Skip to main content

EDITORIAL article

Front. Neurosci., 20 December 2022
Sec. Neurodegeneration
This article is part of the Research Topic Cannabinoids in Neuroinflammation, Neurodegeneration and Pain: Focus on Non-Neuronal Cells View all 5 articles

Editorial: Cannabinoids in neuroinflammation, neurodegeneration and pain: Focus on non-neuronal cells

  • 1Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
  • 2Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL, United States
  • 3Department of Neurology, Norman Fixel Institute for Neurological Diseases, University of Florida Health, Gainesville, FL, United States
  • 4Department of Neurochemistry, Maj Institute of Pharmacology PAS, Kraków, Poland

Cannabinoids have the potential to be therapeutic for neurological and psychiatric diseases associated with neuroinflammation, neurodegeneration and pain (Kelly et al., 2020; Bryk and Starowicz, 2021). The cannabinoid system includes endogenous cannabinoids, phytocannabinoids and synthetic cannabinoids, their target receptors and biosynthetic and degradative enzymes (Morena et al., 2016). Alterations of the cannabinoid system are associated with the inflammatory processes of these conditions, but intriguingly, are also implicated in the alterations in affect which may co-occur (Vecchiarelli et al., 2021). Non-neuronal cells in the central nervous system are in particular related to the pathogenesis, maintenance and/or alleviation of neuroinflammatory, neurodegenerative and pain states (Kelly et al., 2020; Šimončičová et al., 2022; St-Pierre et al., 2022). To further elucidate the role of cannabinoids in these disease contexts, this Research Topic includes a collection of primary research articles and a mini review on the role of cannabinoids in these states in particular in non-neuronal cells.

Two primary research articles in the collection investigate the role of enzymes that metabolize N-acylethanolamines, including the primary endocannabinoid, N-arachidonoylethanolamine (anandamide or AEA). In Duncan et al., human neural precursor cell culture (ReN cells) were exposed to sublethal oxidative stress [tert-butyl hydroperoxide (tBHP)]—which is particularly associated with neurodegenerative diseases. They found that exposure to tBHP increased protein levels of cannabinoid receptors (CB1 and CB2) and of an N-acylethanolamine metabolizing enzyme, fatty acid amide hydrolase (FAAH), which metabolizes AEA, as well as the associated N-acylethanolamine molecules, oleoylethanolamide (OEA) and palmitoylethanolamide (PEA) (Malek and Starowicz, 2016). However, exposure to a lower dose of tBHP increased expression of N-acylethanolamide specific phospholipase D (NAPE-PLD), a synthesizing enzyme for AEA. Intriguingly, they also found that the mild level of oxidative stress also increased neurite outgrowth. It is possible that because AEA can serve to promote neurite outgrowth (Compagnucci et al., 2013), that the oxidative stress-induced increases in NAPE-PLD generated AEA, leading to the observed increased neurite outgrowth. It is possible these cannabinoid increases are protective, as previously demonstrated (Elmazoglu et al., 2020). While Duncan et al., showed an effect of oxidative stress on the N-acylethanolamine metabolizing enzyme, FAAH, Vecchiarelli et al., investigates the effects of a single nucleotide polymorphism (SNP) in FAAH (C385A) mouse model (Dincheva et al., 2015) on basal and colitis-induced alterations in inflammatory mediators in plasma and the amygdala. Carriers of the mutant allele have reduced FAAH activity and show attenuated colitis-induced increases of plasma IL-2, LIF, MCP-1, and TNF, as well as amygdala G-CSF and MCP-1 levels—without altering the colitis-induced disease macroscopic colon damage. Interestingly, following chronic stress, the receptor for MCP-1 is necessary for the development of anxiety-like behavior-inducing monocyte trafficking to the brain (Wohleb et al., 2013). Additionally, a central increase of FAAH activity and decrease of AEA levels can contribute to colitis-induced anxiety (Vecchiarelli et al., 2021); therefore, it is possible that colitis-induced reductions of AEA allow for an increase in MCP-1 contributing to monocyte-trafficking-induced generation of anxiety-like behavior, suggesting AEA is a potential modulator of inflammatory responses. Additionally, Vecchiarelli et al., show that FAAH reduction leads to reduced IL-1α, IL-9, MIP-1β, and MIP-2 levels in the amygdala, indicating that FAAH, or the compounds it metabolizes, may be involved in their baseline regulation centrally. Together, these studies illustrate the effects of FAAH in response to oxidative stress and on neuroinflammation, which may be important for neurodegeneration and the affective symptoms of inflammatory diseases.

The remaining articles in this Research Topic discuss the role of CB2. Honig et al., show the effects of a clinically available CB2 inverse agonist, Raloxifene, on visual system outcomes following focal cranial impact mild traumatic brain injury (mTBI). They found that Raloxifene reversed the effects of mTBI on contrast sensitivity, light aversion, pupillary excessive dilation and optic nerve axonal loss. Furthermore, in the injured optic nerve, Honig et al., show that IBA1+ cell numbers are increased and have a normalized transcriptional profile after 10 mg/kg of Raloxifene following mTBI. Therefore, highlighting a further protective role for CB2 inverse agonism in the regulation of IBA1+ cells (predominantly microglia) following mTBI, which may contribute to the beneficial outcomes seen in visual behavior following mTBI. The protective effects of inverse agonism might seem counterintuitive, as inverse agonists suppress constitutive activity and activation of CB2 has shown a role in cytokine signaling and is generally considered anti-inflammatory (Young and Denovan-Wright, 2021), although this is not always the case, as there may be ligand-specific signaling biases (Oláh et al., 2017). Important to the function of microglia and myeloid cells, activation of CB2 can suppress phagocytosis (Han et al., 2022). Therefore, it is possible that CB2 inverse agonism promotes favorable disease outcomes by accelerating phagocytosis allowing the innate immune system to clear debris while also providing an environment enriched in restorative mediators (Yu et al., 2020; Martinez Ramirez et al., 2022). This indicates that characterization of Raloxifene and other CB2 inverse agonists at microglial CB2 could be of great benefit for therapeutic discovery.

Our final article is a mini-review by Ferranti and Foster, which highlights a role for CB2 in schizophrenia, a disease becoming increasingly understood to possess inflammatory risk factors (Comer et al., 2020a) and microglia-mediated mechanisms (Sekar et al., 2016; Comer et al., 2020b). This article highlights that in addition to, or maybe through, inflammatory mediator signaling, a role for CB2 receptors in microglia on associated behaviors, such as contextual fear memory. Fear memory was enhanced with the overexpression of CB2 in hippocampal CA1 microglia, yet reduced with disrupted microglial CB2 (Li and Kim, 2017). The Ferranti and Foster review reminds us that CB2 receptors have also been described on some neuronal populations. Therefore, a crucial next step in the field is to better understand the cellular distribution of central CB2 and the interplay between neuronal and non-neuronal CB2 under basal physiological conditions and across disease phenotypes.

Further understanding of the complex signaling of this system will hopefully lead to the generation or refinement of therapeutics for a host of neurological and psychiatric diseases and articles in this collection have contributed toward this goal.

Author contributions

HV and VJ wrote the initial draft of the manuscript. All authors contributed to manuscript revision, read, and approved the submitted version.

Funding

HV is supported through a Canadian Institutes of Health Research Fellowship and is a Michael Smith Health Research BC Research Trainee. VJ is supported through an Alzheimer's Association Research Grant (AARG) and a Michael J. Fox Foundation for Parkinson's Research Target Validation grant. KS is supported through the National Science Centre, Poland grant OPUS no. 2016/23/B/NZ7/01143. MT is supported through NIA Grant RF1AG057247-05, NINDS Grant RF1NS128800-01, the Parkinson's Foundation Research Center of Excellence Award PF-RCE-1945, the Weston Family Foundation, the Michael J. Fox Foundation for Parkinson's Research (MJFF-18212, 18891 and 16778) and Aligning Science Across Parkinson's (ASAP-020621), and UF-Fixel Institute Norman and Susan Fixel Endowment.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Bryk, M., and Starowicz, K. (2021). Cannabinoid-based therapy as a future for joint degeneration. Focus on the role of CB2 receptor in the arthritis progression and pain: an updated review. Pharmacol. Rep. 73, 681–699. doi: 10.1007/s43440-021-00270-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Comer, A. L., Carrier, M., Tremblay, M.-È., and Cruz-Martín, A. (2020a). The inflamed brain in schizophrenia: the convergence of genetic and environmental risk factors that lead to uncontrolled neuroinflammation. Front. Cell. Neurosci. 14, 274. doi: 10.3389/fncel.2020.00274

PubMed Abstract | CrossRef Full Text | Google Scholar

Comer, A. L., Jinadasa, T., Sriram, B., Phadke, R. A., Kretsge, L. N., Nguyen, T. P. H., et al. (2020b). Increased expression of schizophrenia-associated gene C4 leads to hypoconnectivity of prefrontal cortex and reduced social interaction. PLoS Biol. 18, e3000604. doi: 10.1371/journal.pbio.3000604

PubMed Abstract | CrossRef Full Text | Google Scholar

Compagnucci, C., Di Siena, S., Bustamante, M. B., Di Giacomo, D., Di Tommaso, M., Maccarrone, M., et al. (2013). Type-1 (CB1) cannabinoid receptor promotes neuronal differentiation and maturation of neural stem cells. PLoS ONE 8, e54271. doi: 10.1371/journal.pone.0054271

PubMed Abstract | CrossRef Full Text | Google Scholar

Dincheva, I., Drysdale, A. T., Hartley, C. A., Johnson, D. C., Jing, D., King, E. C., et al. (2015). FAAH genetic variation enhances fronto-amygdala function in mouse and human. Nat. Commun. 6, 6395. doi: 10.1038/ncomms7395

PubMed Abstract | CrossRef Full Text | Google Scholar

Elmazoglu, Z., Rangel-López, E., Medina-Campos, O. N., Pedraza-Chaverri, J., Túnez, I., Aschner, M., et al. (2020). Cannabinoid-profiled agents improve cell survival via reduction of oxidative stress and inflammation, and Nrf2 activation in a toxic model combining hyperglycemia+Aβ1-42 peptide in rat hippocampal neurons. Neurochem. Int. 140, 104817. doi: 10.1016/j.neuint.2020.104817

PubMed Abstract | CrossRef Full Text | Google Scholar

Han, Q.-W., Shao, Q.-H., Wang, X.-T., Ma, K.-L., Chen, N.-H., and Yuan, Y.-H. (2022). CB2 receptor activation inhibits the phagocytic function of microglia through activating ERK/AKT-Nurr1 signal pathways. Acta Pharmacol. Sin. 43, 2253–2266. doi: 10.1038/s41401-021-00853-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Kelly, R., Joers, V., Tansey, M. G., McKernan, D. P., and Dowd, E. (2020). Microglial phenotypes and their relationship to the cannabinoid system: therapeutic implications for Parkinson's Disease. Molecules 25, E453. doi: 10.3390/molecules25030453

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Y., and Kim, J. (2017). Distinct roles of neuronal and microglial CB2 cannabinoid receptors in the mouse hippocampus. Neuroscience 363, 11–25. doi: 10.1016/j.neuroscience.2017.08.053

PubMed Abstract | CrossRef Full Text | Google Scholar

Malek, N., and Starowicz, K. (2016). Dual-acting compounds targeting endocannabinoid and endovanilloid systems-a novel treatment option for chronic pain management. Front. Pharmacol. 7, 257. doi: 10.3389/fphar.2016.00257

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinez Ramirez, C. E., Ruiz-Pérez, G., Stollenwerk, T. M., Behlke, C., Doherty, A., and Hillard, C. J. (2022). Endocannabinoid signaling in the central nervous system. Glia 71, 5–35. doi: 10.1002/glia.24280

PubMed Abstract | CrossRef Full Text | Google Scholar

Morena, M., Leitl, K. D., Vecchiarelli, H. A., Gray, J. M., Campolongo, P., and Hill, M. N. (2016). Emotional arousal state influences the ability of amygdalar endocannabinoid signaling to modulate anxiety. Neuropharmacology 111, 59–69. doi: 10.1016/j.neuropharm.2016.08.020

PubMed Abstract | CrossRef Full Text | Google Scholar

Oláh, A., Szekanecz, Z., and Bíró, T. (2017). Targeting cannabinoid signaling in the immune system: “high”-ly exciting questions, possibilities, and challenges. Front. Immunol. 8, 1487. doi: 10.3389/fimmu.2017.01487

PubMed Abstract | CrossRef Full Text | Google Scholar

Sekar, A., Bialas, A. R., de Rivera, H., Davis, A., Hammond, T. R., Kamitaki, N., et al. (2016). Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183. doi: 10.1038/nature16549

PubMed Abstract | CrossRef Full Text | Google Scholar

Šimončičová, E., Gonçalves de Andrade, E., Vecchiarelli, H. A., Awogbindin, I. O., Delage, C. I., and Tremblay, M.-È. (2022). Present and future of microglial pharmacology. Trends Pharmacol. Sci. 43, 669–685. doi: 10.1016/j.tips.2021.11.006

PubMed Abstract | CrossRef Full Text | Google Scholar

St-Pierre, M.-K., VanderZwaag, J., Loewen, S., and Tremblay, M.-È. (2022). All roads lead to heterogeneity: the complex involvement of astrocytes and microglia in the pathogenesis of Alzheimer's disease. Front. Cell. Neurosci. 16, 932572. doi: 10.3389/fncel.2022.932572

PubMed Abstract | CrossRef Full Text | Google Scholar

Vecchiarelli, H. A., Morena, M., Keenan, C. M., Chiang, V., Tan, K., Qiao, M., et al. (2021). Comorbid anxiety-like behavior in a rat model of colitis is mediated by an upregulation of corticolimbic fatty acid amide hydrolase. Neuropsychopharmacology 46, 992–1003. doi: 10.1038/s41386-020-00939-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Wohleb, E. S., Powell, N. D., Godbout, J. P., and Sheridan, J. F. (2013). Stress-induced recruitment of bone marrow-derived monocytes to the brain promotes anxiety-like behavior. J. Neurosci. 33, 13820–13833. doi: 10.1523/JNEUROSCI.1671-13.2013

PubMed Abstract | CrossRef Full Text | Google Scholar

Young, A. P., and Denovan-Wright, E. M. (2021). The dynamic role of microglia and the endocannabinoid system in neuroinflammation. Front. Pharmacol. 12, 806417. doi: 10.3389/fphar.2021.806417

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, Y., Li, L., Nguyen, D. T., Mustafa, S. M., Moore, B. M., and Jiang, J. (2020). Inverse agonism of cannabinoid receptor type 2 confers anti-inflammatory and neuroprotective effects following status epileptics. Mol. Neurobiol. 57, 2830–2845. doi: 10.1007/s12035-020-01923-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: cannabinoids, neuroinflammation, neurodegeneration, non-neuronal cells, editorial

Citation: Vecchiarelli HA, Joers V, Tansey MG and Starowicz K (2022) Editorial: Cannabinoids in neuroinflammation, neurodegeneration and pain: Focus on non-neuronal cells. Front. Neurosci. 16:1114775. doi: 10.3389/fnins.2022.1114775

Received: 02 December 2022; Accepted: 07 December 2022;
Published: 20 December 2022.

Edited and reviewed by: Wendy Noble, King's College London, United Kingdom

Copyright © 2022 Vecchiarelli, Joers, Tansey and Starowicz. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Haley A. Vecchiarelli, yes haleyvecchiarelli@uvic.ca; Valerie Joers, yes vjoers@ufl.edu; Malú Gámez Tansey, yes mgtansey@ufl.edu; Katarzyna Starowicz, yes starow@if-pan.krakow.pl

These authors share first authorship

These authors share senior authorship

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.