Cannabigerol Action at Cannabinoid CB1 and CB2 Receptors and at CB1-CB2 Heteroreceptor Complexes

Gemma Navarro, Katia Varani, Irene Reyes-Resina, Verónica Sánchez de Medina, Rafael Rivas-Santisteban, Carolina Sánchez-Carnerero Callado, Fabrizio Vincenzi, Salvatore Casano, Carlos Ferreiro-Vera, Enric I Canela, Pier Andrea Borea, Xavier Nadal, Rafael Franco, Gemma Navarro, Katia Varani, Irene Reyes-Resina, Verónica Sánchez de Medina, Rafael Rivas-Santisteban, Carolina Sánchez-Carnerero Callado, Fabrizio Vincenzi, Salvatore Casano, Carlos Ferreiro-Vera, Enric I Canela, Pier Andrea Borea, Xavier Nadal, Rafael Franco

Abstract

Cannabigerol (CBG) is one of the major phytocannabinoids present in Cannabis sativa L. that is attracting pharmacological interest because it is non-psychotropic and is abundant in some industrial hemp varieties. The aim of this work was to investigate in parallel the binding properties of CBG to cannabinoid CB1 (CB1R) and CB2 (CB2R) receptors and the effects of the compound on agonist activation of those receptors and of CB1-CB2 heteroreceptor complexes. Using [3H]-CP-55940, CBG competed with low micromolar Ki values the binding to CB1R and CB2R. Homogeneous binding in living cells, which is only technically possible for the CB2R, provided a 152 nM Ki value. Also interesting, CBG competed the binding of [3H]-WIN-55,212-2 to CB2R but not to CB1R (Ki: 2.7 versus >30 μM). The phytocannabinoid modulated signaling mediated by receptors and receptor heteromers even at low concentrations of 0.1-1 μM. cAMP, pERK, β-arrestin recruitment and label-free assays in HEK-293T cells expressing the receptors and treated with endocannabinoids or selective agonists proved that CBG is a partial agonist of CB2R. The action on cells expressing heteromers was similar to that obtained in cells expressing the CB2R. The effect of CBG on CB1R was measurable but the underlying molecular mechanisms remain uncertain. The results indicate that CBG is indeed effective as regulator of endocannabinoid signaling.

Keywords: G-protein-coupled receptor; TR-FRET; cannabigerol; cannabinoid receptor; partial agonist; phytocannabinoid.

Figures

FIGURE 1
FIGURE 1
Radioligand binding assays to CB1R and CB2R. (A–D) Saturation curves of either [3H]-CP-55940 or [3H]-WIN-55,212-2 binding on membranes from CHO cells stably expressing human CB1R (A,C) or CB2R (B,D). (E,F) Competition curves for WIN-55,212-2 in radioligand-based assays using either [3H]-CP-55940 (E) or [3H]-WIN-55,212-2 (F) binding on membranes from CHO cells stably expressing human CB1R or CB2R. Data are expressed as the mean ± SEM of five independent experiments performed in duplicate. KD (obtained from saturation isotherms) are shown in Table 1.
FIGURE 2
FIGURE 2
Competition by CBG of agonist binding to CB1R and/or CB2R. (A,B) Competition curves for CBG in radioligand-based assays using either [3H]-CP-55940 (A) or [3H]-WIN-55,212-2 (B) binding on membranes from CHO cells stably expressing human CB1R or CB2R. (C) Scheme of the HTRF-based competitive binding assay. The GPCR of interest with the SNAP-tagged enzyme fused to its N-terminal domain is expressed at the cell surface. SNAP is a commercially available tag consisting of circa 180 amino acids, that can be labeled with fluorophores or other probes in a covalent fashion. The GPCR–SNAP-tagged cells are subsequently labeled with a Tb-containing probe (SNAP-Lumi4-Tb) through a covalent bond between the Tb and the reactive side of the SNAP enzyme. The Tb acts as FRET donor of an acceptor covalently linked to a selective CB2 receptor ligand. Thus, upon binding of a fluorophore-conjugated ligand (FRET acceptor) on the donor-labeled SNAP-tagged/GPCR fusion protein, an HTRF signal from the sensitized acceptor can be detected since the energy transfer can occur only when the donor and the acceptor are in close proximity. In competition binding assays using CM-157, the unlabelled specific ligand competes for receptor binding site with the fluorophore-conjugated ligand, leading to a decrease in the HTRF signal detected. (D–G) HEK-293T were transiently transfected with 1 μg cDNA for SNAP-CB2R in the absence (D,E) or presence of 0.5 μg cDNA for CB1R (F,G). Competition curves of specific binding of 20 nM fluorophore-conjugated CM-157 using CM-157 (0–10 μM) (D,F) or of CBG (0–10 μM) (E,G) as competitors are shown. Data represent the mean ± SEM of five experiments in triplicates.
FIGURE 3
FIGURE 3
Cannabigerol action in cells expressing CB1R or CB2R. HEK-293T cells were transfected with 0.75 μg cDNA for CB1R (red line) or 1 μg cDNA for CB2R (blue line). Dose–effect curves for cAMP production are expressed as % of levels obtained by 0.5 μM forskolin treatment (A). Dose-effect curves for ERK1/2 phosphorylation are expressed as % respect to basal levels (B). Dose-effect curves for β-arrestin recruitment (C) and label-free (D) assays are expressed, respectively, in mBRET units and pm. In β-arrestin-2 recruitment assays cells were transfected with 1 μg cDNA for β-arrestin-Rluc and either 0.75 μg cDNA for CB1R-YFP or 1 μg cDNA for CB2R-YFP. Data are the mean ± SEM of a representative experiment in triplicates (n = 6).
FIGURE 4
FIGURE 4
Effect of CBG on the action of CB1R and CB2R agonists. (A–D) HEK-293T cells were transfected with 0.75 μg cDNA for CB1R and treated with 100 nM AEA or a selective CB1R ligand (100 nM ACEA) in the absence (black bars) or presence of 100 nM (white bars) or 1 μM (gray bars) CBG. (E–H) HEK-293T cells were transfected with 1 μg cDNA for CB2R and treated with 100 nM AEA or a selective CB2R ligand (100 nM JWH133) in the absence (black bars) or presence of 100 nM (white bars) or 1 μM (gray bars) CBG. cAMP production (A,E) is expressed as % of levels obtained by 0.5 μM forskolin. ERK1/2 phosphorylation data are expressed as % respect to basal levels (B,F). In β-arrestin-2 recruitment assays cells were transfected with 1 μg cDNA for β-arrestin-Rluc and either 0.75 μg cDNA for CB1R-YFP or 1 μg cDNA for CB2R-YFP. Data for β-arrestin recruitment (C,G) and label-free (D,H) assays are expressed, respectively, in mBRET units and pm. Data represent the mean ± SEM of six different experiments performed with six replicates. One-way ANOVA and Bonferroni’s multiple comparison post hoc test were used for statistical analysis (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; versus treatment with AEA, ACEA, or JWH133 alone).
FIGURE 5
FIGURE 5
Effect of CBG in cells expressing CB1 and CB2 receptors. (A–D) Effect of CBG in HEK-293T cells transfected with 0.75 μg cDNA for CB1R and 1 μg cDNA for CB2R (A,B,D) or 1 μg cDNA for β-arrestin-Rluc, 0.75 μg cDNA for CB1R and 1 μg cDNA for CB2R-YFP (C). Dose–effect curves for cAMP production are expressed as % of levels obtained by 0.5 μM forskolin treatment (A). Dose-effect curves for ERK1/2 phosphorylation are expressed as % respect of basal levels (B). Dose-effect curves for β-arrestin recruitment (C) and label-free (D) assays are expressed, respectively, in mBRET units and pm. Dotted lines (red and blue) are the same than those shown in Figure 3 and serve as a reference for differential effects in cells coexpressing both receptors. Data are the mean ± SEM of a representative experiment in triplicates (n = 6). (E–H) HEK-293T cells transfected with 0.75 μg cDNA for CB1R and 1 μg cDNA for CB2R (E,F,H) or 1 μg cDNA for β-arrestin-Rluc, 0.75 μg cDNA for CB1R and 1 μg cDNA for CB2R-YFP (G) were treated with 100 nM AEA or a selective CB2R ligand (100 nM JWH133) in the absence (black bars) or presence (white bars) of 100 nM CBG. cAMP production (E) is expressed as % of levels obtained by 0.5 μM forskolin. ERK1/2 phosphorylation data are expressed as % respect of basal levels (F). Data for β-arrestin recruitment (G) and label-free (H) assays are expressed, respectively, in mBRET units and pm. Data represent the mean ± SEM of six different experiments performed with three replicates. One-way ANOVA and Bonferroni’s multiple comparison post hoc test were used for statistical analysis (∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001; versus treatment with AEA, ACEA, or JWH133 alone).

References

    1. Adams A. J., Banister S. D., Irizarry L., Trecki J., Schwartz M., Gerona R. (2017). Zombie” outbreak caused by the synthetic cannabinoid AMB-FUBINACA in New York. N. Engl. J. Med. 376 235–242. 10.1056/NEJMoa1610300
    1. Bayewitch M., Avidor-Reiss T., Levy R., Barg J., Mechoulam R., Vogel Z. (1995). The peripheral cannabinoid receptor: adenylate cyclase inhibition and G protein coupling. FEBS Lett. 375 143–147. 10.1016/0014-5793(95)01207-U
    1. Bolognini D., Grazia A., Parolaro D., Pertwee R. G. (2012). AM630 behaves as a protean ligand at the human cannabinoid CB2 receptor. Br. J. Pharmacol. 165 2561–2574. 10.1111/j.1476-5381.2011.01503.x
    1. Bonner I., Song Z. H., Bonner T. I. (1996). A lysine residue of the cannabinoid receptor is critical for receptor recognition by several agonists but not WIN55212-2. Mol. Pharmacol. 49 891–896.
    1. Borrelli F., Fasolino I., Romano B., Capasso R., Maiello F., Coppola D., et al. (2013). Beneficial effect of the non-psychotropic plant cannabinoid cannabigerol on experimental inflammatory bowel disease. Biochem. Pharmacol. 85 1306–1316. 10.1016/j.bcp.2013.01.017
    1. Callén L., Moreno E., Barroso-Chinea P., Moreno-Delgado D., Cortés A., Mallol J., et al. (2012). Cannabinoid receptors CB1 and CB2 form functional heteromers in brain. J. Biol. Chem. 287 20851–20865. 10.1074/jbc.M111.335273
    1. Cascio M. G., Gauson L. A., Stevenson L. A., Ross R. A., Pertwee R. G. (2010). Evidence that the plant cannabinoid cannabigerol is a highly potent α 2-adrenoceptor agonist and moderately potent 5HT 1A receptor antagonist. Br. J. Pharmacol. 159 129–141. 10.1111/j.1476-5381.2009.00515.x
    1. Cheng H. C. (2001). The power issue: determination of KB or Ki from IC50 - A closer look at the Cheng-Prusoff equation, the Schild plot and related power equations. J. Pharmacol. Toxicol. Methods 46 61–71. 10.1016/S1056-8719(02)00166-1
    1. Chin C. N., Lucas-Lenard J., Abadji V., Kendall D. A. (1998). Ligand binding and modulation of cyclic AMP levels depend on the chemical nature of residue 192 of the human cannabinoid receptor 1. J. Neurochem. 70 366–373.
    1. De Petrocellis L., Ligresti A., Moriello A. S., Allarà M., Bisogno T., Petrosino S., et al. (2011). Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br. J. Pharmacol. 163 1479–1494. 10.1111/j.1476-5381.2010.01166.x
    1. De Petrocellis L., Vellani V., Schiano-Moriello A., Marini P., Magherini P. C., Orlando P., et al. (2008). Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J. Pharmacol. Exp. Ther. 325 1007–1015. 10.1124/jpet.107.134809
    1. Diana M. A., Marty A. (2004). Endocannabinoid-mediated short-term synaptic plasticity: depolarization-induced suppression of inhibition (DSI) and depolarization- induced suppression of excitation (DSE). Br. J. Pharmacol. 142 9–19. 10.1038/sj.bjp.0705726
    1. Fellermeier M., Zenk M. H. (1998). Prenylation of olivetolate by a hemp transferase yields cannabigerolic acid, the precursor of tetrahydrocannabinol. FEBS Lett. 427 283–285. 10.1016/S0014-5793(98)00450-5
    1. Gaoni Y., Mechoulan R. (1964). Structure synthesis of cannabigerol new hashish constituent. Proc. Chem. Soc. Lond. 82 2189–2192.
    1. Gauson L. A., Stevenson L. A., Thomas A., Baillie G. L., Ross R. A., Pertwee R. G. (2007). “Cannabigerol behaves as a partial agonist at both CB1 and CB2 receptors,” in Proceedings of the 17th Annual Symposium on the Cannabinoids, (Burlington, VT: International Cannabinoid Research Society; ), 206.
    1. Georgieva T., Devanathan S., Stropova D., Park C. K., Salamon Z., Tollin G., et al. (2008). Unique agonist-bound cannabinoid CB1 receptor conformations indicate agonist specificity in signaling. Eur. J. Pharmacol. 581 19–29. 10.1016/j.ejphar.2007.11.053
    1. Giacoppo S., Gugliandolo A., Trubiani O., Pollastro F., Grassi G., Bramanti P., et al. (2017). Cannabinoid CB2 receptors are involved in the protection of RAW264.7 macrophages against the oxidative stress: An in vitro study. Eur. J. Histochem. 61 1–13. 10.4081/ejh.2017.2749
    1. Granja A. G., Carrillo-Salinas F., Pagani A., Gómez-Cañas M., Negri R., Navarrete C., et al. (2012). A cannabigerol quinone alleviates neuroinflammation in a chronic model of multiple sclerosis. J. Neuroimmune Pharmacol. 7 1002–1016. 10.1007/s11481-012-9399-3
    1. Grundmann M., Kostenis E. (2015a). Holistic methods for the analysis of cNMP effects. Handb. Exp. Pharmacol. 238 339–357. 10.1007/164_2015_42
    1. Grundmann M., Kostenis E. (2015b). Label-free biosensor assays in GPCR screening. Methods Mol. Biol. 1272 199–213. 10.1007/978-1-4939-2336-6_14
    1. Grunfeld Y., Edery H. (1969). Psychopharmacological activity of the active constituents of hashish and some related cannabinoids. Psychopharmacologia 14 200–210. 10.1007/BF00404218
    1. Hamamoto A., Kobayashi Y., Saito Y. (2015). Identification of amino acids that are selectively involved in Gi/o activation by rat melanin-concentrating hormone receptor 1. Cell. Signal. 27 818–827. 10.1016/j.cellsig.2015.01.008
    1. Hinz S., Navarro G., Borroto-Escuela D., Seibt B. F., Ammon C., de Filippo E., et al. (2018). Adenosine A2A receptor ligand recognition and signaling is blocked by A2B receptors. Oncotarget 9 13593–13611. 10.18632/oncotarget.24423
    1. Kebig A., Kostenis E., Mohr K., Mohr-Andrä M. (2009). An optical dynamic mass redistribution assay reveals biased signaling of dualsteric GPCR activators. J. Recept. Signal Transduct. 29 140–145. 10.1080/10799890903047437
    1. Lanciego J. L., Barroso-Chinea P., Rico A. J., Conte-Perales L., Callén L., Roda E., et al. (2011). Expression of the mRNA coding the cannabinoid receptor 2 in the pallidal complex of Macaca fascicularis. J. Psychopharmacol. 25 97–104. 10.1177/0269881110367732
    1. Laprairie R. B., Bagher A. M., Kelly M. E. M., Denovan-Wright E. M. (2015). Cannabidiol is a negative allosteric modulator of the cannabinoid CB1 receptor. Br. J. Pharmacol. 172 4790–4805. 10.1111/bph.13250
    1. Lu H.-C. C., Mackie K. (2016). an introduction to the endogenous cannabinoid system. Biol. Psychiatry 79 516–525. 10.1016/j.biopsych.2015.07.028
    1. Martínez-Pinilla E., Rabal O., Reyes-Resina I., Zamarbide M., Navarro G., Sanchez-Arias J. A., et al. (2016). Two affinity sites of the cannabinoid subtype 2 receptor identified by a novel homogeneous binding assay. J. Pharmacol. Exp. Ther. 358 580–587. 10.1124/jpet.116.234948
    1. Mcguinness D., Malikzay A., Visconti R., Lin K., Bayne M., Monsma F., et al. (2009). Characterizing cannabinoid CB2 receptor ligands using DiscoveRx PathHunterTM β-arrestin assay. J. Biomol. Screen. 14 49–58. 10.1177/1087057108327329
    1. McPartland J. M., Glass M., Pertwee R. G. (2007). Meta-analysis of cannabinoid ligand binding affinity and receptor distribution: Interspecies differences. Br. J. Pharmacol. 152 583–593. 10.1038/sj.bjp.0707399
    1. Mechoulam R. (2016). Cannabis - The Israeli perspective. J. Basic Clin. Physiol. Pharmacol. 27 181–187. 10.1515/jbcpp-2015-0091
    1. Mechoulam R., Shani A., Edery H., Grunfeld Y. (1970). Chemical basis of hashish activity. Science 169 611–612. 10.1126/science.169.3945.611
    1. Medrano M., Aguinaga D., Reyes-Resina I., Canela E. I., Mallol J., Navarro G., et al. (2017). Orexin A/Hypocretin modulates leptin receptor-mediated signaling by allosteric modulations mediated by the Ghrelin GHS-R1A receptor in hypothalamic neurons. Mol. Neurobiol. 55 4718–4730. 10.1007/s12035-017-0670-8
    1. Merighi S., Simioni C., Gessi S., Varani K., Borea P. A. (2010). Binding thermodynamics at the human cannabinoid CB1 and CB2 receptors. Biochem. Pharmacol. 79 471–477. 10.1016/j.bcp.2009.09.009
    1. Nadal X. (2016). Methods of purifying cannabinoids, compositions and kits thereof. U.S. Patent No 9765000 Washington, DC: U.S. Patent and Trademark Office.
    1. Nadal X., del Río C., Casano S., Palomares B., Ferreiro-Vera C., Navarrete C., et al. (2017). Tetrahydrocannabinolic acid is a potent PPARγ agonist with neuroprotective activity. Br. J. Pharmacol. 174 4263–4276. 10.1111/bph.14019
    1. Navarro G., Borroto-Escuela D., Angelats E., Etayo Í., Reyes-Resina I., Pulido-Salgado M., et al. (2018a). Receptor-heteromer mediated regulation of endocannabinoid signaling in activated microglia. Role of CB1 and CB2 receptors and relevance for Alzheimer’s disease and levodopa-induced dyskinesia. Brain Behav. Immun. 67 139–151. 10.1016/j.bbi.2017.08.015
    1. Navarro G., Cordomí A., Brugarolas M., Moreno E., Aguinaga D., Pérez-Benito L., et al. (2018b). Cross-communication between Gi and Gs in a G-protein-coupled receptor heterotetramer guided by a receptor C-terminal domain. BMC Biol. 16:24. 10.1186/s12915-018-0491-x
    1. Navarro G., Cordomí A., Zelman-Femiak M., Brugarolas M., Moreno E., Aguinaga D., et al. (2016). Quaternary structure of a G-protein-coupled receptor heterotetramer in complex with Gi and Gs. BMC Biol. 14:26. 10.1186/s12915-016-0247-4
    1. Navarro G., Ferré S., Cordomi A., Moreno E., Mallol J., Casadó V., et al. (2010). Interactions between intracellular domains as key determinants of the quaternary structure and function of receptor heteromers. J. Biol. Chem. 285 27346–27359. 10.1074/jbc.M110.115634
    1. Pertwee R. G. (2008). The diverse CB 1 and CB 2 receptor pharmacology of three plant cannabinoids: Δ 9-tetrahydrocannabinol, cannabidiol and Δ 9-tetrahydrocannabivarin. Br. J. Pharmacol. 153 199–215. 10.1038/sj.bjp.0707442
    1. Peters M. F., Scott C. W. (2009). Evaluating cellular impedance assays for detection of GPCR pleiotropic signaling and functional selectivity. J. Biomol. Screen. 14 246–255. 10.1177/1087057108330115
    1. Pollastro F., Taglialatela-Scafati O., Allarà M., Muñoz E., Di Marzo V., De Petrocellis L., et al. (2011). Bioactive prenylogous cannabinoid from fiber hemp (Cannabis sativa). J. Nat. Prod. 74 2019–2022. 10.1021/np200500p
    1. Rosenthaler S., Pöhn B., Kolmanz C., Nguyen Huu C., Krewenka C., Huber A., et al. (2014). Differences in receptor binding affinity of several phytocannabinoids do not explain their effects on neural cell cultures. Neurotoxicol. Teratol. 46 49–56. 10.1016/j.ntt.2014.09.003
    1. Schröder R., Merten N., Mathiesen J. M., Martini L., Kruljac-Letunic A., Krop F., et al. (2009). The C-terminal tail of CRTH2 is a key molecular determinant that constrains Gαi and downstream signaling cascade activation. J. Biol. Chem. 284 1324–1336. 10.1074/jbc.M806867200
    1. Shim J. Y., Howlett A. C. (2006). WIN55212-2 docking to the CB 1 cannabinoid receptor and multiple pathways for conformational induction. J. Chem. Inf. Model. 46 1286–1300. 10.1021/ci0504824
    1. Shim J. Y., Welsh W. J., Howlett A. C. (2003). Homology model of the CB1 cannabinoid receptor: sites critical for nonclassical cannabinoid agonist interaction. Biopolym. Pept. Sci. Sect. 71 169–189. 10.1002/bip.10424
    1. Sierra S., Luquin N., Rico A. J., Gómez-Bautista V., Roda E., Dopeso-Reyes I. G., et al. (2015). Detection of cannabinoid receptors CB1 and CB2 within basal ganglia output neurons in macaques: changes following experimental parkinsonism. Brain Struct. Funct. 220 2721–2738. 10.1007/s00429-014-0823-8
    1. Solymosi K., Kofalvi A. (2017). Cannabis: a treasure trove or pandora’s box? Mini Rev. Med. Chem. 17 1223–1291. 10.2174/1389557516666161004162133
    1. Turner S. E., Williams C. M., Iversen L., Whalley B. J. (2017). Molecular pharmacology of phytocannabinoids. Prog. Chem. Org. Nat. Prod. 103 61–101. 10.1007/978-3-319-45541-9_3
    1. Van Der Lee M. M. C., Blomenröhr M., Van Der Doelen A. A., Wat J. W. Y., Smits N., Hanson B. J., et al. (2009). Pharmacological characterization of receptor redistribution and β-arrestin recruitment assays for the cannabinoid receptor 1. J. Biomol. Screen. 14 811–823. 10.1177/1087057109337937
    1. Wartmann M., Campbell D., Subramanian A., Burstein S. H., Davis R. J. (1995). The MAP kinase signal transduction pathway is activated by the endogenous cannabinoid anandamide. FEBS Lett. 359 133–136. 10.1016/0014-5793(95)00027-7
    1. Weinstein A. M., Rosca P., Fattore L., London E. D. (2017). Synthetic cathinone and cannabinoid designer drugs pose a major risk for public health. Front. Psychiatry 8:156. 10.3389/fpsyt.2017.00156

Source: PubMed

3
Předplatit