Abstract
Cell fate events are regulated by different endogenous developmental factors such as the cell micro‐environment, external or remote signals and epigenetic factors. Among the many regulatory factors, endocannabinoid‐associated signalling pathways are known to conduct several of these events in the developing nervous system and in the adult brain. Interestingly, endocannabinoids exert modulatory actions in both physiological and pathological conditions. Endocannabinoid signalling can promote cell survival by acting on non‐transformed brain cells (neurons, astrocytes or oligodendrocytes) and can have either a protumoural or antitumoural effect on transformed cells. Moreover, endocannabinoids are able to attenuate the detrimental effects on neurogenesis and neuroinflammation associated with ageing. Thus, the endocannabinoid system emerges as an important regulator of cell fate, controlling cell survival/cell death decisions depending on the cell type and its environment.
Linked Articles
This article is part of a themed section on 8th European Workshop on Cannabinoid Research. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.10/issuetoc
Abbreviations
- 2‐AG
2‐arachidonoylglycerol
- ACEA
arachidonyl‐2′‐chloroethylamide
- AEA
anandamide
- CB
cannabinoids
- DAGL
DAG lipase
- eCB
endocannabinoid
- GBM
glioblastoma
- IL‐1ra
IL‐1 receptor antagonist
- LTD
long‐term depression
- MBP
myelin basic protein
- met‐AEA
R(+)‐methanandamide
- mTOR
mammalian target of rapamycin
- NSC
neural stem/progenitor cells
- OPC
oligodendrocyte progenitor
- THC
Δ9‐tetrahydrocannabinol
- TRPV1
transient receptor potential vanilloid 1
Introduction
During embryogenesis, development and tissue turnover, some cells undergo different processes, which include proliferation, migration and differentiation to achieve their final cell identity and perform their specific functions, while other die by apoptosis (Sears and Nevins, 2002). In order to maintain tissue homeostasis and normal cell function, these processes must be tightly regulated; otherwise, uncontrolled proliferation or cell death can appear giving rise to different disorders like cancer or neurodegeneration (Flusberg and Sorberg, 2015). Cell fate events are regulated by different endogenous developmental factors such as cell micro‐environment, external or remote signals and epigenetic factors. Among the many regulatory factors, endocannabinoid (eCB)‐associated signalling pathways are known to mediate several of these events in the developing nervous system and in the adult brain (Guzman, 2005; Maccarrone et al., 2014).
The endogenous cannabinoid (CB) system, or eCB system, is a lipid‐based signalling system involved in the control of neuronal function, inflammation and other physiological processes in mammals (Zou and Kumar, 2018). It is composed of GPCRs (CB1 and CB2 receptors), their endogenous ligands (eCBs), and the proteins involved in the synthesis and degradation of those ligands. The two main eCBs are N‐arachidonoyl‐ethanolamine (AEA; anandamide) and 2‐arachidonoylglycerol (2‐AG), which are both derivatives of arachidonic acid (Pertwee et al., 2010). Both endocannabinods are synthetized on demand. AEA is produced from N‐acyl‐phosphatidylethanolamine (NAPE) by NAPE‐specific PLD and is degraded by fatty acid amide hydrolase (Pacher et al., 2006), whereas 2‐AG is synthetized from DAG by DAG lipase α (DAGLα) or DAGLβ and is degraded by monoacylglycerol lipase (Murataeva et al., 2014).
The eCB system is phylogenetically widespread, with CB1 and CB2 receptors present in all chordates and the enzymes responsible for the synthesis and degradation of eCBs appearing throughout the animal kingdom (Elphick, 2012). The eCB system is highly conserved among species with strong genomic and non‐genomic effects, acting on cell fate decisions, in a time‐ and dose‐dependent modulatory manner. Low doses enhance cell cycle activity (self‐renewal/division) while high doses induce cell cycle arrest/apoptosis (Guzmán et al., 2002). Furthermore, their effects on cell fate are different in non‐transformed cells (without genetic alterations) or in transformed cells (i.e. tumoural cells) (Guzmán et al., 2002). The binding of eCBs to CB1 or CB2 receptors mediate synaptic plasticity or progenitor cell fate in the CNS (Elphick, 2012) to promote brain self‐repair (Molina‐Holgado and Molina‐Holgado, 2010). Moreover, the orphan GPCRs GPR18, GPR55 and GPR119 have been described as CB receptor‐like receptors (Irving et al., 2017). There is also evidence that the protective actions of eCBs are mediated by CB receptor‐independent mechanisms (García‐Arencibia et al., 2007; Wen et al., 2018). For example, several CB compounds interact with the nuclear hormone receptors PPARs (Pistis and O'Sullivan, 2017) and can also exert their actions through the transient receptor potential vanilloid 1 (TRPV1) (Fatahi et al., 2018; Fonseca et al., 2018).
Effects of the endocannabinoid system on transformed and non‐transformed cells
Effects of the endocannabinoid system on non‐transformed neural cells
Neuroimmune signalling and the bioavailability of brain eCBs contribute to the regulation and maintenance of neurogenesis, as well as the differentiation of neural progenitors and their integration in the brain circuitry (Jiang et al., 2007;Goncalves et al., 2008; Molina‐Holgado and Molina‐Holgado, 2010; Maccarrone et al., 2014). CBs are immunomodulatory and neuroprotective in vivo and in vitro and can modify the production and action of inflammatory mediators such as cytokines and chemokines (García‐Ovejero et al., 2013). These are important neuroinflammatory mediators that are involved in the pathological processes resulting from brain trauma, ischaemia and chronic neurodegenerative diseases (Denes et al., 2010). The eCB system has also been shown to be a key participant in the determination of cell fate in remote cell death and its associated mechanisms (Viscomi et al., 2010). However, eCBs are also involved in brain repair and recovery. Compelling evidence obtained, in vivo and in vitro, establishes a dynamic interplay between the eCB system, the immune system and neural stem/progenitor cells (NSC) in order to promote brain self‐repair (Molina‐Holgado and Molina‐Holgado, 2010). Crosstalk between inflammatory mediators and NSC might have important consequences for neural development and brain repair.
eCBs are well‐stablished neuroprotective agents in the CNS as demonstrated in vivo and in vitro. Recently, it has been demonstrated that long‐term depression (LTD) induced by endogenous CBs produces neuroprotection via astroglial CB1 receptors after stroke in rodents (Wang et al., 2018a). eCBs block the astroglial glutamate transporter GLT1 (also known as EAAT2) and activate GluN2B receptors (also known as NR2B), inducing postsynaptic AMPA receptor endocytosis to trigger astroglial eCB‐LTD. Thus, the LTD preconditioning induced by endogenous CBs protects neurons against subsequent lethal ischaemia (Wang et al., 2018a). However, the role of the eCB system in ischaemia is more complicated, as CB1 receptor antagonists have been demonstrated to be neuroprotective too (Reichenbach et al., 2016).
The presence of CB1 receptors in intracellular compartments, such as mitochondria, offers new strategies to pharmacologically act on this receptor in different pathologies (Hebert‐Chatelain et al., 2016). Importantly, eCB protection against mitochondrial dysfunction and oxidative stress is one of the proposed mechanisms to induce neuroprotection (Rangel‐López et al., 2015; Cai et al., 2017). Specifically, the mitochondrial CB1 receptor is involved in arachidonyl‐2′‐chloroethylamide (ACEA)‐induced protective effects on neurons and mitochondrial functions (Ma et al., 2015). Recently, it has been described as a new pathway for the pool of CB1 receptors located on corticostriatal terminals that efficiently safeguards dopamine D1receptors from different insults, and this is particularly relevant in neurodegenerative diseases (Ruiz‐Calvo et al., 2018).
Effects of the endocannabinoid system on oligodendrocytes
The function of CB receptors in oligodendrocytes has received less attention even though classical autoradiographic studies demonstrated that CB receptors were expressed in several white matter regions of the CNS (Herkenham et al., 1991). Later reports presented evidence that functional CB receptors are expressed in oligodendrocyte cultures, in the postnatal and adult corpus callosum and in the spinal cord white matter (Molina‐Holgado et al., 2002; Arevalo‐Martín et al., 2007; Garcia‐Ovejero et al., 2009). Thus, post‐natal rats treated with the CB1/CB2 agonist WIN 55,212‐2 up‐regulated myelin basic protein (MBP)‐immunoreactivity in the subcortical white matter, an effect overridden with CB1 or CB2 antagonists (Arevalo‐Martín et al., 2007). In oligodendrocyte cultures, the synthetic CB agonists, ACEA, JWH133 and HU210, were potent at increasing MBP levels, a marker of oligodendrocyte differentiation, mediated by the PI3K/Akt and mammalian target of rapamycin (mTOR) signalling pathways (Gomez et al., 2011). Moreover, we demonstrated that constitutive release of 2‐AG by late oligodendrocyte progenitors (OPCs) permits oligodendrocyte maturation through the ERK signalling cascade activated by CB receptors (Gomez et al., 2010). However, a basal level of 2‐AG synthesized by OPCs acting via CB1 and CB2 receptors is required to maintain proliferation of early progenitors in culture (Gomez et al., 2015). Indeed, OPC proliferation appears to be mediated by the activation of the PI3K/Akt/mTOR signalling pathways. In addition, inhibition of 2‐AG synthesis or antagonist binding to CB receptors seems to induce cell cycle arrest as evidenced by the down‐regulation of Ki67 or by the increase in the cell cycle inhibitor p27Kip1 (Gomez et al., 2015).
Effects of the endocannabinoid system on tumoural brain cell fate
The in vivo or in vitro effects of eCBs in tumoural cell fate are an open debate in the scientific community that needs deeper investigation. Conflicting reports, suggesting a role for CB signalling in tumour generation and progression or anticancer activity, have been published recently (Velasco et al., 2012; Malfitano et al., 2012; Proto et al., 2012; Pérez‐Gómez et al., 2015; Velasco et al., 2016a) (Figure 1).
Figure 1.

Schematic depiction of the effect of the endocannabinoid system on brain cell survival/cell death decision.
Increased levels of eCBs and their receptors are associated with tumour aggressiveness (Malfitano et al., 2012). In support of the above, a dysregulation of eCB levels, which produced a modified responsiveness to specific ligands, has been shown in different cancer cell lines (Proto et al., 2012). In contrast, intracranially administered Δ9‐tetrahydrocannabinol (THC) has been demonstrated to have an antiproliferative action in a pilot clinical study in patients with recurrent study glioblastoma (GBM) (Guzmán et al., 2006). Interestingly, in the above study, CB receptors were present in GBM biopsies, with no correlation between receptor‐type expression and survival (Guzmán et al., 2006). THC induced both a down‐regulation of CB1 receptors (but not CB2 receptors) and decreased the number of viable cells. These antiproliferative effects of THC were due to apoptosis. However, other studies have suggested that THC induces proliferation of cells in vitro by a CB‐mediated inhibition of host antitumour activation (McKallip et al., 2005). Interestingly, the peripheral CB receptor CB2 has been characterized as a novel murine proto‐oncogen and confirmed to have a role in leukaemia development (Alberich‐Jorda et al., 2003) and in the promotion of renal cell carcinoma prognosis and progression (Wang et al., 2018b). Other studies have implicated CB2 receptors as regulators of HER2 pro‐oncogenic signalling, demonstrating that genetic inactivation of the CB2 receptor impairs tumour generation and progression in MMTV‐neu mice (Pérez‐Gómez et al., 2015).
Furthermore, pharmacological blockade of the CB2 receptor signalling pathway with a selective antagonist/inverse agonist (AM630) impairs the proliferation of human glioblastoma (U87) cells, suggesting that the CB2 receptor is somehow involved in the promoting the proliferation of these glioblastoma cells (Nikoloudakou and Molina‐Holgado, 2017).
Effect of the endocannabinoid signalling pathways on cell death (apoptosis and necrosis)
The eCB system has emerged as an important neuroprotective system through the control of glutamatergic excitotoxicity, calcium influx, oxidative stress, inflammation and autophagy (Fernández‐Ruiz et al., 2015). In contrast, other evidence indicates that the eCB system can also induce cell death in neural and non‐neural cells (Table 1). Thus, the eCB system can control cell fate by determining whether a cell survives or dies.
Table 1.
Signalling mechanisms by which the activation of the endocannabonoid system can cause different types of cell death
| Cell death mechanism | Signalling | References |
|---|---|---|
| Apoptosis | Cell cycle arrest via CB1/CB2 activation | Melck et al., 1999; Caffarel et al., 2006 |
| Cell cycle arrest independent of CB receptor activation | Almada et al., 2017 | |
| Inhibition of cell survival signalling cascades via CB1/CB2 receptor activation | Greenhough et al., 2007; Jia et al., 2006; Orellana‐Serradell et al., 2015 | |
| De novo synthesis of ceramide via CB1/CB2 activation | Galve‐Roperh et al., 2000; Herrera et al., 2006 | |
| NF‐κB‐dependent apoptosis via CB1/CB2 activation | Do et al., 2004 | |
| Superoxide anion formation and activation of caspase‐3 via CB1/CB2 activation | Sarker et al., 2000 | |
| AEA induction of oxidative stress, calcium influx and caspase activation via TRPV1 receptor | Maccarone and Finazzi‐Agró, 2003 | |
| met‐AEA induction of prostaglandins synthesis via PPARγ | Eichele et al., 2009 | |
| AEA‐mediated recruitment and activation of the Fas/FasL death complex via GPR55 receptor | Huang et al., 2011 | |
| AEA and met‐AEA receptor‐independent production of prostaglandins | Hinz et al., 2004b; Soliman and Van Dross, 2016 | |
| Necrosis | Endocannabinoid action on lipid rafts | Fonseca et al., 2009; Fonseca et al., 2010 |
| AEA induction of ROS production and intracellular calcium | Siegmund et al., 2005 | |
| Autophagy | ER stress via CB1/CB2 receptor activation | Velasco et al., 2016b |
eCBs have been shown to induce apoptosis through different signalling mechanisms in several cell types. One of the most common mechanisms is by inhibiting the cell cycle progression, as has been demonstrated, among others, in human breast carcinoma cells where activation of CB1 receptors by AEA causes an inhibition of cAMP production leading to cell cycle arrest at the G1/S phase (Melck et al., 1999), while activation of CB2 receptors by THC in these same cells leads to their arrest in G2‐M (Caffarel et al., 2006). Interestingly, AEA has recently been demonstrated to have the ability to arrest cell cycle through a receptor‐independent pathway, inducing G2/M arrest in a human‐derived endometrial cell line (St‐T1b) directly and by inhibiting the Akt survival pathway (Almada et al., 2017).
This effect on the cell survival signalling cascades emerges as another common mechanism by which the eCB system induces apoptosis. Thus, CB1 receptor activation by THC inhibits both PI3K/Akt and RAS‐MAPK/ERK survival pathways in colorectal cancer cells (Greenhough et al., 2007). Similarly, activation of either CB1 or CB2 receptors with THC was demonstrated to induce an inhibitory effect on ERK and Akt in human Jurkat leukaemia T cells (Jia et al., 2006). However, in prostate cancer cells, the apoptosis induced by eCBs (either AEA or 2‐AG) acting on CB1 receptors was found to be the result of an inhibitory effect on the Akt pathway whereas the ERK signalling cascade was activated (Orellana‐Serradell et al., 2015).
Another major mechanism by which the eCB system can cause apoptosis is by stimulating the de novo synthesis of ceramide, a pro‐apoptotic sphingolipid. This can be due to activation of CB1 receptors in, for example, C6 glioma cells (Galve‐Roperh et al., 2000) and primary astrocytes (Blazquez et al., 2000) where it then causes a sustained accumulation of ceramide that activates the JNK/p38 MAPK pathway leading to cell death. Activation of CB2 receptors, which can also cause ceramide accumulation, has been shown to occur in human Jurkat leukaemia T cells (Herrera et al., 2006), and to lead to mitochondrial‐related apoptosis and in human pancreatic tumour cells by a mechanism that involves endoplasmic reticulum stress related genes (Carracedo et al., 2006). However, in colon cancer cells TNF‐α has been shown to mediate the de novo synthesis of ceramide, induced by activation of either CB1 or CB2 receptors, which then induces apoptosis (Cianchi et al., 2008).
Moreover, activation of CB1 and CB2 receptors on dentritic cells by AEA or THC induces NF‐κB‐dependent apoptosis (Do et al., 2004). When acting through the CB1 receptor, AEA can cause apoptotic cell death of rat phaeochromocytoma PC‐12 cells by increasing superoxide anion formation and activating caspase‐3 (Sarker et al., 2000).
However, eCB‐mediated apoptosis is not exclusively through CB receptors; apoptosis induced by AEA can occur be mediated by stimulation of TRPV1, increasing oxidative stress, calcium influx and caspase activation (Maccarone and Finazzi‐Agró, 2003). R(+)‐methanandamide (met‐AEA), a non‐hydrolysable AEA derivative, induce the synthesis of prostaglandins that lead to the induction of apoptosis in human cervical carcinoma cells by activating PPARγ (Eichele et al., 2009). GPR55, an orphan receptor proposed as a novel CB receptor, may mediate the apoptosis induced by AEA in cholangiocarcinoma cells, which involves the recruitment and activation of the Fas/FasL death complex in lipid rafts (Huang et al., 2011).
eCBs can also induce apoptosis by receptor‐independent effects. In human glioma cells, treatment with met‐AEA causes apoptosis by a mechanism involving the up‐regulation of COX‐2 expression via a pathway linked to lipid raft microdomains (Hinz et al., 2004a) and leading to the production of PGE2 (Hinz et al., 2004b). AEA can be directly metabolized by COX‐2 to form different prostamides (PGE2, PGF2α and PD2). Recently, a novel series of AEA‐derived prostamides, prostamides J, have been demonstrated to induce apoptosis in a receptor‐independent way (Soliman and Van Dross, 2016). Some of the effects previously mentioned to be responsible for the apoptosis induced by AEA, such as ER stress, oxidative stress and action on lipid raft microdomains, have now been reported to be receptor‐independent (Soderstrom et al., 2017).
Besides this ability to induce apoptosis, eCBs can also induce necrosis in some circumstances. Thus, AEA and 2‐AG have a dual effect on rat primary decidual cells. At low concentrations, the eCBs induce apoptosis via CB1 receptors, while at high concentrations they can cause necrosis through a mechanism involving lipid rafts, as methyl‐β‐cyclodextrin (a membrane cholesterol depletor) can block this effect (Fonseca et al., 2009; Fonseca et al., 2010). In primary hepatic stellate cells, high concentrations of AEA cause necrosis, an effect dependent on membrane cholesterol and the induction of ROS and intracellular calcium (Siegmund et al., 2005).
Effect of the endocannabinoid signalling pathways on autophagy
Macroautophagy, hereafter referred to as autophagy, is an evolutionary conserved process in which double‐membrane bound structures called autophagosomes engulf portions of cytoplasm that are delivered to lysosomes for degradation. Basal autophagy plays an important role in maintaining homeostasis, acting during starvation to restore nutrient levels in the cell, and in pathogen infections, removing long‐lived proteins, protein aggregates and damaged organelles. Autophagy dysfunction has been associated with various diseases (Ravikumar et al., 2010).
Autophagy malfunction can cause ‘type II programmed cell death’, which is different from apoptosis or ‘type I programmed cell death’ (Liu and Levine, 2015). Autophagy can be a survival mechanism against apoptosis or can promote apoptotic cell death.
The eCB system can up‐regulate autophagy in transformed and non‐transformed cells. While this up‐regulation generally leads to cell death in cancer cells, it can have therapeutic potential in the case of non‐cancer cells. AEA activation of CB1 receptors induces autophagy in fully differentiated Caco‐2 cells, a model of mature intestinal epithelium, leading to a reduction of the regulatory protein SOCS3 levels (Koay et al., 2014). In contrast, activation of CB2 receptors with the synthetic agonist HU‐308 in the spinal cord of mice with experimental autoimmune encephalomyelitis promotes autophagy, which leads to an inhibition of NLRP3 inflammasome activation, thus alleviating the pathogenesis of this disease in this animal model (Shao et al., 2014). Activation of both CB1 and CB2 receptors by Sativex®, a mixture of THC and cannabidiol, stimulates autophagy in a mouse model of tauopathy, which is associated with decreased levels of tau and amyloid proteins in the brain (Casarejos et al., 2013).
Activation of CB1 and CB2 receptors in different tumour cell lines and animal models of cancer can cause autophagic cell death. The general mechanism involves activation of ER stress‐related proteins such as p8, ATF‐4 and CHOP, which enhances the interaction of tribbles pseudokinase 3 (TRIB3) with Akt, leading to inhibition of the mTOR complex 1 (mTORC1) and the stimulation of autophagy causing cell death (Velasco et al., 2016b). In hepatocellular carcinoma cell lines, activation of the CB2 receptor causes ER stress‐dependent activation of AMP‐activated protein kinase, which inhibits mTORC1 and promotes autophagic cell death in addition to the p8/TRIB3 pathway (Vara et al., 2011).
Neuroimmune interactions of endocannabinoids and neural cell fate decisions
Recent work has highlighted that there is a synergy between the brain immune response and eCB signalling to promote functional recovery, as immune cells help to maintain neurogenesis in germinal centres of the adult CNS even under non‐pathological conditions (Molina‐Holgado and Molina‐Holgado, 2010). Thus, the eCB system, which has neuroprotective and immunomodulatory actions mediated by different signalling cascades in the brain, could assist the process of proliferation and differentiation of embryonic or adult neural stem cells inducing cellular responses by modulating the expression of a number of different genes (Galve‐Roperh et al., 2013). Embryonic/adult neurogenesis is at present one of the most exciting phenomena in neuroscience, but a lack of knowledge about how it works has made it difficult to understand why neurogenesis fails and hence to identify the missing factors that would form the basis for both induction and implantation of cell‐based therapies. Such therapies could be beneficial towards reversing/treating all manner of neurodegenerative diseases as well as providing important insights as to how the brain functions.
Although the role of several cytokines has been examined in neuroinflammation and neurodegeneration, a possible bidirectional interaction between the immune system and brain eCB in cell fate remains to be established. Evidence is emerging that the IL‐1 receptor antagonist (IL‐1ra), an endogenous antagonist for the actions of IL‐1 in the brain, is a potent signal to induce neural stem cell proliferation and migration, enhances neuroblast migration and the number of newly born neurons after cerebral ischaemia (Pradillo et al., 2017). IL‐1ra induces NSC proliferation in vivo and in vitro and mediates anti‐inflammatory and neuroprotective actions of CBs in neurons and glia (Molina‐Holgado et al., 2003; García‐Ovejero et al., 2013).
Similarly, a bi‐directional crosstalk between the TNF‐α and eCB signalling pathways is required to stimulate NSC proliferation (Rubio‐Araiz et al., 2008). However, although TNF‐α is detrimental to neurons after a stroke, it is involved in the repair process, since infusion of anti‐TNF‐α antibodies reduces the survival of newly formed neuroblasts (Heldmann et al., 2005; Katakowski et al., 2007). The above findings support the idea that activation of TNF‐α acting via its receptor TNFR2 leads to 2‐AG synthesis, which may act on the cells' own CB1 and CB2 receptors directing the migration of the progenitor cells to the site of injury. This is consistent with previous findings where DAGLα expression is dramatically down‐regulated when neural stem cells are differentiated (Walker et al., 2010), suggesting that TNF‐α‐induced neural stem proliferation depends on eCB production, release and binding to CB receptors.
The eCB system establishes interactions with other signalling systems that are relevant to the modulation of neural progenitor proliferation or neuritogenesis. Thus, activation of FGF receptors in rat cerebellar granule neurons promotes DAGL synthesis of 2‐AG, which activates CB1 receptors and promotes axonal growth (Williams et al., 2003). Co‐activation of opioid μ receptors and CB1 receptors in Neuro‐2A mouse neuroblastoma cells causes an attenuation of the neurite outgrowth seen upon the activation of each receptor type alone (Rios et al., 2006). Co‐stimulation of CB1 receptor and D2 dopamine receptors induce the formation of heterodimers of both types of receptor, which affects CB1 receptor signalling (Kearn et al., 2005). Finally, eicosapentaenoic acid, an ω‐3 polyunsaturated fatty acid, increases the proliferation of neural stem cells by increasing the levels of 2‐AG, causing the activation of CB1 and CB2 receptors (Dyall et al., 2016).
Conclusions
Cell fate decisions are key in the homeostatic maintenance of the cellular milieu as well as in the survival of tissues and organisms. The eCB network is an important regulator of brain cell fate determination in health and in pathological conditions. Signal transduction via cannabinoid receptors leads to the proliferation, differentiation and cell death events of brain cells, and this has important consequences for neural development and brain repair. Thus, eCB signalling‐mediated maintenance of synaptic plasticity and neuronal cell function provide potential opportunities for endogenous stem cell‐based neuroreparative strategies. The effect of the eCB system on cell death makes it a promising target to treat different brain tumours, although more studies are needed to avoid the pro‐tumoural effects shown in some models, especially when the CB2 receptor is activated. Furthermore, the bi‐directional crosstalk of the brain eCB system with the immune system drives neuroprotective and anti‐inflammatory actions that could assist the process of proliferation and differentiation of embryonic or adult neural stem cells.
Nomenclature of targets and ligands
Key protein targets and ligands in this article are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Harding et al., 2018), and are permanently archived in the Concise Guide to PHARMACOLOGY 2017/18 (Alexander et al., 2017a,b,c,d,e,f,g).
Conflict of interest
The authors declare no conflicts of interest.
Garcia‐Arencibia, M. , Molina-Holgado, E. , and Molina‐Holgado, F. (2019) Effect of endocannabinoid signalling on cell fate: life, death, differentiation and proliferation of brain cells. British Journal of Pharmacology, 176: 1361–1369. 10.1111/bph.14369.
References
- Alberich‐Jorda M, Rayman N, Valk P, De Wee E, Del Wel R (2003). Identification, characterization, and function of a novel oncogene: the peripheral cannabinoid receptor CB2. Ann N Y Acad Sci 996: 10–16. [DOI] [PubMed] [Google Scholar]
- Alexander SPH, Christopoulos A, Davenport AP, Kelly E, Marrion NV, Peters JA et al (2017a). The Concise Guide to PHARMACOLOGY 2017/18: G protein‐coupled receptors. Br J Pharmacol 174: S17–S129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Cidlowski JA, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017b). The Concise Guide to PHARMACOLOGY 2017/18: Nuclear hormone receptors. Br J Pharmacol 174: S208–S224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017c). The Concise Guide to PHARMACOLOGY 2017/18: Catalytic receptors. Br J Pharmacol 174: S225–S271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Fabbro D, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017d). The Concise Guide to PHARMACOLOGY 2017/18: Enzymes. Br J Pharmacol 174: S272–S359. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Kelly E, Marrion NV, Peters JA, Faccenda E, Harding SD et al (2017e). The Concise Guide to PHARMACOLOGY 2017/18: Transporters. Br J Pharmacol 174: S360–S446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Peters JA, Kelly E, Marrion NV, Faccenda E, Harding SD et al (2017f). The Concise Guide to PHARMACOLOGY 2017/18: Ligand‐gated ion channels. Br J Pharmacol 174: S130–S159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alexander SPH, Striessnig J, Kelly E, Marrion NV, Peters JA, Faccenda E et al (2017g). The Concise Guide To PHARMACOLOGY 2017/18: Voltage‐gated ion channels. Br J Pharmacol 174: S160–S194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almada M, Fonseca BM, Amaral C, Diniz‐da‐Costa M, Correia‐da‐Silva G, Teixeira N (2017). Anandamide oxidative metabolism‐induced endoplasmic reticulum stress and apoptosis. Apoptosis 22: 816–826. [DOI] [PubMed] [Google Scholar]
- Arevalo‐Martín A, Garcia‐Ovejero D, Rubio‐Araiz A, Gomez O, Molina‐Holgado F, Molina‐Holgado E (2007). Cannabinoids modulate Olig2 and polysialylated neural cell adhesion molecule expression in the subventricular zone of post‐natal rats through cannabinoid receptor 1 and cannabinoid receptor 2. Eur J Neurosci 26: 1548–1559. [DOI] [PubMed] [Google Scholar]
- Blazquez C, Galve‐Roperh I, Guzman M (2000). De novo‐synthesized ceramide signals apoptosis in astrocytes via extracellular signal‐regulated kinase. FASEB J 14: 2315–2322. [DOI] [PubMed] [Google Scholar]
- Caffarel MM, Sarrio D, Palacios J, Guzman M, Sanchez C (2006). Delta9‐tetrahydrocannabinol inhibits cell cycle progression in human breast cancer cells through Cdc2 regulation. Cancer Res 66: 6615–6621. [DOI] [PubMed] [Google Scholar]
- Cai M, Yang Q, Li G, Sun S, Chen Y, Tian L et al (2017). Activation of cannabinoid receptor 1 is involved in protection against mitochondrial dysfunction and cerebral ischaemic tolerance induced by isoflurane preconditioning. Br J Anaesth 119: 1213–1223. [DOI] [PubMed] [Google Scholar]
- Casarejos MJ, Perucho J, Gomez A, Muñoz MP, Fernandez‐Estevez M, Sagredo O et al (2013). Natural cannabinoids improve dopamine neurotransmission and tau and amyloid pathology in a mouse model of tauopathy. J Alzheimers Dis 35: 525–539. [DOI] [PubMed] [Google Scholar]
- Carracedo A, Gironella M, Lorente M, Garcia S, Guzman M, Velasco G et al (2006). Cannabinoids induce apoptosis of pancreatic tumor cells via endoplasmic reticulum stress‐related genes. Cancer Res 66: 6748–6755. [DOI] [PubMed] [Google Scholar]
- Cianchi F, Papucci L, Schiavone N, Lulli M, Magnelli L, Vinci MC et al (2008). Cannabinoid receptor activation induces apoptosis through tumor necrosis factor alpha‐mediated ceramide de novo synthesis in colon cancer cells. Clin Cancer Res 14: 7691–7700. [DOI] [PubMed] [Google Scholar]
- Denes A, Thornton P, Rothwell NJ, Allan SM (2010). Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain Behav Immun 24: 708–723. [DOI] [PubMed] [Google Scholar]
- Do Y, McKallip RJ, Nagarkatti M, Nagarkatti PS (2004). Activation through cannabinoid receptors 1 and 2 on dendritic cells triggers NF‐kappaB‐dependent apoptosis: novel role for endogenous and exogenous cannabinoids in immunoregulation. J Immunol 173: 2373–2382. [DOI] [PubMed] [Google Scholar]
- Dyall SC, Mandhair HK, Fincham REA, Kerr DM, Roche M, Molina‐Holgado F (2016). Distinctive effects of eicosapentaenoic and docosahexaenoic acids in regulating neural stem cell fate are mediated via endocannabinoid signalling pathways. Neuropharmacology 107: 387–395. [DOI] [PubMed] [Google Scholar]
- Eichele K, Ramer R, Hinz B (2009). R(+)‐methanandamide‐induced apoptosis of human cervical carcinoma cells involves a cyclooxygenase‐2‐ dependent pathway. Pharm Res 26: 346–355. [DOI] [PubMed] [Google Scholar]
- Elphick MR (2012). The evolution and comparative neurobiology of endocannabinoid signalling. Philos Trans R Soc Lond B Biol Sci 367: 3201–3215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fatahi Z, Reisi Z, Rainer G, Haghparast A, Khani A (2018). Cannabinoids induce apathetic and impulsive patterns of choice through CB1 receptors and TRPV1 channels. Neuropharmacology 133: 75–84. [DOI] [PubMed] [Google Scholar]
- Fernández‐Ruiz J, Romero J, Ramos JA (2015). Endocannabinoids and neurodegenerative disorders: Parkinson's disease, Huntington's chorea, Alzheimer's disease, and others. Handb Exp Pharmacol 231: 233–259. [DOI] [PubMed] [Google Scholar]
- Flusberg DA, Sorberg PK (2015). Surviving apoptosis: life‐death signaling in single cells. Trends Cell Biol 25: 446–458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fonseca BM, Correia‐da‐Silva G, Teixeira NA (2009). Anandamide‐induced cell death: dual effects in primary rat decidual cell cultures. Placenta 30: 686–692. [DOI] [PubMed] [Google Scholar]
- Fonseca BM, Correia‐da‐Silva G, Taylor AH, Lam PM, Marczylo TH, Bell SC et al (2010). The endocannabinoid 2‐arachidonoylglycerol (2‐AG) and metabolizing enzymes during rat fetoplacental development: a role in uterine remodelling. Int J Biochem Cell Biol 42: 1884–1892. [DOI] [PubMed] [Google Scholar]
- Fonseca BM, Correia‐da‐Silva G, Teixeira NA (2018). Cannabinoid‐induced cell death in endometrial cancer cells: involvement of TRPV1 receptors in apoptosis. J Physiol Biochem 74: 261–272. [DOI] [PubMed] [Google Scholar]
- Galve‐Roperh I, Chiurchiù V, Díaz‐Alonso J, Bari M, Guzmán M, Maccarrone M (2013). Cannabinoid receptor signaling in progenitor/stem cell proliferation and differentiation. Prog Lipid Res 52 (4): 633–650. [DOI] [PubMed] [Google Scholar]
- Galve‐Roperh I, Sanchez C, Cortes ML, del Pulgar TG, Izquierdo M, Guzman M (2000). Anti‐tumoral action of cannabinoids: involvement of sustained ceramide accumulation and extracellular signal‐regulated kinase activation. Nat Med 6: 313–319. [DOI] [PubMed] [Google Scholar]
- García‐Arencibia M, González S, de Lago E, Ramos JA, Mechoulam R, Fernández‐Ruiz J (2007). Evaluation of the neuroprotective effect of cannabinoids in a rat model of Parkinson's disease: importance of antioxidant and cannabinoid receptor‐independent properties. Brain Res 1134: 162–170. [DOI] [PubMed] [Google Scholar]
- García‐Ovejero D, Arévalo‐Martín Á, Navarro‐Galve B, Pinteaux E, Molina‐Holgado E, Molina‐Holgado F (2013). Neuroimmmune interactions of cannabinoids in neurogenesis: focus on interleukin‐1β (IL‐1β) signalling. Biochem Soc Trans 41: 1577–1582. [DOI] [PubMed] [Google Scholar]
- Garcia‐Ovejero D, Arevalo‐Martin A, Petrosino S, Docagne F, Hagen C, Bisogno T et al (2009). The endocannabinoid system is modulated in response to spinal cord injury in rats. Neurobiol Dis 33: 57–71. [DOI] [PubMed] [Google Scholar]
- Gomez O, Arevalo‐Martin A, Garcia‐Ovejero D, Ortega‐Gutierrez S, Cisneros JA, Almazan G et al (2010). The constitutive production of the endocannabinoid 2‐arachidonoylglycerol participates in oligodendrocyte differentiation. Glia 58: 1913–1927. [DOI] [PubMed] [Google Scholar]
- Gomez O, Sanchez‐Rodriguez A, Le M, Sanchez‐Caro C, Molina‐Holgado F, Molina‐Holgado E (2011). Cannabinoid receptor agonists modulate oligodendrocyte differentiation by activating PI3K/Akt and the mammalian target of rapamycin (mTOR) pathways. B J Pharmacol 163: 1520–1532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomez O, Sanchez‐Rodriguez MA, Ortega‐Gutierrez S, Vazquez‐Villa H, Guaza C, Molina‐Holgado F et al (2015). A basal tone of 2‐arachidonoylglycerol contributes to early oligodendrocyte progenitor proliferation by activating phosphatidylinositol 3‐kinase (PI3K)/AKT and the mammalian target of rapamycin (MTOR) pathways. J Neuroimmune Pharmacol 10: 309–317. [DOI] [PubMed] [Google Scholar]
- Goncalves MB, Suetterlin P, Yip P, Molina‐Holgado F, Walker DJ, Oudin MJ et al (2008). A diacylglycerol lipase‐CB2 cannabinoid pathway regulates adult subventricular zone neurogenesis in an age‐dependent manner. Mol Cell Neurosci 38: 526–536. [DOI] [PubMed] [Google Scholar]
- Greenhough A, Patsos HA, Williams AC, Paraskeva C et al (2007). The cannabinoid delta(9)‐tetrahydrocannabinol inhibits RAS‐MAPK and PI3K‐AKT survival signalling and induces BAD‐mediated apoptosis in colorectal cancer cells. Int J Cancer 121: 2172–2180. [DOI] [PubMed] [Google Scholar]
- Guzmán M, Sánchez C, Galve‐Roperh I (2002). Cannabinoids and cell fate. Pharmacol Ther 95: 175–184. [DOI] [PubMed] [Google Scholar]
- Guzman M (2005). Effects on cell viability. Handb Exp Pharmacol 168: 627–642. [DOI] [PubMed] [Google Scholar]
- Guzmán M, Duarte MJ, Blázquez C, Ravina J, Rosa MC, Galve‐Roperh I et al (2006). A pilot clinical study of Delta9‐tetrahydrocannabinol in patients with recurrent glioblastoma multiforme. Br J Cancer 95: 197–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harding SD, Sharman JL, Faccenda E, Southan C, Pawson AJ, Ireland S et al (2018). The IUPHAR/BPS Guide to PHARMACOLOGY in 2018: updates and expansion to encompass the new guide to IMMUNOPHARMACOLOGY. Nucl Acids Res 46: D1091–D1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hebert‐Chatelain E, Desprez T, Serrat R, Bellocchio L, Soria‐Gomez E, Busquets‐Garcia A et al (2016). A cannabinoid link between mitochondria and memory. Nature 539: 555–559. [DOI] [PubMed] [Google Scholar]
- Heldmann U, Thored P, Claasen JH, Arvidsson A, Kokaia Z, Lindvall O (2005). TNFalpha antibody infusion impairs survival of stroke‐generated neuroblasts in adult rat brain. Exp Neurol 196: 204–208. [DOI] [PubMed] [Google Scholar]
- Herkenham M, Lynn AB, Johnson MR, Melvin LS, de Costa BR, Rice KC (1991). Characterization and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic study. J Neurosci 11: 563–583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrera B, Carracedo A, Diez‐Zaera M, delPulgar TG, Guzman M, Velasco G (2006). The CB2 cannabinoid receptor signals apoptosis via ceramide‐dependent activation of the mitochondrial intrinsic pathway. Exp Cell Res 312: 2121–2131. [DOI] [PubMed] [Google Scholar]
- Hinz B, Ramer R, Eichele K, Weinzierl U, Brune K (2004a). R(+)‐methanandamide‐induced cyclooxygenase‐2 expression in H4 human neuroglioma cells: possible involvement of membrane lipid rafts. Biochem Biophys Res Commun 324: 621–626. [DOI] [PubMed] [Google Scholar]
- Hinz B, Ramer R, Eichele K, Weinzierl U, Brune K (2004b). Up‐regulation of cyclooxygenase‐2 expression is involved in R(+)‐methanandamide‐induced apoptotic death of human neuroglioma cells. Mol Pharmacol 66: 1643–1651. [DOI] [PubMed] [Google Scholar]
- Huang L, Ramirez JC, Frampton GA, Golden LE, Quinn MA, Pae HY et al (2011). Anandamide exerts its antiproliferative actions on cholangiocarcinoma by activation of the GPR55 receptor. Lab Invest 91: 1007–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Irving A, Abdulrazzaq G, Chan SLF, Penman J, Harvey J, Alexander SPH (2017). Cannabinoid receptor‐related orphan G protein‐coupled receptors. Adv Pharmacol 80: 223–247. [DOI] [PubMed] [Google Scholar]
- Jia W, Hegde VL, Singh NP, Sisco D, Grant S, Nagarkatti M et al (2006). Delta9‐tetrahydrocannabinol‐induced apoptosis in Jurkat leukemia T cells is regulated by translocation of bad to mitochondria. Mol Cancer Res 4: 549–562. [DOI] [PubMed] [Google Scholar]
- Jiang S, Fu Y, Williams J, Wood J, Pandarinathan L, Avraham S et al (2007). Expression and function of cannabinoid receptors CB1 and CB2 and their cognate cannabinoid ligands in murine embryonic stem cells. PLoS One 2: e641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Katakowski M, Chen J, Zhang ZG, Santra M, Wang Y, Chopp M (2007). Stroke‐induced subventricular zone proliferation is promoted by tumor necrosis factor‐alpha converting enzyme protease activity. J Cereb Blood Flow Metab 27: 669–678. [DOI] [PubMed] [Google Scholar]
- Kearn CS, Blake‐Palmer K, Daniel E, Mackie K, Glass M (2005). Concurrent stimulation of cannabinoid CB1 and dopamine D2 receptors enhances heterodimer formation: a mechanism for receptor cross‐talk? Mol Pharmacol 67: 1697–1704. [DOI] [PubMed] [Google Scholar]
- Koay LC, Rigby RJ, Wright KL (2014). Cannabinoid‐induced autophagy regulates suppressor of cytokine signaling‐3 in intestinal epithelium. Am J Physiol Gastrointest Liver Physiol 307: G140–G148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Levine B (2015). Autosis and autophagic cell death: the dark side of autophagy. Cell Death Differ 22: 367–376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma L, Jia J, Niu W, Jiang T, Zhai Q, Yang L et al (2015). Mitochondrial CB1 receptor is involved in ACEA‐induced protective effects on neurons and mitochondrial functions. Sci Rep 5: 12440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maccarone M, Finazzi‐Agró A (2003). The endocannabinoid system, anandamide and the regulation of mammalian cell apoptosis. Cell Death Differ 10: 946–955. [DOI] [PubMed] [Google Scholar]
- Maccarrone M, Guzmán M, Mackie K, Doherty P, Harkany T (2014). Programming of neural cells by (endo)cannabinoids: from physiological rules to emerging therapies. Nat Rev Neurosci 15: 786–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malfitano AM, Laezza C, Galgani M, Matarese G, D'Alessandro A, Gazzerro P et al (2012). The CB1 receptor antagonist rimonabant controls cell viability and ascitic tumour growth in mice. Pharmacol Res 65: 365–371. [DOI] [PubMed] [Google Scholar]
- McKallip RJ, Nagarkatti M, Nagarkatti PS (2005). Delta‐9‐tetrahydrocannabinol enhances breast cancer growth and metastasis by suppression of the antitumor immune response. J Immunol 174: 3281–3289. [DOI] [PubMed] [Google Scholar]
- Melck D, Rueda D, Galve‐Roperh I, De Petrocellis L, Guzman M, Di Marzo V (1999). Involvement of the cAMP/protein kinase A pathway and of mitogen‐activated protein kinase in the anti‐proliferative effects of anandamide in human breast cancer cells. FEBS Lett 463: 235–240. [DOI] [PubMed] [Google Scholar]
- Molina‐Holgado E, Molina‐Holgado F (2010). Mending the broken brain: neuroimmune interactions in neurogenesis. J Neurochem 114: 1277–1290. [DOI] [PubMed] [Google Scholar]
- Molina‐Holgado F, Pinteaux E, Moore JD, Molina‐Holgado E, Guaza C, Gibson RM et al (2003). Endogenous interleukin‐1 receptor antagonist mediates anti‐inflammatory and neuroprotective actions of cannabinoids in neurons and glia. J Neurosci 23: 6470–6474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Molina‐Holgado E, Vela JM, Arevalo‐Martın A, Almazan G, Molina‐Holgado F, Borrell J et al (2002). Cannabinoids promote oligodendrocyte progenitor survival: Involvement of cannabinoid receptors and phosphatidylinositol‐3kinase/Akt signaling. J Neurosci 22: 9742–9753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murataeva N, Straiker A, Mackie K (2014). Parsing the players: 2‐arachidonoylglycerol synthesis and degradation in the CNS. Br J Pharmacol 171: 1379–1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nikoloudakou A, Molina‐Holgado F (2017). AM630, a CB2 cannabinoid receptor antagonist, inhibits proliferation of human U87 glioblastoma cells by targeting the mitochondrial unfolded protein response. Proceedings of the British Pharmacological Society at 8th European Workshop on Cannabinoid Research 17–1
- Orellana‐Serradell O, Poblete CE, Sanchez C, Castellon EA, Gallegos I, Huidobro C et al (2015). Proapoptotic effect of endocannabinoids in prostate cancer cells. Oncol Rep 33: 1599–1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pacher P, Batkai S, Kunos G (2006). The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev 58: 389–462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pérez‐Gómez E, Andradas C, Blasco‐Benito S, Caffarel MM, García‐Taboada E, Villa‐Morales M et al (2015). Role of cannabinoid receptor CB2 in HER2 pro‐oncogenic signalling in breast cancer. J Natl Cancer Inst 107: djv077. [DOI] [PubMed] [Google Scholar]
- Pertwee RG, Howlett AC, Abood ME, Alexander SP, di Marzo V, Elphick MR et al (2010). International union of basic and clinical pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB1and CB2. Pharmacol Rev 62: 588–631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pistis M, O'Sullivan SE (2017). The role of nuclear hormone receptors in cannabinoid function. Adv Pharmacol 80: 291–328. [DOI] [PubMed] [Google Scholar]
- Pradillo JM, Murray KN, Coutts GA, Moraga A, Oroz‐Gonjar F, Boutin H et al (2017). Reparative effects of interleukin‐1 receptor antagonist in young and aged/co‐morbid rodents after cerebral ischemia. Brain Behav Immun 61: 117–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Proto MC, Gazzerro P, Di Croce L, Santoro A, Malfitano AM, Pisanti S et al (2012). Interaction of endocannabinoid system and steroid hormones in the control of colon cancer cell growth. J Cell Physiol 227: 250–258. [DOI] [PubMed] [Google Scholar]
- Rangel‐López E, Colín‐González AL, Paz‐Loyola AL, Pinzón E, Torres I, Serratos IN et al (2015). Cannabinoid receptor agonists reduce the short‐term mitochondrial dysfunction and oxidative stress linked to excitotoxicity in the rat brain. Neuroscience 285: 97–106. [DOI] [PubMed] [Google Scholar]
- Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia‐Arencibia M, Green‐Thompson ZW et al (2010). Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90: 1383–1435. [DOI] [PubMed] [Google Scholar]
- Reichenbach ZW, Li H, Ward SJ, Tuma RF (2016). The CB1 antagonist, SR141716A, is protective in permanent photothrombotic cerebral ischemia. Neurosci Lett 630: 9–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rios C, Gomes I, Devi LA (2006). Mu opioid and CB1 cannabinoid receptor interactions: reciprocal inhibition of receptor signaling and neuritogenesis. Br J Pharmacol 148: 387–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rubio‐Araiz A, Arévalo‐Martín A, Gómez‐Torres O, Navarro‐Galve B, García‐Ovejero D, Suetterlin P et al (2008). The endocannabinoid system modulates a transient TNF pathway that induces neural stem cell proliferation. Mol Cell Neurosci 38: 374–380. [DOI] [PubMed] [Google Scholar]
- Ruiz‐Calvo A, Maroto IB, Bajo‐Grañeras R, Chiarlone A, Gaudioso Á, Ferrero JJ et al (2018). Pathway‐specific control of striatal neuron vulnerability by corticostriatal cannabinoid CB1 receptors. Cereb Cortex 28: 307–322. [DOI] [PubMed] [Google Scholar]
- Sarker KP, Obara S, Nakata M, Kitajima I, Maruyama I (2000). Anandamide induces apoptosis of PC‐12 cells: involvement of superoxide and caspase‐3. FEBS Lett 472: 39–44. [DOI] [PubMed] [Google Scholar]
- Sears RC, Nevins JR (2002). Signaling networks that link cell proliferation and cell fate. J Cell Biol 277: 11617–11620. [DOI] [PubMed] [Google Scholar]
- Shao BZ, Wei W, Ke P, Xu ZQ, Zhou JX, Liu C (2014). Activating cannabinoid receptor 2 alleviates pathogenesis of experimental autoimmune encephalomyelitis via activation of autophagy and inhibiting NLRP3 inflammasome. CNS NeurosciTher 20: 1021–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Siegmund SV, Uchinami H, Osawa Y, Brenner DA, Schwabe RF (2005). Anandamide induces necrosis in primary hepatic stellate cells. Hepatology 41: 1085–1095. [DOI] [PubMed] [Google Scholar]
- Soliman E, Van Dross R (2016). Anandamide‐induced endoplasmic reticulum stress and apoptosis are mediated by oxidative stress in nonmelanoma skin cancer: receptor‐independent endocannabinoid signaling. Mol Carcinog 55: 1807–1821. [DOI] [PubMed] [Google Scholar]
- Soderstrom K, Soliman E, van Dross R (2017). Cannabinoids modulate neuronal activity and cancer by CB1 and CB2 receptor‐independent mechanisms. Front Pharmacol 8: 720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vara D, Salazar M, Olea‐Herrero N, Guzmán M, Velasco G, Díaz‐Laviada I (2011). Anti‐tumoral action of cannabinoids on hepatocellular carcinoma: role of AMPK‐dependent activation of autophagy. Cell Death Differ 18: 1099–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Velasco G, Sánchez C, Guzmán M (2012). Towards the use of cannabinoids as antitumouragents. Nat Rev Cancer 12: 436–444. [DOI] [PubMed] [Google Scholar]
- Velasco G, Hernández‐Tiedra S, Dávila D, Lorente M (2016a). The use of cannabinoids as anticancer agents. Prog Neuropsychopharmacol Biol Psychiatry 64: 259–266.26071989 [Google Scholar]
- Velasco G, Sánchez C, Guzmán M (2016b). Anticancer mechanisms of cannabinoids. Curr Oncol 23: S23–S32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Viscomi MT, Oddi S, Latini L, Bisicchia E, Maccarrone M, Molinari M (2010). The endocannabinoid system: a new entry in remote cell death mechanisms. Exp Neurol 224: 56–65. [DOI] [PubMed] [Google Scholar]
- Walker DJ, Suetterlin P, Reisenberg M, Williams G, Doherty P (2010). Down‐regulation of diacylglycerol lipase‐alpha during neural stem cell differentiation: identification of elements that regulate transcription. J Neurosci Res : 735–745. [DOI] [PubMed] [Google Scholar]
- Wang F, Han J, Higashimori H, Wang J, Liu J, Tong L et al (2018a). Long‐term depression induced by endogenous cannabinoids produces neuroprotection via astroglial CB1R after stroke in rodents. J Cereb Blood Flow Metab. 10.1177/0271678X18755661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang J, Xu Y, Zhu L, Zou Y, Kong W, Dong B et al (2018b). Cannabinoid receptor 2 as a novel target for promotion of renal cell carcinoma prognosis and progression. J Cancer Res Clin Oncol 144: 39–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wen J, Jones M, Tanaka M, Selvaraj P, Symes AJ, Cox B et al (2018). WWL70 protects against chronic constriction injury‐induced neuropathic pain in mice by cannabinoid receptor‐independent mechanisms. J Neuroinflammation 15: 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams EJ, Walsh FS, Doherty P (2003). The FGF receptor uses the endocannabinoid signaling system to couple to an axonal growth response. J Cell Biol 17: 481–486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou S, Kumar U (2018). Cannabinoid receptors and the endocannabinoid system: signaling and function in the central nervous system. Int J Mol Sci 19: 833. [DOI] [PMC free article] [PubMed] [Google Scholar]
