3.1. Selective Agonists of Cannabinoid Receptor 1 (CB1R)
The research by Crunfli et al. [
36] explored the therapeutic potential of the CB
1R agonist arachidonyl-2'-chloroethylamide (ACEA) against sporadic AD, which was explored through various in vitro in neuro-2a neuroblastoma cells and in vivo experiments in adult male Wistar rats, following streptozotocin (STZ) infusion. It was observed that intraperitoneal (i.p.) treatment with 3 mg/kg ACEA restored cognitive impairment, improving both short-term (33.07%) and long-term (44.94%) memory in comparison to STZ-treated rats. It was also shown that the STZ+ACEA group showed a significant increase in the levels of insulin receptors (92.13%) and antiapoptotic proteins Bcl-2 (110.4%), while at the same time a significant decrease in the activity of protein kinase B (Akt) (≈61%) and extracellular signal-regulated kinase (ERK) (144.9%) was observed. Finally, administration of ACEA to STZ-treated cells increased their viability by 34.18%±10.93%.
In their study, Moreira-Silva et al. [
37] investigated the impact of intracerebroventricular (i.c.v.) injection of AEA (100 ng) in a rat model induced with STZ, simulating sporadic AD dementia. Cognitive performance was evaluated using the Novel Object Recognition (NOR) test and the escape latency index in the elevated plus maze (EPM) test. AEA demonstrated a preventive effect against STZ-induced impairments in recognition and non-associative emotional memory. Moreover, STZ-induced cerebroventricular enlargement was mitigated by AEA administration. Notably, key components of synaptic transmission, such as synaptophysin and syntaxin, which were reduced by STZ, showed a reversal after AEA treatment.
Khavandi et al. [
38] explored the impact of specific endocannabinoids—AEA, noladin, and O-arachidonylethanolamine (OAE)—on the accumulation and toxicity of Aβ
42. Employing in vitro techniques with mouse hippocampal HT22 cells and human CB
1R-expressing hamster ovary CHO cells, they found significant inhibition of Aβ
42 accumulation by AEA (93.3%), noladin (72.9%), and arachidonic acid (AA) (94.5%) at a concentration of 10μM. Moreover, they demonstrated the capacity of AEA (10μM), noladin (10μM), OAE (1μM, 10μM), and AA (5μM, 10μM) to enhance the viability of HT22 cells via CB
1R agonism, with only AEA showing no efficacy in CHO cells at concentrations of 1μM, 5μM, and 10μM.
In their study, Hosseininia et al. [
39] investigated the impact of chronic corticolimbic microinjection of the selective CB
1R agonist arachidonylcyclopropylamide (ACPA; 10 ng/0.5 μL) and lentiviral particles containing miRNA-137 (miR-137) or miR-let-7a on memory-impaired animal models exposed to i.c.v. STZ. Following ACPA microinjection, the step-through latency of STZ rats in the passive avoidance (PA) test significantly increased compared to controls, indicating enhanced memory function in various brain regions including the hippocampal CA1 region (≈90%), central amygdala (CeA) (≈75%), and medial prefrontal cortex (mPFC) (≈95%). Notably, this effect highlighted the cannabinoid's action across different injection sites. Additionally, the expression levels of the
MAGL gene, directly linked to the ECS, were observed to decrease (resulting in increased endocannabinoids) in all targeted regions of STZ rat brains receiving miRNA-137 or -let-7a-carrying lentivirus particles, effectively reversing the amnesic effects of STZ.
In another study, Zhang et al. [
40] explored the effects of CB
1R peptide agonists (m)RVD-hemopressin (RVD) and (m)VD-hemopressin (VD), on memory in Aβ
1-42-lesioned mice. Through i.c.v. injection of RVD or VD (5 nmol), they found restoration of Aβ
1-42-induced memory impairment, with mice displaying significantly higher discrimination indices in both the NOR test and the Object Location Recognition (OLR) task. Building upon their prior research, the same group [
41] conducted a similar experiment, this time using a mouse model of neurodegeneration affected by an i.p. injection of scopolamine. The results resembled those of the previous study, with RVD and VD (1, 2.5, 5 nmol) demonstrating a dose-dependent restoration of memory function in the NOR and OLR tests, as expected. In an attempt to elucidate the mechanism underlying VD's role in memory restoration, they revealed that VD countered the Aβ
1-42-induced elevation of reactive oxidant species (ROS) and malondialdehyde (MDA) in mouse hippocampal neurons. Concurrently, there was an increase in the levels of antioxidant enzymes such as catalase (CAT) and glutathione peroxidase (GPx). Moreover, VD administration resulted in a decrease in the expression of the pro-apoptotic protein Bax, alongside an increase in the expression of the anti-apoptotic protein Bcl-2, shedding light on its apoptotic regulatory mechanisms [
42]. In their latest attempt to elucidate the mechanism of action of RVD [
43], they observed similar mechanisms of action, this time in HT22 cells treated with scopolamine. Notably, RVD prevented the dysfunction of the brain-derived neurotrophic factor (BDNF)/Tropomyosin receptor kinase B (TrkB)/protein kinase B (Akt) signaling pathway, concurrently boosting the expression of synapsin-1 and postsynaptic density protein 95 (PSD-95) proteins. Adding to these findings, prior research [
44] showcased RVD's efficacy in mitigating Aβ
1-42-induced TAU protein phosphorylation by inhibiting protein kinase A (PKA) and GSK-3β enzyme activity, as well as curtailing neuronal growth in SH-SY5Y cells. These actions of both RVD and VD, facilitated through CB
1R activation, position them as promising candidates for therapeutic interventions aimed at mitigating the characteristic pathogenesis of AD.
Finally, the research conducted by Velikova et al. [
45] focused on examining the role of CB
1R in memory and learning processes. Through their study, they administered either the CB
1R agonist HU-210 (5 μg/day), or the CB
1R antagonist SR 141716A (3 μg/day), via i.c.v. injection to olfactory bulbectomy rats (OBX) (
Scheme 1). Their findings revealed that HU-210 improved memory function in OBX rats, whereas SR 141716A exacerbated the memory deficits induced by bulbectomy, as evidenced by active/passive avoidance tests. This underscores the direct involvement of the CB
1R in memory function in OBX rats, serving as a model simulating AD symptomatology.
The main outcomes of the studies involving selective CB
1R agonists are summarized in
Table 1.
3.2. Selective Agonists of Cannabinoid Receptor 2 (CB2R)
The study conducted by Jayant et al. [
46] explored the impact of the CB
2R agonist, 1-phenylsatin, in a mouse model of NA induced by either STZ or aluminum chloride (AlCl
3) + D-galactose (D-Gal). Oral administration of 20mg/kg 1-phenylsatin restored escape latency and time spent in the target quadrant to normal levels more effectively than donepezil in the Morris Water Maze (MWM) test. In the attentional set shifting test, 1-phenylsatin reduced efforts in the reversal 1 (REV1) and extra-dimensional (EV) stages, while also mitigating biochemical (oxidative stress, AChE activity, etc.) and structural (accumulation of Aβ plaques) brain lesions.
Cheng et al. [
47] conducted a study to investigate the anti-inflammatory effects of the CB
2R agonist β-caryophyllene (BCP) in APP/PS1 mice, focusing on its action via the PPARγ pathway. Following oral administration of 48mg/kg BCP, significant improvements in escape latency and distance traveled were observed in the MWM test starting from the 3rd day. Moreover, the time spent in the target quadrant closely resembled that of healthy mice. BCP administration also dose-dependently reduced Aβ deposition in the cerebral cortex and hippocampus. Additionally, the study measured reduced levels of astrogliosis, microglial activation, cyclooxygenase-2 (COX-2), and of the expression of pro-inflammatory cytokines TNF-a and IL-1β. Overall, there was restoration of spatial memory and cognitive functions by BCP.
Del Cerro et al. [
48] demonstrated the effectiveness of the innovative CB
2R agonist, PGN33 (
Scheme 2), in influencing the viability of lymphoblasts isolated from late-onset AD patients. PGN33 administration at varying doses (2.5nM, 5nM, 7.5nM, 10nM) dose-dependently decreased the viability of NA-lymphoblasts, limiting their uncontrolled proliferation with a potency similar to that of the agonist JWH-133. This mechanism is achieved by impeding Ca
2+/CaM-dependent activation of phosphoinositide 3-kinase (PI3K)/Akt signaling, which typically triggers cell cycle activation. Additionally, PGN33 demonstrated efficacy in mitigating Aβ-induced death of SH-SY5Y neuroblastoma cells.
Abd El-Rahman and Fayed [
49] aimed to investigate the potential protective effects and mechanisms of action of the CB
2R agonist AM1241 (
Scheme 2) in alleviating cognitive and learning deficits in a model of AD in female rats (oophorectomy + D-gal). Administration of AM1241 led to a dose-dependent increase in the discrimination index [0.66±0.02 and 0.95±0.04 for 3 and 6 mg (i.p.), respectively] as well as the preference index (1.06±0.05 at 6mg) compared to the AD-model values in the NOR test. Concurrently, 6mg AM1241 significantly reduced the escape latency of the rats compared to the AD-model (10.38±0.95 vs. 39.08±3.52) in the MWM test. These improvements were accompanied by a decrease in inflammatory signaling via the toll-like receptor 4 (TLR4)/nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) p65 pathway, evidenced by downregulation of the adapter protein myeloid differentiation primary response 88 (Myd88) and enhancement of cAMP response element-binding protein (CREB)/ brain-derived neurotrophic factor (BDNF) expression. Additionally, AM1241 inhibited astrocyte and microglial cell activation, highlighting the importance of CB
2R activation in AD treatment.
Li et al. [
50] investigated the effects of CB
2R activation via i.p. administration of 0.5 mg/kg of the selective CB
2R agonist JWH-015 (
Scheme 2) in transgenic APP/PS1 mice. Following the NOR and MWM tests, CB
2R activation was found to enhance novel object recognition but did not ameliorate the impaired hippocampus-dependent spatial memory in mice. Additionally, using ionized calcium-binding adaptor molecule 1 (Iba1) immunofluorescence and real-time PCR techniques, the researchers observed that the agonist prevented the activation of cortical microglia while simultaneously promoting their conversion from M1 to M2 phenotype, demonstrating the immunoprotective effect of activated CB
2R. Moreover, Golgi staining revealed improvements in dendritic complexity in the cortex of APP/PS1 mice.
The present study conducted by Çakır et al. [
51] describes an experiment utilizing a rat model with hyperphosphorylated TAU induced by okadaic acid (OKA) administration, wherein the CB
2R agonist JWH-133 (0.2 mg/kg, i.p.) (
Scheme 2) was administered. During the 1st to 4th day of the MWM test, reductions were observed in both escape latency (85sec - 45sec vs. 95sec - 35sec) and distance traveled (1500cm - 900cm vs. 1550cm - 600cm) in the OKA and OKA+JWH-133 groups, respectively. Additionally, decreased levels of caspase-3, phosphorylated TAU, Aβ, TNF-a, and IL-1β were observed in the cerebral cortex and hippocampus of the OKA+JWH-133 group compared to the OKA group. Overall, the agonist JWH-133 prevented the decline of spatial memory, limited the inflammatory response, and consequently reduced neuronal apoptosis, positioning it as a promising novel therapeutic agent.
The main outcomes of the studies involving selective CB
2R agonists are summarized in
Table 2.
3.3. Agonists of Cannabinoid Receptor 2 (CB2R) Associated with Cholinergic Pathways
In their investigation, Marta et al. [
52] explored the relationship between cholinergic pathways and the CB
2R agonist JWH-133, as well as their impact on mouse memory following scopolamine administration. Utilizing PA tests in mice, they demonstrated that combining a suboptimal dose of JWH-133 (0.25 mg/kg) with a suboptimal dose of a cholinergic receptor agonist (nicotine, 0.05 mg/kg) significantly improved cognitive performance in mice. Likewise, the co-administration of JWH-133 with an effective dose of a cholinergic receptor antagonist (scopolamine, 1 mg/kg) mitigated the cognitive impairment induced by the antagonist. These findings suggest a significant involvement of CB
2R activation in memory processes associated with cholinergic pathways.
Montanari et al. [
53] outlined a procedure for synthesizing 2-arylbenzofuran derivatives, examining their interactions with cholinesterases and cannabinoid receptors. Among these derivatives, compound 8 (5 μM) (
Scheme 3) emerged as a promising candidate for a comprehensive treatment approach. It not only acted as a butyrylcholinesterase (BChE) inhibitor, addressing cholinergic impairment, but also displayed neuroprotective properties against Aβ
1-42 oligomers. Furthermore, it exhibited potent and selective agonism toward CB
2R, offering immunoregulatory effects by prompting a shift in microglial cells from the inflammatory (M1) to the neuroprotective (M2) phenotype. Conversely, compound 10 (5 μM) (
Scheme 3) demonstrated robust immunomodulatory activity as an inverse agonist of CB
2R, suggesting its potential as a foundational model for the development of other immunomodulatory pharmaceutical agents.
Spatz et al. [
54] focused on synthesizing and investigating hybrid molecules acting as CB
2R agonists and BChE antagonists. Among the compounds they synthesized, 15d (with IC
50 BChE= 0.62 µM, EC
50 CB
2R= 244 nM) and 21d (with IC
50 BChE= 0.15 µM, EC
50 CB
2R= 1.3 µM) (
Scheme 3) demonstrated considerable promise. These compounds exhibited CB
2R-dependent immunomodulatory effects, particularly by attenuating the inflammatory M1 phenotype in lipopolysaccharide (LPS)-treated N9 microglial cells. Moreover, when administered i.p. to mice challenged with Aβ
25–35 oligomers, compound 15d (at doses of 0.3–3 mg/kg/day) effectively prevented learning impairments in both the spontaneous alternation Y-maze and PA tests. The dual action of these substances, coupled with their ability to cross the blood-brain barrier (BBB), suggests their potential therapeutic utility against AD.
In their study, Scheiner et al. [
55] attempted to merge the therapeutic benefits of AChE inhibitors with the neuroprotective properties of CB
2R agonists by designing hybrid synthetic analogs of tacrine and a selective CB
2R agonist. Among these compounds, 3e, 4a, and 8 (
Scheme 3) exhibited neuroprotective effects in a cellular model of neuronal oxidative stress, with compound 8 (at 5 µM) proving to be the most potent, while compound 4a displayed toxicity at doses exceeding 5 µM. Subsequently, in AD mouse models, administration of compounds 3e (at 0.3 mg/kg), 4a (at 1 mg/kg), and 8 (at 0.3 mg/kg) effectively prevented Aβ
25-35 infusion-induced memory impairments and learning deficits. Importantly, these compounds demonstrated significantly greater efficacy compared to the parent molecules, with the ability to penetrate the BBB.
The main outcomes of the studies involving compounds targeting CB
2R and cholinergic pathways are summarized in
Table 3.
3.4. Non-Selective Agonists of Cannabinoid Receptors 1 and 2 (CB1R and CB2R)
The study conducted by Cao et al. [
56] explored the potential therapeutic benefits of THC in N2a/AβPPswe cells. Through a series of experiments, they established a time-dependent (6h, 24h, 48h) and dose-dependent (0.25nM, 2.5nM, 25nM, 250nM, 2500nM) relationship between THC administration and the reduction of Aβ
40 levels. This reduction occurred alongside the inhibition of Aβ
40 aggregation, all without compromising the normal immune response. Additionally, repeated THC administration over 24 h led to further decreases in Aβ protein production. Furthermore, a dose-dependent correlation emerged between THC and the reduction of GSK-3β levels, as well as phosphorylated TAU.
In their study, Gugliandolo et al. [
57] investigated the impact of pretreating SH-SY5Y cells with 20μM Δ8-THC before exposure to retinoic acid and subsequent treatment with Aβ
1-42. They observed that pretreatment with Δ8-THC elevated cell viability from approximately 75% to about 87%, without inducing any cytotoxic effects upon direct administration. Further analysis revealed that this effect was partly attributed to the attenuation of endoplasmic reticulum (ER) stress induced by Aβ
1-42. Specifically, Δ8-THC restored proteostasis by upregulating the expression of proteasome subunits (PSMB5) and ubiquitin, while concurrently suppressing the unfolded protein response (UPR). Additionally, a reduction in Bax protein levels accompanied by an increase in Bcl-2 levels was noted.
Due to the promising outcomes observed in prior studies involving THC, a recent randomized controlled trial [
58] focused on the oral administration of THC (1.5 mg thrice daily) or a placebo over a span of 21 days. This trial involved 50 patients with dementia, including AD, and accompanying neuropsychiatric symptoms (NPS). Despite earlier encouraging findings, this study revealed no statistically significant disparities in both the total Neuropsychiatric Inventory (NPI) score and its various subscales (such as agitation/aggression and aberrant motor behavior) between the THC and placebo cohorts (mean difference in NPI total = 3.2, 95% CI -0.8 to 1.9). Similar trends were observed across other assessment metrics like the Cohen-Mansfield Agitation Inventory (CMAI), Quality of Life-Alzheimer's Disease, and the Barthel Index. Additionally, no severe adverse effects attributable to the treatment were documented. Hence, while THC administration was well-tolerated, its therapeutic efficacy for managing dementia-related NPS did not demonstrate discernible benefits.
In their specific research, Coles et al. [
59] conducted a series of experiments in female APPxPS1 mice. They demonstrated that the chronic daily administration of a moderate dose (5mg/kg, i.p.) of CBD could have beneficial effects in AD. These experiments aimed to observe potential improvements in AD symptoms such as anxiety, disinterest in exploration, hyperactivity, cognitive and motor dysfunction, as well as sensorimotor impairment. Despite encountering unsuccessful outcomes in the majority of experiments, CBD administration seemed to restore both spatial learning speed and perseveration, along with the ability to recognize novel objects.
In this study [
60], CBD administration was explored as a potential safeguard for synaptic plasticity in slices from the CA1 region of the hippocampus in C57Bl/6 mice. The researchers utilized hippocampal long-term potentiation (LTP), a marker of synaptic strength limited by Aβ, to assess the impact. Treating hippocampal slices with CBD before Aβ injection seemed to prevent the decline of LTP, restoring it to levels comparable to the control group. Through the use of various antagonists, it was determined that CBD's specific therapeutic effect is attributed to the activation of the PPAR-γ receptor, with the CB
1R not directly involved.
In the study conducted by Amini and Abdolmaleki [
61], the efficacy of nano-chitosan-coated CBD administration was evaluated in AD-model rats to assess its impact on learning and memory. Results from the MWM test indicated a significant decrease in both escape latency and distance traveled. Notably, rats treated with 120mg/kg CBD spent more time on the exit platform. Concurrently, there was an observed increase in the expression of CB
1R and CB
2R in the hippocampus. These findings underscore the potential of coating CBD with nano-chitosan to enhance memory and learning processes.
Hao and Feng [
62] focused on uncovering the mechanisms behind CBD's effects on AD by analyzing gene expression data from RNA sequencing experiments conducted on APP/PS1 mice following cannabinoid administration. Their analysis of differentially expressed genes (DEGs) revealed an up-regulation of factors involved in both the immune response and the cellular autophagy pathway. This suggests that CBD's therapeutic actions stem from two primary mechanisms: firstly, by curbing neuroinflammation through bolstering the immune response, and secondly, by impeding AD's pathological processes through the induction of cellular recycling mechanisms.
Building upon the encouraging outcomes mentioned earlier, Alexandri et al. [
63] conducted the present study to assess the efficacy of CBD compared to standard treatment over a span of 6 months for managing dementia-related NPS. The administration of 3% CBD resulted in a significant reduction in the NPI index, indicating an improvement in the behavioral and psychological symptoms of dementia (BPSD) among the 20 patients involved. Conversely, classical treatment showed limited or no effects during this timeframe. These findings suggest that CBD might offer a more effective and safer alternative for treating BPSD, although larger clinical studies are warranted to validate these results.
In a limited-sized cohort study [
64], the administration of cannabis extract dissolved in oil (22% THC, 0.5% CBD, maximum dose 1ml/day) was investigated in 30 patients diagnosed with moderate to severe AD. The findings from the NPI questionnaire revealed a noteworthy improvement in key behavioral issues, including restlessness, irritability, sleep disturbances, and apathy. Moreover, both physical and verbal aggression behaviors exhibited notable reductions, as evidenced by the results of the CMAI questionnaire. Remarkably, following the administration of the cannabis extract, 45% of patients achieved a Mini Mental State Examination (MMSE) score indicative of mild to moderate cognitive improvement (15-17). Despite these promising results, the study's limited sample size underscores the necessity for larger-scale investigations to ascertain the efficacy of cannabis in the symptomatic treatment of AD.
In the specific case report by Ruver-Martins et al. [
28], the focus is on a 75-year-old patient diagnosed with mild AD, presenting with primary symptoms of memory impairment and spatiotemporal disorientation. Over a period of 22 months, the patient underwent treatment with small doses of cannabis extract, characterized by a THC:CBD ratio of 8:1. Throughout this treatment duration, the patient underwent evaluation using the MMSE and the Alzheimer's Disease Assessment Scale-Cognitive Subscale (ADAS-COG), revealing significant improvement, as evidenced by notable increases and decreases in respective scores. The doses of the cannabis extracts varied over the months, with a dosage of 500 μg being the most frequently administered. Remarkably, the treatment led to marked improvement in symptoms, corroborated both by the test results and the patient's subjective experience. The patient opted to continue the treatment, maintaining stable test results (MMSE
→ 24, ADAS-COG
→ 10).
In their study, Kim et al. [
65] explored the impact of intrahippocampal injection of cannabidiol acid (CBDA, 6µM, 3 µL) and tetrahydrocannabinolic acid (THCA, 12µM, 3 µL) in Aβ
1-42-injected ICR mice. Notably, mice exhibited a significant improvement in escape latency during day 4 of the MWM test when administered either THCA (≈35 s) or CBDA (≈25 s), compared to the Aβ group (≈45 s). Additionally, both cannabinoids demonstrated an equivalent increase in the discrimination index in the NOR test. Furthermore, there was a noteworthy reduction in the levels of Aβ polymers (Aβ group: 202%, CBDA: 66%, THCA: 81%, vs. control) and p-TAU (Aβ group: 160%, CBDA: 116%, THCA: 105%, vs. control) in the hippocampus of mice. These findings suggest that cannabinoids achieved restoration of cognitive functions, coupled with neuroprotective properties and ability to penetrate the BBB.
In another case report [
66], a 69-year-old woman diagnosed with AD presented severe NPS including depression and paranoid perceptions. From 2008 to 2019, her condition was regularly monitored, during which she underwent treatment with six different psychotropic medications, yielding no improvement. In the final two years of observation, the patient was prescribed dronabinol (Δ9-THC) drops at a dosage ranging from 4.9mg to 6.7mg per day. Remarkably, this intervention enabled the simultaneous discontinuation of three out of the six previously administered psychotropic drugs. Consequently, an improvement in the patient's emotional state was noted, alongside a reduction in disruptive behavior, aggression, and sedation, with no adverse effects reported.
In their study, Long et al. [
67] administered the AEA analog, N-linoleyltyrosine (NlTyr), to APP/PS1 mice. Notably, on the 7th day of the Rotarod test (RRT), mice treated with NlTyr exhibited increased time spent on the rod (control: 106.6 ± 3.7 s, APP/PS1: 66.25 ± 7.29 s, NlTyr 60 mg/kg: 101.75 ± 5.56 s), indicating a restoration of motor coordination. Concurrently, NlTyr-treated mice displayed improved cognitive and learning abilities, as evidenced by a return to normal escape latency values on the 6th day of the MWM test (control: 42.38 ± 7.73 s, APP/PS1: 53.00 ± 4.41 s, NlTyr 60 mg/kg: 43.13 ± 5.41 s) and enhanced time spent in the target quadrant. Moreover, through the induction of cannabinoid receptor-mediated autophagy, NlTyr effectively reduced Aβ
42 levels in the hippocampal CA1 region (APP/PS1: 26.33 ± 8.19 ng/g, NlTyr 30 mg/kg: 15 ± 1.63 ng/g), thereby mitigating neuronal injury.
In another study [
68], the objective was to explore the potential anti-inflammatory effects of the cannabinoid agonist β-amyrin in rat microglial cells treated with LPS/interferon-γ (IFN-γ). Results revealed that β-amyrin, at concentrations ranging from 4 to 16 µM, not only enhanced cell survival rates without inducing any toxicity but also elicited a significant reduction in the levels and expression of pro-inflammatory cytokines such as TNF-a, IL-1β, IL-6, and prostaglandin E2 (PGE-2). Additionally, it downregulated the expression of COX-2. Interestingly, β-amyrin also modulated the gene expression ratio of arginase-1/inducible nitric oxide synthase (iNOS) and the urea/nitric oxide (NO) ratio, indicating a shift towards an M2 (anti-inflammatory) state in microglial cells.
In their randomized double-blind crossover clinical trial, Herrmann et al. [
69] showcased the efficacy of nabilone, a synthetic cannabinoid derivative (administered at 1-2mg daily for 6 weeks), in addressing restlessness observed in patients with moderate to severe AD, despite standard treatments, when compared with a placebo. Utilizing various assessment tools including the CMAI (with a reduction of b = -4.0), the Neuropsychiatric Inventory-Nursing Home (NPI-NH) (with a reduction of b = -4.6), the Mini-Nutritional Assessment Short-Form (MNA-SF) (with a reduction of b = -1.7), and the Standardized Mini-Mental State Examination (sMMSE) (with an increase of b = +1.1), they observed the superiority of nabilone over placebo. However longer trials are definitely needed.
In a different study [
70], the effect of the synthetic CBR agonist WIN 55,212-2 (
Scheme 4) on astrocytes from the cerebral cortex of rats was investigated, particularly when exposed to the toxic peptide Aβ
1-42. Remarkably, pre-treatment of the cells with 10µM WIN 55,212-2 significantly enhanced their viability while preventing the Aβ
1-42-induced surge in proinflammatory cytokines IL-1β and TNF-a. Concurrently, the application of this agonist notably diminished the expression of the p65 protein and inflammatory proteins COX-2 and iNOS, while increasing the expression of the transcription factor PPAR-γ and the antioxidant enzyme Cu/Zn superoxide dismutase (SOD).
In their study, Mahdi et al. [
71] investigated the therapeutic potential of WIN55,212-2 at varying doses (0.5, 1, 2mg/kg) in an AD rat model, induced by injection of aluminum chloride (AlCl
3) and D-galactose (D-Gal). Notably, on days 4-5, statistically significant differences were observed in escape latency and time spent in the target quadrant among the different groups in the MWM test. Biochemical analyses of their brains revealed a significant reduction in MDA levels, along with a restoration of the antioxidant molecules glutathione (GSH) and SOD to higher levels. Additionally, administration of WIN55,212-2 mitigated cellular abnormalities in the hippocampus of rats by promoting the production of the proteins nestin and glial fibrillary acidic protein (GFAP), which serve as markers of neurogenesis.
In their study, Gajardo-Gómez et al. [
72] investigated the potential of single administrations (5μM) of synthetic and endogenous cannabinoid agonists, including WIN-55,212-2, 2-AG, and methanandamide, to mitigate neuronal death triggered by the opening of astroglial Cx43 hemichannels—an event linked to the excitotoxic release of ATP and glutamate. Their findings revealed that these agonists effectively prevented the increase in the number of surface Cx43 hemichannels, thereby significantly reducing their activity in both astroglia and hippocampal pyramidal cells incubated with Aβ
25-35. Moreover, the same agonists demonstrated a notable decrease in the secretion of glutamate (from 200 to 24, 25 and 27 pmol/mg, respectively) and ATP (from 86 to 12, 13 and 14 pmol/mg, respectively) in pyramidal cells, resulting in reduced rates of pyramidal neuron death. These actions were primarily mediated by CB
1R.
In the study conducted by Soto-Mercado et al. [
73], the action of the non-selective CBR agonist, CP55-940 (
Scheme 4), in PSEN1 E280A cells—a model of familial AD—was elucidated. CP55-940 demonstrated the capability to inhibit both intracellular sAβPPβf aggregation and TAU phosphorylation, while restoring mitochondrial membrane potential (ΔΨm) to normal levels, particularly when used in combination with the CB
1R inverse agonist, SR141716. Simultaneously, this combination effectively curtailed the formation of ROS and suppressed the activation of transcription factors p53 and c-Jun, as well as the expression of p53 upregulated modulator of apoptosis (PUMA) protein and caspase-3, which are markers of apoptosis. However, the combination failed to reverse the dysfunction of ACh-induced Ca
2+ influx, crucial for neuronal function, which was only achieved after co-administration of an anti-Aβ
42 antibody. Thus, the synergistic approach of combining cannabinoids with CB
1R inverse agonists and anti-Aβ
42 antibodies holds promise as a decisive therapeutic strategy for treating familial AD.
The main outcomes of the studies involving non-selective agonists of CB
1R and CB
2R are summarized in
Table 4.