The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies
Abstract
1. Introduction
2. Molecular Biology of the CB2 Receptor
3. Epigenetic and Transcriptional Regulation of the CB2 Receptor
4. CB2 Receptor Pharmacology
5. The Role of CB2 Receptor in Immune Regulation
6. CB2 Receptor and Microglia in Neuroinflammation
7. CB2 Receptor in Depression and Psychiatric Disorders
8. CB2 Receptor in Chronic Pain and Neuropathy
9. CB2 Receptor in Schizophrenia and Dopaminergic Circuits
10. Crosstalk Within the eCBome
11. Conclusions
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
Abbreviations
| 2-AG | 2-Arachidonoylglycerol |
| AEA | Anandamide (N-Arachidonoyl-Ethanolamine) |
| AKT/PKB | Protein Kinase B |
| Aβ | β-Amyloid Peptide |
| CB1 | Cannabinoid Receptor Type 1 |
| CB2 | Cannabinoid Receptor Type 2 |
| CBG | Cannabigerol |
| CBD | Cannabidiol |
| CNS | Central Nervous System |
| DAGLα | Diacylglycerol Lipase Alpha |
| DAGLβ | Diacylglycerol Lipase Beta |
| DRG | Dorsal Root Ganglia |
| EGFP | Enhanced Green Fluorescent Protein |
| ERK | Extracellular Signal-Regulated Kinase |
| ECS | Endocannabinoid System |
| eCBome | Endocannabinoidome |
| FAAH | Fatty Acid Amide Hydrolase |
| GPCRs | G Protein-Coupled Receptors |
| GRKs | G Protein-Coupled Receptor Kinases |
| Gαi/o | G Protein Alpha Subunits Gi/Go |
| IFN-γ | Interferon-Gamma |
| IL-1β | Interleukin-1 Beta |
| IL-6 | Interleukin-6 |
| IL-10 | Interleukin-10 |
| MAGL | Monoacylglycerol Lipase |
| M1 | Macrophage Phenotype 1 |
| M2 | Macrophage Phenotype 2 |
| MeCP | Methyl-CpG-Binding Proteins |
| MeCP2 | Methyl-CpG-Binding Protein 2 |
| miRNAs | MicroRNAs |
| NAMs | Negative Allosteric Modulators |
| NAPE-PLD | N-Acyl Phosphatidylethanolamine-Specific Phospholipase D |
| NF-κB | Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells |
| PAMs | Positive Allosteric Modulators |
| PPARγ | Peroxisome Proliferator-Activated Receptor-Gamma |
| PPARs | Peroxisome Proliferator-Activated Receptors |
| ROS | Reactive Oxygen Species |
| TET1/2/3 | Ten-Eleven Translocation Enzymes 1/2/3 |
| THC | Δ9-Tetrahydrocannabinol |
| TNF-α | Tumor Necrosis Factor-Alpha |
| TRP | Transient Receptor Potential Channels |
| TRPM8 | Transient Receptor Potential Melastatin Subtype 8 |
| TRPV1 | Transient Receptor Potential Vanilloid 1 |
References
- Gaoni, Y.; Mechoulam, R. Isolation, Structure, and Partial Synthesis of an Active Constituent of Hashish. J. Am. Chem. Soc. 1964, 86, 1646–1647. [Google Scholar] [CrossRef]
- Mechoulam, R.; Shvo, Y. Hashish—I: The structure of Cannabidiol. Tetrahedron 1963, 19, 2073–2078. [Google Scholar] [CrossRef]
- Turcotte, C.; Chouinard, F.; Lefebvre, J.S.; Flamand, N. Regulation of inflammation by cannabinoids, the endocannabinoids 2-arachidonoyl-glycerol and arachidonoyl-ethanolamide, and their metabolites. J. Leukoc. Biol. 2015, 97, 1049–1070. [Google Scholar] [CrossRef]
- Simard, M.; Archambault, A.-S.; Lavoie, J.-P.C.; Dumais, É.; Di Marzo, V.; Flamand, N. Biosynthesis and metabolism of endocannabinoids and their congeners from the monoacylglycerol and N-acyl-ethanolamine families. Biochem. Pharmacol. 2022, 205, 115261. [Google Scholar] [CrossRef] [PubMed]
- Galiegue, S.; Mary, S.; Marchand, J.; Dussossoy, D.; Carriere, D.; Carayon, P.; Bouaboula, M.; Shire, D.; Le Fur, G.; Casellas, P. Expression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulations. Eur. J. Biochem. 1995, 232, 54–61. [Google Scholar] [CrossRef] [PubMed]
- Matsuda, L.A.; Lolait, S.J.; Brownstein, M.J.; Young, A.C.; Bonner, T.I. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature 1990, 346, 561–564. [Google Scholar] [CrossRef] [PubMed]
- Simard, M.; Rakotoarivelo, V.; Di Marzo, V.; Flamand, N. Expression and Functions of the CB2 Receptor in Human Leukocytes. Front. Pharmacol. 2022, 13, 826400. [Google Scholar] [CrossRef]
- Munro, S.; Thomas, K.L.; Abu-Shaar, M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993, 365, 61–65. [Google Scholar] [CrossRef]
- Rakotoarivelo, V.; Mayer, T.Z.; Simard, M.; Flamand, N.; Di Marzo, V. The Impact of the CB2 Cannabinoid Receptor in Inflammatory Diseases: An Update. Molecules 2024, 29, 3381. [Google Scholar] [CrossRef]
- Turcotte, C.; Blanchet, M.-R.; LaViolette, M.; Flamand, N. The CB2 receptor and its role as a regulator of inflammation. Cell. Mol. Life Sci. 2016, 73, 4449–4470. [Google Scholar] [CrossRef]
- Brown, S.M.; Wager-Miller, J.; Mackie, K. Cloning and molecular characterization of the rat CB2 cannabinoid receptor. Biochim. Biophys. 2002, 1576, 255–264. [Google Scholar] [CrossRef]
- Shire, D.; Calandra, B.; Rinaldi-Carmona, M.; Oustric, D.; Pessegue, B.; Bonnin-Cabanne, O.; Le Fur, G.; Caput, D.; Ferrara, P. Molecular cloning, expression and function of the murine CB2 peripheral cannabinoid receptor. Biochim. Biophys. Acta (BBA) Gene Struct. Expr. 1996, 1307, 132–136. [Google Scholar] [CrossRef]
- Griffin, G.; Tao, Q.; Abood, M.E. Cloning and pharmacological characterization of the rat CB(2) cannabinoid receptor. J. Pharmacol. Exp. Ther. 2000, 292, 886–894. [Google Scholar] [CrossRef] [PubMed]
- Iannotti, F.A.; Di Marzo, V.; Petrosino, S. Endocannabinoids and endocannabinoid-related mediators: Targets, metabolism and role in neurological disorders. Prog. Lipid Res. 2016, 62, 107–128. [Google Scholar] [CrossRef] [PubMed]
- Olabiyi, B.F.; Schmoele, A.-C.; Beins, E.C.; Zimmer, A. Pharmacological blockade of cannabinoid receptor 2 signaling does not affect LPS/IFN-γ-induced microglial activation. Sci. Rep. 2023, 13, 11105. [Google Scholar] [CrossRef]
- Grabon, W.; Ruiz, A.; Gasmi, N.; Degletagne, C.; Georges, B.; Belmeguenai, A.; Bodennec, J.; Rheims, S.; Marcy, G.; Bezin, L. CB2 expression in mouse brain: From mapping to regulation in microglia under inflammatory conditions. J. Neuroinflamm. 2024, 21, 206. [Google Scholar] [CrossRef]
- Holloman, B.L.; Nagarkatti, M.; Nagarkatti, P. Epigenetic Regulation of Cannabinoid-Mediated Attenuation of Inflammation and Its Impact on the Use of Cannabinoids to Treat Autoimmune Diseases. Int. J. Mol. Sci. 2021, 22, 7302. [Google Scholar] [CrossRef]
- Börner, C.; Martella, E.; Höllt, V.; Kraus, J. Regulation of Opioid and Cannabinoid Receptor Genes in Human Neuroblastoma and T Cells by the Epigenetic Modifiers Trichostatin A and 5-Aza-2′-Deoxycytidine. Neuroimmunomodulation 2012, 19, 180–186. [Google Scholar] [CrossRef]
- Ghosh, K.; Zhang, G.-F.; Chen, H.; Chen, S.-R.; Pan, H.-L. Cannabinoid CB2 receptors are upregulated via bivalent histone modifications and control primary afferent input to the spinal cord in neuropathic pain. J. Biol. Chem. 2022, 298, 101999. [Google Scholar] [CrossRef]
- Meccariello, R.; Santoro, A.; D’Angelo, S.; Morrone, R.; Fasano, S.; Viggiano, A.; Pierantoni, R. The Epigenetics of the Endocannabinoid System. Int. J. Mol. Sci. 2020, 21, 1113. [Google Scholar] [CrossRef]
- Weizman, N.F.; A Wyse, B.; Szaraz, P.; Defer, M.; Jahangiri, S.; Librach, C.L. Cannabis alters epigenetic integrity and endocannabinoid signalling in the human follicular niche. Hum. Reprod. 2021, 36, 1922–1931. [Google Scholar] [CrossRef]
- Smith, R.C.; Sershen, H.; Janowsky, D.S.; Lajtha, A.; Grieco, M.; Gangoiti, J.A.; Gertsman, I.; Johnson, W.S.; Marcotte, T.D.; Davis, J.M. Changes in Expression of DNA-Methyltransferase and Cannabinoid Receptor mRNAs in Blood Lymphocytes After Acute Cannabis Smoking. Front. Psychiatry 2022, 13, 887700. [Google Scholar] [CrossRef]
- Innocenzi, E.; De Domenico, E.; Ciccarone, F.; Zampieri, M.; Rossi, G.; Cicconi, R.; Bernardini, R.; Mattei, M.; Grimaldi, P. Paternal activation of CB2 cannabinoid receptor impairs placental and embryonic growth via an epigenetic mechanism. Sci. Rep. 2019, 9, 17034. [Google Scholar] [CrossRef] [PubMed]
- Nan, X.; Ng, H.-H.; Johnson, C.A.; Laherty, C.D.; Turner, B.M.; Eisenman, R.N.; Bird, A. Transcriptional repression by the methyl-CpG-binding protein MeCP2 involves a histone deacetylase complex. Nature 1998, 393, 386–389. [Google Scholar] [CrossRef]
- Bird, A. DNA methylation patterns and epigenetic memory. Genes Dev. 2002, 16, 6–21. [Google Scholar] [CrossRef] [PubMed]
- Jones, P.L.; Veenstra, G.J.C.; Wade, P.A.; Vermaak, D.; Kass, S.U.; Landsberger, N.; Strouboulis, J.; Wolffe, A.P. Methylated DNA and MeCP2 recruit histone deacetylase to repress transcription. Nat. Genet. 1998, 19, 187–191. [Google Scholar] [CrossRef] [PubMed]
- Kouzarides, T. Histone methylation in transcriptional control. Curr. Opin. Genet. Dev. 2002, 12, 198–209. [Google Scholar] [CrossRef]
- Fuks, F.; Hurd, P.J.; Wolf, D.; Nan, X.; Bird, A.P.; Kouzarides, T. The methyl-CpG-binding protein MeCP2 links DNA methylation to histone methylation. J. Biol. Chem. 2003, 278, 4035–4040. [Google Scholar] [CrossRef]
- Basavarajappa, B.S.; Subbanna, S. Molecular Insights into Epigenetics and Cannabinoid Receptors. Biomolecules 2022, 12, 1560. [Google Scholar] [CrossRef]
- Aguado, T.; Carracedo, A.; Julien, B.; Velasco, G.; Milman, G.; Mechoulam, R.; Alvarez, L.; Guzmán, M.; Galve-Roperh, I. Cannabinoids Induce Glioma Stem-like Cell Differentiation and Inhibit Gliomagenesis. J. Biol. Chem. 2007, 282, 6854–6862. [Google Scholar] [CrossRef]
- Kikuchi, M.; Morita, S.; Wakamori, M.; Sato, S.; Uchikubo-Kamo, T.; Suzuki, T.; Dohmae, N.; Shirouzu, M.; Umehara, T. Epigenetic mechanisms to propagate histone acetylation by p300/CBP. Nat. Commun. 2023, 14, 4103. [Google Scholar] [CrossRef]
- Zhang, T.; Cooper, S.; Brockdorff, N. The interplay of histone modifications—Writers that read. EMBO Rep. 2015, 16, 1467–1481. [Google Scholar] [CrossRef]
- Ghare, S.S.; Joshi-Barve, S.; Moghe, A.; Patil, M.; Barker, D.F.; Gobejishvili, L.; Brock, G.N.; Cave, M.; McClain, C.J.; Barve, S.S. Coordinated histone H3 methylation and acetylation regulates physiologic and pathologic Fas Ligand gene expression in human CD4+ T cells. J. Immunol. 2014, 193, 412–421. [Google Scholar] [CrossRef]
- Dou, Y.; Milne, T.A.; Tackett, A.J.; Smith, E.R.; Fukuda, A.; Wysocka, J.; Allis, C.D.; Chait, B.T.; Hess, J.L.; Roeder, R.G. Physical association and coordinate function of the H3 K4 methyltransferase MLL1 and the H4 K16 acetyltransferase MOF. Cell 2005, 121, 873–885. [Google Scholar] [CrossRef] [PubMed]
- Mozzetta, C.; Pontis, J.; Fritsch, L.; Robin, P.; Portoso, M.; Proux, C.; Margueron, R.; Ait-Si-Ali, S. The Histone H3 Lysine 9 Methyltransferases G9a and GLP Regulate Polycomb Repressive Complex 2-Mediated Gene Silencing. Mol. Cell 2014, 53, 277–289. [Google Scholar] [CrossRef]
- Yu, J.; Yang, W.; Wang, W.; Wang, Z.; Pu, Y.; Chen, H.; Wang, F.; Qian, J. Involvement of miR-665 in protection effect of dexmedetomidine against Oxidative Stress Injury in myocardial cells via CB2 and CK. Biomed. Pharmacother. 2019, 115, 108894. [Google Scholar] [CrossRef] [PubMed]
- Weis, F.; Beiras-Fernandez, A.; Sodian, R.; Kaczmarek, I.; Reichart, B.; Beiras, A.; Schelling, G.; Kreth, S. Substantially altered expression pattern of cannabinoid receptor 2 and activated endocannabinoid system in patients with severe heart failure. J. Mol. Cell. Cardiol. 2010, 48, 1187–1193. [Google Scholar] [CrossRef] [PubMed]
- Möhnle, P.; Schütz, S.V.; Schmidt, M.; Hinske, C.; Hübner, M.; Heyn, J.; Beiras-Fernandez, A.; Kreth, S. MicroRNA-665 is involved in the regulation of the expression of the cardioprotective cannabinoid receptor CB2 in patients with severe heart failure. Biochem. Biophys. Res. Commun. 2014, 451, 516–521. [Google Scholar] [CrossRef] [PubMed]
- Hashiesh, H.M.; Sharma, C.; Goyal, S.N.; Jha, N.K.; Ojha, S. Pharmacological Properties, Therapeutic Potential and Molecular Mechanisms of JWH133, a CB2 Receptor-Selective Agonist. Front. Pharmacol. 2021, 12, 702675. [Google Scholar] [CrossRef]
- Murataeva, N.; Mackie, K.; Straiker, A. The CB2-preferring agonist JWH015 also potently and efficaciously activates CB1 in autaptic hippocampal neurons. Pharmacol. Res. 2012, 66, 437–442. [Google Scholar] [CrossRef][Green Version]
- Hanuš, L.; Breuer, A.; Tchilibon, S.; Shiloah, S.; Goldenberg, D.; Horowitz, M.; Pertwee, R.G.; Ross, R.A.; Mechoulam, R.; Fride, E. HU-308: A specific agonist for CB2, a peripheral cannabinoid receptor. Proc. Natl. Acad. Sci. USA 1999, 96, 14228–14233. [Google Scholar] [CrossRef]
- An, D.; Peigneur, S.; Tytgat, J. WIN55,212-2, a Dual Modulator of Cannabinoid Receptors and G Protein-Coupled Inward Rectifier Potassium Channels. Biomedicines 2021, 9, 484. [Google Scholar] [CrossRef]
- Pertwee, R.G.; Howlett, A.C.; Abood, M.E.; Alexander, S.P.H.; Di Marzo, V.; Elphick, M.R.; Greasley, P.J.; Hansen, H.S.; Kunos, G.; Mackie, K.; et al. International Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid Receptors and Their Ligands: Beyond CB1 and CB2. Pharmacol. Rev. 2010, 62, 588–631. [Google Scholar] [CrossRef]
- Polini, B.; Cervetto, C.; Carpi, S.; Pelassa, S.; Gado, F.; Ferrisi, R.; Bertini, S.; Nieri, P.; Marcoli, M.; Manera, C. Positive Allosteric Modulation of CB1 and CB2 Cannabinoid Receptors Enhances the Neuroprotective Activity of a Dual CB1R/CB2R Orthosteric Agonist. Life 2020, 10, 333. [Google Scholar] [CrossRef]
- Petrucci, V.; Chicca, A.; Glasmacher, S.; Paloczi, J.; Cao, Z.; Pacher, P.; Gertsch, J. Pepcan-12 (RVD-hemopressin) is a CB2 receptor positive allosteric modulator constitutively secreted by adrenals and in liver upon tissue damage. Sci. Rep. 2017, 7, 333. [Google Scholar] [CrossRef] [PubMed]
- Gado, F.; Mohamed, K.A.; Meini, S.; Ferrisi, R.; Bertini, S.; Digiacomo, M.; D’aNdrea, F.; Stevenson, L.A.; Laprairie, R.B.; Pertwee, R.G.; et al. Variously substituted 2-oxopyridine derivatives: Extending the structure-activity relationships for allosteric modulation of the cannabinoid CB2 receptor. Eur. J. Med. Chem. 2021, 211, 113116. [Google Scholar] [CrossRef] [PubMed]
- Pandey, P.; Roy, K.K.; Doerksen, R.J. Negative Allosteric Modulators of Cannabinoid Receptor 2: Protein Modeling, Binding Site Identification and Molecular Dynamics Simulations in the Presence of an Orthosteric Agonist. J. Biomol. Struct. Dyn. 2019, 38, 32–47. [Google Scholar] [CrossRef]
- Rzeczycki, P.; Rasner, C.; Lammlin, L.; Junginger, L.; Goldman, S.; Bergman, R.; Redding, S.; Knights, A.; Elliott, M.; Maerz, T. Cannabinoid receptor type 2 is upregulated in synovium following joint injury and mediates anti-inflammatory effects in synovial fibroblasts and macrophages. Osteoarthr. Cartil. 2021, 29, 1720–1731. [Google Scholar] [CrossRef] [PubMed]
- Tarique, A.A.; Evron, T.; Zhang, G.; Tepper, M.A.; Morshed, M.M.; Andersen, I.S.; Begum, N.; Sly, P.D.; Fantino, E. Anti-inflammatory effects of lenabasum, a cannabinoid receptor type 2 agonist, on macrophages from cystic fibrosis. J. Cyst. Fibros. 2020, 19, 823–829. [Google Scholar] [CrossRef]
- Gaffal, E.; Kemter, A.M.; Scheu, S.; Dantas, R.L.; Vogt, J.; Baune, B.; Tüting, T.; Zimmer, A.; Alferink, J. Cannabinoid Receptor 2 Modulates Maturation of Dendritic Cells and Their Capacity to Induce Hapten-Induced Contact Hypersensitivity. Int. J. Mol. Sci. 2020, 21, 475. [Google Scholar] [CrossRef]
- Adhikary, S.; Kocieda, V.P.; Yen, J.-H.; Tuma, R.F.; Ganea, D. Signaling through cannabinoid receptor 2 suppresses murine dendritic cell migration by inhibiting matrix metalloproteinase 9 expression. Blood 2012, 120, 3741–3749. [Google Scholar] [CrossRef] [PubMed]
- Kong, W.; Li, H.; Tuma, R.F.; Ganea, D. Selective CB2 receptor activation ameliorates EAE by reducing Th17 differentiation and immune cell accumulation in the CNS. Cell. Immunol. 2014, 287, 3741–3749. [Google Scholar] [CrossRef]
- Sido, J.M.; Nagarkatti, P.S.; Nagarkatti, M. Production of endocannabinoids by activated T cells and B cells modulates inflammation associated with delayed-type hypersensitivity. Eur. J. Immunol. 2016, 46, 1472–1479. [Google Scholar] [CrossRef]
- Khalil, M.; Alliger, K.; Weidinger, C.; Yerinde, C.; Wirtz, S.; Becker, C.; Engel, M.A. Functional Role of Transient Receptor Potential Channels in Immune Cells and Epithelia. Front. Immunol. 2018, 9, 174. [Google Scholar] [CrossRef]
- Perry, V.H.; Teeling, J. Microglia and macrophages of the central nervous system: The contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin. Immunopathol. 2013, 35, 601–612. [Google Scholar] [CrossRef] [PubMed]
- Grabon, W.; Rheims, S.; Smith, J.; Bodennec, J.; Belmeguenai, A.; Bezin, L. CB2 receptor in the CNS: From immune and neuronal modulation to behavior. Neurosci. Biobehav. Rev. 2023, 150, 105226. [Google Scholar] [CrossRef] [PubMed]
- Gallelli, C.A.; Calcagnini, S.; Romano, A.; Koczwara, J.B.; De Ceglia, M.; Dante, D.; Villani, R.; Giudetti, A.M.; Cassano, T.; Gaetani, S. Modulation of the Oxidative Stress and Lipid Peroxidation by Endocannabinoids and Their Lipid Analogues. Antioxidants 2018, 7, 93. [Google Scholar] [CrossRef]
- Tanaka, M.; Sackett, S.; Zhang, Y. Endocannabinoid Modulation of Microglial Phenotypes in Neuropathology. Front. Neurol. 2020, 11, 87. [Google Scholar] [CrossRef]
- Askari, V.R.; Shafiee-Nick, R. The protective effects of β-caryophyllene on LPS-induced primary microglia M1/M2 imbalance: A mechanistic evaluation. Life Sci. 2019, 219, 40–73. [Google Scholar] [CrossRef]
- Ma, L.; Niu, W.; Lv, J.; Jia, J.; Zhu, M.; Yang, S. PGC-1α-Mediated Mitochondrial Biogenesis is Involved in Cannabinoid Receptor 2 Agonist AM1241-Induced Microglial Phenotype Amelioration. Cell. Mol. Neurobiol. 2018, 38, 1529–1537. [Google Scholar] [CrossRef]
- Malek, N.; Popiolek-Barczyk, K.; Mika, J.; Przewlocka, B.; Starowicz, K. Anandamide, Acting via CB2 Receptors, Alleviates LPS-Induced Neuroinflammation in Rat Primary Microglial Cultures. Neural Plast. 2015, 2015, 130639. [Google Scholar] [CrossRef] [PubMed]
- Vuic, B.; Milos, T.; Tudor, L.; Konjevod, M.; Perkovic, M.N.; Jembrek, M.J.; Erjavec, G.N.; Strac, D.S. Cannabinoid CB2 Receptors in Neurodegenerative Proteinopathies: New Insights and Therapeutic Potential. Biomedicines 2022, 10, 3000. [Google Scholar] [CrossRef] [PubMed]
- Aso, E.; Juvés, S.; Maldonado, R.; Ferrer, I. CB2 Cannabinoid Receptor Agonist Ameliorates Alzheimer-Like Phenotype in AβPP/PS1 Mice. J. Alzheimer’s Dis. 2013, 35, 847–858. [Google Scholar] [CrossRef]
- Navarro-Dorado, J.; Villalba, N.; Prieto, D.; Brera, B.; Martín-Moreno, A.M.; Tejerina, T.; de Ceballos, M.L. Vascular Dysfunction in a Transgenic Model of Alzheimer’s Disease: Effects of CB1R and CB2R Cannabinoid Agonists. Front. Neurosci. 2016, 10, 422. [Google Scholar] [CrossRef] [PubMed]
- Köfalvi, A.; Lemos, C.; Martín-Moreno, A.M.; Pinheiro, B.S.; García-García, L.; Pozo, M.A.; Valério-Fernandes, Â.; Beleza, R.O.; Agostinho, P.; Rodrigues, R.J.; et al. Stimulation of brain glucose uptake by cannabinoid CB2 receptors and its therapeutic potential in Alzheimer’s disease. Neuropharmacology 2016, 110 Pt A, 519–529. [Google Scholar] [CrossRef]
- Long, C.-M.; Zheng, Q.-X.; Zhou, Y.; Liu, Y.-T.; Gong, L.-P.; Zeng, Y.-C.; Liu, S. N-linoleyltyrosine exerts neuroprotective effects in APP/PS1 transgenic mice via cannabinoid receptor-mediated autophagy. J. Pharmacol. Sci. 2021, 147, 315–324. [Google Scholar] [CrossRef]
- Rivas-Santisteban, R.; Lillo, A.; Lillo, J.; Rebassa, J.-B.; Contestí, J.S.; Saura, C.A.; Franco, R.; Navarro, G. N-Methyl-D-aspartate (NMDA) and cannabinoid CB2 receptors form functional complexes in cells of the central nervous system: Insights into the therapeutic potential of neuronal and microglial NMDA receptors. Alzheimer’s Res. Ther. 2021, 13, 184. [Google Scholar] [CrossRef]
- Li, C.; Shi, J.; Wang, B.; Li, J.; Jia, H. CB2 cannabinoid receptor agonist ameliorates novel object recognition but not spatial memory in transgenic APP/PS1 mice. Neurosci. Lett. 2019, 707, 134286. [Google Scholar] [CrossRef]
- Wu, J.; Bie, B.; Yang, H.; Xu, J.J.; Brown, D.L.; Naguib, M. Activation of the CB2 receptor system reverses amyloid-induced memory deficiency. Neurobiol. Aging 2013, 34, 791–804. [Google Scholar] [CrossRef]
- Wu, J.; Hocevar, M.; Foss, J.F.; Bie, B.; Naguib, M. Activation of CB2 receptor system restores cognitive capacity and hippocampal Sox2 expression in a transgenic mouse model of Alzheimer’s disease. Eur. J. Pharmacol. 2017, 811, 12–20. [Google Scholar] [CrossRef]
- López, A.; Aparicio, N.; Pazos, M.R.; Grande, M.T.; Barreda-Manso, M.A.; Benito-Cuesta, I.; Vázquez, C.; Amores, M.; Ruiz-Pérez, G.; García-García, E.; et al. Cannabinoid CB2 receptors in the mouse brain: Relevance for Alzheimer’s disease. J. Neuroinflamm. 2018, 15, 158. [Google Scholar] [CrossRef] [PubMed]
- Esteban, S.R.d.M.; Benito-Cuesta, I.; Terradillos, I.; Martínez-Relimpio, A.M.; Arnanz, M.A.; Ruiz-Pérez, G.; Korn, C.; Raposo, C.; Sarott, R.C.; Westphal, M.V.; et al. Cannabinoid CB2 Receptors Modulate Microglia Function and Amyloid Dynamics in a Mouse Model of Alzheimer’s Disease. Front. Pharmacol. 2022, 13, 841766. [Google Scholar] [CrossRef]
- Foley, P.B.; Hare, D.J.; Double, K.L. A brief history of brain iron accumulation in Parkinson disease and related disorders. J. Neural Transm. 2022, 129, 505–520. [Google Scholar] [CrossRef]
- Kibret, B.G.; Ishiguro, H.; Horiuchi, Y.; Onaivi, E.S. New Insights and Potential Therapeutic Targeting of CB2 Cannabinoid Receptors in CNS Disorders. Int. J. Mol. Sci. 2022, 23, 975. [Google Scholar] [CrossRef]
- García, M.C.; Cinquina, V.; Palomo-Garo, C.; Rábano, A.; Fernández-Ruiz, J. Identification of CB2 receptors in human nigral neurons that degenerate in Parkinson’s disease. Neurosci. Lett. 2015, 587, 1–4. [Google Scholar] [CrossRef]
- Concannon, R.M.; Okine, B.N.; Finn, D.P.; Dowd, E. Upregulation of the cannabinoid CB2 receptor in environmental and viral inflammation-driven rat models of Parkinson’s disease. Exp. Neurol. 2016, 283, 204–212. [Google Scholar] [CrossRef]
- Wi, R.; Chung, Y.C.; Jin, B.K. Functional Crosstalk between CB and TRPV1 Receptors Protects Nigrostriatal Dopaminergic Neurons in the MPTP Model of Parkinson’s Disease. J. Immunol. Res. 2020, 2020, 5093493. [Google Scholar] [CrossRef]
- Yu, H.; Liu, X.; Chen, B.; Vickstrom, C.R.; Friedman, V.; Kelly, T.J.; Bai, X.; Zhao, L.; Hillard, C.J.; Liu, Q.-S. The Neuroprotective Effects of the CB2 Agonist GW842166x in the 6-OHDA Mouse Model of Parkinson’s Disease. Cells 2021, 10, 3548. [Google Scholar] [CrossRef]
- Ni, R.; Mu, L.; Ametamey, S. Positron emission tomography of type 2 cannabinoid receptors for detecting inflammation in the central nervous system. Acta Pharmacol. Sin. 2018, 40, 351–357. [Google Scholar] [CrossRef] [PubMed]
- Farooq, R.K.; Asghar, K.; Kanwal, S.; Zulqernain, A. Role of inflammatory cytokines in depression: Focus on interleukin-1β. Biomed. Rep. 2016, 6, 15–20. [Google Scholar] [CrossRef] [PubMed]
- Sălcudean, A.; Bodo, C.-R.; Popovici, R.-A.; Cozma, M.-M.; Păcurar, M.; Crăciun, R.-E.; Crisan, A.-I.; Enatescu, V.-R.; Marinescu, I.; Cimpian, D.-M.; et al. Neuroinflammation—A Crucial Factor in the Pathophysiology of Depression—A Comprehensive Review. Biomolecules 2025, 15, 502. [Google Scholar] [CrossRef]
- Maes, M.; Rachayon, M.; Jirakran, K.; Sughondhabirom, A.; Almulla, A.F.; Sodsai, P. Role of T and B lymphocyte cannabinoid type 1 and 2 receptors in major depression and suicidal behaviours. Acta Neuropsychiatr. 2023, 36, 287–298. [Google Scholar] [CrossRef]
- García-Gutiérrez, M.S.; Manzanares, J. Overexpression of CB2 cannabinoid receptors decreased vulnerability to anxiety and impaired anxiolytic action of alprazolam in mice. J. Psychopharmacol. 2010, 25, 111–120. [Google Scholar] [CrossRef]
- Stempel, A.V.; Stumpf, A.; Zhang, H.-Y.; Özdoğan, T.; Pannasch, U.; Theis, A.-K.; Otte, D.-M.; Wojtalla, A.; Rácz, I.; Ponomarenko, A.; et al. Cannabinoid Type 2 Receptors Mediate a Cell Type-Specific Plasticity in the Hippocampus. Neuron 2016, 90, 795–809. [Google Scholar] [CrossRef]
- Hwang, E.-S.; Kim, H.-B.; Lee, S.; Kim, M.-J.; Kim, K.-J.; Han, G.; Han, S.-Y.; Lee, E.-A.; Yoon, J.-H.; Kim, D.-O.; et al. Antidepressant-like effects of β-caryophyllene on restraint plus stress-induced depression. Behav. Brain Res. 2020, 380, 112439. [Google Scholar] [CrossRef]
- Galán-Ganga, M.; del Río, R.; Jiménez-Moreno, N.; Díaz-Guerra, M.; Lastres-Becker, I. Cannabinoid CB2 Receptor Modulation by the Transcription Factor NRF2 is Specific in Microglial Cells. Cell. Mol. Neurobiol. 2019, 40, 167–177. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Daksh, R.; Khanna, S.; Mudgal, J.; Lewis, S.A.; Arora, D.; Nampoothiri, M. Microglial cannabinoid receptor 2 and epigenetic regulation: Implications for the treatment of depression. Eur. J. Pharmacol. 2025, 995, 177422. [Google Scholar] [CrossRef] [PubMed]
- Gallego-Landin, I.; García-Baos, A.; Castro-Zavala, A.; Valverde, O. Reviewing the Role of the Endocannabinoid System in the Pathophysiology of Depression. Front. Pharmacol. 2021, 12, 762738. [Google Scholar] [CrossRef] [PubMed]
- Manzanares, J.; Julian, M.D.; Carrascosa, A. Role of the Cannabinoid System in Pain Control and Therapeutic Implications for the Management of Acute and Chronic Pain Episodes. Curr. Neuropharmacol. 2006, 4, 239–257. [Google Scholar] [CrossRef]
- Guindon, J.; Hohmann, A.G. Cannabinoid CB2 receptors: A therapeutic target for the treatment of inflammatory and neuropathic pain. Br. J. Pharmacol. 2008, 153, 319–334. [Google Scholar] [CrossRef]
- Ibrahim, M.M.; Deng, H.; Zvonok, A.; Cockayne, D.A.; Kwan, J.; Mata, H.P.; Vanderah, T.W.; Lai, J.; Porreca, F.; Makriyannis, A.; et al. Activation of CB2 cannabinoid receptors by AM1241 inhibits experimental neuropathic pain: Pain inhibition by receptors not present in the CNS. Proc. Natl. Acad. Sci. USA 2003, 100, 10529–10533. [Google Scholar] [CrossRef]
- Curto-Reyes, V.; Boto, T.; Hidalgo, A.; Menéndez, L.; Baamonde, A. Antinociceptive effects induced through the stimulation of spinal cannabinoid type 2 receptors in chronically inflamed mice. Eur. J. Pharmacol. 2011, 668, 184–189. [Google Scholar] [CrossRef]
- Brownjohn, P.; Ashton, J. Spinal cannabinoid CB2 receptors as a target for neuropathic pain: An investigation using chronic constriction injury. Neuroscience 2012, 203, 180–193. [Google Scholar] [CrossRef]
- Ashton, J.C.; Glass, M. The Cannabinoid CB2 Receptor as a Target for Inflammation-Dependent Neurodegeneration. Curr. Neuropharmacol. 2007, 5, 73–80. [Google Scholar] [CrossRef]
- Shiue, S.-J.; Peng, H.-Y.; Lin, C.-R.; Wang, S.-W.; Rau, R.-H.; Cheng, J.-K. Continuous Intrathecal Infusion of Cannabinoid Receptor Agonists Attenuates Nerve Ligation–Induced Pain in Rats. Reg. Anesthesia Pain Med. 2017, 42, 499–506. [Google Scholar] [CrossRef] [PubMed]
- Beltramo, M.; Bernardini, N.; Bertorelli, R.; Campanella, M.; Nicolussi, E.; Fredduzzi, S.; Reggiani, A. CB2 receptor-mediated antihyperalgesia: Possible direct involvement of neural mechanisms. Eur. J. Neurosci. 2006, 23, 1530–1538. [Google Scholar] [CrossRef] [PubMed]
- Racz, I.; Nadal, X.; Alferink, J.; Baños, J.E.; Rehnelt, J.; Martín, M.; Pintado, B.; Gutierrez-Adan, A.; Sanguino, E.; Manzanares, J.; et al. Crucial Role of CB2 Cannabinoid Receptor in the Regulation of Central Immune Responses during Neuropathic Pain. J. Neurosci. 2008, 28, 12125–12135. [Google Scholar] [CrossRef] [PubMed]
- Romero-Sandoval, A.; Nutile-McMenemy, N.; DeLeo, J.A. Spinal microglial and perivascular cell cannabinoid receptor type 2 activation reduces behavioral hypersensitivity without tolerance after peripheral nerve injury. Anesthesiology 2008, 108, 722–734. [Google Scholar] [CrossRef]
- Duffy, S.S.; Hayes, J.P.; Fiore, N.T.; Moalem-Taylor, G. The cannabinoid system and microglia in health and disease. Neuropharmacology 2021, 190, 108555. [Google Scholar] [CrossRef]
- Yao, B.B.; Hsieh, G.C.; Frost, J.M.; Fan, Y.; Garrison, T.R.; Daza, A.V.; Grayson, G.K.; Zhu, C.Z.; Pai, M.; Chandran, P.; et al. In vitro and in vivo characterization of A-796260: A selective cannabinoid CB2 receptor agonist exhibiting analgesic activity in rodent pain models. Br. J. Pharmacol. 2008, 153, 390–401. [Google Scholar] [CrossRef]
- Fu, W.; Taylor, B.K. Activation of Cannabinoid CB2 receptors Reduces Hyperalgesia in an Experimental Autoimmune Encephalomyelitis Mouse Model of Multiple Sclerosis. Neurosci. Lett. 2015, 595, 1–6. [Google Scholar] [CrossRef]
- Klauke, A.-L.; Racz, I.; Pradier, B.; Markert, A.; Zimmer, A.; Gertsch, J.; Zimmer, A. The cannabinoid CB2 receptor-selective phytocannabinoid beta-caryophyllene exerts analgesic effects in mouse models of inflammatory and neuropathic pain. Eur. Neuropsychopharmacol. 2014, 24, 608–620. [Google Scholar] [CrossRef] [PubMed]
- Nomura, D.K.; Morrison, B.E.; Blankman, J.L.; Long, J.Z.; Kinsey, S.G.; Marcondes, M.C.G.; Ward, A.M.; Hahn, Y.K.; Lichtman, A.H.; Conti, B.; et al. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science 2011, 334, 809–813. [Google Scholar] [CrossRef]
- Cristino, L.; Bisogno, T.; Di Marzo, V. Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat. Rev. Neurol. 2019, 16, 9–29. [Google Scholar] [CrossRef]
- De Petrocellis, L.; Ligresti, A.; Moriello, A.S.; Allarà, M.; Bisogno, T.; Petrosino, S.; Stott, C.G.; Di Marzo, V. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br. J. Pharmacol. 2011, 163, 1479–1494. [Google Scholar] [CrossRef]
- Navarro, G.; Varani, K.; Reyes-Resina, I.; Sánchez de Medina, V.; Rivas-Santisteban, R.; Sánchez-Carnerero Callado, C.; Vincenzi, F.; Casano, S.; Ferreiro-Vera, C.; Canela, E.I.; et al. Cannabigerol Action at Cannabinoid CB1 and CB2 Receptors and at CB1–CB2 Heteroreceptor Complexes. Front. Pharmacol. 2018, 9, 632. [Google Scholar] [CrossRef]
- Vernail, V.L.; Bingaman, S.S.; Silberman, Y.; Raup-Konsavage, W.M.; Vrana, K.E.; Arnold, A.C. Acute Cannabigerol Administration Lowers Blood Pressure in Mice. Front. Physiol. 2022, 13, 871962. [Google Scholar] [CrossRef] [PubMed]
- Killestein, J.; Hoogervorst, E.L.; Reif, M.; Kalkers, N.F.; van Loenen, A.C.; Staats, P.G.; Gorter, R.W.; Uitdehaag, B.M.; Polman, C.H. Safety, tolerability, and efficacy of orally administered cannabinoids in MS. Neurology 2002, 58, 1404–1407. [Google Scholar] [CrossRef] [PubMed]
- Zajicek, J.P.; Sanders, H.P.; E Wright, D.; Vickery, P.J.; Ingram, W.M.; Reilly, S.M.; Nunn, A.J.; Teare, L.J.; Fox, P.J.; Thompson, A.J. Cannabinoids in multiple sclerosis (CAMS) study: Safety and efficacy data for 12 months follow up. J. Neurol. Neurosurg. Psychiatry 2005, 76, 1664–1669. [Google Scholar] [CrossRef]
- Vaney, C.; Heinzel-Gutenbrunner, M.; Jobin, P.; Tschopp, F.; Gattlen, B.; Hagen, U.; Schnelle, M.; Reif, M. Efficacy, safety and tolerability of an orally administered cannabis extract in the treatment of spasticity in patients with multiple sclerosis: A randomized, double-blind, placebo-controlled, crossover study. Mult. Scler. J. 2004, 10, 417–424. [Google Scholar] [CrossRef]
- Wade, D.T.; Makela, P.M.; House, H.; Bateman, C.; Robson, P. Long-term use of a cannabis-based medicine in the treatment of spasticity and other symptoms in multiple sclerosis. Mult. Scler. J. 2006, 12, 639–645. [Google Scholar] [CrossRef]
- Collin, C.; Davies, P.; Mutiboko, I.K.; Ratcliffe, S.; for the Sativex Spasticity in MS Study Group. Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis. Eur. J. Neurol. 2007, 14, 290–296. [Google Scholar] [CrossRef] [PubMed]
- Novotna, A.; Mares, J.; Ratcliffe, S.; Novakova, I.; Vachova, M.; Zapletalova, O.; Gasperini, C.; Pozzilli, C.; Cefaro, L.; Comi, G.; et al. A randomized, double-blind, placebo-controlled, parallel-group, enriched-design study of nabiximols* (Sativex®), as add-on therapy, in subjects with refractory spasticity caused by multiple sclerosis. Eur. J. Neurol. 2011, 18, 1122–1131. [Google Scholar] [CrossRef]
- Wade, D.T.; Makela, P.; Robson, P.; House, H.; Bateman, C. Do cannabis-based medicinal extracts have general or specific effects on symptoms in multiple sclerosis? A double-blind, randomized, placebo-controlled study on 160 patients. Mult. Scler. J. 2004, 10, 434–441. [Google Scholar] [CrossRef]
- Collin, C.; Ehler, E.; Waberzinek, G.; Alsindi, Z.; Davies, P.; Powell, K.; Notcutt, W.; O’Leary, C.; Ratcliffe, S.; Nováková, I.; et al. A double-blind, randomized, placebo-controlled, parallel-group study of Sativex, in subjects with symptoms of spasticity due to multiple sclerosis. Neurol. Res. 2010, 32, 451–459. [Google Scholar] [CrossRef]
- Langford, R.M.; Mares, J.; Novotna, A.; Vachova, M.; Novakova, I.; Notcutt, W.; Ratcliffe, S. A double-blind, randomized, placebo-controlled, parallel-group study of THC/CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis. J. Neurol. 2012, 260, 984–997. [Google Scholar] [CrossRef] [PubMed]
- Russo, M.; Calabrò, R.S.; Naro, A.; Sessa, E.; Rifici, C.; D’aLeo, G.; Leo, A.; De Luca, R.; Quartarone, A.; Bramanti, P. Sativex in the Management of Multiple Sclerosis-Related Spasticity: Role of the Corticospinal Modulation. Neural Plast. 2015, 2015, 656582. [Google Scholar] [CrossRef] [PubMed]
- Trépanier, M.O.; Hopperton, K.E.; Mizrahi, R.; Mechawar, N.; Bazinet, R.P. Postmortem evidence of cerebral inflammation in schizophrenia: A systematic review. Mol. Psychiatry 2016, 21, 1009–1026. [Google Scholar] [CrossRef]
- Canseco-Alba, A.; Schanz, N.; Sanabria, B.; Zhao, J.; Lin, Z.; Liu, Q.-R.; Onaivi, E.S. Behavioral effects of psychostimulants in mutant mice with cell-type specific deletion of CB2 cannabinoid receptors in dopamine neurons. Behav. Brain Res. 2019, 360, 286–297. [Google Scholar] [CrossRef]
- Zhang, H.; Gao, M.; Shen, H.; Bi, G.; Yang, H.; Liu, Q.; Wu, J.; Gardner, E.L.; Bonci, A.; Xi, Z. Expression of Functional Cannabinoid CB2 Receptor in VTA Dopamine Neurons in Rats. Addict. Biol. 2016, 22, 752–765. [Google Scholar] [CrossRef]
- Zhang, H.Y.; Gao, M.; Liu, Q.-R.; Bi, G.-H.; Li, X.; Yang, H.-J.; Gardner, E.L.; Wu, J.; Xi, Z.-X. Cannabinoid CB2 receptors modulate midbrain dopamine neuronal activity and dopamine-related behavior in mice. Proc. Natl. Acad. Sci. USA 2014, 111, E5007–E5015. [Google Scholar] [CrossRef]
- Liu, Q.-R.; Canseco-Alba, A.; Zhang, H.-Y.; Tagliaferro, P.; Chung, M.; Dennis, E.; Sanabria, B.; Schanz, N.; Escosteguy-Neto, J.C.; Ishiguro, H.; et al. Cannabinoid type 2 receptors in dopamine neurons inhibits psychomotor behaviors, alters anxiety, depression and alcohol preference. Sci. Rep. 2017, 7, 17410. [Google Scholar] [CrossRef]
- Jordan, C.J.; Xi, Z.-X. Progress in Brain Cannabinoid CB2 Receptor Research: From Genes to Behavior. Neurosci. Biobehav. Rev. 2019, 98, 208–220. [Google Scholar] [CrossRef]
- Delis, F.; Polissidis, A.; Poulia, N.; Justinova, Z.; Nomikos, G.G.; Goldberg, S.R.; Antoniou, K. Attenuation of Cocaine-Induced Conditioned Place Preference and Motor Activity via Cannabinoid CB2 Receptor Agonism and CB1 Receptor Antagonism in Rats. Int. J. Neuropsychopharmacol. 2016, 20, 269–278. [Google Scholar] [CrossRef][Green Version]
- Xi, Z.-X.; Peng, X.Q.; Li, X.; Song, R.; Zhang, H.Y.; Liu, Q.-R.; Yang, H.-J.; Bi, G.-H.; Li, J.; Gardner, E.L. Brain cannabinoid CB2 receptors modulate cocaine’s actions in mice. Nat. Neurosci. 2011, 14, 1160–1166. [Google Scholar] [CrossRef]
- Aracil-Fernández, A.; Trigo, J.M.; García-Gutiérrez, M.S.; Ortega-Álvaro, A.; Ternianov, A.; Navarro, D.; Robledo, P.; Berbel, P.; Maldonado, R.; Manzanares, J. Decreased Cocaine Motor Sensitization and Self-Administration in Mice Overexpressing Cannabinoid CB2 Receptors. Neuropsychopharmacology 2012, 37, 1749–1763. [Google Scholar] [CrossRef]
- Legge, S.E.; Jones, H.J.; Kendall, K.M.; Pardiñas, A.F.; Menzies, G.; Bracher-Smith, M.; Escott-Price, V.; Rees, E.; Davis, K.A.S.; Hotopf, M.; et al. Association of Genetic Liability to Psychotic Experiences with Neuropsychotic Disorders and Traits. JAMA Psychiatry 2019, 76, 1256–1265. [Google Scholar] [CrossRef]
- Horcajadas, F.A.; Piriz, J.R.D.; González, A.P.; Romero, S.S.; Sánchez-Morla, E.; Sánchez, I.A.; Atance, J.A.R. Cannabinoid receptor type 2 gene is associated with comorbidity of schizophrenia and cannabis dependence and fatty acid amide hydrolase gene is associated with cannabis dependence in the Spanish population. Adicciones 2021, 35, 33–46. [Google Scholar] [CrossRef] [PubMed]
- Ishiguro, H.; Horiuchi, Y.; Ishikawa, M.; Koga, M.; Imai, K.; Suzuki, Y.; Morikawa, M.; Inada, T.; Watanabe, Y.; Takahashi, M.; et al. Brain Cannabinoid CB2 Receptor in Schizophrenia. Biol. Psychiatry 2010, 67, 974–982. [Google Scholar] [CrossRef] [PubMed]
- Lazary, J.; Eszlari, N.; Juhasz, G.; Bagdy, G. A functional variant of CB2 receptor gene interacts with childhood trauma and FAAH gene on anxious and depressive phenotypes. J. Affect. Disord. 2019, 257, 716–722. [Google Scholar] [CrossRef] [PubMed]
- Onaivi, E.S.; Ishiguro, H.; Gong, J.-P.; Patel, S.; Meozzi, P.A.; Myers, L.; Perchuk, A.; Mora, Z.; Tagliaferro, P.A.; Gardner, E.; et al. Brain Neuronal CB2 Cannabinoid Receptors in Drug Abuse and Depression: From Mice to Human Subjects. PLoS ONE 2008, 3, e1640. [Google Scholar] [CrossRef] [PubMed]
- Singh, T.; I Kurki, M.; Curtis, D.; Purcell, S.M.; Crooks, L.; McRae, J.; Suvisaari, J.; Chheda, H.; Blackwood, D.; Breen, G.; et al. Rare loss-of-function variants in SETD1A are associated with schizophrenia and developmental disorders. Nat. Neurosci. 2016, 19, 571–577. [Google Scholar] [CrossRef]
- Hannon, E.; Spiers, H.; Viana, J.; Pidsley, R.; Burrage, J.; Murphy, T.M.; Troakes, C.; Turecki, G.; O’DOnovan, M.C.; Schalkwyk, L.C.; et al. Methylation QTLs in the developing brain and their enrichment in schizophrenia risk loci. Nat. Neurosci. 2015, 19, 48–54. [Google Scholar] [CrossRef]
- Gusev, A.; Mancuso, N.; Won, H.; Kousi, M.; Finucane, H.K.; Reshef, Y.; Song, L.; Safi, A.; McCarroll, S.; Neale, B.M.; et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat. Genet. 2018, 50, 538–548. [Google Scholar] [CrossRef]
- Abdolmaleky, H.M.; Nohesara, S.; Ghadirivasfi, M.; Lambert, A.W.; Ahmadkhaniha, H.; Ozturk, S.; Wong, C.K.; Shafa, R.; Mostafavi, A.; Thiagalingam, S. DNA hypermethylation of serotonin transporter gene promoter in drug naïve patients with schizophrenia. Schizophr. Res. 2014, 152, 373–380. [Google Scholar] [CrossRef] [PubMed]
- Ikegame, T.; Bundo, M.; Sunaga, F.; Asai, T.; Nishimura, F.; Yoshikawa, A.; Kawamura, Y.; Hibino, H.; Tochigi, M.; Kakiuchi, C.; et al. DNA methylation analysis of BDNF gene promoters in peripheral blood cells of schizophrenia patients. Neurosci. Res. 2013, 77, 208–214. [Google Scholar] [CrossRef]
- Mill, J.; Tang, T.; Kaminsky, Z.; Khare, T.; Yazdanpanah, S.; Bouchard, L.; Jia, P.; Assadzadeh, A.; Flanagan, J.; Schumacher, A.; et al. Epigenomic profiling reveals DNA-methylation changes associated with major psychosis. Am. J. Hum. Genet. 2008, 82, 696–711. [Google Scholar] [CrossRef]
- Foster, R.; Kandanearatchi, A.; Beasley, C.; Williams, B.; Khan, N.; Fagerhol, M.K.; Everall, I.P. Calprotectin in microglia from frontal cortex is up-regulated in schizophrenia: Evidence for an inflammatory process? Eur. J. Neurosci. 2006, 24, 3561–3566. [Google Scholar] [CrossRef]
- E Laskaris, L.; A Di Biase, M.; Everall, I.; Chana, G.; Christopoulos, A.; Skafidas, E.; Cropley, V.L.; Pantelis, C. Microglial activation and progressive brain changes in schizophrenia. Br. J. Pharmacol. 2016, 173, 666–680. [Google Scholar] [CrossRef] [PubMed]
- Bloomfield, P.S.; Selvaraj, S.; Veronese, M.; Rizzo, G.; Bertoldo, A.; Owen, D.; Bloomfield, M.; Bonoldi, I.; Kalk, N.; Turkheimer, F.; et al. Microglial Activity in People at Ultra High Risk of Psychosis and in Schizophrenia: An [(11)C]PBR28 PET Brain Imaging Study. Am. J. Psychiatry 2016, 173, 44–52. [Google Scholar] [CrossRef]
- Ortega-Alvaro, A.; Aracil-Fernández, A.; García-Gutiérrez, M.S.; Navarrete, F.; Manzanares, J. Deletion of CB2 Cannabinoid Receptor Induces Schizophrenia-Related Behaviors in Mice. Neuropsychopharmacology 2011, 36, 1489–1504. [Google Scholar] [CrossRef]
- Cortez, I.L.; Silva, N.R.; Rodrigues, N.S.; Pedrazzi, J.F.C.; Del Bel, E.A.; Mechoulam, R.; Gomes, F.V.; Guimarães, F.S. HU-910, a CB2 receptor agonist, reverses behavioral changes in pharmacological rodent models for schizophrenia. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2022, 117, 110553. [Google Scholar] [CrossRef]
- Ribeiro, M.A.; Aguiar, R.P.; Scarante, F.F.; Fusse, E.J.; de Oliveira, R.M.W.; Guimaraes, F.S.; Campos, A.C. Spontaneous Activity of CB2 Receptors Attenuates Stress-Induced Behavioral and Neuroplastic Deficits in Male Mice. Front. Pharmacol. 2022, 12, 805758. [Google Scholar] [CrossRef]
- Cortez, I.L.; da Silva, N.R.; Guimarães, F.S.; Gomes, F.V. Are CB2 Receptors a New Target for Schizophrenia Treatment? Front. Psychiatry 2020, 11, 587154. [Google Scholar] [CrossRef]
- De Marchi, N.; De Petrocellis, L.; Orlando, P.; Daniele, F.; Fezza, F.; Di Marzo, V. Endocannabinoid signalling in the blood of patients with schizophrenia. Lipids Health Dis. 2003, 2, 5. [Google Scholar] [CrossRef]
- D’Addario, C.; Micale, V.; Di Bartolomeo, M.; Stark, T.; Pucci, M.; Sulcova, A.; Palazzo, M.; Babinska, Z.; Cremaschi, L.; Drago, F.; et al. A preliminary study of endocannabinoid system regulation in psychosis: Distinct alterations of CNR1 promoter DNA methylation in patients with schizophrenia. Schizophr. Res. 2017, 188, 132–140. [Google Scholar] [CrossRef]
- Bioque, M.; García-Bueno, B.; MacDowell, K.S.; Meseguer, A.; Saiz, P.A.; Parellada, M.; Gonzalez-Pinto, A.; Rodriguez-Jimenez, R.; Lobo, A.; Leza, J.C.; et al. Peripheral endocannabinoid system dysregulation in first-episode psychosis. Neuropsychopharmacology 2013, 38, 2568–2577. [Google Scholar] [CrossRef]
- Qi, A.; Han, X.; Quitalig, M.; Wu, J.; Christov, P.P.; Jeon, K.; Jana, S.; Kim, K.; Engers, D.W.; Lindsley, C.W.; et al. The cannabinoid CB2 receptor positive allosteric modulator EC21a exhibits complicated pharmacology in vitro. J. Recept. Signal Transduct. 2024, 44, 151–159. [Google Scholar] [CrossRef] [PubMed]
- Gado, F.; Di Cesare Mannelli, L.; Lucarini, E.; Bertini, S.; Cappelli, E.; Digiacomo, M.; Stevenson, L.A.; Macchia, M.; Tuccinardi, T.; Ghelardini, C.; et al. Identification of the First Synthetic Allosteric Modulator of the CB2 Receptors and Evidence of Its Efficacy for Neuropathic Pain Relief. J. Med. Chem. 2018, 62, 276–287. [Google Scholar] [CrossRef] [PubMed]
- Gado, F.; Arena, C.; La Fauci, C.; Reynoso-Moreno, I.; Bertini, S.; Digiacomo, M.; Meini, S.; Poli, G.; Macchia, M.; Tuccinardi, T.; et al. Modification on the 1,2-dihydro-2-oxo-pyridine-3-carboxamide core to obtain multi-target modulators of endocannabinoid system. Bioorg. Chem. 2020, 94, 103353. [Google Scholar] [CrossRef] [PubMed]
- Ishiguro, H.; Horiuchi, Y.; Tabata, K.; Liu, Q.-R.; Arinami, T.; Onaivi, E.S. Cannabinoid CB2 Receptor Gene and Environmental Interaction in the Development of Psychiatric Disorders. Molecules 2018, 23, 1836. [Google Scholar] [CrossRef]
- Gutiérrez, G.; Pérez-Ortiz, J.; Gutiérrez-Adán, A.; Manzanares, J. Depression-resistant endophenotype in mice overexpressing cannabinoid CB2 receptors. Br. J. Pharmacol. 2010, 160, 1773–1784. [Google Scholar] [CrossRef]
- Stark, T.; Ruda-Kucerova, J.; Iannotti, F.A.; D’Addario, C.; Di Marco, R.; Pekarik, V.; Drazanova, E.; Piscitelli, F.; Bari, M.; Babinska, Z.; et al. Peripubertal cannabidiol treatment rescues behavioral and neurochemical abnormalities in the MAM model of schizophrenia. Neuropharmacology 2019, 146, 212–221. [Google Scholar] [CrossRef]
- García, C.; Palomo-Garo, C.; García-Arencibia, M.; Ramos, J.; Pertwee, R.; Fernández-Ruiz, J. Symptom-relieving and neuroprotective effects of the phytocannabinoid Δ9-THCV in animal models of Parkinson’s disease. Br. J. Pharmacol. 2011, 163, 1495–1506. [Google Scholar] [CrossRef] [PubMed]
- Price, D.A.; Martinez, A.A.; Seillier, A.; Koek, W.; Acosta, Y.; Fernandez, E.; Strong, R.; Lutz, B.; Marsicano, G.; Roberts, J.L.; et al. WIN55,212-2, a cannabinoid receptor agonist, protects against nigrostriatal cell loss in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Eur. J. Neurosci. 2009, 29, 2177–2186. [Google Scholar] [CrossRef]
- Gomes, T.M.; da Silva, D.D.; Carmo, H.; Carvalho, F.; Silva, J.P. Epigenetics and the endocannabinoid system signaling: An intricate interplay modulating neurodevelopment. Pharmacol. Res. 2020, 162, 105237. [Google Scholar] [CrossRef] [PubMed]
- Di Marzo, V. Targeting the endocannabinoid system: To enhance or reduce? Nat. Rev. Drug Discov. 2008, 7, 438–455. [Google Scholar] [CrossRef] [PubMed]
- Qu, Y.; Fu, Y.; Liu, Y.; Liu, C.; Xu, B.; Zhang, Q.; Jiang, P. The role of TRPV1 in RA pathogenesis: Worthy of attention. Front. Immunol. 2023, 14, 1232013. [Google Scholar] [CrossRef]
- Niu, M.; Zhao, F.; Chen, R.; Li, P.; Bi, L. The transient receptor potential channels in rheumatoid arthritis: Need to pay more attention. Front. Immunol. 2023, 14, 1127277. [Google Scholar] [CrossRef]
- Muller, C.; Morales, P.; Reggio, P.H. Cannabinoid Ligands Targeting TRP Channels. Front. Mol. Neurosci. 2019, 11, 487. [Google Scholar] [CrossRef]
- Rossi, F.; Bellini, G.; Tortora, C.; Bernardo, M.E.; Luongo, L.; Conforti, A.; Starc, N.; Manzo, I.; Nobili, B.; Locatelli, F.; et al. CB(2) and TRPV(1) receptors oppositely modulate in vitro human osteoblast activity. Pharmacol. Res. 2015, 99, 194–201. [Google Scholar] [CrossRef] [PubMed]
- Wilkerson, J.L.; Alberti, L.B.; Thakur, G.A.; Makriyannis, A.; Milligan, E.D. Peripherally administered cannabinoid receptor 2 (CB2R) agonists lose anti-allodynic effects in TRPV1 knockout mice, while intrathecal administration leads to anti-allodynia and reduced GFAP, CCL2 and TRPV1 expression in the dorsal spinal cord and DRG. Brain Res. 2022, 1774, 147721. [Google Scholar] [CrossRef] [PubMed]
- Peterson, C.; Ratan, R.; Shelanski, M.; Goldman, J. Changes in Calcium Homeostasis during Aging and Alzheimer’s Diseasea. Ann. N. Y. Acad. Sci. 1989, 568, 262–270. [Google Scholar] [CrossRef]
- Sheehan, J.P.; Swerdlow, R.H.; Miller, S.W.; Davis, R.E.; Parks, J.K.; Parker, W.D.; Tuttle, J.B. Calcium homeostasis and reactive oxygen species production in cells transformed by mitochondria from individuals with sporadic Alzheimer’s disease. J. Neurosci. 1997, 17, 4612–4622. [Google Scholar] [CrossRef]
- Ham, S.J.; Yoo, H.; Woo, D.; Lee, D.H.; Park, K.-S.; Chung, J. PINK1 and Parkin regulate IP3R-mediated ER calcium release. Nat. Commun. 2023, 14, 5202. [Google Scholar] [CrossRef]
- Morales, P.; Muller, C.; Jagerovic, N.; Reggio, P.H. Targeting CB2 and TRPV1: Computational Approaches for the Identification of Dual Modulators. Front. Mol. Biosci. 2022, 9, 841190. [Google Scholar] [CrossRef] [PubMed]
- Borrelli, F.; Pagano, E.; Romano, B.; Panzera, S.; Maiello, F.; Coppola, D.; De Petrocellis, L.; Buono, L.; Orlando, P.; Izzo, A.A. Colon carcinogenesis is inhibited by the TRPM8 antagonist cannabigerol, a Cannabis-derived non-psychotropic cannabinoid. Carcinogenesis 2014, 35, 2787–2797. [Google Scholar] [CrossRef]
- Fernandez-Boyanapalli, R.; Frasch, S.C.; Riches, D.W.H.; Vandivier, R.W.; Henson, P.M.; Bratton, D.L. PPARγ activation normalizes resolution of acute sterile inflammation in murine chronic granulomatous disease. Blood 2010, 116, 4512–4522. [Google Scholar] [CrossRef]
- Iannotti, F.A.; Vitale, R.M. The Endocannabinoid System and PPARs: Focus on Their Signalling Crosstalk, Action and Transcriptional Regulation. Cells 2021, 10, 586. [Google Scholar] [CrossRef]
- Petrosino, S.; Moriello, A.S.; Cerrato, S.; Fusco, M.; Puigdemont, A.; De Petrocellis, L.; Di Marzo, V. The anti-inflammatory mediator palmitoylethanolamide enhances the levels of 2-arachidonoyl-glycerol and potentiates its actions at TRPV1 cation channels. Br. J. Pharmacol. 2015, 173, 1154–1162. [Google Scholar] [CrossRef]
- Zygmunt, P.M.; Petersson, J.; Andersson, D.A.; Chuang, H.-H.; Sørgård, M.; Di Marzo, V.; Julius, D.; Högestätt, E.D. Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature 1999, 400, 452–457. [Google Scholar] [CrossRef]
- DI Marzo, V.; Melck, D.; Orlando, P.; Bisogno, T.; Zagoory, O.; Bifulco, M.; Vogel, Z.; DE Petrocellis, L. Palmitoylethanolamide inhibits the expression of fatty acid amide hydrolase and enhances the anti-proliferative effect of anandamide in human breast cancer cells. Biochem. J. 2001, 358, 249–255. [Google Scholar] [CrossRef] [PubMed]
- Ho, W.; A Barrett, D.; Randall, M.D. ‘Entourage’ effects of N-palmitoylethanolamide and N-oleoylethanolamide on vasorelaxation to anandamide occur through TRPV1 receptors. Br. J. Pharmacol. 2008, 155, 837–846. [Google Scholar] [CrossRef] [PubMed]
- Komorowska-Müller, J.A.; Schmöle, A.-C. CB2 Receptor in Microglia: The Guardian of Self-Control. Int. J. Mol. Sci. 2020, 22, 19. [Google Scholar] [CrossRef] [PubMed]
- Qin, X.; Wang, W.; Wu, H.; Liu, D.; Wang, R.; Xu, J.; Jiang, H.; Pan, F. PPARγ-mediated microglial activation phenotype is involved in depressive-like behaviors and neuroinflammation in stressed C57BL/6J and ob/ob mice. Psychoneuroendocrinology 2020, 117, 104674. [Google Scholar] [CrossRef]
- Hill, J.D.; Zuluaga-Ramirez, V.; Gajghate, S.; Winfield, M.; Sriram, U.; Rom, S.; Persidsky, Y. Activation of GPR55 induces neuroprotection of hippocampal neurogenesis and immune responses of neural stem cells following chronic, systemic inflammation. Brain. Behav. Immun. 2019, 76, 165–181. [Google Scholar] [CrossRef]
- Wu, C.-S.; Chen, H.; Sun, H.; Zhu, J.; Jew, C.P.; Wager-Miller, J.; Straiker, A.; Spencer, C.; Bradshaw, H.; Mackie, K.; et al. GPR55, a G-protein coupled receptor for lysophosphatidylinositol, plays a role in motor coordination. PLoS ONE 2013, 8, e60314. [Google Scholar] [CrossRef]
- Medina-Vera, D.; Zhao, H.; Bereczki, E.; Rosell-Valle, C.; Shimozawa, M.; Chen, G.; de Fonseca, F.R.; Nilsson, P.; Tambaro, S. The Expression of the Endocannabinoid Receptors CB2 and GPR55 Is Highly Increased during the Progression of Alzheimer’s Disease in AppNL-G-F Knock-In Mice. Biology 2023, 12, 805. [Google Scholar] [CrossRef]
- Wu, M.; Wu, L.; Wu, W.; Zhu, M.; Li, J.; Wang, Z.; Li, J.; Ding, R.; Liang, Y.; Li, L.; et al. Phagocytosis of Glioma Cells Enhances the Immunosuppressive Phenotype of Bone Marrow–Derived Macrophages. Cancer Res. 2023, 83, 771–785. [Google Scholar] [CrossRef]
- Henstridge, C.M.; Balenga, N.A.B.; Ford, L.A.; Ross, R.A.; Waldhoer, M.; Irving, A.J. The GPR55 ligand L-α-lysophosphatidylinositol promotes RhoA-dependent Ca2+ signaling and NFAT activation. FASEB J. 2008, 23, 183–193. [Google Scholar] [CrossRef]
- McHugh, D.; Hu, S.S.; Rimmerman, N.; Juknat, A.; Vogel, Z.; Walker, J.M.; Bradshaw, H.B. N-arachidonoyl glycine, an abundant endogenous lipid, potently drives directed cellular migration through GPR18, the putative abnormal cannabidiol receptor. BMC Neurosci. 2010, 11, 44. [Google Scholar] [CrossRef] [PubMed]
- Penumarti, A.; Abdel-Rahman, A.A. The Novel Endocannabinoid Receptor GPR18 Is Expressed in the Rostral Ventrolateral Medulla and Exerts Tonic Restraining Influence on Blood Pressure. J. Pharmacol. Exp. Ther. 2014, 349, 29–38. [Google Scholar] [CrossRef] [PubMed]
- Grabiec, U.; Hohmann, T.; Ghadban, C.; Rothgänger, C.; Wong, D.; Antonietti, A.; Groth, T.; Mackie, K.; Dehghani, F. Protective Effect of N-Arachidonoyl Glycine-GPR18 Signaling after Excitotoxical Lesion in Murine Organotypic Hippocampal Slice Cultures. Int. J. Mol. Sci. 2019, 20, 1266. [Google Scholar] [CrossRef]
- Landén, N.X.; Li, D.; Ståhle, M. Transition from inflammation to proliferation: A critical step during wound healing. Cell. Mol. Life Sci. 2016, 73, 3861–3885. [Google Scholar] [CrossRef]
- Di Marzo, V. New approaches and challenges to targeting the endocannabinoid system. Nat. Rev. Drug Discov. 2018, 17, 623–639. [Google Scholar] [CrossRef] [PubMed]




| Compound | Chemical Class | Ki CB1 (nM) | Ki CB2 (nM) | Key Notes | Ref. |
|---|---|---|---|---|---|
| JWH-133 | Dimethylbutyl-deoxy-Δ8-THC | 677 | 3.4 | Widely used CB2 agonist; strong in vivo efficacy; off-target at high doses. | [39] |
| JWH-015 | 1-Propyl-2-methyl-3-(1-naphthoyl) indole | 383 | 13.8 | CB2-preferring; retains CB1 activity in neural systems. | [40] |
| HU-308 | bicyclo [3.1.1]hept-2-ene | >10,000 | 22.7 | Peripherally restricted CB2 agonist; strong immune/inflammation applications. | [41] |
| WIN55,212-2 | aminoalkylindole analog | 1.89–123 | 0.28–16.2 | Potent dual cannabinoid receptor agonist. | [42] |
| AEA | N-acylethanolamine | 61–543 | 279–1940 | Endogenous ligand; partial agonist; rapidly hydrolyzed by FAAH. | [43] |
| 2-AG | monoacyl-glycerol | 58.3–472 | 145–1400 | Full agonist; hydrolyzed by MAGL; critical in physiology. | [43] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kalkan, H.; Flamand, N. The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies. DNA 2025, 5, 59. https://doi.org/10.3390/dna5040059
Kalkan H, Flamand N. The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies. DNA. 2025; 5(4):59. https://doi.org/10.3390/dna5040059
Chicago/Turabian StyleKalkan, Hilal, and Nicolas Flamand. 2025. "The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies" DNA 5, no. 4: 59. https://doi.org/10.3390/dna5040059
APA StyleKalkan, H., & Flamand, N. (2025). The CB2 Receptor in Immune Regulation and Disease: Genetic Architecture, Epigenetic Control, and Emerging Therapeutic Strategies. DNA, 5(4), 59. https://doi.org/10.3390/dna5040059

