More Information

Submitted: April 22, 2021 | Approved: May 31, 2021 | Published: June 03, 2021

How to cite this article: Dutta R. Endocannabinoidome and its role in neurological disorders-A comprehensive update of existing literature. J Neurosci Neurol Disord. 2021; 5: 034-047.

DOI: 10.29328/journal.jnnd.1001049

ORCiD: orcid.org/0000-0002-6129-1038

Copyright License: © 2021 Dutta R. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Keywords: Cannabinoid; CB1; CB2; Receptors; Neurological disorders

 FullText PDF

Endocannabinoidome and its role in neurological disorders-A comprehensive update of existing literature

Rajib Dutta*

MD, Neurology, India

*Address for Correspondence: Rajib Dutta, MD, Neurology, India, Email: rajibdutta808@gmail.com

Medical benefits of cannabis and related compounds is widely known. Discovery of psychotropic plant cannabinoid Δ9-tetrahydrocannabinol have urged researchers to study more about the cannabinoid system and related therapeutics in the field of neurology and medicine. Where activation of cannabinoid receptor type 1 (CB1R) yielded in unwanted and serious side effects, discovery of cannabinoid receptor type 2 (CB2R) and its ligands gave a new hope. Till now there is limited success in this field because of complex expanded endocannabinoid system comprising of receptors, ligands and enzymes. In this review we will update about the role of endocannabinoidome relevant to neurological disorders.

Anecdotal evidence and recent case reports talked about the therapeutic effect of cannabis sativa [1]. First cannabis derived compound, dronabinol was approved in clinical practice in late 20th century. In 2011, it was approved for treating spasticity in multiple sclerosis (MS) [2-4]. Two specific cannabinoids discovered in the 1960s were Δ9-tetrahydrocannabinol (THC) and the non-euphoric cannabidiol (CBD) [5]. However, mechanism of action of THC, a psychotropic component of marijuana gave us the insight and discovery of the receptors in 1990s [6,7], consequently, of endogenous ligands of these receptors, endocannabinoid [8].

Endocannabinoids and cannabinoid receptors are signaling molecules which are mainly pleiotropic in nature. They are responsible in maintaining homeostatic milieu in central and peripheral nervous system after a significant pathological insult. This ultimately leads to treatment opportunities of complicated neurological disorders as well as diseases of the peripheral organ systems [9-13]. Study in animal models showed that this signaling system is altered in neurological diseases [14]. Nabiximols (Sativex®), a combination of THC and CBD, was an important breakthrough drug for MS patients in treating pain and moderate to severe spasticity [3].

Endocannabinoid system consists of CB1, CB2, two endocannabinoids especially 2-arachidonoylglycerol (2-AG) and anandamide, and anabolic and catabolic enzymes of the endocannabinoid family. Endocannabinoid signaling is related to multiple neurological diseases. It participates in development of brain, release of neurotransmitters, neuronal plasticity, and cytokine release from microglial cells thereby maintaining homeostasis at a cellular level [15].

A new expanded signaling system, called as endocannabinoidome was coined because endocannabinoids activated different receptors and their anabolic/biosynthetic and catabolic pathways were often shared with other mediators. A lot of interest has been generated recently in research community in decoding the role of endocannabinoidome, anabolic/catabolic enzymes, allosteric modulators or inhibitors in terms of brain and especially neuronal pathology [16].

THC was considered to be a psychotropic agent but many unique compounds are developed now and almost none are psychotropic. Central and peripheral targets have been identified and being studied [17]. CBD modulates the activity of several proteins whereas THC induces psycho-activity thereby giving CBD an edge [18,19].

CB1 receptor is found in both peripheral and central nervous system. CB2 is mainly responsible for psychoactive properties of THC, active agent in medical cannabis and most importantly in determining endocannabinoid-mediated presynaptic-inhibition efficacy. Both of them are G protein- coupled receptor (GPCR) based [6,7].

Inhibition of fatty acid amide hydrolase (FAAH) controls degradation of endocannabinoid. The mechanism is complex and associated with activation of transient receptor potential cation channel subfamily V member 1 (TRPV1), G protein-coupled receptor 119 (GPCR 119/GPR119), orphan G protein-coupled receptor 55 (GPR55), and peroxisome proliferator-activated receptor-alpha (PPARα) [14].

TRPV1, GPR 55/119, and PPARα often functions opposite to those of cannabinoid receptors [20-23]. 2-AG is a precursor of arachidonic acid and pro-inflammatory prostanoids, so beneficial effects of monoacylglycerol lipase (MAGL) inhibitors, particularly those seen in experimental models of Alzheimer’s disease (AD) and Parkinson’s disease (PD) might be mediated by inhibition of prostanoid receptor signaling [24,25].

The super complex signaling system comprises of endocannabinoid-related molecules extending to several long- chain N- acyl-amides mainly N- acyl-taurines, serotonin, dopamine, and fatty acid primary amide [26].

The most likely location of CB1is presynaptic in both excitatory and inhibitory neurons [27,28]. CB1 controls vesicular release of gamma aminobutyric acid (GABA) or glutamate by inhibiting voltage-gated Ca2+ channels [27]. Endocannabinoids, particularly 2-AG, are inhibitory retrograde neuromodulators [29]. Slow self-inhibition of neocortical interneurons is mediated by postsynaptic CB1 receptors [30].

A small proportion of postsynaptic CB1 is located in the external membrane of mitochondria [31], where it inhibits electron transport and the respiratory chain, thereby affecting brain metabolism and memory formation [32]. Activation of CB1 also stimulates proliferation of adult progenitor stem cells and their differentiation into neurons or astrocytes [33], a role that could be relevant to neurodegenerative disorders.

CB2 is expressed in microglia in diseases such as amyotrophic lateral sclerosis (ALS), MS, AD as evident from pathological studies of human brain samples [34]. CB2 activation also stimulates adult neurogenesis [35], and plays an active role in regulating blood–brain barrier (BBB) permeability [36]. CB2 is expressed at very low levels in healthy neurons and that their activation has the opposite effects to CB1 activation [37,38]. One study has suggested that activation of postsynaptic CB2 reduces neuronal excitability in the CA3 and CA2 regions of the hippocampus through functional coupling with the sodium bicarbonate transporter [39].

The most studied of the receptors involved in the wider endocannabinoidome are TRPV1, peroxisome proliferator-activated receptor -gamma (PPARγ) and PPARα, although some work has addressed the role of two orphan GPCRs, GPR55 and GPR18. TRPV1 is found in glutamatergic, GABAergic terminals, and neuronal soma in the hippocampus and cerebellum [40,41].

PPARγ and PPARα are expressed in brain cells like microglia and astrocytes and even neurons. They exhibit neuroprotective effects and inhibit neuroinflammation during any acute or chronic insults in the form of traumatic brain injury, ischemic insult, neurodegenerative diseases like AD and MS [42].

The role of GPR 55 is controversial but evidence suggests that its activation stimulates excitatory hippocampal neurons [43] and only little information is available of GPR 18. However, presence of GPR 18 in microglia has neuroinflammatory function [44].

Neurological disorders involved with endocannabinoi-dome system

Parkinson’s Disease (PD): TRPV1 and CB1R are responsible for CBD-induced analgesic effect by increasing endogenous anandamide levels. Reduction of parkinsonism-induced nociceptive threshold can be achieved by CBD [45].

Palmitoylethanolamide (PEA) is a nutraceutical endocannabinoid found in egg yolks. It is responsible for targeting non classical cannabinoid receptors and shows no excitatory modulation of CB1 and CB2. Classical receptors of cannabinoid system are only activated by entourage effect [46]. In a mouse-model of PD, PEA restored tyrosine hydroxylase activity in the SNPc, thereby improving dopamine neurotransmission [198,199]

In MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced PD model intraperitoneal PEA injections, maintained expression of tyrosine hydroxylase activity, prevented parkinsonian like behaviors, and had a blunted effect on upregulation of a-synuclein [200]. Addition of ultramicronized PEA to PD patients already receiving levodopa therapy significantly reduced most of the motor and non-motor symptoms [50].

Another study reported cannabinoid receptor antagonist (CBR), SR141716, was responsible for almost full neuroprotection in CBR-expressing cells even when a selective agonist, arachidonyl-2’-chloroethylamide (ACEA), was present. However, in cells expressing CB/GPR55 heteromeric complex, SR141716 was not found to be effective. In addition, an agonist of GPR55, CID1792197, did not enhance neuroprotection in GPR55-expressing cells [47].

In an exploratory, double- blind trial of CBD in patients with PD, the highest dose tested (300 mg daily) improved quality of life [48]. In another pilot study, both THC and nabilone, a synthetic analogue of THC, reduced levodopa-induced dyskinesia in PD [49]. A recent study found medical marijuana to be effective in alleviating motor symptoms and non-motor symptoms mainly reducing sleep disturbances and pain [201].

Alzheimer’s Disease (AD): Severity of agitation may be associated with oxidative stress and neuroinflammation, while nabilone may have anti-inflammatory effects [51]. Heteromer-specific feature suggests that adenosine receptor antagonists would potentiate, via microglia, the neuroprotective action of endocannabinoids with implications for AD therapy [52].

Montanari, et al. in their study evaluated the role of unique 2-arylbenzofuran derivatives towards cannabinoid receptors and cholinesterases. Both of them had neuroprotective and immunomodulatory properties in treatment of AD [53].

Novel insight into the network mechanisms underlying cognitive decline in AD and suggest TRPV1 activation as a novel therapeutic target [54]. Link between the endocannabinoid system and interleukin-1β in the context of AD is also reported [55]. Night-time agitation, eating behavior and aggressiveness, have yielded positive results [56].

Takkinen and colleagues used inverse agonist for CB1R, [18 F] FMPEP-d 2 and PET (Positron Emission Tomography) imaging in a mouse model of AD. They showed genotype-and age-dependent impairments in the availability of CB1R [57]. Beneficial effect of CB2-deficiency in APP transgenic mice. CB2 appears to be part of a protective system that may be detrimental when engaged continuously [58]. CB1R desensitization may be a plausible strategy to improve behavior alterations associated with genetic risk factors for developing AD [59].

THC and nabilone have been tested in controlled clinical trials for the treatment of anxiety, agitation, and depression in AD individuals. THC was found to be ineffective in controlled clinical trials against neuropsychiatric symptoms. However, there was some beneficial effects on gait and balance. Nabilone reduced the severity of agitation [60-63].

Huntington’s Disease (HD): Studies have found reduced expression of CB1R in the striatum in HD patients and animal models [202,203]. Endocanabinoid signaling have a significant role in maintaining plasticity within striatum and at the corticostriatal synapse, thereby controlling goal-oriented behaviour [64,204]. Positive allosteric modulation of the CB1R reduces signs and symptoms of HD in the R6/2 mouse model [65].

A recent study involving a Q175 mouse model of HD reported rectification of motivational and dopaminergic deficits once degradation of endocannabinoid is inhibited. Neuropsychiatric symptoms of HD may benefit from CBR based therapies [66].

Recently, a research focused on several methods to quantify cell signaling and GPCR receptor ligand bias characterization of drugs that target the endocannabinoid receptors in HD [67]. Sepers, et al. in their study with a mouse model of HD have showed anandamide exhibiting a selective deficit in synaptic plasticity over 2-AG [68].

In humans with HD, FAAH activity is decreased and, consequently, anandamide levels are increased in lymphocytes [69]. A trial with 26 patients with HD, nabiximols was well tolerated but did not improve disease outcome over time [70], although in a subsequent study of seven patients with early- onset HD, it reduced dystonia [71].

CBD has been tested in 15 patients with HD, but no therapeutic effect was seen, even with a high dose (700 mg daily) [72]. Nabilone has also been tested for the treatment of motor symptoms in patients with HD, with contrasting results [73-76].

Traumatic Brain Injury (TBI): Mild TBI (mTBI) induces a strain-specific CB1-dependent bone anabolic response in the skull, probably mediated by anandamide, but seemingly unrelated to inflammation [77]. Changes in the CNS lipidome associated with mTBI likely play a role in headache and in long-term neurodegenerative effects of repeated mTBI [78]. Boosting endocannabinoid tone post-TBI may represent a viable therapeutic strategy for TBI-related psychiatric comorbidities such as alcohol use disorder and anxiety [79].

AM281, a CB1 receptor antagonist, reversed the TBI-reduced N-Methyl-D-aspartate (NMDA) receptor subunits NR2B in the hippocampus and ameliorate the spatial learning and memory impairment at 7d post-TBI, suggesting CB1 receptor is involved in the TBI-induced hippocampal-dependent spatial learning and memory impairment [80].

Post head injury in animal models, partial recovery of corticospinal tract, inhibition of tumor necrosis factor-α production in vivo, increased synaptic plasticity and neuro behavioral recovery was seen in CB2 agonists [81].

In ischemic brain insult post-TBI, cannabinoid receptor signaling partially mediates protective effects of the MAGL inhibitor. TBI can cause neuroinflammation and cerebral edema caused by BBB dysfunction. MAGL inhibitors can serve as potential therapeutics [82]. Neuroinflammation after TBI can be reduced by selective activation of CB2R via alternative macrophage polarization [83].

In a mouse model of TBI, levels of 2-AG were increased between 1 h and 24 h after injury in the same ipsilateral hemisphere. In the same model, administration of 2-AG protected the BBB, reduced inflammation and oedema and improved clinical recovery via CB1-mediated mechanisms [84,85].

Inhibition of endocannabinoid degradation by blocking FAAH, MAGL or α/β-hydrolase domain-containing 6 (ABHD6) reduced neurodegeneration and inflammation, protected BBB integrity and improved motor impairments, memory deficits and anxiety behavior in different TBI models [86,87].

In mouse models, PEA and N-arachidonoyl-l-serine had beneficial effects in TBI. Reduction of gliosis, cerebral edema, and behavioral deficits was seen with enhanced neurogenesis [88,89].

In several studies in experimental TBI, dexanabinol (also known as HU-211) - an enantiomer of the ultra- potent synthetic CB1 and CB2 ligand HU-210 that is inactive at cannabinoid receptors - exhibited potent neuroprotective activity, probably by inhibiting the NMDA receptor but was never bought to clinical practice because results were negative in phase III trial [90,91].

Seizure and epilepsy: Increase in anandamide levels in brain reduces hyperexcitability. Inhibitors of anandamide hydrolysis may help in reducing convulsions [92]. Cannabidiol has shown and anti-inflammatory and anti-epileptic properties, and it shows promise for epilepsy treatment [93]. Reduced CB2R activity is associated with increased seizure susceptibility [94]. Various models of temporal lobe epilepsy, an agonist of CB1 and CB2 had antiepileptogenic effects [95-97]. CB1 and CB2 blockade had pro- epileptogenic effects [98,99].

Anandamide and 2-AG are released after neuronal hyperexcitability to counteract glutamate excitotoxicity during seizures [100]. FAAH inhibitors protected against seizures induced by pentylenetetrazole or kainic acid [101,102]. CBD had anti- convulsant effects in the pilocarpine model of temporal lobe epilepsy via GPR55 antagonism and TRPV1 desensitization [5,103-104]. It also rescued morphological anomalies in interneurons induced by epilepsy [105].

Two double- blind, placebo- controlled phase III trials have shown CBD to be effective in controlling seizures in Lennox-Gastaut syndrome and Dravet syndrome [19,106]. CBD is also used in rare epileptic disorders including Doose syndrome, Cyclin Dependent Kinase Like 5 (CDKL5) deficiency disorder, Dup15q syndrome, Aicardi syndrome, and treatment- resistant pediatric epilepsies [107,108].

Tourette’s Syndrome (TS): THC has proved effective in Tourette syndrome in multiple studies [109-112]. ABX-1431 is an experimental drug that was well-tolerated and found to be safe in phase 1 clinical studies. Data indicates that it has the potential to treat symptoms of adult patients with Tourette’s [113]. Improvement in motor and vocal tics was seen with intake of Sativex in two patients [193]. Very limited evidence for THC’s role in symptomatic management of TS is only known from some small clinical trials [194].

A recently conducted double blind randomized clinical trial with 96 adult patients showed treatment with the cannabis extract nabiximols is superior to placebo in patients with chronic tic disorders (TS or chronic motor tic disorder) [195].

A recent polish study has shown C allele of rs2023239 polymorphism of the CNR1 gene is a risk factor of TS in Polish population. This unique variant can be possibly associated with abnormal endocannabinoid transmission, a known cause of TS [196]. Cannabis-based medicine might be safe in tics and adults with TS. THC-rich cannabis, dronabinol and nabiximols all can be tried according to preference. Entourage effect might be seen with nabiximols and dronabinol [197].

Stroke and neonatal ischemia: Stimulation with PEA or 2-AG did not change the expression of PPARα but distribution was altered. Co-application of PEA and 2-AG did not provide neuroprotection rather abolished activity was seen. Both exerted opposite effects on function and morphology of microglial cells. This might be because of contrary polarization of microglial cells [114].

Cannabidiol is a publicly available cannabinoid. It does not bind to CB1R and CB2R but neuroprotective properties are observed in stroke [115]. Post ischemic stroke, attenuation of astrocytic scar formation and improvement of motor function can be seen with oleoylethanolamide (OEA). OEA inhibits glial activation via moudulating PPARα [116].

OEA-SPC NPs is a novel neuroprotective nanoformation was found to have neuroprotective effect in vivo. Reduction in infarct volume and cerebral edema was also observed. OEA-SPC NPs was also shown to inhibit the inflammation of reperfusion to a very slight level [117].

In the early pro-inflammatory phase after CNS injury, activation of CB2R can be protective. This activation inhibits neuro-inflammation and, thereby attenuates severity of CNS injury-induced immunodepression (CIDS). However, in later phases inhibition of CB2R can restore immune function, which may be a promising pharmacological strategy [118].

WIN 55212-2 is a potent cannabinoid receptor agonist and a potent analgesic. In permanent and transient middle cerebral artery occlusion models of anoxic stroke, reduced brain tissues damage was observed. Maturation of oligodendroglia and increased oligodendrocyte progenitor cells (OPC) proliferation was seen with WIN 55212-2 [119].

CBD was reported to reduce infarct size in a permanent unilateral middle cerebral artery occlusion. CBD has antagonist like activity towards sigma 1 receptor thereby reducing the negative effects N-methyl-D-aspartate receptor (NMDAR) over activity [120].

Several preclinical models of neonatal hypoxia and ischemic damage improved with CBD derived molecules [121]. Neonatal anoxia-ischemia was shown to be associated with decreased locomotory and impairment of spatial memory. PEA decreased astrogliosis and neuroinflammation [122].

In acute stroke activation of CB1R decreases cerebral edema, reduces infarcted tissue volume, and maintains BBB integrity. Activation of CB1R induces hypothermia like state which is neuroprotective, effects that are all usually reversed by CB1 antagonists [123-125]. Stroke severity was increased in CB1 knockout mice [126].

One study has suggested that CB1 antagonists could be protective in transient or permanent cerebral artery occlusion [127]. In mice with middle cerebral artery occlusion, CB2 activation reduced infarct volume and improved neurological outcome and cerebral microcirculatory function [128,129].

After hypoxia induced ischemia in newborn pigs, CBD reduced cerebral edema and convulsive seizures [130], and brain damage was reversed after 72 h from treatment [131], CBD also has a neuroprotective effect that is partly mediated by adenosine 2A receptors [132].

Nabiximols is currently being tested as an add- on therapy for post- stroke spasticity [133]. Previously, PEA with luteolin was tested in patients with stroke during rehabilitation. It improved pain, spasticity, cognitive dysfunction, and bought independence in daily living activities [134].

Amyotrophic Lateral Sclerosis (ALS): CBRs are elevated in the motor cortex of motor neuron disease (MND) patients associated with the reactive gliosis. This phenomenon is previous to neuronal losses. CB receptors in cortical and spinal motor neurons. These observations support that targeting this receptor may serve for developing neuroprotective therapies in MNDs [135].

Elevating 2-AG levels by MAGL inhibition is a therapeutic target in ALS and demonstrate that the endocannabinoid defense mechanisms [136]. Marked upregulation of CB receptors in the spinal cord in canine degenerative myelopathy (DM), which is concentrated in activated astrocytes was observed [137]. CB1 unlikely to be beneficial as in superoxide dismutase 1 (SOD1) model as it was downregulated [138].

CB2 was upregulated in the spinal cord of SOD1 mice and in activated microglia in the spine of TAR- DNA binding protein 43 (TDP43) mutant mice [139,140]. CB2 is known to be upregulated in post- mortem spinal cord and primary motor cortex samples in patients with ALS. Furthermore, a selective CB2 agonist slowed disease progression in SOD1 mice, and these findings together suggest that CB2 has a protective role in ALS [139,141].

In SOD1 mice, anandamide and 2-AG concentrations are increased in the lumbar spinal cord [142]. Genetic knockout of FAAH in SOD1 mice prevented development of symptoms without prolonging survival [143]. Administration of an MAGL inhibitor delayed disease onset, slowed progression and increased survival [144], suggesting that the increases in anandamide and, in particular, 2-AG are neuroprotective.

In human gingiva- derived mesenchymal stromal cells, CBD modulated expression of genes associated with ALS [145] and nabiximols- like combinations of THC and CBD slightly delayed disease progression in SOD1 mice [146] In patients with ALS, PEA slowed reductions in forced vital capacity over time-suggesting that it can improve pulmonary function in this disease and improved the clinical condition of one patient [147,148].

Multiple Sclerosis (MS): TRPV1 regulates cytokine release by activated microglial cells which in turn influences central inflammation in MS [149]. For MS treatment, TRPV1 could be a prime target. Treatment with 2-AG increases the clearance of myelin debris by microglia and OPC differentiation. This results in thickening of the myelin sheath and complete remyelination [150].

In Chronic relapsing experimental allergic encephalomyelitis (CREAE) mice model, CB1 agonists ameliorated tremor and spasticity, whereas antagonists worsened them [151,152]. In CREAE mice, CBD potentiated the anti- spasticity effects of THC [153]. Nabiximols is approved for treatment-resistant spasticity in patients with MS and neuropathic pain that comes along with it in several countries. Clinical practice has confirmed that nabiximols is useful for MS spasticity [154] as an add- on therapy with other anti- spastic agents [155].

Several neurophysiological studies have showed that nabiximols to have a beneficial effect on spinal and cortical excitability. It also has a metaplastic effect on the motor cortex but upper motor neurons are spared. This is the main reason behind its analgesic property, improving sensory responses and evoked potentials [156-159]. However, in an Italian population of patients with MS, around 40% of patients were resistant to the anti- spastic action of the drug [160].

Nabiximols has immunomodulatory effects in MS, raising the possibility that it could be used to alter disease progression [161]. Ultra-micronized PEA has also been tested in patients with MS. The treatment reduced circulating levels of pro-inflammatory cytokines and reduced the adverse effects of interferon-β1a treatment for relapsing–remitting MS [162].

Glioblastoma Multiforme (GBM): CB1 and CB2 agonists decreased the size of tumor and increased survival by reducing angiogenesis in xenograft models with human glioma cells [163-166]. CB2 activation induced differentiation and inhibited gliomagenesis of glioma derived stem- like cells, which express all elements of the endocannabinoid system [167].

Anandamide suppressed proliferation, adhesion, migration and invasion of temozolomide- resistant human U251 gliomablastoma cells [168]. The protective role of TRPV1, the 5′-untranslated regions of human TRPV1 generate a stable transcript that encodes TRPV1v3, a variant of the channel that is very highly expressed in human glioblastoma tissue and stem- like cells and is associated with longer survival of patients [169].

CBD inhibits glioma cell proliferation and migration in vitro; these effects are independent of CB1 but at least partly mediated by CB2 [170]. THC had concentration- dependent effects on xenografts of temozolomide- resistant human glioblastoma T98G cells in mice -low doses stimulated proliferation and high doses inhibited proliferation [171]. CBD was shown to potentiate the anti-proliferative effect of THC, and administration of THC, CBD and temozolomide or radiation greatly increased glioma cell death [172,173].

Finally, CBD increased uptake of chemotherapeutic drugs and caused cytotoxicity in human glioma cells by activating TRPV2 [174], and promoted differentiation while reducing proliferation of glioma-derived stem-like cells by upregulating acute myeloid leukemia 1, a driver of tumor initiation that promotes TRPV2 expression [175].

Sugimoto, et al. investigated the effects of corticosterone on the endocannabinoid system in malignant glioblastoma cells in vitro. They found corticosterone inhibited the CBR agonist-induced decrease in cell viability by downregulating the mRNA and protein expressions of CB1 in glioblastoma cells and thereby exhibit anti-tumor properties [176]

Phytanoyl-CoA 2-hydroxylase-interacting protein-like gene PHYHIPL may be a target gene for the treatment and prognosis of GBM, good prognosis accompanied by upregulated PHYHIPL may be the result of retrograde endocannabinoid signaling and the cAMP signaling pathway [177].

Spinal cord injury (SCI): Synthetic cannabinoid WIN55212-2 improves the functional recovery after SCI via inhibition of glyceraldehyde 3-phosphate dehydrogenase (GAPDH/Siah1) cascades in a CB2 receptor dependent manner, indicative of its therapeutic potential for traumatic SCI [178]. A recent systematic review concluded that cannabinoid can be beneficial in reducing pain and spasticity in individuals with SCI, but intricate details like clinical significance and effect magnitude are unclear [186].

A recent animal study by Na, et al. found out CB2R agonist JWH-133 induced exogeneous activation of CB2R improved neurological deficit and blood spinal cord barrier disruption after SCI via inhibition of matrix metallopeptidase 9/toll-like receptors 4 (MMP9/TLR4) expression [187]. A descriptive qualitative study cannabis reduces pain after SCI and increased activities of daily living without having drowsiness, seen with opioids [188].

Post SCI, initiation of nuclear transportation of Siah1 and GADPH happens along with complex formation. This activation complex was inhibited by WIN. Treatment with WIN55212-2 improved survival of neurons in the spinal cord, decreased inflammation and apoptosis thereby improving neurological scores and outcome [189]. In a mice model, CBD was found to decrease heat sensitivity following SCI and this might protect against pathological invasion of T-cell [190].

Na and colleagues showed that activation and upregulation of CB1 and CB2 by remote ischemic preconditioning has protective effect on ischemic-reperfusion injury of spinal cord and maintaining integrity of blood spinal cord barrier [191]. A spanish study by Castellote, et al. showed improvement of spasticity in chronic SCI individuals by Sativex, but side-effects have to borne in mind while using it [192].

Neuropathic pain: CB1 has an important analgesic property which can be an alternative to opioids in treating chronic neuropathic pain [179]. Sharon et al. has reported cannabinoids for otherwise unresponsive pain but care should be taken in frail clinical populations [180]. CB2-specifc compounds, peripherally restricted CB1 compounds, and phytocannabinoids such as CBD are also efficacious in various preclinical models, and may avoid the psychoactive effect associated with centrally acting CB1 receptor agonists, such as ∆9-THC in treatment of chemotherapeutic agent-induced neuropathic pain [181].

A recent paper reported CBD and CBD+THC combination exhibits a predominant anti-inflammatory effect in vivo, however, THC alone could not reduce pro-inflammatory or increase anti-inflammatory cytokines [182].

A systemically administered AM404, endocannabinoid reuptake inhibitor, fully activated endocannabinoid system and showed antinociceptive effects. The authors also found out anti-hyperalgesic effects of AM404 was mainly via CB1 and not CB2 and could be promising in treatment of neuropathic pain [183].

A recent literature also reported about the role of GPR55 in the periaqueductal gray of the brainstem responsible in mitigating neuropathic pain. They demonstrated the role of GPR55 in the descending pain control system [184]. On contrary, Canavan, et al. found ineffectiveness of cannabinoids in neuropathic pain [185].

In this article, we have discussed in details about the endocannabinoidome and its role in neurological disorders. Evidence suggests this has excellent potential in treating diseases of CNS. This potential depends on their ability to modulate an endogenous signaling network made of lipid signals i.e, endocannabinoid, their receptor targets, metabolic enzymes, and intracellular/transmembrane/extracellular transporters. Most studies of endocannabinoidome targeting have been preclinical and more on animal models. More double blind and placebo controlled human studies are required in future for developing new neurotherapeutic agents.

Disclosure

The author has no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

Thanks to my mentor Prof. Hui Fang Shang, for constant support and Dr. Swati, for PubMed literature screening.

  1. Alexander SP. Therapeutic potential of cannabis related drugs. Prog. Neuropsychopharmacol. Biol Psychiatry. 2016; 64: 157–166. PubMed: https://pubmed.ncbi.nlm.nih.gov/26216862/
  2. Plasse TF. Clinical use of dronabinol. J Clin Oncol. 2016; 9: 2079–2080. PubMed: https://pubmed.ncbi.nlm.nih.gov/1658243/b
  3. Novotna A, Mares J, Ratcliffe S, Novakova I, Vachova M, 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. PubMed: https://pubmed.ncbi.nlm.nih.gov/21362108/
  4. Keating GM. Delta-9-tetrahydrocannabinol/ cannabidiol oromucosal spray (Sativex®): a review in multiple sclerosis- related spasticity. Drugs. 2017; 77: 563–574. PubMed: https://pubmed.ncbi.nlm.nih.gov/28293911/
  5. Mechoulam R, Shvo Y. The structure of cannabidiol. Tetrahedron. 1963; 19: 2073–2078. PubMed: https://pubmed.ncbi.nlm.nih.gov/5879214/
  6. Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI, et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990; 346: 561–564. PubMed: https://pubmed.ncbi.nlm.nih.gov/2165569/
  7. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993; 365: 61–65. PubMed: https://pubmed.ncbi.nlm.nih.gov/7689702/
  8. Maccarrone M. Missing Pieces to the Endocannabinoid Puzzle. Trends Mol Med. 2020; 26: 263-272. PubMed: https://pubmed.ncbi.nlm.nih.gov/31822395/
  9. Mazzola C, Micale V, Drago F. Amnesia induced by beta- amyloid fragments is counteracted by cannabinoid CB1 receptor blockade. Eur J Pharmacol. 2003; 47: 219–225. PubMed: https://pubmed.ncbi.nlm.nih.gov/14522360/
  10. Cerri S, Levandis G, Ambrosi G, Montepeloso E, Antoninetti GF, et al. Neuroprotective potential of adenosine A2A and cannabinoid CB1 receptor antagonists in an animal model of Parkinson disease. J Neuropathol Exp Neurol. 2014; 73: 414–424. PubMed: https://pubmed.ncbi.nlm.nih.gov/24709676/
  11. Lunn CA. Updating the chemistry and biology of cannabinoid CB2 receptor-specific inverse agonists. Curr Top Med Chem. 2010; 10: 768-778. PubMed: https://pubmed.ncbi.nlm.nih.gov/20370714/
  12. Lunn CA, Reich EP, Fine JS, Lavey B, Kozlowski JA, et al. Biology and therapeutic potential of cannabinoid CB2receptor inverse agonists. Br J Pharmacol. 2008; 153: 226–239. PubMed: https://pubmed.ncbi.nlm.nih.gov/17906679/
  13. Nguyen T, Li JX, Thomas BF, Wiley JL, Kenakin TP, et al. Allosteric Modulation: An Alternate Approach Targeting the Cannabinoid CB1 Receptor. Med Res Rev. 2016; 37L 441–474. PubMed: https://pubmed.ncbi.nlm.nih.gov/27879006/
  14. Di Marzo V. New approaches and challenges to targeting the endocannabinoid system. Nature Rev Drug Discov. 2018; 17: 623-639. PubMed: https://pubmed.ncbi.nlm.nih.gov/30116049/
  15. Sergio O, Scipioni L, Maccarrone M. Endocannabinoid system and adult neurogenesis: a focused review. Curr Opinion Pharmacol. 2020; 50: 25-32.
  16. Arturo IF, Fabiana P. Endocannabinoidome. 2018.
  17. Turner SE, Williams CM, Iversen L, Whalley BJ. Molecular pharmacology of phytocannabinoids. Prog Chem Org Nat Prod. 2017; 103: 61–101. PubMed: https://pubmed.ncbi.nlm.nih.gov/28120231/
  18. Devinsky O, Marsh E, Friedman D, Thiele E, Laux L, et al. Cannabidiol in patients with treatment- resistant epilepsy: an open- label interventional trial. Lancet Neurol. 2016; 15: 270–278. PubMed: https://pubmed.ncbi.nlm.nih.gov/26724101/
  19. Devinsky O, Cross JH, Laux L, Marsh E, Miller I,  et al. Trial of cannabidiol for drug resistant seizures in the Dravet syndrome. N Engl J Med. 2017; 376: 2011–2020. PubMed: https://pubmed.ncbi.nlm.nih.gov/28538134/
  20. Benito C, Tolón RM, Castillo AI, Ruiz-Valdepeñas L, Martínez-Orgado JA, et al. beta- Amyloid exacerbates inflammation in astrocytes lacking fatty acid amide hydrolase through a mechanism involving PPAR- alpha, PPAR- gamma and TRPV1, but not CB(1) or CB(2) receptors. Br J Pharmacol. 2012; 166: 1474–1489. PubMed: https://pubmed.ncbi.nlm.nih.gov/22321194/
  21. Luchicchi A, Lecca S, Carta S, Pillolla G, Muntoni AL,  et al. Effects of fatty acid amide hydrolase inhibition on neuronal responses to nicotine, cocaine and morphine in the nucleus accumbens shell and ventral tegmental area: involvement of PPAR- alpha nuclear receptors. Addict Biol. 2010; 15: 277–288. PubMed: https://pubmed.ncbi.nlm.nih.gov/20477753/
  22. Hansen HS, Rosenkilde MM, Holst JJ, Schwartz TW, et al. GPR119 as a fat sensor. Trends Pharmacol Sci. 2012; 33: 374–381. PubMed: https://pubmed.ncbi.nlm.nih.gov/22560300/
  23. Kawahara H, Drew GM, Christie MJ, Vaughan CW. Inhibition of fatty acid amide hydrolase unmasks CB1 receptor and TRPV1 channel mediated modulation of glutamatergic synaptic transmission in midbrain periaqueductal grey. Br J Pharmacol. 2011; 163: 1214–1222. PubMed: https://pubmed.ncbi.nlm.nih.gov/21175570/
  24. Nomura DK, Lombardi DP, Chang JW, Niessen S, Ward AM, et al. Monoacylglycerol lipase exerts dual control over endocannabinoid and fatty acid pathways to support prostate cancer. Chem Biol. 2011; 18: 846–856. PubMed: https://pubmed.ncbi.nlm.nih.gov/21802006/
  25. Piro JR, Benjamin DI, Duerr JM, Pi Y, Gonzales C,  et al. A dysregulated endocannabinoid eicosanoid network supports pathogenesis in a mouse model of Alzheimer’s disease. Cell Rep. 2012; 1: 617–623. PubMed: https://pubmed.ncbi.nlm.nih.gov/22813736/
  26. Iannotti FA, Di Marzo V, Petrosino S. Endocannabinoids and endocannabinoid-related mediators: Targets, metabolism and role in neurological disorders. Progr Lipid Res. 2016; 62: 107–128. PubMed: https://pubmed.ncbi.nlm.nih.gov/26965148/
  27. Katona I, Freund TF. Endocannabinoid signaling as a synaptic circuit breaker in neurological disease. Nat Med. 2008; 14: 923–930. PubMed: https://pubmed.ncbi.nlm.nih.gov/18776886/
  28. Matyas F, Urbán GM, Watanabe M, Mackie K, Zimmer A,  et al. Identification of the sites of 2-arachidonoylglycerol synthesis and action imply retrograde endocannabinoid signaling at both GABAergic and glutamatergic synapses in the ventral tegmental area. Neuropharmacology. 2008; 54: 95–107. PubMed: https://pubmed.ncbi.nlm.nih.gov/17655884/
  29. Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001; 410: 588–592. PubMed: https://pubmed.ncbi.nlm.nih.gov/11279497/
  30. Marinelli S, Pacioni S, Bisogno T, Di Marzo V, Prince DA, et al. The endocannabinoid 2-arachidonoylglycerol is responsible for the slow self- inhibition in neocortical interneurons. J Neurosci. 2008; 28: 13532–13541. PubMed: https://pubmed.ncbi.nlm.nih.gov/19074027/
  31. Benard G, Massa F, Puente N, Lourenço J, Bellocchio L,  et al. Mitochondrial CB(1) receptors regulate neuronal energy metabolism. Nat Neurosci. 2012; 15: 558–564. PubMed: https://pubmed.ncbi.nlm.nih.gov/22388959/
  32. Hebert-Chatelain E, Desprez T, Serrat R, Bellocchio L, Soria-Gomez E, et al. A cannabinoid link between mitochondria and memory. Nature. 2016; 539: 555–559. PubMed: https://pubmed.ncbi.nlm.nih.gov/27828947/
  33. Prenderville JA, Kelly ÁM, Downer EJ. The role of cannabinoids in adult neurogenesis. Br J Pharmacol. 2015; 172: 3950–3963. PubMed: https://pubmed.ncbi.nlm.nih.gov/25951750/
  34. Aymerich MS, Aso E, Abellanas MA, Tolon RM, Ramos JA, et al. Cannabinoid pharmacology/ therapeutics in chronic degenerative disorders affecting the central nervous system. Biochem Pharmacol. 2018; 157: 67–84. PubMed: https://pubmed.ncbi.nlm.nih.gov/30121249/
  35. Palazuelos J, Ortega Z, Díaz-Alonso J, Guzmán M, Galve-Roperh I. CB2 cannabinoid receptors promote neural progenitor cell proliferation via mTORC1 signaling. J Biol Chem. 2012; 287: 1198–1209. PubMed: https://pubmed.ncbi.nlm.nih.gov/22102284/
  36. Chung YC, Shin WO, Baek JY, Cho EJ, Baik HH, et al. CB2 receptor activation prevents glial- derived neurotoxic mediator production, BBB leakage and peripheral immune cell infiltration and rescues dopamine neurons in the MPTP model of Parkinson’s disease. Exp Mol Med. 2016; 48: e205. PubMed: https://pubmed.ncbi.nlm.nih.gov/27534533/
  37. Marchalant Y, Brownjohn PW, Bonnet A, Kleffmann T, Ashton JC. Validating antibodies to the cannabinoid CB2 receptor: antibody sensitivity is not evidence of antibody specificity. J Histochem Cytochem. 2014; 62: 395–404. PubMed: https://pubmed.ncbi.nlm.nih.gov/24670796/
  38. Soethoudt M, Grether U, Fingerle J, Grim TW, Fezza F, et al. Cannabinoid CB2 receptor ligand profiling reveals biased signalling and off- target activity. Nat Commun. 2017; 8: 13958. PubMed: https://pubmed.ncbi.nlm.nih.gov/28045021/
  39. Stempel AV, Stumpf A, Zhang HY, Özdoğan T, Pannasch U, et al. Cannabinoid type 2 receptors mediate a cell type- specific plasticity in the hippocampus. Neuron. 2016; 90: 795–809. PubMed: https://pubmed.ncbi.nlm.nih.gov/27133464/
  40. Cristino L, de Petrocellis L, Pryce G, Baker D, Guglielmotti V, et al. Immunohistochemical localization of cannabinoid type 1 and vanilloid transient receptor potential vanilloid type 1 receptors in the mouse brain. Neuroscience. 2006; 139: 1405–1415. PubMed: https://pubmed.ncbi.nlm.nih.gov/16603318/
  41. Cristino L, Starowicz K, De Petrocellis L, Morishita J, Ueda N,  et al. Immunohistochemical localization of anabolic and catabolic enzymes for anandamide and other putative endovanilloids in the hippocampus and cerebellar cortex of the mouse brain. Neuroscience. 2008; 151: 955–968. PubMed: https://pubmed.ncbi.nlm.nih.gov/18248904/
  42. Villapol S. Roles of peroxisome proliferator activated receptor gamma on brain and peripheral inflammation. Cell Mol. Neurobiol. 2018; 38: 121–132. PubMed: https://pubmed.ncbi.nlm.nih.gov/28975471/
  43. Sylantyev S, Jensen TP, Ross RA, Rusakov DA. Cannabinoid- and lysophosphatidyl inositol- sensitive receptor GPR55 boosts neurotransmitter release at central synapses. Proc Natl Acad Sci. USA. 2013; 110: 5193–5198. PubMed: https://pubmed.ncbi.nlm.nih.gov/23472002/
  44. McHugh D, Wager-Miller J, Page J, Bradshaw HB. siRNA knockdown of GPR18 receptors in BV-2 microglia attenuates N- arachidonoyl glycine- induced cell migration. J Mol Signal. 2012; 7: 10. PubMed: https://pubmed.ncbi.nlm.nih.gov/22834922/
  45. Crivelaro do Nascimento G, Ferrari DP, Guimaraes FS, Del Bel EA, Bortolanza M, et al. Cannabidiol increases the nociceptive threshold in a preclinical model of Parkinson’s disease. Neuropharmacology. 2019; 163: 107808. PubMed: https://pubmed.ncbi.nlm.nih.gov/31706993/
  46. Davis MP, Behm B, Mehta Z, Fernandez C. The Potential Benefits of Palmitoylethanolamide in Palliation: A Qualitative Systematic Review. Am J Hosp Palliat Med. 2019; 36: 1134-1154. PubMed: https://pubmed.ncbi.nlm.nih.gov/31113223/
  47. Martínez-Pinilla E, Aguinaga D, Navarro G, Rico AJ, Oyarzábal J, et al. Targeting CB1 and GPR55 Endocannabinoid Receptors as a Potential Neuroprotective Approach for Parkinson’s Disease. Mole Neurobiol. 2019; 56: 5900-5910. PubMed: https://pubmed.ncbi.nlm.nih.gov/30687889/
  48. Chagas MH, Zuardi AW, Tumas V, Pena-Pereira MA, Sobreira ET, et al. Effects of cannabidiol in the treatment of patients with Parkinson’s disease: an exploratory double- blind trial. J Psychopharmacol. 2014; 28: 1088–1098. PubMed: https://pubmed.ncbi.nlm.nih.gov/25237116/
  49. Sieradzan KA, Fox SH, Hill M, Dick JP, Crossman AR, et al. Cannabinoids reduce levodopa induced dyskinesia in Parkinson’s disease: a pilot study. Neurology. 2001; 57: 2108–2111. PubMed: https://pubmed.ncbi.nlm.nih.gov/11739835/
  50. Brotini S, Schievano C, Guidi L. Ultramicronized palmitoylethanolamide: an efficacious adjuvant therapy for Parkinson’s disease. CNS Neurol Disord Drug Targets. 2017; 16: 705–713. PubMed: https://pubmed.ncbi.nlm.nih.gov/28325153/
  51. Ruthirakuhan M, Herrmann N, Andreazza AC, Verhoeff NP, Gallagher D, et al. Agitation, Oxidative Stress, and Cytokines in Alzheimer Disease: Biomarker Analyses From a Clinical Trial With Nabilone for Agitation. J Geriatric Psychiatry Neurol. 2019; 2020; 33: 175-184. PubMed: https://pubmed.ncbi.nlm.nih.gov/31547752/
  52. Franco R, Reyes‐Resina I, Aguinaga D, Lillo A, Jiménez J, Raïch I, et al. Potentiation of cannabinoid signaling in microglia by adenosine A 2A receptor antagonists. Glia. 2019; 67: 2410-2423.  PubMed: https://pubmed.ncbi.nlm.nih.gov/31429130/
  53. Montanari S, Mahmoud AM, Pruccoli L, Rabbito A, Naldi M, et al. Discovery of novel benzofuran-based compounds with neuroprotective and immunomodulatory properties for Alzheimer’s disease treatment. Eur J Med Chem. 2019. PubMed: https://pubmed.ncbi.nlm.nih.gov/31185414/
  54. Balleza-Tapia H, Crux S, Andrade-Talavera Y, Dolz-Gaiton P, Papadia D, et al. TrpV1 receptor activation rescues neuronal function and network gamma oscillations from Aβ-induced impairment in mouse hippocampus in vitro. Elife, 2018; 7: e37703. PubMed: https://pubmed.ncbi.nlm.nih.gov/30417826/
  55. Aparicio N, Grande MT, Ruiz de Martín Esteban S, López A, Ruiz-Pérez G, et al. Role of Interleukin 1-Beta In The Inflammatory Response In A Fatty Acid Amide Hydrolase-Knockout Mouse Model of Alzheimer’s Disease. Biochem Pharmacol. 2018; 157: 202-209. PubMed: https://pubmed.ncbi.nlm.nih.gov/30195729/
  56. Talarico G, Trebbastoni A, Bruno G, de Lena C. Modulation of the Cannabinoid System: A New Perspective for the Treatment of the Alzheimer’s Disease. Curr Neuropharmacol. 2019; 17: 176-183. PubMed: https://pubmed.ncbi.nlm.nih.gov/29962346/
  57. Takkinen JS, López-Picón FR, Kirjavainen AK, Pihlaja R, Snellman A, et al. 18 F]FMPEP- d 2 PET imaging shows age- and genotype-dependent impairments in the availability of cannabinoid receptor 1 in a mouse model of Alzheimer’s disease. Neurobiol Aging. 2018; 69: 199–208. PubMed: https://pubmed.ncbi.nlm.nih.gov/29909177/
  58. Schmöle AC, Lundt R, Toporowski G, Hansen JN, Beins E, et al. Cannabinoid Receptor 2-Deficiency Ameliorates Disease Symptoms in a Mouse Model with Alzheimer’s Disease-Like Pathology. J Alzheimers Dis. 2018; 64: 379–392. PubMed: https://pubmed.ncbi.nlm.nih.gov/29865078/
  59. Llorente-Ovejero A, Manuel I, Lombardero L, Giralt MT, Ledent C, et al. Endocannabinoid and Muscarinic Signaling Crosstalk in the 3xTg-AD Mouse Model of Alzheimer’s Disease. J Alzheimers Dis. 2018; 64: 117–136. PubMed: https://pubmed.ncbi.nlm.nih.gov/29865071/
  60. Passmore MJ. The cannabinoid receptor agonist nabilone for the treatment of dementia- related agitation. Int J Geriatr Psychiatry. 2008; 23: 116–117. PubMed: https://pubmed.ncbi.nlm.nih.gov/18081000/
  61. van den Elsen GAH. Ahmed AIA, Verkes RJ, Kramers C, Feuth T, et al. Tetrahydrocannabinol for neuropsychiatric symptoms in dementia: a randomized controlled trial. Neurology. 2015; 84: 2338–2346. PubMed: https://pubmed.ncbi.nlm.nih.gov/25972490/
  62. van den Elsen GAH, Ahmed AIA, Verkes RJ, Feuth T, van der Marck MA. et al. Tetrahydrocannabinol in behavioral disturbances in dementia: a crossover randomized controlled trial. Am J Geriatr Psychiatry. 2015; 23: 1214–1224. PubMed: https://pubmed.ncbi.nlm.nih.gov/26560511/
  63. van den Elsen GAH, Tobben L, Ahmed AI, Verkes RJ, Kramers  C, et al. Effects of tetrahydrocannabinol on balance and gait in patients with dementia: a randomised controlled crossover trial. J Psychopharmacol. 2017; 31: 184–191. PubMed: https://pubmed.ncbi.nlm.nih.gov/27624148/
  64. Smith‐Dijak AI, Sepers MD, Raymond LA. Alterations in Synaptic Function and Plasticity in Huntington Disease. J Neurochem. 2019; 150: 346-365.  PubMed: https://pubmed.ncbi.nlm.nih.gov/31095731/
  65. Laprairie RB, Bagher AM, Rourke JL, Zrein A, Cairns EA, et al. Positive allosteric modulation of the type 1 cannabinoid receptor reduces the signs and symptoms of Huntington’s disease in the R6/2 mouse model. Neuropharmacology. 2019; 151: 1-12. PubMed: https://pubmed.ncbi.nlm.nih.gov/30940536/
  66. Covey DP, Dantrassy HM, Yohn SE, Castro A, Conn PJ, et al. Inhibition of endocannabinoid degradation rectifies motivational and dopaminergic deficits in the Q175 mouse model of Huntington’s disease. Neuropsychopharmacology. 2018; 43: 2056–2063. PubMed: https://pubmed.ncbi.nlm.nih.gov/29925886/
  67. Bagher AM, Laprairie RB, Kelly MEM, Denovan-Wright EM. Methods to Quantify Cell Signaling and GPCR Receptor Ligand Bias: Characterization of Drugs that Target the Endocannabinoid Receptors in Huntington’s Disease. Methods Mol Biol. 2018; 549–571. PubMed: https://pubmed.ncbi.nlm.nih.gov/29856035/
  68. Sepers MD, Smith-Dijak A, LeDue J, Kolodziejczyk K, Mackie K, et al. Endocannabinoid-Specific Impairment in Synaptic Plasticity in Striatum of Huntington’s Disease Mouse Model. J Neurosci. 2017; 38: 544–554. PubMed: https://pubmed.ncbi.nlm.nih.gov/29192125/
  69. Battista N, Bari M, Tarditi A, Mariotti C, Bachoud-Lévi AC, Zuccato C,  et al. Severe deficiency of the fatty acid amide hydrolase (FAAH) activity segregates with the Huntington’s disease mutation in peripheral lymphocytes. Neurobiol Dis. 2007; 27: 108–116. PubMed: https://pubmed.ncbi.nlm.nih.gov/17553686/
  70. Lopez-Sendon M, Caldentey JC, Cubillo PT, Romero CR, Ribas GG, et al. A double- blind, randomized, cross- over, placebo- controlled, pilot trialwith Sativex in Huntington’s disease. J Neurol. 2016; 263: 1390–1400. PubMed: https://pubmed.ncbi.nlm.nih.gov/27159993/
  71. Saft C, von Hein SM, Lücke T, Thiels C, Peball M, et al. Cannabinoids for treatment of dystonia in Huntington’s disease. J Huntingt Dis. 2018; 7: 167–173. PubMed: https://pubmed.ncbi.nlm.nih.gov/29562549/
  72. Consroe P, Laguna J, Allender J, Snider S, Stern L, et al. Controlled clinical trial of cannabidiol in Huntington’s disease. Pharmacol Biochem Behav. 1991; 40: 701–708. PubMed: https://pubmed.ncbi.nlm.nih.gov/1839644/
  73. Curtis A, Mitchell I, Patel S, Ives N, Rickards H. A pilot study using nabilone for symptomatic treatment in Huntington’s disease. Mov Disord. 2009; 24: 2254–2259. PubMed: https://pubmed.ncbi.nlm.nih.gov/19845035/
  74. Armstrong MJ, Miyasaki JM. Evidence-based guideline: Pharmacologic treatment of chorea in Huntington disease: Report of the Guideline Development Subcommittee of the American Academy of Neurology. Neurology. 2012; 79: 597–603. PubMed: https://pubmed.ncbi.nlm.nih.gov/22815556/
  75. Curtis A, Rickards H. Nabilone Could Treat Chorea and Irritability in Huntington’s Disease. J Neuropsychiatry Clin Neurosci. 2006; 18: 553–554. PubMed: https://pubmed.ncbi.nlm.nih.gov/17135385/
  76. Muller-Vahl KR, Schneider U, Emrich HM. Nabilone increases choreatic movements in Huntington’s disease. Mov Disord. 1999; 14: 1038–1040. PubMed: https://pubmed.ncbi.nlm.nih.gov/10584686/ 
  77. Eger M, Bader M, Bree D, Hadar R, Nemerovski A, et al. Bone Anabolic Response in the Calvaria Following Mild Traumatic Brain Injury is Mediated by the Cannabinoid-1 Receptor. Sci Rep. 2019; 9: 16196. PubMed: https://pubmed.ncbi.nlm.nih.gov/31700010/
  78. Honn KV, Zeldin DC. The Role of Bioactive Lipids in Cancer, Inflammation and Related Diseases. Adv Experimen Med Biol. 2019.
  79. Fucich EA, Mayeux JP, McGinn MA, Gilpin NW, Edwards S, et al. A Novel Role for the Endocannabinoid System in Ameliorating Motivation for Alcohol Drinking and Negative Behavioral Affect Following Traumatic Brain Injury in Rats. J Neurotrauma. 2019; : 36: 1847-1855. PubMed: https://pubmed.ncbi.nlm.nih.gov/30638118/
  80. Xu X, Jiang S, Xu E, Wu X, Zhao R. Inhibition of CB1 receptor ameliorates spatial learning and memory impairment in mice with traumatic brain injury. Neurosci Lett. 2019; 696: 127-131. PubMed: https://pubmed.ncbi.nlm.nih.gov/30576711/
  81. Magid L, Heymann S, Elgali M, Avram L, Cohen Y, et al. The Role of CB2 Receptor in the Recovery of Mice after Traumatic Brain Injury. J Neurotrauma. 2018; 36: 1836-1846. PubMed: https://pubmed.ncbi.nlm.nih.gov/30489198/
  82. Piro JR, Suidan GL, Quan J, Pi Y, O’Neill SM, et al. Inhibition of 2-AG hydrolysis differentially regulates blood brain barrier permeability after injury. J Neuroinflammation. 2018; 15.
  83. Braun M, Khan ZT, Khan MB, Kumar M, Ward A, et al. Selective activation of cannabinoid receptor-2 reduces neuroinflammation after traumatic brain injury via alternative macrophage polarization. Brain Behav Immun. 2017; 68: 224–237. PubMed: https://pubmed.ncbi.nlm.nih.gov/29079445/
  84. Panikashvili D, Simeonidou C, Ben-Shabat S, Hanus L, Breuer A, et al. An endogenous cannabinoid (2-AG) is neuroprotective after brain injury. Nature. 2001; 413: 527–531. PubMed: https://pubmed.ncbi.nlm.nih.gov/11586361/
  85. Panikashvili D, Shein NA, Mechoulam R, Trembovler V, Kohen R, et al. The endocannabinoid 2-AG protects the blood- brain barrier after closed head injury and inhibits mRNA expression of proinflammatory cytokines. Neurobiol Dis. 2006; 22: 257–264. PubMed: https://pubmed.ncbi.nlm.nih.gov/16364651/
  86. Mayeux J, Katz P, Edwards S, Middleton JW, Molina PE, et al. Inhibition of endocannabinoid degradation improves outcomes from mild traumatic brain injury: a mechanistic role for synaptic hyperexcitability. J Neurotrauma. 2017; 34: 436–443. PubMed: https://pubmed.ncbi.nlm.nih.gov/27189876/
  87. Katz PS, Sulzer JK, Impastato RA, Teng SX, Rogers EK, et al. Endocannabinoid degradation inhibition improves neurobehavioral function, bloodbrain barrier integrity, and neuroinflammation following mild traumatic brain injury. J Neurotrauma. 2015; 32: 297–306. PubMed: https://pubmed.ncbi.nlm.nih.gov/25166905/
  88. Ahmad A, Crupi R, Impellizzeri D, Campolo M, Marino A, et al. Administration of palmitoylethanolamide (PEA) protects the neurovascular unit and reduces secondary injury after Traumatic brain injury in mice. Brain Behav. Immun. 2012; 26: 1310–1321. PubMed: https://pubmed.ncbi.nlm.nih.gov/22884901/ 
  89. Cohen- Yeshurun A, Willner D, Trembovler V, Alexandrovich A, Mechoulam R, et al. N- arachidonoyl-L- serine (AraS) possesses proneurogenic properties in vitro and in vivo after traumatic brain injury. J Cereb Blood Flow Metab. 2013; 33: 1242–1250. PubMed: https://pubmed.ncbi.nlm.nih.gov/23695434/
  90. Feigenbaum JJ, Bergmann F, Richmond SA, Mechoulam R, Nadler V, et al. Nonpsychotropic cannabinoid acts as a functional N-methyl-D-aspartate receptor blocker. Proc Natl Acad Sci. USA. 1989; 86: 9584–9587. PubMed: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC298542/
  91. Maas AI, Murray G, Henney H, Kassem N, Legrand V, et al. Efficacy and safety of dexanabinol in severe traumatic brain injury: results of a phase III randomised, placebo- controlled, clinical trial. Lancet Neurol. 2006; 5: 38–45. PubMed: https://pubmed.ncbi.nlm.nih.gov/16361021/
  92. Laila A, Iglesias LP, De Oliveira AC, Moraes MFD, Moreira FA. Exploiting cannabinoid and vanilloid mechanisms for epilepsy treatment. Epilepsy Behavior. 2019; 106832. PubMed: https://pubmed.ncbi.nlm.nih.gov/31839498/
  93. Cheung KAK, Peiris H, Wallace G, Holland OJ, Mitchell MD. The Interplay between the Endocannabinoid System, Epilepsy and Cannabinoids. Int J Mole Sci. 2019; 20: 6079. PubMed: https://pubmed.ncbi.nlm.nih.gov/31810321/
  94. Shapiro L, Wong JC, Escayg A. Reduced cannabinoid 2 receptor activity increases susceptibility to induced seizures in mice. Epilepsia. 2019; 60: 2359-2369. PubMed: https://pubmed.ncbi.nlm.nih.gov/31758544/
  95. Vinogradova LV, van Rijn CM. Long- term disease- modifying effect of the endocannabinoid agonist WIN55,212-2 in a rat model of audiogenic epilepsy. Pharmacol Rep. 2015; 67: 501–503. PubMed: https://pubmed.ncbi.nlm.nih.gov/25933961/
  96. Di Maio R, Cannon JR, Greenamyre JT. Poststatus epilepticus treatment with the cannabinoid agonist WIN 55,212-2 prevents chronic epileptic hippocampal damage in rats. Neurobiol Dis. 2015; 73: 356–365. PubMed: https://pubmed.ncbi.nlm.nih.gov/25447228/ 
  97. Bhaskaran MD, Smith BN. Cannabinoid mediated inhibition of recurrent excitatory circuitry in the dentate gyrus in a mouse model of temporal lobe epilepsy. Plos One. 2010; 5: e10683. PubMed: https://pubmed.ncbi.nlm.nih.gov/20498848/
  98. Wallace MJ, Blair RE, Falenski KW, Martin BR, DeLorenzo RJ. The endogenous cannabinoid system regulates seizure frequency and duration in a model of temporal lobe epilepsy. J Pharmacol Exp Ther. 2003; 307: 129–137. PubMed: https://pubmed.ncbi.nlm.nih.gov/12954810/
  99. Vinogradova LV, Shatskova AB, van Rijn CM. Pro- epileptic effects of the cannabinoid receptor antagonist SR141716 in a model of audiogenic epilepsy. Epilepsy Res. 2011; 96: 250–256. PubMed: https://pubmed.ncbi.nlm.nih.gov/21733658/
  100. Marsicano G, Goodenough S, Monory K, Hermann H, Eder M, et al. CB1 cannabinoid receptors and on- demand defense against excitotoxicity. Science. 2003; 302: 84–88. PubMed: https://pubmed.ncbi.nlm.nih.gov/14526074/
  101. Vilela LR, Medeiros DC, Rezende GHS, de Oliveira ACP, Moraes MFD, et al. Effects of cannabinoids and endocannabinoid hydrolysis inhibition on pentylenetetrazole- induced seizure and electroencephalographic activity in rats. Epilepsy Res. 2013; 104; 195–202. PubMed: https://pubmed.ncbi.nlm.nih.gov/23352737/
  102. Shubina L, Aliev R, Kitchigina V. Attenuation of kainic acid- induced status epilepticus by inhibition of endocannabinoid transport and degradation in guinea pigs. Epilepsy Res. 2015; 111: 33–44. PubMed: https://pubmed.ncbi.nlm.nih.gov/25769371/
  103. Jones NA, Hill AJ, Smith J, Bevan SA, Williams CM, et al. Cannabidiol displays antiepileptiform and antiseizure properties in vitro and in vivo. J Pharmacol Exp Ther. 2010; 332: 569–577. PubMed: https://pubmed.ncbi.nlm.nih.gov/19906779/
  104. Jones NA, Glyn SE, Akiyama S, Hill TDM, Hill AJ, et al. Cannabidiol exerts anti- convulsant effects in animal models of temporal lobe and partial seizures. Seizure. 2012; 21: 344–352. PubMed: https://pubmed.ncbi.nlm.nih.gov/22520455/
  105. Khan AA, Shekh-Ahmad T, Khalil A, Walker MC, Ali AA, et al. Cannabidiol exerts antiepileptic effects by restoring hippocampal interneuron functions in a temporal lobe epilepsy model. Br J Pharmacol. 2018; 175: 2097–2115. PubMed: https://pubmed.ncbi.nlm.nih.gov/29574880/ 
  106. Thiele EA, Marsh ED, French JA, Mazurkiewicz-Beldzinska M, Benbadis SR, et al. Cannabidiol in patients with seizures associated with Lennox- Gastaut syndrome (GWPCARE4): a randomised, double- blind, placebo-controlled phase 3 trial. Lancet. 2018; 391: 1085–1096. PubMed: https://pubmed.ncbi.nlm.nih.gov/29395273/
  107. Devinsky O, Verducci C, Thiele EA, Laux LC, Patel AD, et al. Open- label use of highly purified CBD (Epidiolex®) in patients with CDKL5 deficiency disorder and Aicardi, Dup15q, and Doose syndromes. Epilepsy Behav. 2018; 86: 131–137. PubMed: https://pubmed.ncbi.nlm.nih.gov/30006259/
  108. Gofshteyn JS. Cannabidiol as a potential treatment for febrile infection- related epilepsy syndrome (FIRES) in the acute and chronic phases. J Child Neurol. 2017; 32: 35–40. PubMed: https://pubmed.ncbi.nlm.nih.gov/27655472/
  109. Abi-Jaoude E, Chen L, Cheung P, Bhikram T, Sandor P. Preliminary evidence on cannabis effectiveness and tolerability for adults with Tourette syndrome. J Neuropsychiatry Clin Neurosci. 2017; 29: 391–400. PubMed: https://pubmed.ncbi.nlm.nih.gov/28464701/
  110. Muller-Vahl KR, Schneider U, Kolbe H, Emrich HM. Treatment of Tourette’s syndrome with delta-9-tetrahydrocannabinol. Am J Psychiatry. 1999; 156: 495. PubMed: https://pubmed.ncbi.nlm.nih.gov/10080574/
  111. Muller-Vahl KR. Treatment of Tourette syndrome with cannabinoids. Behav Neurol. 2013; 27: 119–124. PubMed: https://pubmed.ncbi.nlm.nih.gov/23187140/
  112. Kanaan A, Jakubovski E, Müller-Vahl K. Significant Tic Reduction in An Otherwise Treatment-Resistant Patient with Gilles de la Tourette Syndrome Following Treatment with Nabiximols. Brain Sci. 2017; 7: 47. PubMed: https://pubmed.ncbi.nlm.nih.gov/28445405/
  113. Jiang M, van der Stelt M. Activity-Based Protein Profiling Delivers Selective Drug Candidate ABX-1431, a Monoacylglycerol Lipase Inhibitor, To Control Lipid Metabolism in Neurological Disorders. J Med Chem. 2018; 61: 9059-9061. PubMed: https://pubmed.ncbi.nlm.nih.gov/30354159/
  114. Hohmann U, Pelzer M, Kleine J, Hohmann T, Ghadban C, et al. Opposite Effects of Neuroprotective Cannabinoids, Palmitoylethanolamide, and 2-Arachidonoylglycerol on Function and Morphology of Microglia. Front Neurosci. 2019; 13: 1180. PubMed: https://pubmed.ncbi.nlm.nih.gov/31787870/
  115. Rajah G, Kolb B, Saber H, Fadel H. The endocannabinoid system and stroke: A focused review. Brain Circulation. 2019; 5: 1-7. PubMed: https://pubmed.ncbi.nlm.nih.gov/31001593/
  116. Luo D, Zhang Y, Yuan X, Pan Y, Yang L, et al. Oleoylethanolamide inhibits glial activation via moudulating PPARα and promotes motor function recovery after brain ischemia. Pharmacological Res. 2019; 141: 530-540. PubMed: https://pubmed.ncbi.nlm.nih.gov/30660821/
  117. Yang X, Xu L, Zhou J, Ge Y, Wu S, et al. Integration of phospholipid-complex nanocarrier assembly with endogenous N-oleoylethanolamine for efficient stroke therapy. J Nanobiotechnol. 2019; 17: 8. PubMed: https://pubmed.ncbi.nlm.nih.gov/30660200/
  118. Zhou J, Noori H, Burkovskiy I, Lafreniere J, Kelly M, et al. Modulation of the Endocannabinoid System Following Central Nervous System Injury. Int J Mole Sci. 2019; 20: 388. PubMed: https://pubmed.ncbi.nlm.nih.gov/30658442/
  119. Ilyasov AA, Milligan CE, Pharr EP, Howlett AC. The Endocannabinoid System and Oligodendrocytes in Health and Disease. Front Neurosci. 2018; 12; 733. PubMed: https://pubmed.ncbi.nlm.nih.gov/30416422/
  120. Rodríguez-Muñoz M, Onetti Y, Cortés-Montero E, Garzón J, Sánchez-Blázquez P. Cannabidiol enhances morphine antinociception, diminishes NMDA-mediated seizures and reduces stroke damage via the sigma 1 receptor. Molecular Brain. 2018; 11: 51. PubMed: https://pubmed.ncbi.nlm.nih.gov/30223868/
  121. Sagredo O, Palazuelos J, Gutierrez-Rodriguez A, Satta V, Galve-Roperh I, et al. Cannabinoid signalling in the immature brain: encephalopathies and neurodevelopmental disorders. Biochem Pharmacol. 2018; 157: 85-96. PubMed: https://pubmed.ncbi.nlm.nih.gov/30118663/
  122. Holubiec MI, Romero JI, Suárez J, Portavella M, Fernández-Espejo E, et al. Palmitoylethanolamide prevents neuroinflammation, reduces astrogliosis and preserves recognition and spatial memory following induction of neonatal anoxia-ischemia. Psychopharmacology. 2018; 235: 2929-2945. PubMed: https://pubmed.ncbi.nlm.nih.gov/30058012/
  123. Chi OZ, Barsoum S, Grayson J, Hunter C, Liu X, et al. Effects of cannabinoid receptor agonist WIN 55,212-2 on blood- brain barrier disruption in focal cerebral ischemia in rats. Pharmacology. 2012; 89: 333–338. PubMed: https://pubmed.ncbi.nlm.nih.gov/22678129/
  124. Mauler F, Hinz V, Augstein KH, Fassbender M, Horváth E. Neuroprotective and brain edema reducing efficacy of the novel cannabinoid receptor agonist BAY 38-7271. Brain Res. 2003; 989: 99–111. PubMed: https://pubmed.ncbi.nlm.nih.gov/14519516/
  125. Hayakawa K, Mishima K, Nozako M, Hazekawa M, Ogata A, et al. Delta 9-tetrahydrocannabinol (delta9-THC) prevents cerebral infarction via hypothalamic- independent hypothermia. Life Sci. 2007; 80: 1466–1471. PubMed: https://pubmed.ncbi.nlm.nih.gov/17289082/
  126. Parmentier-Batteur S, Jin K, Mao XO, Xie L, Greenberg DA. Increased severity of stroke in CB1 cannabinoid receptor knock- out mice. J Neurosci. 2002; 22: 9771–9775. PubMed: https://pubmed.ncbi.nlm.nih.gov/12427832/
  127. Muthian S, Rademacher DJ, Roelke CT, Gross GJ, Hillard CJ. Anandamide content is increased and CB1 cannabinoid receptor blockade is protective during transient, focal cerebral ischemia. Neuroscience. 2004; 129: 743–750. PubMed: https://pubmed.ncbi.nlm.nih.gov/15541895/
  128. Zarruk JG, Fernández-López D, García-Yébenes I, García-Gutiérrez MS, Vivancos J,  et al. Cannabinoid type 2 receptor activation downregulates stroke- induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke. 2012; 43: 211–219. PubMed: https://pubmed.ncbi.nlm.nih.gov/22020035/
  129. Zhang M, Adler MW, Abood ME, Ganea D, Jallo J, et al. CB2 receptor activation attenuates microcirculatory dysfunction during cerebral ischemic/ reperfusion injury. Microvasc Res. 2009; 78: 86–94. PubMed: https://pubmed.ncbi.nlm.nih.gov/19332079/
  130. Alvarez FJ, Lafuente H, Rey-Santano MC, Mielgo VE, Gastiasoro E, et al. Neuroprotective effects of the nonpsychoactive cannabinoid cannabidiol in hypoxicischemic newborn piglets. Pediatr Res. 2008; 64: 653–658. PubMed: https://pubmed.ncbi.nlm.nih.gov/18679164/
  131. Lafuente H, Alvarez FJ, Pazos MR, Alvarez A, Rey-Santano MC, et al. Cannabidiol reduces brain damage and improves functional recovery after acute hypoxia ischemia in newborn pigs. Pediatr Res. 2011; 70; 272–277. PubMed: https://pubmed.ncbi.nlm.nih.gov/21654550/
  132. Castillo A, Tolón MR, Fernández-Ruiz J, Romero J, Martinez-Orgado J. The neuroprotective effect of cannabidiol in an in vitro model of newborn hypoxicischemic brain damage in mice is mediated by CB(2) and adenosine receptors. Neurobiol Dis. 2010; 37: 434–440. PubMed: https://pubmed.ncbi.nlm.nih.gov/19900555/
  133. Marinelli L, Maurizio Balestrino, Laura Mori, Luca Puce, Gian Marco Rosa, et al. A randomised controlled cross- over double- blind pilot study protocol on THC: CBD oromucosal spray efficacy as an add- on therapy for post- stroke spasticity. BMJ Open. 2017; 7: e016843. PubMed: https://pubmed.ncbi.nlm.nih.gov/28882919/
  134. Caltagirone C, Carlo Cisari, Carlo Schievano, Rosanna Di Paola, Marika Cordaro, et al. Co-ultramicronized palmitoylethanolamide/luteolin in the treatment of cerebral ischemia: from rodent to man. Transl Stroke Res. 2016; 7: 54-69. PubMed: https://pubmed.ncbi.nlm.nih.gov/26706245/
  135. Espejo-Porras F, Fernández-Ruiz J, de Lago E. Analysis of endocannabinoid receptors and enzymes in the post-mortem motor cortex and spinal cord of amyotrophic lateral sclerosis patients. Amyotrophic Lateral Scler Frontotemporal Degener. 2018; 19: 377-386. PubMed: https://pubmed.ncbi.nlm.nih.gov/29334787/
  136. Pasquarelli N, Engelskirchen M, Hanselmann J, Endres S, Porazik C, et al. Evaluation of monoacylglycerol lipase as a therapeutic target in a transgenic mouse model of ALS. Neuropharmacology. 2017; 124: 157-169. PubMed: https://pubmed.ncbi.nlm.nih.gov/28373073/
  137. Fernández-Trapero M, Espejo-Porras F, Rodríguez-Cueto C, Coates JR, Pérez-Díaz C, et al. Upregulation of CB2receptors in reactive astrocytes in canine degenerative myelopathy, a disease model of amyotrophic lateral sclerosis. Dis Model Mech. 2017; 10: 551-558. PubMed: https://pubmed.ncbi.nlm.nih.gov/28069688/
  138. Zhao P, Ignacio S, Beattie EC, Abood ME. Altered presymptomatic AMPA and cannabinoid receptor trafficking in motor neurons of ALS model mice: implications for excitotoxicity. Eur J Neurosci. 2008; 27: 572-579. PubMed: https://pubmed.ncbi.nlm.nih.gov/18279310/
  139. Shoemaker JL, Kathryn A Seely, Ronald L Reed, John P Crow, Paul L Prather. The CB2 cannabinoid agonist AM-1241 prolongs survival in a transgenic mouse model of amyotrophic lateral sclerosis when initiated at symptom onset. J Neurochem. 2007; 101: 87-98. PubMed: https://pubmed.ncbi.nlm.nih.gov/17241118/
  140. Espejo- Porras F, Fabiana Piscitelli, Roberta Verde, José A Ramos, Vincenzo Di Marzo, et al. Changes in the endocannabinoid signaling system in CNS structures of TDP-43 transgenic mice: relevance for a neuroprotective therapy in TDP-43-related disorders. J Neuroimmune Pharmacol. 2015; 10: 233-244. PubMed: https://pubmed.ncbi.nlm.nih.gov/25819934/
  141. Espejo- Porras F, Fernandez- Ruiz J, de Lago E. Analysis of endocannabinoid receptors and enzymes in the post- mortem motor cortex and spinal cord of amyotrophic lateral sclerosis patients. Amyotroph Lateral Scler Frontotemporal Degener. 2018; 19: 377-386. PubMed: https://pubmed.ncbi.nlm.nih.gov/29334787/
  142. Witting A, Patrick Weydt, Soyon Hong, Michel Kliot, Thomas Moller, et al. Endocannabinoids accumulate in spinal cord of SOD1 G93A transgenic mice. J Neurochem. 2004; 89: 1555-1557. PubMed: https://pubmed.ncbi.nlm.nih.gov/15189359/
  143. Bilsland LG, James RT Dick, Pryce G, Petrosino S, Di Marzo V, et al. Increasing cannabinoid levels by pharmacological and genetic manipulation delay disease progression in SOD1 mice. FASEB J. 2006; 20: 1003–1005. PubMed: https://pubmed.ncbi.nlm.nih.gov/16571781/
  144. Pasquarelli N, Engelskirchen M, Hanselmann J, Endres S, Porazik C, et al. Evaluation of monoacylglycerol lipase as a therapeutic target in a transgenic mouse model of ALS. Neuropharmacology. 2017; 124: 157-169. PubMed: https://pubmed.ncbi.nlm.nih.gov/28373073/
  145. Rajan TS, Scionti D, Diomede F, Grassi G, Pollastro F, et al. Gingival stromal cells as an in vitro model: cannabidiol modulates genes linked with amyotrophic lateral sclerosis. J Cell Biochem. 2017; 118: 819-828. PubMed: https://pubmed.ncbi.nlm.nih.gov/27714895/
  146. Moreno-Martet M, Espejo-Porras F, Fernández-Ruiz J, de Lago E. Changes in endocannabinoid receptors and enzymes in the spinal cord of SOD1(G93A) transgenic mice and evaluation of a Sativex®-like combination of phytocannabinoids: interest for future therapies in amyotrophic lateral sclerosis. CNS Neurosci Ther. 2014; 20: 809-815. PubMed: https://pubmed.ncbi.nlm.nih.gov/24703394/
  147. Palma E, Reyes-Ruiz JM, Lopergolo D, Roseti C, Bertollini C, et al. Acetylcholine receptors from human muscle as pharmacological targets for ALS therapy. Proc Natl Acad Sci USA. 2016; 113: 3060-3065. PubMed: https://pubmed.ncbi.nlm.nih.gov/26929355/
  148. Clemente S. Amyotrophic lateral sclerosis treatment with ultramicronized palmitoylethanolamide: a case report. CNS Neurol Disord Drug Targets. 2012; 11: 933-936. PubMed: https://pubmed.ncbi.nlm.nih.gov/22998138/
  149. Stampanoni Bassi M, Gentile A, Iezzi E, Zagaglia S, Musella A, et al. Transient Receptor Potential Vanilloid 1 Modulates Central Inflammation in Multiple Sclerosis. Front Neurol. 2019; 10: 30. PubMed: https://pubmed.ncbi.nlm.nih.gov/30761069/
  150. Mecha M, Yanguas-Casás N, Feliú AA, Mestre L, Carrillo-Salinas F, et al. The endocannabinoid 2-AG enhances spontaneous remyelination by targeting microglia. Brain, Behav Immun. 2019; 77: 110-126. PubMed: https://pubmed.ncbi.nlm.nih.gov/30582962/
  151. Baker D, G Pryce, J L Croxford, P Brown, R G Pertwee, et al. Cannabinoids control spasticity and tremor in a multiple sclerosis model. Nature. 2000; 404: 84-87. PubMed: https://pubmed.ncbi.nlm.nih.gov/10716447/
  152. Baker D, G Pryce, J L Croxford, P Brown, R G Pertwee, et al. Endocannabinoids control spasticity in a multiple sclerosis model. FASEB J. 2001; 15: 300-302. PubMed: https://pubmed.ncbi.nlm.nih.gov/11156943/
  153. Hilliard A, C Stott, S Wright, G Guy, G Pryce, et al. Evaluation of the effects of Sativex (THC BDS: CBD BDS) on inhibition of spasticity in a chronic relapsing experimental allergic autoimmune encephalomyelitis: a model of multiple sclerosis. ISRN Neurol. 2012; 2012: 802649. PubMed: https://pubmed.ncbi.nlm.nih.gov/22928118/
  154. Giacoppo S, Bramanti P, Mazzon E. Sativex in the management of multiple sclerosis- related spasticity: an overview of the last decade of clinical evaluation. Mult Scler Relat Disord. 2017; 17: 22-31. PubMed: https://pubmed.ncbi.nlm.nih.gov/29055461/
  155. Markova J, Ute Essner, Bülent Akmaz, Marcella Marinelli, Christiane Trompke, et al. Sativex® as add- on therapy vs. further optimized first- line ANTispastics (SAVANT) in resistant multiple sclerosis spasticity: a double- blind, placebo- controlled randomised clinical trial. Int J Neurosci. 2018; 129: 119-128. PubMed: https://pubmed.ncbi.nlm.nih.gov/29792372/
  156. Koch G, Francesco Mori, Claudia Codecà, Hajime Kusayanagi, Fabrizia Monteleone, et al. Cannabis- based treatment induces polarity- reversing plasticity assessed by theta burst stimulation in humans. Brain Stimul. 2009; 2: 229-233. PubMed: https://pubmed.ncbi.nlm.nih.gov/20633421/
  157. Carotenuto A, Iodice R, Petracca M, Inglese M, Cerillo I, et al. Upper motor neuron evaluation in multiple sclerosis patients treated with Sativex®. Acta Neurol Scand. 2017; 135: 442-448. PubMed: https://pubmed.ncbi.nlm.nih.gov/27500463/
  158. Russo M, Calabrò RS, Naro A, Sessa E, Rifici C, et al. Sativex in the management of multiple sclerosis- related spasticity: role of the corticospinal modulation. Neural Plast. 2015; 2015: 656582. PubMed: https://pubmed.ncbi.nlm.nih.gov/25699191/
  159. Turri M, Francesco T, Francesco D, Giampietro Z, Valeria T, et al. Pain modulation after oromucosal cannabinoid spray (SATIVEX®) in patients with multiple sclerosis: a study with quantitative sensory testing and laser- evoked potentials. Medicines. 2018; 5: 59. PubMed: https://pubmed.ncbi.nlm.nih.gov/29933552/
  160. Messina S, Solaro C, Righini I, Bergamaschi R, Bonavita S, et al. Sativex in resistant multiple sclerosis spasticity: discontinuation study in a large population of Italian patients (SA.FE. study). PLOS ONE. 2017; 12:  e0180651. PubMed: https://pubmed.ncbi.nlm.nih.gov/28763462/
  161. Sorosina M, Clarelli F, Ferrè L, Osiceanu AM , Unal NT, et al. Clinical response to Nabiximols correlates with the downregulation of immune pathways in multiple sclerosis. Eur J Neurol. 2018; 25: 934–e70. PubMed: https://pubmed.ncbi.nlm.nih.gov/29528549/
  162. Orefice NS, Alhouayek M, Carotenuto A, Montella S, Barbato F, et al. Oral palmitoylethanolamide treatment is associated with reduced cutaneous adverse effects of interferon- beta1a and circulating proinflammatory cytokines in relapsing- remitting multiple sclerosis. Neurotherapeutics. 2016; 13: 428-438. PubMed: https://pubmed.ncbi.nlm.nih.gov/26857391/
  163. Galve-Roperh I, Sánchez C, Cortés ML, del Pulgar TD, Izquierdo M, et al. Anti- tumoral action of cannabinoids: involvement of sustained ceramide accumulation and extracellular signal- regulated kinase activation. Nat Med. 2000; 6: 313-319. PubMed: https://pubmed.ncbi.nlm.nih.gov/10700234/
  164. Blazquez C, M Llanos Casanova, Anna Planas, Teresa Gómez Del Pulgar, Concepción Villanueva, et al. Inhibition of tumor angiogenesis by cannabinoids. FASEB J. 2003; 17: 529-531. PubMed: https://pubmed.ncbi.nlm.nih.gov/12514108/
  165. 165.Gurley SN, Ammaar H Abidi, Patrick Allison, Peihong Guan, Christopher Duntsch, et al. Mechanism of anti- glioma activity and in vivo efficacy of the cannabinoid ligand KM-233. J Neurooncol. 2012; 110: 163-177. PubMed: https://pubmed.ncbi.nlm.nih.gov/22875710/
  166. 166.Sanchez C, M L de Ceballos, T Gomez del Pulgar, D Rueda, C Corbacho, et al. Inhibition of glioma growth in vivo by selective activation of the CB(2) cannabinoid receptor. Cancer Res. 2001; 61: 5784-5789. PubMed: https://pubmed.ncbi.nlm.nih.gov/11479216/
  167. Aguado T, Arkaitz Carracedo, Boris Julien, Guillermo Velasco, Garry Milman, et al. Cannabinoids induce glioma stem- like cell differentiation and inhibit gliomagenesis. J Biol Chem. 2007; 282: 6854-6862. PubMed: https://pubmed.ncbi.nlm.nih.gov/17202146/
  168. Chao Ma, Ting-Ting Wu, Pu-Cha Jiang, Zhi-Qiang Li, Xin-Jun Chen, et al. Anti- carcinogenic activity of anandamide on human glioma in vitro and in vivo. Mol Med Rep. 2016; 13: 1558-1562. PubMed: https://pubmed.ncbi.nlm.nih.gov/26707955/
  169. Massimo Nabissi, Maria Beatrice Morelli, Antonietta Arcella, Claudio Cardinali, Matteo Santoni, et al. Post- transcriptional regulation of5′-untranslated regions of human transient receptor potential vanilloid type-1 (TRPV-1) channels: role in the survival of glioma patients. Oncotarget. 2016; 7: 81541–81554. PubMed: https://pubmed.ncbi.nlm.nih.gov/27829230/
  170. Vaccani A, Massi P, Colombo A, Rubino T, Parolaro D. Cannabidiol inhibits human glioma cell migration through a cannabinoid receptor independent mechanism. Br J Pharmacol. 2005; 144: 1032-1036. PubMed: https://pubmed.ncbi.nlm.nih.gov/15700028/
  171. Moreno E, Clara A, Mireia M, María M C, Eduardo Pérez-Gómez, et al. Targeting CB2-GPR55 receptor heteromers modulates cancer cell signaling. J Biol Chem. 2014; 289: 21960-21972. PubMed: https://pubmed.ncbi.nlm.nih.gov/24942731/
  172. Scott KA, Dalgleish AG, Liu WM. The combination of cannabidiol and delta9- tetrahydrocannabinol enhances the anticancer effects of radiation in an orthotopic murine glioma model. Mol Cancer Ther. 13, 2955-2967. PubMed: https://pubmed.ncbi.nlm.nih.gov/25398831/
  173. Torres S, Lorente M, Rodríguez-Fornés F, Hernández-Tiedra S, Salazar M, et al. A combined preclinical therapy of cannabinoids and temozolomide against glioma. Mol Cancer Ther. 2011; 10: 90-103. PubMed: https://pubmed.ncbi.nlm.nih.gov/21220494/
  174. Nabissi M, Morelli MB, Santoni M, Santoni G. Triggering of the TRPV2 channel by cannabidiol sensitizes glioblastoma cells to cytotoxic chemotherapeutic agents. Carcinogenesis. 2013; 34: 48–57. PubMed: https://pubmed.ncbi.nlm.nih.gov/23079154/
  175. Nabissi M, Morelli MB, Amantini C, Liberati S, Santoni M, et al. Cannabidiol stimulates Aml-1adependent glial differentiation and inhibits glioma stem- like cells proliferation by inducing autophagy in a TRPV2-dependent manner. Int J Cancer. 2015; 137: 1855-1869. PubMed: https://pubmed.ncbi.nlm.nih.gov/25903924/
  176. Sugimoto N, Ishibashi H, Ueda Y, Nakamura H, Yachie A, et al. Corticosterone inhibits the expression of cannabinoid receptor‑1 and cannabinoid receptor agonist‑induced decrease in cell viability in glioblastoma cells. Oncol Lett. 2019; 18: 1557-1563. PubMed: https://pubmed.ncbi.nlm.nih.gov/31423223/
  177. Fu H, Ge B, Chen D, Wu Y, Luo Q, et al. Phytanoyl-CoA 2-Hydroxylase-Interacting Protein-Like Gene Is a Therapeutic Target Gene for Glioblastoma Multiforme. Med Sci Monit. 2019; 25: 2583-2590. PubMed: https://pubmed.ncbi.nlm.nih.gov/30962415/
  178. Su B-X, Chen X, Huo J, Guo S.-Y, Ma R, et al. The synthetic cannabinoid WIN55212-2 ameliorates traumatic spinal cord injury via inhibition of GAPDH/Siah1 in a CB2-receptor dependent manner. Brain Res. 2017; 1671: 85–92. PubMed: https://pubmed.ncbi.nlm.nih.gov/28716633/
  179. Milligan AL, Szabo-Pardi TA, Burton MD. Cannabinoid Receptor Type 1 and Its Role as an Analgesic: An Opioid Alternative? J Dual Diagn. 2019; 16: 106-119. PubMed: https://pubmed.ncbi.nlm.nih.gov/31596190/
  180. Haggai S, Brill S. Cannabis-based medicines for chronic pain management: current and future prospects. Curr Opin Anesthesiol. 2019; 32: 623-628. PubMed: https://pubmed.ncbi.nlm.nih.gov/31356363/
  181. Blanton HL, Brelsfoard J, DeTurk N, Pruitt K, Narasimhan M, et al. Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain. Drugs. 2019; 79: 969-995. PubMed: https://pubmed.ncbi.nlm.nih.gov/31127530/
  182. Henshaw FR, Dewsbury LS, Lim CK, Steiner GZ. The Effects of Cannabinoids on Pro- and Anti-Inflammatory Cytokines: A Systematic Review of In Vivo Studies. Cannabis Cannabinoid Res. 2021. PubMed: https://pubmed.ncbi.nlm.nih.gov/33998900/
  183. Haranishi Y, Hara K, Terada T. Inhibitory effect of intrathecally administered AM404, an endocannabinoid reuptake inhibitor, on neuropathic pain in a rat chronic constriction injury model. Pharmacol Rep. 2021. PubMed: https://pubmed.ncbi.nlm.nih.gov/33783763/
  184. Armin S, Muenster S, Abood M, Benamar K. GPR55 in the brain and chronic neuropathic pain. Behav Brain Res. 2021; 406: 113248. PubMed: https://pubmed.ncbi.nlm.nih.gov/33745983/
  185. Canavan C, Inoue T, McMahon S, Doody C, Blake C, et al. The Efficacy, Adverse Events & Withdrawal Rates of the Pharmacological Management of Chronic Spinal Cord Injury Pain: A Systematic Review & Meta-Analysis. Pain Med. 2021; pnab140. PubMed: https://pubmed.ncbi.nlm.nih.gov/33844010/
  186. Nabata KJ, Tse EK, Nightingale TE, Lee AHX, Eng JJ, et al. The Therapeutic Potential and Usage Patterns of Cannabinoids in People with Spinal Cord Injuries: A Systematic Review. Curr Neuropharmacol. 2021; 19: 402-432. PubMed: https://pubmed.ncbi.nlm.nih.gov/32310048/
  187. Jing N, Fang B, Li Z, Tian A. Exogenous activation of cannabinoid-2 receptor modulates TLR4/MMP9 expression in a spinal cord ischemia reperfusion rat model. J Neuroinflammation. 2020; 17: 101. PubMed: https://pubmed.ncbi.nlm.nih.gov/32248810/
  188. Bourke JA, Catherwood VJ, Nunnerley JL, Martin RA, Levack WMM, et al. Using cannabis for pain management after spinal cord injury: a qualitative study. Spinal Cord Ser Cases. 2019; 5: 82. PubMed: https://pubmed.ncbi.nlm.nih.gov/31632740/
  189. Huo J, Ma R, Chai X, Liang HJ, Jiang P, et al. Inhibiting a spinal cord signaling pathway protects against ischemia injury in rats. J Thorac Cardiovasc Surg. 2019; 157: 494-503. PubMed: https://pubmed.ncbi.nlm.nih.gov/30195603/
  190. Li H, Kong W, Chambers CR, Yu D, Ganea D, et al. The non-psychoactive phytocannabinoid cannabidiol (CBD) attenuates pro-inflammatory mediators, T cell infiltration, and thermal sensitivity following spinal cord injury in mice. Cell Immunol. 2018; 329: 1-9. PubMed: https://pubmed.ncbi.nlm.nih.gov/29784129/
  191. Jing N, Fang B, Wang ZL, Ma H. Remote Ischemia Preconditioning Attenuates Blood-Spinal Cord Barrier Breakdown in Rats Undergoing Spinal Cord Ischemia Reperfusion Injury: Associated with Activation and Upregulation of CB1 and CB2 Receptors. Cell Physiol Biochem. 2017; 43: 2516-2524. PubMed: https://pubmed.ncbi.nlm.nih.gov/29130941/
  192. Grao-Castellote C, Torralba-Collados F, Gonzalez LM, Giner-Pascual M. Delta-9-tetrahydrocannabinol-cannabidiol in the treatment of spasticity in chronic spinal cord injury: a clinical experience. Rev Neurol. 2017; 65: 295-302. PubMed: https://pubmed.ncbi.nlm.nih.gov/28929471/
  193. Mouhamed Y, Vishnyakov A, Qorri B, Sambi M, Frank SS, et al. Therapeutic potential of medicinal marijuana: an educational primer for health care professionals. Drug Healthc Patient Saf. 2018; 10: 45-66. PubMed: https://pubmed.ncbi.nlm.nih.gov/29928146/
  194. Abramovici H. Information for Health Care Professionals: Cannabis (Marihuana, Marijuana) and the Cannabinoids; Health Canada: Ottawa, ON, Canada, 2018.
  195. Jakubovski E, Pisarenko A, Fremer C, Haas M, May M, et al. The CANNA-TICS Study Protocol: A Randomized Multi-Center Double-Blind Placebo Controlled Trial to Demonstrate the Efficacy and Safety of Nabiximols in the Treatment of Adults With Chronic Tic Disorders. Front Psychiatry. 2020; 11: 575826. PubMed: https://pubmed.ncbi.nlm.nih.gov/33324255/
  196. Szejko N, Fichna JP, Safranow K, Dziuba T, Żekanowski C, et al. Association of a Variant of CNR1 Gene Encoding Cannabinoid Receptor 1 With Gilles de la Tourette Syndrome. Front Genet. 2020; 11: 125. PubMed: https://pubmed.ncbi.nlm.nih.gov/32194619/
  197. Milosev LM, Psathakis N, Szejko N, Jakubovski E, Müller-Vahl KR. Treatment of Gilles de la Tourette Syndrome with Cannabis-Based Medicine: Results from a Retrospective Analysis and Online Survey. Cannabis Cannabinoid Res. 2019; 4: 265-274. PubMed: https://pubmed.ncbi.nlm.nih.gov/31872061/
  198. Avagliano C, Russo R, De Caro C, Cristiano C, La Rana G, et al. Palmitoylethanolamide protects mice against 6-OHDA-induced neurotoxicity and endoplasmic reticulum stress: In vivo and in vitro evidence. Pharmacol Res. 2016; 113: 276-289. PubMed: https://pubmed.ncbi.nlm.nih.gov/27616549/
  199. Esposito E, Impellizzeri D, Mazzon E, Paterniti I, Cuzzocrea S. Neuroprotective activities of palmitoylethanolamide in an animal model of Parkinson's disease. PLoS One. 2012; 7: e41880. PubMed: https://pubmed.ncbi.nlm.nih.gov/22912680/
  200. Crupi R, Impellizzeri D, Cordaro M, Siracusa R, Casili G, et al. N-palmitoylethanolamide Prevents Parkinsonian Phenotypes in Aged Mice. Mol Neurobiol. 2018; 55: 8455-8472. PubMed: https://pubmed.ncbi.nlm.nih.gov/29552727/
  201. Lotan I, Treves TA, Roditi Y, Djaldetti R. Cannabis (medical marijuana) treatment for motor and non-motor symptoms of Parkinson disease: an open-label observational study. Clin Neuropharmacol. 2014; 37: 41-44. PubMed: https://pubmed.ncbi.nlm.nih.gov/24614667/
  202. Van Laere K, Casteels C, Dhollander I, Goffin K, Grachev I, et al. Widespread decrease of type 1 cannabinoid receptor availability in Huntington disease in vivo. J Nucl Med. 2010; 51: 1413-1417. PubMed: https://pubmed.ncbi.nlm.nih.gov/20720046/
  203. Horne EA, Coy J, Swinney K, Fung S, Cherry AE, et al. Down regulation of cannabinoid receptor 1 from neuropeptide Y interneurons in the basal ganglia of patients with Huntington's disease and mouse models. Eur J Neurosci. 2013; 37: 429-440. PubMed: https://pubmed.ncbi.nlm.nih.gov/23167744/
  204. Augustin SM, Lovinger DM. Functional Relevance of Endocannabinoid-Dependent Synaptic Plasticity in the Central Nervous System. ACS Chem Neurosci. 2018; 9: 2146-2161. PubMed: https://pubmed.ncbi.nlm.nih.gov/29400439/