Devinsky et al., Reference Devinsky, Cilio, Cross, Fernández-Ruiz, French, Hill, Katz, Di Marzo, Jutras-Aswad, Notcutt, Martínez-Orgado, Robson, Rohrback, Thiele, Whalley and Friedman2014). Although its translation to clinical trials is somewhat limited to date, the successful case of Epidiolex®, an oral solution based on a botanical extract containing purified CBD, is notable. Epidiolex® was approved by the US Food and Drug Administration in 2018 for the treatment of Lennox-Gastaut and Dravet syndromes, two rare and debilitating genetic forms of epilepsy in children. Additionally, CBD is currently under clinical evaluation for other conditions, including different forms of pain, obsessive-compulsive disorders, and behavioural problems associated with intellectual disability or autism, among others (ClinicalTrials.gov database).

Despite growing interest in its potential clinical applications, the mechanism(s) of action of CBD require further exploration. CBD has a very low affinity for the orthosteric site of CB1 and CB2 receptors, the main G protein-coupled receptors (GPCRs) that belong to the endogenous cannabinoid system (McPartland et al., Reference McPartland, Duncan, Di Marzo and Pertwee2015). Alternatively, CBD can act on multiple targets, including TRPV1 channels and PPARγ, adenosine A2A, 5-HT1A, α3-glycine, α1-adrenal, dopamine D2, GABAA, μ- and δ-opioid receptors (McPartland et al., Reference McPartland, Duncan, Di Marzo and Pertwee2015). Additionally, CBD can inhibit the activity of GPR55 (Ryberg et al., Reference Ryberg, Larsson, Sjögren, Hjorth, Hermansson, Leonova, Elebring, Nilsson, Drmota and Greasley2007), an effect that has been associated with its antiepileptic activity (Sylantyev et al., Reference Sylantyev, Jensen, Ross and Rusakov2013). In the present study, we probed the putative direct effects of CBD on adenosine A2A receptors (A2ARs). The relevant role that A2ARs play in several of the neuropsychiatric disorders in which CBD could offer beneficial effects (i.e. dementia, schizophrenia, epilepsy, depression, anxiety) supports this interest (Domenici et al., Reference Domenici, Ferrante, Martire, Chiodi, Pepponi, Tebano and Popoli2019). Furthermore, previous preclinical evidence supports the participation of A2AR in CBD-mediated effects. Thus, A2AR antagonists blocked the anti-inflammatory effects of CBD (Liou et al., Reference Liou, Auchampach, Hillard, Zhu, Yousufzai, Mian, Khan and Khalifa2008; Ribeiro et al., Reference Ribeiro, Ferraz-de-Paula, Pinheiro, Vitoretti, Mariano-Souza, Quinteiro-Filho, Akamine, Almeida, Quevedo, Dal-Pizzol, Hallak, Zuardi, Crippa and Palermo-Neto2012; Mecha et al., Reference Mecha, Feliú, Iñigo, Mestre, Carrillo-Salinas and Guaza2013; Oláh et al., Reference Oláh, Tóth, Borbíró, Sugawara, Szöllõsi, Czifra, Pál, Ambrus, Kloepper, Camera, Ludovici, Picardo, Voets, Zouboulis, Paus and Bíró2014), or the ability of CBD to blunt Δ9-THC-induced cognitive impairment (Aso et al., Reference Aso, Fernández-Dueñas, López-Cano, Taura, Watanabe, Ferrer, Luján and Ciruela2019). Similarly, the genetic deletion of A2AR reduced the CBD-induced potentiation of the cataleptic and anxiolytic properties of Δ9-THC (Stollenwerk et al., Reference Stollenwerk, Pollock and Hillard2021). This A2AR-dependent activity of CBD was proposed to depend on the ability of CBD to bind to the equilibrative nucleoside transporter (ENT). Thus, inhibition of adenosine uptake would lead to indirect activation of A2AR (Pandolfo et al., Reference Pandolfo, Silveirinha, dos Santos-Rodrigues, Venance, Ledent, Takahashi, Cunha and Köfalvi2011). However, a direct effect of CBD on A2AR has not been further investigated. Here we aimed to evaluate the capacity of CBD to bind to the orthosteric site of A2AR and/or to modify its intrinsic activity by using state-of-the-art luminescence-based assays.

Materials and methods

Reagents

The ligands used were CGS21680, ZM241385, and CBD (Tocris Bioscience, Bristol, United Kingdom). MRS7396, a fluorescent selective A2AR orthosteric antagonist derived from SCH442416 and containing a BODIPY630/650 fluorophore, was previously described (Duroux et al., Reference Duroux, Ciancetta, Mannes, Yu, Boyapati, Gizewski, Yous, Ciruela, Auchampach, Gao and Jacobson2017). Other reagents used were Dulbecco’s modified Eagle’s medium (DMEM; Sigma Aldrich, St Louis, MO, USA), geneticin (Santa Cruz Biotechnology, Dallas, TX, USA), adenosine deaminase (ADA; Roche Diagnostics GmbH, Mannheim, Germany), zardaverine (Calbiochem, San Diego, CA, USA), and coelenterazine 400a (NanoLight Technologies, Pinetop, AZ, USA).

Plasmid constructs

To perform bioluminescence resonance energy transfer (BRET) experiments and cAMP accumulation assays, we used the A2AR NanoLuciferase (NanoLuc) sensor (A2ARNL), previously described (Lanznaster et al., Reference Lanznaster, Massari, Marková, Šimková, Duroux, Jacobson, Fernández-Dueñas, Tasca and Ciruela2019). To perform the NanoBiT™ assay, the cDNA encoding human A2AR was cloned at the BamHI/EcoRV restriction enzyme sites of pIREShyg3-SmBiT (Promega, Madison, WI, USA), as previously described (Sarasola et al., Reference Sarasola, Del Torrent, Pérez-Arévalo, Argerich, Casajuana-Martín, Chevigné, Fernández-Dueñas, Ferré, Pardo and Ciruela2022). The construct (A2ARSmBiT) was verified by DNA sequencing. The plasmid encoding the mini-Gαs (engineered GTPase domain of Gα subunit; LgBiTmini-Gαs) linked to LgBiT was previously described (Wan et al., Reference Wan, Okashah, Inoue, Nehmé, Carpenter, Tate and Lambert2018; Meyrath et al., Reference Meyrath, Palmer, Reynders, Vanderplasschen, Ollert, Bouvier, Szpakowska and Chevigné2021).

Cell culture and transfection

Human embryonic kidney (HEK)-293T cells were grown in DMEM supplemented with 1 mM sodium pyruvate (Biowest, Nuaillé, France), 2 mM L-glutamine (Biowest), 100 U/mL streptomycin (Biowest), 100 mg/mL penicillin (Biowest), and 5% (v/v) foetal bovine serum (Invitrogen Corporation, Camarillo, CA, USA) at 37°C and in an atmosphere of 5% CO2. Cells were transiently transfected with the indicated cDNA construct using polyethylenimine (PEI, 1 mg/mL, Sigma Aldrich), as previously described (Longo et al., Reference Longo, Kavran, Kim and Leahy2013). Finally, HEK-293T cells stably expressing A2ARNL were grown in the presence of geneticin (1 mg/mL).

NanoBRET experiments

The NanoBRET assay was performed as previously described (Lanznaster et al., Reference Lanznaster, Massari, Marková, Šimková, Duroux, Jacobson, Fernández-Dueñas, Tasca and Ciruela2019). Briefly, HEK-293T cells expressing the A2ARNL construct were resuspended in Hank’s balanced salt solution (HBSS; Thermo Fisher, Waltham, MA, USA) containing ADA (0.5 U/mL) and plated on white 96-well plates coated with poly-ornithine (Corning, Corning, NY, USA) at a density of 20,000 cells/well. After 24 h, cells were challenged with the fluorescent A2AR antagonist (MRS7396) in the absence/presence of ZM241385 or CBD and incubated for 1 h at 37°C. Subsequently, coelenterazine 400a was added at a final concentration of 1 μM, and the readings were performed after 15 min using a CLARIOStar microplate reader (BMG Labtech, Durham, NC, USA). Donor and acceptor emission were measured at 490 ± 10 nm and 650 ± 40 nm, respectively. The raw NanoBRET ratio was calculated by dividing the 650 nm emission by the 490 nm emission and the values fitted by nonlinear regression using GraphPad Prism 9 (GraphPad Software, La Jolla, CA, USA). The results were expressed as a percentage of the maximum signal obtained (mBU; miliBRET units).

NanoBiT assay

The NanoBiT™ assay (Promega) was performed as previously described (Sarasola et al., Reference Sarasola, Del Torrent, Pérez-Arévalo, Argerich, Casajuana-Martín, Chevigné, Fernández-Dueñas, Ferré, Pardo and Ciruela2022). Briefly, transient transfected HEK-293T cells with A2ARSmBiT and LgBiTmini-Gαs were resuspended in HBSS containing ADA (0.5 U/mL) and transferred (90 µl) into white 96-well plates (Corning). Subsequently, coelenterazine 400a was added (1 μM) to each well. After 15-minute incubation, basal luminescence was determined using a CLARIOstar plate reader (BMG Labtech). Immediately after the initial measurement (basal), the ligands were added, and the luminescent signal was measured every 5 min for 30 min. The luminescence signal (RLU) was normalised as follows: (RLUsample − RLUbasal) / RLUbasal.

cAMP assay

cAMP accumulation was measured using the LANCE® Ultra cAMP Kit (PerkinElmer, Waltham, MA, USA) as previously described (Lanznaster et al., Reference Lanznaster, Massari, Marková, Šimková, Duroux, Jacobson, Fernández-Dueñas, Tasca and Ciruela2019). Briefly, HEK-293T cells stably expressing the A2ARNL construct were first incubated for 1 h at 37°C with stimulation buffer (BSA 0.1%, ADA 0.5 units/mL, zardaverine 2 µM; in serum-free DMEM) and later with CGS21680 (100 nM) and increasing concentrations of ZM241385 or CBD for 30 min at 37°C. Subsequently, cells were transferred (1000 cells/well) into white 384-well plates (Corning), in which reagents were added following the manufacturer’s instructions. After 1 h at room temperature, time-resolved fluorescence resonance energy transfer (TR-FRET) was determined by measuring light emission at 620 nm and 665 nm using a CLARIOstar plate reader (BMG Labtech).

Statistics

Data are represented as mean ± standard error of mean (SEM) with statistical significance set at P < 0.05. The number of samples (n) in each experimental condition is indicated in the legend of the corresponding figure. Outliers were assessed using the ROUT method (Motulsky & Brown, Reference Motulsky and Brown2006); thus, no sample was excluded assuming a Q value of 1% in GraphPad Prism 9. Comparisons between experimental groups were performed using one-way factor analysis of variance (ANOVA) followed by Dunnett’s multiple comparisons post hoc test using GraphPad Prism 9 as indicated.

Results

To assess the impact of CBD on A2AR functionality, we initially evaluated whether CBD modified the binding of MRS7396, a fluorescent A2AR antagonist. To this end, we took advantage of a previously reported NanoBRET-based A2AR binding assay (Fig. 1a) (Lanznaster et al., Reference Lanznaster, Massari, Marková, Šimková, Duroux, Jacobson, Fernández-Dueñas, Tasca and Ciruela2019). HEK-293T cells permanently expressing the A2ARNL construct were challenged with increasing concentrations of MRS7396, which upon binding to the receptor can act as a compatible acceptor in a BRET process (Fig. 1a). As expected, a saturable hyperbolic curve was obtained for the total binding of MRS7396, which was blocked upon incubation with a saturating concentration (1 µM) of the unlabelled A2AR antagonist ZM241385 (nonspecific binding; Fig. 1b). The analysis of the specific binding of MRS7396 revealed a dissociation constant (K D) of 8.5 ± 3.2 nM and a maximum binding capacity (B max) of 98.9 ± 8.4 %. Next, upon the same experimental conditions, we examined the ability of CBD to attenuate MRS7396 binding. Differently from ZM241385, CBD (1 µM) was unable to modify the specific binding of MRS7396 to the A2AR (Fig. 1b). Accordingly, no significant differences in affinity (K D) and receptor capacity (B max) were found in the presence of CBD (KD = 8.2 ± 2.7 nM; Bmax = 96.9 ± 7.2 %; P = 0.992, F (2, 34) = 0.0079). In addition, we also assessed whether CBD could modulate orthosteric binding of A2AR by performing a competition binding assay with a fixed concentration of MRS7396 (30 nM). Again, while ZM241385 blocked A2AR binding, CBD did not significantly modify the NanoBRET signal (Fig. 1c; P = 0.269, F (4, 10) = 1.518). Collectively, these results indicate that CBD does not bind orthosterically to A2AR.

Figure 1. Determination of CBD effects on A2AR ligand binding affinity. (a) Schematic representation of the NanoBRET assay. A nanoluciferase is linked to the N-terminal part of the A2AR (A2ARNL). When the nanoluciferase substrate coelenterazine is added, A2ARNL (donor) emits light at 490–10 nm. Light excites the fluorescent selective A2AR ligand, MRS7396 (acceptor), which subsequently emits fluorescence at 650-80 nm. (b) NanoBRET saturation binding curves obtained by challenging A2ARNL expressing HEK-293T cells with increasing concentrations of MRS7396 in the absence/presence of CBD (1 µM) or ZM241385 (1 µM). (c) NanoBRET signals obtained by challenging A2ARNL expressing HEK-293T cells with a fixed concentration of MRS7396 (30 nM, normalised to 100%) in the presence of increasing concentrations of CBD or ZM241385. The represented data are mean ± SEM of three independent experiments each performed in triplicate.

Subsequently, we aimed to determine whether CBD affected A2AR signalling. To this end, we first evaluated the interaction of A2AR with mini-Gαs protein using the NanoBiT™ complementation assay (Sarasola et al., Reference Sarasola, Del Torrent, Pérez-Arévalo, Argerich, Casajuana-Martín, Chevigné, Fernández-Dueñas, Ferré, Pardo and Ciruela2022). Accordingly, cells were transiently transfected with the A2ARSmBiT and LgBiTmini-Gαs constructs, which once expressed allow the reconstitution of the split nanoluciferase and real-time recordings of receptor-effector coupling induced by agonists (Fig. 2a). Cells were challenged with the selective A2AR agonist CGS21680 (100 nM), which induced a rapid increase in the luminescent signal, reaching a peak at 15 min that remained stable for 30 min. This effect was absent in cells challenged with CBD alone (Fig. 2b). Notably, this agonist-dependent A2AR interaction with mini-Gαs protein was completely blocked when co-incubating cells with ZM241385 (50 nM, Fig. 2b). Next, we assessed CGS21680-induced A2AR coupling to Gαs protein in the presence of increasing concentrations of CBD. Interestingly, CBD dose-dependently blocked A2AR coupling to Gαs protein, both decreasing the maximum peak and the density of the effect (Fig. 2b). Of note, differently from ZM241385, CBD only led to a partial blockade of CGS21680-mediated effects.

Figure 2. Assessment of CBD effects on A2AR intrinsic activity. (a) Schematic representation of the NanoBIT™-based assay. The two fragments of nanoluciferase, small (SmBIT) and large (LgBIT), are fused to A2AR and mini-Gαs protein, respectively. Then, upon agonist binding, A2AR intrinsic activity is assessed by receptor recruitment of Gαs, which induces an increase on luminescence due to nanoluciferase reconstitution. (b) Representative time-course of A2AR agonist-mediated Gαs recruitment. The selective A2AR agonist CGS21680 was challenged to A2ARSmBiT and LgBiTmini-Gαs expressing HEK-293T cells in the absence/presence of increasing concentrations of CBD or ZM241385. The luminescent signal obtained after reconstitution of the nanoluciferase was assessed by calculating the area under the curve for each condition. Data are shown as mean ± SEM of three independent experiments with five replicates. *P < 0.05, ***P < 0.001, one-way ANOVA with Dunnett’s post hoc test. (c) cAMP accumulation was assessed on HEK-293T cells permanently expressing the A2ARNL. Cells were challenged with the selective A2AR agonist CGS21680 (100 nM, normalised to 100% of effect) in the absence/presence of increasing concentrations of CBD. Data are expressed as mean ± SEM of four independent experiments performed in triplicates. *P < 0.05, one-way ANOVA with Dunnett’s post hoc test.

Finally, to further characterise the effects of CBD on the intrinsic activity of A2AR, we evaluated agonist-induced cAMP accumulation in cells permanently expressing the A2ARNL construct and challenged with CGS21680 in the presence/absence of CBD. Interestingly, although CBD itself did not modify cAMP levels, it was able to dose-dependently block CGS21680-induced cAMP levels (Fig. 2c). Again, differently from ZM241385, a full-antagonist, CBD partially blocked A2AR agonist increase of cAMP levels. Overall, these results are compatible with the notion that CBD acts as a weak A2AR negative allosteric modulator.

Discussion

CBD is a promising drug for several pathologies (ElSohly et al., Reference ElSohly, Radwan, Gul, Chandra, Galal, Kinghorn, Falk, Gibbons and Kobayashi)2017). Accordingly, unravelling its precise mechanism of action is relevant in the progress towards its clinical development. Here, we reveal that CBD does not affect the binding of an A2AR orthosteric ligand, but it is capable of negatively modulating agonist-induced interaction with the Gαs protein at sub-micromolar concentrations (≥100 nM), thus reducing receptor signalling (i.e. cAMP generation). Therefore, we disclose a new non-competitive interaction of CBD with A2AR.

The effect of CBD on A2AR could operate through a new allosteric site at the receptor. However, further experiments (i.e. using labelled CBD) would be needed to confirm this hypothesis. On the other hand, we cannot rule out other mechanisms of action for CBD different from classical allosteric drugs. In this sense, previous evidence indicates that other lipids, including the endogenous cannabinoid anandamide at micromolar concentrations, might act as allosteric modulators of other GPCRs through a membrane-perturbing effect that is sensitive to receptor conformation (Lanzafame et al., Reference Lanzafame, Guida and Christopoulos2004; Van der Westhuizen et al., Reference Van der Westhuizen, Valant, Sexton and Christopoulos2015). Further studies are needed to assess this putative CBD-mediated membrane effect on A2AR-Gαs protein coupling. Similarly, CBD could indirectly modify A2AR functioning by interacting with equilibrative nucleoside transporter 1 (ENT1), as was previously demonstrated in striatal synaptosomes (Pandolfo et al., Reference Pandolfo, Silveirinha, dos Santos-Rodrigues, Venance, Ledent, Takahashi, Cunha and Köfalvi2011). However, this hypothetical CBD effect on ENT seems not to play a relevant role in vivo, since a recent study demonstrated that CBD lacks the ability to substantially raise endogenous adenosine levels by using the hypothermia mouse model (Xiao et al., Reference Xiao, Gavrilova, Liu, Lewicki, Reitman and Jacobson2023). These discrepancies between in vitro and in vivo studies could be also explained by the fact that A2AR can form heteromers with other GPCRs, including CB1R (Carriba et al., Reference Carriba, Ortiz, Patkar, Justinova, Stroik, Themann, Müller, Woods, Hope, Ciruela, Casadó, Canela, Lluis, Goldberg, Moratalla, Franco and Ferré2007; Ferré et al., Reference Ferré, Lluís, Justinova, Quiroz, Orru, Navarro, Canela, Franco and Goldberg2010; Aso et al., Reference Aso, Fernández-Dueñas, López-Cano, Taura, Watanabe, Ferrer, Luján and Ciruela2019), in physiological conditions different from that obtained in heterologous expression systems. The assembly of A2AR-containing heteromers leads to changes in the agonist recognition, signalling, and trafficking, which might result in different A2AR activity in the presence of CBD.

Although we evaluated the effects of CBD in cultured cells expressing A2AR, these results could be relevant for many disorders in which A2AR activity increases. For example, in certain inflammatory processes and cardiovascular diseases, but also in pathological conditions that affect the central nervous system, such as Alzheimer’s disease, Parkinson’s disease, attention deficit hyperactivity disorder, fragile X syndrome, depression, or anxiety (Domenici et al., Reference Domenici, Ferrante, Martire, Chiodi, Pepponi, Tebano and Popoli2019). A2ARs, which are widely expressed both in neurons and glia, are mainly found in the dorsal and ventral striatum and other nuclei of the basal ganglia, where they play a key role in the control of voluntary movements, as well as in motivational, emotional and cognitive processes (Sebastião and Ribeiro, Reference Sebastião and Ribeiro2009). In this way, A2ARs are involved in regulating the release of neurotransmitters and contribute to the homeostatic control of synaptic transmission and brain function (Sebastião and Ribeiro, Reference Sebastião and Ribeiro2009). In general, our results are consistent with the positive effects reported for CBD in various brain disorders that can be associated with an exacerbated A2AR function, where CBD would tone down A2AR hyperactivity.

Overall, the present study provides evidence on the ability of CBD to negatively modulate A2AR signalling. The CBD-mediated negative modulation of A2AR function is restricted to the receptor-effector coupling and does not interfere with the binding of the orthosteric ligand. Accordingly, we provide a new and genuine pharmacological way to modulate the adenosinergic system in pathological conditions in which A2AR function is increased.

Acknowledgements

We thank Centres de Recerca de Catalunya Programme/Generalitat de Catalunya for IDIBELL institutional support and Maria de Maeztu MDM-2017-0729 to Institut de Neurociencies, Universitat de Barcelona. We are also grateful to the CannaLatan network members (CYTED-Programa Iberoamericano de Ciencia y Tecnología para el Desarrollo) for the fruitful discussion about the results.

Author contribution

EA, VFD, and FC conceived and designed the study and wrote the manuscript. NSF performed all the experiments and contributed to the manuscript preparation. FC and AC designed the cDNA constructs used in this study. LGA, LIS, and JA cloned and validated cDNA constructs. KAJ provided the fluorescent selective A2AR antagonist.

Financial support

This study was supported by Ministerio de Ciencia, Innovación y Universidades–Agencia Estatal de Investigación-FEDER funds/European Regional Development Fund – ‘a way to build Europe’ grant RTI2018-097773-A-I00 to EA and PID2020-118511RB-I00 to FC. Founded by MCIN/AEI /10.13039/501100011033 ‘ESF Investing in your future’ grant PRE2018–084480 to JA, grant PRE2019-088153 to NSF and grant FPU19/03142 to LGA. Also supported by ‘Acció instrumental de formació de científics i tecnòlegs’ (SLT017/20/000114) of the Departament de Salut de la Generalitat de Catalunya to LIS. The study was also supported by the Luxembourg Institute of Health (LIH), Luxembourg National Research Fund (INTER/FNRS grants 20/15084569 to AC) and the National Institute of Diabetes and Digestive and Kidney Diseases NIDDK Intramural Research Program (ZIADK031117 to KAJ).

Competing interests

None of the authors declare any conflict of interest.

Ethical standard

The authors assert that all procedures contributing to this work comply with the ethical standards of the relevant national and institutional committees on experimentation with cells and DNA constructs.

References

Aso, E, Fernández-Dueñas, V, López-Cano, M, Taura, J, Watanabe, M, Ferrer, I, Luján, R and Ciruela, F (2019) Adenosine A2A-cannabinoid CB1 receptor heteromers in the hippocampus: cannabidiol blunts Δ9-tetrahydrocannabinol-induced cognitive impairment. Molecular Neurobiology 56(8), 53825391.CrossRefGoogle Scholar
Carriba, P, Ortiz, O, Patkar, K, Justinova, Z, Stroik, J, Themann, A, Müller, C, Woods, AS, Hope, BT, Ciruela, F, Casadó, V, Canela, EI, Lluis, C, Goldberg, SR, Moratalla, R, Franco, R and Ferré, S (2007) Striatal adenosine A2A and cannabinoid CB1 receptors form functional heteromeric complexes that mediate the motor effects of cannabinoids. Neuropsychopharmacology 32(11), 22492259.CrossRefGoogle ScholarPubMed
ClinicalTrials.gov database. The National Library of Medicine. Available at https://clinicaltrials.gov/ (accessed 16 December 2022).Google Scholar
Devinsky, O, Cilio, MR, Cross, H, Fernández-Ruiz, J, French, J, Hill, C, Katz, R, Di Marzo, V, Jutras-Aswad, D, Notcutt, WG, Martínez-Orgado, J, Robson, PJ, Rohrback, BG, Thiele, E, Whalley, B and Friedman, D (2014) Cannabidiol: pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia 55(6), 791802.CrossRefGoogle ScholarPubMed
Domenici, MR, Ferrante, A, Martire, A, Chiodi, V, Pepponi, R, Tebano, MT and Popoli, P (2019) Adenosine A2A receptor as potential therapeutic target in neuropsychiatric disorders. Pharmacological Research 147, 104338.CrossRefGoogle ScholarPubMed
Duroux, R, Ciancetta, A, Mannes, P, Yu, J, Boyapati, S, Gizewski, E, Yous, S, Ciruela, F, Auchampach, JA, Gao, ZG and Jacobson, KA (2017) Bitopic fluorescent antagonists of the A2A adenosine receptor based on pyrazolo[4,3-: E] [1,2,4]triazolo[1,5- c] pyrimidin-5-amine functionalized congeners. Medicinal Chemistry Communications 8(8), 16591667.CrossRefGoogle Scholar
ElSohly, MA, Radwan, MM, Gul, W, Chandra, S and Galal, A (2017) Phytochemistry of Cannabis sativa L . In Kinghorn, AD, Falk, H, Gibbons, S and Kobayashi), J (ed), Phytocannabinoids: unraveling the complex chemistry and pharmacology of Cannabis sativa. Cham, Switzerland: Springer, pp. 136.Google Scholar
Ferré, S, Lluís, C, Justinova, Z, Quiroz, C, Orru, M, Navarro, G, Canela, EI, Franco, R and Goldberg, SR (2010) Adenosine-cannabinoid receptor interactions. Implications for striatal function. British Journal Pharmacology 160(3), 443453.CrossRefGoogle ScholarPubMed
Lanzafame, A, Guida, E and Christopoulos, A (2004) Effects of anandamide on the binding and signaling properties of M1 muscarinic acetylcholine receptors. Biochemical Pharmacology 68(11), 22072219.CrossRefGoogle ScholarPubMed
Lanznaster, D, Massari, CM, Marková, V, Šimková, T, Duroux, R, Jacobson, KA, Fernández-Dueñas, V, Tasca, CI and Ciruela, F (2019) Adenosine A1-A2A receptor-receptor interaction: contribution to guanosine-mediated effects. Cells 8(12), 1630.CrossRefGoogle Scholar
Liou, GI, Auchampach, JA, Hillard, CJ, Zhu, G, Yousufzai, B, Mian, S, Khan, S and Khalifa, Y (2008) Mediation of cannabidiol anti-inflammation in the retina by equilibrative nucleoside transporter and A2A adenosine receptor. Investigative Ophthalmology & Visual Science 49(12), 55265531.CrossRefGoogle ScholarPubMed
Longo, PA, Kavran, JM, Kim, MS and Leahy, DJ (2013) Transient mammalian cell transfection with polyethylenimine (PEI). Methods Enzymology 529, 227240.CrossRefGoogle ScholarPubMed
McPartland, JM, Duncan, M, Di Marzo, V and Pertwee, RG (2015) Are cannabidiol and Δ(9)-tetrahydrocannabivarin negative modulators of the endocannabinoid system? A systematic review. British Journal Pharmacology 172(3), 737753.CrossRefGoogle ScholarPubMed
Mecha, M, Feliú, A, Iñigo, PM, Mestre, L, Carrillo-Salinas, FJ and Guaza, C (2013) Cannabidiol provides long-lasting protection against the deleterious effects of inflammation in a viral model of multiple sclerosis: a role for A2A receptors. Neurobiology Disease 59, 141150.CrossRefGoogle Scholar
Meyrath, M, Palmer, CB, Reynders, N, Vanderplasschen, A, Ollert, M, Bouvier, M, Szpakowska, M and Chevigné, A (2021) Proadrenomedullin N-terminal 20 peptides (PAMPs) are agonists of the chemokine scavenger receptor ACKR3/CXCR7. ACS Pharmacology Translational Science 4(2), 813823.CrossRefGoogle ScholarPubMed
Motulsky, HJ, Brown, RE (2006) Detecting outliers when fitting data with nonlinear regression - a new method based on robust nonlinear regression and the false discovery rate. BMC Bioinformatics 7, 123.CrossRefGoogle ScholarPubMed
Oláh, A, Tóth, BI, Borbíró, I, Sugawara, K, Szöllõsi, AG, Czifra, G, Pál, B, Ambrus, L, Kloepper, J, Camera, E, Ludovici, M, Picardo, M, Voets, T, Zouboulis, CC, Paus, R and Bíró, T (2014) Cannabidiol exerts sebostatic and antiinflammatory effects on human sebocytes. Journal Clinical Investigation 124(9), 37133724.CrossRefGoogle ScholarPubMed
Pacher, P, Kogan, NM and Mechoulam, R (2020) Beyond THC and endocannabinoids. Annual Review Pharmacology Toxicology 60, 637659.CrossRefGoogle ScholarPubMed
Pandolfo, P, Silveirinha, V, dos Santos-Rodrigues, A, Venance, L, Ledent, C, Takahashi, RN, Cunha, RA and Köfalvi, A (2011) Cannabinoids inhibit the synaptic uptake of adenosine and dopamine in the rat and mouse striatum. European Journal Pharmacology 655(1-3), 3845.CrossRefGoogle ScholarPubMed
Ribeiro, A, Ferraz-de-Paula, V, Pinheiro, ML, Vitoretti, LB, Mariano-Souza, DP, Quinteiro-Filho, WM, Akamine, AT, Almeida, VI, Quevedo, J, Dal-Pizzol, F, Hallak, JE, Zuardi, AW, Crippa, JA and Palermo-Neto, J. (2012) Cannabidiol, a non-psychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A2A receptor. European Journal Pharmacology 678(1-3), 7885.CrossRefGoogle Scholar
Ryberg, E, Larsson, N, Sjögren, S, Hjorth, S, Hermansson, NO, Leonova, J, Elebring, T, Nilsson, K, Drmota, T and Greasley, PJ (2007) The orphan receptor GPR55 is a novel cannabinoid receptor. British Journal Pharmacology 152(7), 10921101.CrossRefGoogle ScholarPubMed
Sarasola, LI, Del Torrent, CL, Pérez-Arévalo, A, Argerich, J, Casajuana-Martín, N, Chevigné, A, Fernández-Dueñas, V, Ferré, S, Pardo, L and Ciruela, F (2022) The ADORA1 mutation linked to early-onset Parkinson’s disease alters adenosine A1-A2A receptor heteromer formation and function. Biomedicine and Pharmacotherapy 156, 113896.CrossRefGoogle Scholar
Sebastião, AM, Ribeiro, JA (2009) Adenosine receptors and the central nervous system. Handbook Experimental Pharmacology 193, 471534.CrossRefGoogle Scholar
Stollenwerk, TM, Pollock, S and Hillard, CJ (2021) Contribution of the adenosine 2A receptor to behavioral effects of tetrahydrocannabinol, cannabidiol and PECS-101. Molecules 26(17), 5354.CrossRefGoogle ScholarPubMed
Sylantyev, S, Jensen, TP, Ross, RA and Rusakov, DA (2013) Cannabinoid- and lysophosphatidylinositol-sensitive receptor GPR55 boosts neurotransmitter release at central synapses. Proceedings of the National Academy of Sciences of the United States of America 110(13), 51935198.CrossRefGoogle ScholarPubMed
Van der Westhuizen, ET, Valant, C, Sexton, MC and Christopoulos, A (2015) Endogenous allosteric modulators of G protein-coupled receptors. Journal Pharmacology Experimental Therapeutics 353(2), 246260.CrossRefGoogle ScholarPubMed
Wan, Q, Okashah, N, Inoue, A, Nehmé, R, Carpenter, B, Tate, CG and Lambert, NA (2018) Mini G protein probes for active G protein-coupled receptors (GPCRs) in live cells. Journal Biological Chemistry 293(19), 74667473.CrossRefGoogle Scholar
Xiao, C, Gavrilova, O, Liu, N, Lewicki, SA, Reitman, ML and Jacobson, KA (2023) In vivo phenotypic validation of adenosine receptor-dependent activity of non-adenosine drugs. Purinergic Signal (in press).CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Determination of CBD effects on A2AR ligand binding affinity. (a) Schematic representation of the NanoBRET assay. A nanoluciferase is linked to the N-terminal part of the A2AR (A2ARNL). When the nanoluciferase substrate coelenterazine is added, A2ARNL (donor) emits light at 490–10 nm. Light excites the fluorescent selective A2AR ligand, MRS7396 (acceptor), which subsequently emits fluorescence at 650-80 nm. (b) NanoBRET saturation binding curves obtained by challenging A2ARNL expressing HEK-293T cells with increasing concentrations of MRS7396 in the absence/presence of CBD (1 µM) or ZM241385 (1 µM). (c) NanoBRET signals obtained by challenging A2ARNL expressing HEK-293T cells with a fixed concentration of MRS7396 (30 nM, normalised to 100%) in the presence of increasing concentrations of CBD or ZM241385. The represented data are mean ± SEM of three independent experiments each performed in triplicate.

Figure 1

Figure 2. Assessment of CBD effects on A2AR intrinsic activity. (a) Schematic representation of the NanoBIT™-based assay. The two fragments of nanoluciferase, small (SmBIT) and large (LgBIT), are fused to A2AR and mini-Gαs protein, respectively. Then, upon agonist binding, A2AR intrinsic activity is assessed by receptor recruitment of Gαs, which induces an increase on luminescence due to nanoluciferase reconstitution. (b) Representative time-course of A2AR agonist-mediated Gαs recruitment. The selective A2AR agonist CGS21680 was challenged to A2ARSmBiT and LgBiTmini-Gαs expressing HEK-293T cells in the absence/presence of increasing concentrations of CBD or ZM241385. The luminescent signal obtained after reconstitution of the nanoluciferase was assessed by calculating the area under the curve for each condition. Data are shown as mean ± SEM of three independent experiments with five replicates. *P < 0.05, ***P < 0.001, one-way ANOVA with Dunnett’s post hoc test. (c) cAMP accumulation was assessed on HEK-293T cells permanently expressing the A2ARNL. Cells were challenged with the selective A2AR agonist CGS21680 (100 nM, normalised to 100% of effect) in the absence/presence of increasing concentrations of CBD. Data are expressed as mean ± SEM of four independent experiments performed in triplicates. *P < 0.05, one-way ANOVA with Dunnett’s post hoc test.