Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2025 Aug 27;62(4):e70229. doi: 10.1111/ejn.70229

Cannabidiol and Alzheimer Disease: A Comprehensive Review and In Silico Insights Into Molecular Interactions

João V Mello‐Hortega 1, Carolina S de Oliveira 1, Vitoria S de Araujo 1, Lupe Furtado‐Alle 1, Luciane V Tureck 1, Ricardo L R Souza 1,
PMCID: PMC12381694  PMID: 40859865

ABSTRACT

Alzheimer's disease (ad) is a neurodegenerative disorder characterized by a set of multifactorial conditions that progressively impair memory processing and cognitive function. The study of this pathology is particularly challenging due to its complex etiology, which involves several pathological hallmarks, including amyloid plaque formation, tau protein hyperphosphorylation, neuroinflammation, oxidative stress, and other contributing factors—all leading to neuronal loss. The primary therapeutic approach for AD involves the use of anticholinesterase agents; however, these treatments are associated with adverse effects, and their efficacy has been increasingly questioned. Against this backdrop, researchers have investigated cannabidiol (CBD) as a potential complementary treatment for AD. This study compiles and synthesizes current evidence regarding the therapeutic effects of CBD in the context of AD, examining its impact on the amyloid cascade, tau phosphorylation, neuroinflammation, oxidative stress, the cholinergic pathway, glucose and lipid metabolism, behavioral alterations, and physiological changes. In addition, an in silico analysis was conducted based on studies that identified differential gene expression in response to CBD. Through this analysis, we mapped the gene network and biological pathways involved in CBD's mechanism of action in ad, contributing to the identification of potential gene targets for further research and providing deeper insight into its therapeutic potential.

Keywords: Cannabinoids, Dementia, Enrichment Pathways, Neuroprotection


Cannabidiol improves several Alzheimer's‐related aspects and regulates disease‐associated genes. Literature review and Over‐Representation Analysis of 64 genes confirm its effects on key mechanisms such as amyloid‐β and tau processing, oxidative stress, inflammation, and lipid metabolism. These findings highlight signaling pathways as targets and support CBD's therapeutic potential in Alzheimer's disease.

graphic file with name EJN-62-0-g004.jpg


Abbreviations

amyloid‐β

AChE

acetylcholinesterase

AD

Alzheimer's disease

AKT

protein kinase 3

APP

amyloid precursor protein

ARRB2

beta‐arrestin‐2

BACH1

BTB and CNC homology 1

BChE

butyrylcholinesterase

BPSD

behavioral and physiological symptoms of dementia

CB1

cannabinoid receptor 1

CBD

cannabidiol

CDK

cyclin‐dependent kinase

CNPq

Conselho Nacional de Desenvolvimento Científico e Tecnológico

CNS

central nervous system

DNM1L

hippocampal dynamin 1 like

ERKs

extracellular signal‐regulated kinases

FAAH

fatty acid amide hydrolase

GFAP

glial fibrillary acidic protein

GSK3

glycogen synthase kinase 3

HMOX1

heme oxygenase 1

HSPs

heat shock proteins

iNOS

inducible nitric oxide synthase

LTP

long‐term potentiation

MAPK

mitogen‐activated protein kinase

NRF2

nuclear factor erythroid 2–related factor 2

ORA

over‐representation analysis

PET

positron emission tomography

PPAR‐γ

peroxisome proliferator‐activated receptor gamma

RNS

reactive nitrogen species

ROS

reactive oxygen species

TREM2

triggering receptor expressed on myeloid cells 2

TRPV2

transient receptor potential cation channel subfamily V member 2

1. Introduction

With the increase in life expectancy worldwide aided by modern medicine, dementia has emerged as a global health challenge. In 2022, it is estimated that 55.2 million individuals will be affected globally, with 60% to 80% of cases triggered by Alzheimer's disease (ad) (Prince 2015; “2022 Alzheimer's disease facts and figures” 2022).

ad is a neurodegenerative disease that results in a set of disorders that progressively affect functions related to memory capacity, clear reasoning, and can affect the functional performance of activities considered complex (Corey‐Bloom et al. 1995; Abreu et al. 2005; Williams et al. 2010).

The development of ad can be influenced by factors such as genetic variants, aging, smoking, vascular problems, obesity, among others (Silva et al. 2019; Zhang et al. 2021). The genetic factors associated with the disease include variants in the amyloid precursor protein (gene APP) and the presenilin 1 and 2 genes (PSEN1 and PSEN2), which are commonly linked to early‐onset or familial ad, while late‐onset ad is primarily associated with variants in the gene encoding apolipoprotein E (APOE) (Tanzi 2012; Pereira et al. 2021).

In addition to the factors that may influence the development of ad, different pathological pathways are observed as the disease progresses, contributing to the complexity associated with this condition. Despite efforts to understand it, studies have identified various pathological hallmarks that lead to neuronal loss. Among them, the amyloid‐β (Aβ) and tau protein pathways are the most described and well established in association with ad. In the ad brain, the exacerbated expression of Aβ proteins results in accumulation between neurons, challenging the proper functioning of synapses and resulting in cognitive deficits. In addition to the formation of Aβ plaques, there are also neurofibrillary tangles, caused by abnormal expression and tau protein hyperphosphorylation (Reitz et al. 2011). This protein plays an important role in maintaining neuronal structure and, when abnormal, clusters in a disorganized way inside cells, disrupting tissue homeostasis. These processes are the most commonly AD pathological hallmarks, being one of the main diagnostic criteria for the pathology (Mariani 2004).

Neuroinflammation and oxidative stress are other processes that appear to mediate the development of AD, resulting in the activation of microglia and astroglia as a form of defense of brain tissue. These mechanisms are mediated by the release of pro‐inflammatory cytokines and the excessive production of reactive oxygen species (ROS) that can cause neuronal death and the consequent cognitive deficit observed in AD (Klegeris et al. 1994; Meda et al. 1995; Hashioka et al. 2021). In addition to these mechanisms, other hypotheses have contributed to a better understanding of the pathology. Notable among them are the metal ion hypothesis, the abnormal autophagy hypothesis, the microbiota–gut–brain axis hypothesis, the glutamate excitotoxicity hypothesis, and the cholinergic hypothesis (Zhang et al. 2024).

The cholinergic hypothesis plays a relevant role in the treatment of patients, since neurochemical changes are observed in brains diagnosed with ad, such as a decrease in the enzyme responsible for the production of acetylcholine in brains with the presence of neuritic plaques (Perry et al. 1978; Cummings and Back 1998; Hampel et al. 2018). Therefore, currently one of the only palliative treatments for ad consists of cholinesterase inhibitors use, which act on this system by blocking the cholinesterases, responsible for metabolizing acetylcholine, aiming to increase neurotransmitter levels in synaptic clefts, thus enabling better communication between neurons (Weinreb et al. 2009). However, this method presents undesirable side effects, such as nausea, circulatory problems, dizziness, insomnia, among others, and its effectiveness has been questioned in some studies, highlighting the need for the development of alternative therapeutic approaches (Hampel et al. 2018; Han et al. 2019; Xu et al. 2021).

Considering the need for the development of new therapeutic approaches, many substances have already been tested in pre‐clinical and clinical studies for ad treatment (Li et al. 2023; Smith and Ownby 2024). Among these substances, cannabidiol (CBD) stands out, as it is a common cannabinoid that does not produce psychodysleptic effects and exhibits anti‐inflammatory and antioxidant properties, among other effects (Hampson et al. 1998; Woelfl et al. 2020). Due to its therapeutic potential, many studies have sought to evaluate the effects of CBD in the context of AD, as well as to identify the pathways through which this cannabinoid exerts its action.

In this context, the aim of the present study was to obtain a comprehensive understanding of CBD therapy in ad and to identify the main biological pathways affected by treatment with this cannabinoid, through an in silico analysis based on genes modulated by CBD. Thus, we sought to gather information on the potential therapeutic applications of CBD in ad and to provide insights into the molecular interactions underlying its effects on the disease.

2. Methodology

To provide a comprehensive overview of CBD therapeutics, a narrative review was conducted based on searches for scientific papers in the PubMed and Web of Science databases, using the keywords “Cannabidiol” and “Alzheimer's.” At this stage, no filters were applied regarding the year of publication or article type.

The initial search retrieved 144 papers, which were subsequently screened to exclude review articles or those that only mentioned the search terms without directly addressing the topic. After this filtering process, 45 original research articles were selected to examine the effects of CBD on the pathogenesis of ad (Supplementary Table S1). These studies encompass a wide range of evidence from in vivo, in vitro, and in silico models, highlighting various biological pathways modulated by CBD. An in silico analysis was conducted with the aim of mapping the interactions between genes and CBD and subsequently identifying the biological pathways modulated by the gene products affected by this compound. For this purpose, we selected from the 45 reviewed studies those that evaluated gene expression in response to CBD treatment using RNA‐seq or microarray technologies. From this selection, two articles (Aso et al. 2014; Libro et al. 2016) were used to generate a gene set consisting of 64 genes modulated by CBD treatment (see Supplementary Table S2). This list includes all differentially expressed genes listed in these works related to ad. These studies were chosen for pathway enrichment analysis because they provided gene lists associated with ad and modulated by CBD treatment, obtained through hypothesis‐free analytical methods. This approach reduces bias and increases the reliability of identifying relevant biological pathways.

An Over‐Representation Analysis (ORA) was then performed using the clusterProfiler package (Yu et al. 2012) in R, with the KEGG gene set database. This technique evaluates whether specific biological pathways or processes are over‐represented or enriched in a list of experimentally identified genes compared to what would be expected by random chance. The parameters set were as follows: pvalueCutoff = 0.05, qvalueCutoff = 0.05, universe = org.Hs.eg.db, minGSSize = 10, maxGSSize = 500.

3. Results and Discussion

3.1. CBD and AD Pathways

Findings in the literature demonstrate the beneficial effects of CBD for the treatment of ad. This evidence was grouped into nine categories that contemplate the effects of this cannabinoid reported in the literature, represented in Figure 1.

FIGURE 1.

FIGURE 1

Alzheimer's disease (ad) factors affected by cannabidiol (CBD) treatment: production and maintenance of beta amyloid, phosphorylation of tau protein, oxidative stress, neuroinflammation, cholinergic system, insulin resistance and lipid metabolism, brain glucose metabolism, behavioral changes, and physiological changes.

Among the 45 articles analyzed, different study methods were applied in order to better understand the effects of CBD on ad. In these works, outcomes were identified that demonstrate the beneficial effect of CBD in the treatment of ad. Studies involving analysis of mechanisms of the amyloid pathway, oxidative stress, and neuroinflammation were the most abundant, as well as behavioral analysis in in vivo models treated with CBD. The proportion of outcomes related to each category of the disease addressed can be seen in Figure 2, as well as the proportion of the type of analysis used for these inferences.

FIGURE 2.

FIGURE 2

Proportion of outcomes of the effects of CBD in AD therapy separated by categories and type of study. The analysis of the 45 papers found in the literature made it possible to gather a set of 61 outcomes that experimentally demonstrate the beneficial effect of CBD treatment in AD therapy. (a) Proportion of outcomes of the effects of CBD in AD therapy separated by categories, such as: production and maintenance of Aβ, phosphorylation of tau protein, oxidative stress, neuroinflammation, cholinergic system, insulin resistance and lipid metabolism, brain glucose metabolism, behavioral changes, and physiological changes. Only percentages > 5% are shown in the figure. (b) Proportion of study type by category, separated by in vivo, in vitro and in silico studies.

3.1.1. CBD in Aβ Production, Aggregation, and Immune Response

The Aβ pathway is well recognized in ad and most studies have evaluated CBD effects on the production and accumulation of the proteins and the immune response linked to the presence of these aggregates.

A study using mesenchymal stem cells found, after the treatment with CBD, lower expression of genes encoding for secretases, enzymes responsible for cleaving the APP, resulting in the formation of smaller Aβ fragments (De Strooper et al. 2010; Libro et al. 2016). CBD was described as a potential inhibitor of the β‐secretase activity (Vallée et al. 2017). In relation to other mechanisms of Aβ production, the regulation of the Wnt/β‐catenin pathway and Peroxisome Proliferator‐Activated Receptor Gamma (PPAR‐γ) also appear to be affected by CBD treatment, resulting in decreased production of the Aβ precursor; besides that, CBD acts as an inverse agonist, inhibiting the beta‐arrestin‐2 (ARRB2) recruitment and also in the ubiquitination of APP, decreasing Aβ production (Scuderi et al. 2014; Vallée et al. 2017; Woelfl et al. 2020).

In addition to acting on the Aβ production, CBD has an important effect on the accumulation of these proteins, which form the protein aggregates. Several studies using in vivo models have demonstrated the role of CBD in reducing processes related to the Aβ accumulation, such as decreasing levels of soluble Aβ and reduced deposition in the hippocampus and cerebral cortex (Casarejos et al. 2013; Aso et al. 2014; Watt et al. 2020; Amini and Abdolmaleki 2022; Zhang et al. 2022). An in silico study reinforces the idea of CBD's regulatory role in reducing the aggregation of these peptides at certain concentrations of use (Chrobak et al. 2021). One of the hypotheses for the reduction in protein aggregation is related to CBD's ability to scavenge ROS. This process may be related to the protective function of the Aβ protein, so that, with the reduction in stress caused by ROS, the signaling for this protein production is reduced (Zhang et al. 2022).

Still in relation to these aggregates, but now considering the effects associated with its presence, the major impact is on neuronal functioning; but also, immune response aiming at re‐establishing tissue homeostasis is closely associated. In this context, microglia play an important role in the response to ad pathology. Microglia comprise a group of cells in the central nervous system responsible for the maintenance and defense of neurons. Activation of these cells is commonly due to infections, injuries, and neurodegenerative diseases (Prinz et al. 2019; Cserép et al. 2020). According to the functions performed, microglia play an important role in combating the protein aggregates. Studies have shown that CBD decreases the expression of genes related to specific actions of microglia. These actions are related to the promotion of an immune response through pathways related to autophagy‐related process and phagocytosis, which occur through the activation of the transient receptor potential cation channel subfamily V member 2 (TRPV2), in order to reduce the neurotoxicity promoted by Aβ (Hao and Feng 2021; Yang et al. 2022; Duan et al. 2023; Marsh et al. 2023).

3.1.2. CBD Modulates Tau Protein Aggregation and Phosphorylation

The tau protein pathway is a well‐established ad pathway. Studies have already shown that CBD has a beneficial effect in reducing tau aggregation, since this compound spontaneously binds to the protein, hindering the process of protein accumulation and deposition in the cortex and hippocampus (Casarejos et al. 2013; Alali et al. 2021). Regarding protein phosphorylation, in vitro studies have shown the influence of CBD on the downregulation of genes encoding kinases, enzymes responsible for the phosphorylation of various molecules, including the tau protein. An important kinase in the tau protein phosphorylation pathway is glycogen synthase kinase 3 (GSK3), which phosphorylates tau on most of the hyperphosphorylated serine and threonine residues in paired helical filaments; GSK3 activity contributes to both Aβ production and Aβ‐mediated neuronal death. This enzyme is regulated by several mechanisms, including the Wnt signaling pathway mediated by β‐catenin, which explains other results found in studies that indicate a regulatory role of CBD in this signaling pathway, decreasing the phosphorylation of the tau protein (Esposito, De Filippis, Carnuccio, et al. 2006; Wu and Pan 2010; Hernandez et al. 2012; Aso et al. 2014; Libro et al. 2016).

3.1.3. CBD as a Modulator of Neuroinflammation

Neuroinflammation is another important mechanism related to the ad pathogenesis, especially in the development and worsening of associated dementia (Zhang et al. 2013). Given the CBD mechanisms, several researchers have made efforts to verify inflammation‐related therapeutic effects in models of the disease (Esposito, De Filippis, Carnuccio, et al. 2006; Esposito et al. 2007, 2011; Martín‐Moreno et al. 2011; Khodadadi et al. 2021; Wang et al. 2021; Landucci et al. 2022). Some associations have been identified between CBD treatments and the activation of microglia, a process in which these cells switch from their “resting” state (due to disturbances in brain homeostasis) to an active state to counteract these disturbing factors (Kettenmann et al. 2011).

Activated microglia, regarding in vitro models, can be classified in two ways: the M1 phenotype presenting pro‐inflammatory and neurotoxic activity; the M2 phenotype plays a role in attenuating the inflammatory process by releasing anti‐inflammatory cytokines and growth factors, eliminating debris by inducing phagocytic activity, and remodeling the tissue (Cherry et al. 2014; Hu et al. 2015; Orihuela et al. 2016; Simon et al. 2017). An in vitro study demonstrates the effect of CBD in preventing the transition of microglia from the M0 (resting) stage to the M1 stage, blocking the activation of these cells and reducing the pro‐inflammatory effect on the disease (Landucci et al. 2022).

CBD acts on decreasing the release of cytokines (such as proinflammatory interleukins) and other proteins such as inducible nitric oxide synthetase (iNOS). The latter releases nitric oxide in certain stress conditions, which can result in the release of reactive nitrogen species, causing further damage to the tissue (Esposito, De Filippis, Maiuri, et al. 2006; Esposito et al. 2007; Martín‐Moreno et al. 2011; Khodadadi et al. 2021). In addition to iNOS, glial fibrillary acidic protein (GFAP) also had its expression decreased with CBD treatment (Esposito et al. 2007). In a study that evaluated neuroinflammation and the expression of this protein in rats subjected to a hypercaloric diet, an exacerbated GFAP expression was related to the impairment in short‐ and long‐term memory (Bondan et al. 2019). This indicates a possible relationship between the expression of GFAP and the deficiency in memory processing observed in ad.

Corroborating the anti‐inflammatory effects through the molecules expression inhibition, in studies based on the Caenorhabditis elegans ad model, CBD promoted greater expression of the gene encoding the triggering receptor expressed on myeloid cells 2 (TREM2); in order to increase the phagocytic response in the tissue, as well as increasing the expression of fatty acid amide hydrolase (FAAH) and the cannabinoid receptor 1 (CB1) (Esposito et al. 2007; Khodadadi et al. 2021; Wang et al. 2021).

In addition to activating and modulating microglia, CBD also has an anti‐inflammatory effect through the PPAR‐γ receptor. This receptor is overexpressed in patients diagnosed with ad and acts in the regulation of lipid and insulin metabolisms, demonstrating anti‐inflammatory effects in other types of central nervous system (CNS) diseases (Landreth et al. 2008). (Landreth et al. 2008). An in vivo ad study model showed that CBD acted as an agonist of the PPAR‐γ receptor, reducing the inflammatory state, consequent neuronal damage, and has been shown to promote hippocampal neurogenesis (Esposito et al. 2011).

3.1.4. Antioxidant Effects of CBD and Modulation of Oxidative Stress

Like neuroinflammation, oxidative stress has been increasingly studied in the context of ad, and it consists of dysregulation in the production of ROS, reactive nitrogen species (RNS), and antioxidant defense mechanisms. Due to this imbalance, these unstable and reactive molecules can transform other molecules into which they collide, causing detrimental effects (Halliwell and Gutteridge 2015). This damage has been linked to the worsening of ad‐related dementia, making oxidative stress‐promoting pathways a promising target for combating the progression of the disease (Halliwell and Gutteridge 2015).

Some studies have already shown a positive effect of CBD in regulating pathways related to the production of free radicals, reducing ROS without inducing the overexpression of oxidative genes (Iuvone et al. 2004; Esposito et al. 2011; Casarejos et al. 2013; Scuderi et al. 2014; Frandsen and Narayanasamy 2022; Vanin et al. 2022; Wang et al. 2023). In a C. elegans model, a preventive CBD effect on cell damage caused by methylglyoxal, a reactive compound resulting from the glyoxalase neural pathway, was found (Frandsen and Narayanasamy 2022). In PC12 and SH‐SY5Y cell lines, a beneficial nature of CBD was observed in relation to cell viability in response to Tert‐Butyl Hydroperoxide, an oxidizing compound (Harvey et al. 2012). CBD also has a role in reducing oxidative stress related to the dopamine metabolism pathway, the Wnt/β‐catenin pathway, and the PPAR‐γ receptor (Scuderi et al. 2014; Vallée et al. 2017). Furthermore, studies using a CBD derivative report its action on the axis of the transcription factor NF‐E2‐related factor 2 (NRF2) and the transcription factor BTB and CNC homology 1 (BACH1). This derivative acts by inhibiting the repressive action of BACH1 in the promoter region of the gene responsive to oxidative stress, heme oxygenase 1 (HMOX1), conferring an antioxidant and cytoprotective effect on cells (Casares et al. 2020).

The antioxidant role of CBD, in addition to acting directly on pathways that produce reactive molecules, also plays an important part in modulating genes related to mitochondrial dynamics (Iuvone et al. 2004; Harvey et al. 2012; da Silva et al. 2014; Rajan et al. 2017). This cannabinoid was able to rescue effects caused by the induction of damage using iron in a model of neurodegenerative diseases using rats with brain iron overload, recovering normal levels of hippocampal dynamin 1 like (DNM1L), caspase 3, and synaptophysin levels (da Silva et al. 2014). The DNM1L gene is responsible for encoding Dynamin‐1, a protein active in the process of mitochondrial fission that has its expression negatively affected in the studied model. In this sense, in addition to CBD returning the protein's basal expression levels, it also decreased the expression of caspase 3, which plays a role in apoptosis, and decreased the expression levels of synaptophysin, a protein that presents high levels of expression and is associated with low scores in tests that evaluate neuropsychological measures in patients with AD (Sze et al. 1997; da Silva et al. 2014; Lee et al. 2017).

3.1.5. CBD Modulates the Cholinergic System

In ad, the cholinergic system comprises an important mechanism. In this pathway, there are two important cholinesterase enzymes, butyrylcholinesterase (BChE) and acetylcholinesterase (AChE); the latter is more expressed in the brain and regulates brain synapses by acetylcholine hydrolysis. Dysfunctions in the activity of these enzymes are commonly described in studies using ad models, such as a report of animals induced to develop a dementia‐like condition that resembles ad, through the cerebroventricular infusion of streptozotocin (Silva et al. 2023). The association between the disruption of the cholinergic system disturbance and ad has increased since significant symptomatic improvement has been observed in patients with AD treated with cholinesterase inhibitors (Summers et al. 1986). However, although still in use today, drugs that inhibit these enzymes have significant adverse reactions and only act to slow down the progression of the disease (Hampel et al. 2018; Han et al. 2019; Xu et al. 2021).

Given the necessity to formulate new treatment strategies, CBD has emerged as an interesting possibility not only for its neuroprotective effects, but also for its anticholinesterase properties. This hypothesis is supported by studies showing a decrease in AChE activity after treatment with CBD in cell culture, C. elegans , and Zebrafish models (Mooko et al. 2022; Vanin et al. 2022). In addition to proving the effect of CBD on AChE, the main cholinesterase enzyme related to the modulation of brain cholinergic synapses, treatments based on this cannabinoid also had an effect on decreasing BChE activity (Jiang et al. 2021; Patil et al. 2023).

3.1.6. CBD Effects on Glucose and Lipid Metabolism

Other pathways related to AD include glucose and lipid metabolism. In Alzheimer's brains, positron emission tomography (PET) scans indicate low glucose metabolism. The effect of intraperitoneal CBD treatment on glucose metabolism in AD brains was first described in a study using PET imaging and rats induced to develop the disease by the use of streptozotocin, in which it was concluded that CBD maintains regular glucose metabolism. At the same time, untreated animals showed regions of hypometabolism near the striatum, motor cortex, hippocampus, and thalamus, further proposing the feasibility of CBD treatment as a possible method of early treatment for dementia (de Paula Faria et al. 2022).

In the context of lipid metabolism, insulin, an important regulator of lipid storage and processing in organisms, has an influence on the development of AD, since insulin resistance can lead to oxidative stress and inflammation (De Felice et al. 2014; Kandimalla et al. 2017). In vivo and in vitro studies indicate CBD as a reducing agent of cerebral insulin resistance, mainly by modulating the de novo synthesis of ceramides and acting on salvage pathways. In addition, results obtained in these studies reinforce the antioxidant role of CBD, since the cannabinoid decreased lipid peroxidation and has a protective effect through the decrease in tau protein phosphorylation (Charytoniuk et al. 2021; Vanin et al. 2022).

3.1.7. CBD‐Induced Functional and Cognitive Improvements

In summary, CBD has numerous molecular interactions in important pathways related to AD. The result of this complex network of interactions can be seen as the basis for the broad physiological changes resulting from treatment with this compound. In humans, it was observed that treatment with CBD increased cerebral blood flow in key regions involved in processing memories, such as the hippocampus (Bloomfield et al. 2020). Another study, carried out on an in vitro model, indicated that pre‐treatment with CBD rescued the deficit caused by Aβ in long‐term potential (LTP), a phenomenon involved in signal transmission between two neurons, thereby stimulating them (Hughes and Herron 2019). LTP is one of the parameters related to synaptic plasticity and is believed to act in the encoding of memories in the brain, so it is considered the basis for learning and memory processes (Bliss and Collingridge 1993; Cooke 2006).

Regarding the changes in clinical aspects resulting from CBD administration, many reports address the drug's ability to improve conditions related to cognition and memory processing. Study models of ad, using rats and mice treated with CBD for different periods and with various concentrations, showed positive results in terms of memory processing and exploratory behavior, compared to control animals (Aso et al. 2014, 2016; Cheng, Low, et al. 2014; Cheng, Spiro, et al. 2014; Watt et al. 2020; Amini and Abdolmaleki 2022; Chesworth et al. 2022; Coles et al. 2022; de Paula Faria et al. 2022; Kreilaus et al. 2022). In addition to animal models, one research study evaluated the effect of a treatment based on the daily use of 3% CBD on human patients listed in the Alzheimer Hellas database with severe behavioral and physiological symptoms of dementia (BPSD). In this analysis, the CBD‐treated group showed a significant improvement in BPSD, compared to the conventional treatment group, making CBD a possible treatment method for these conditions as soon as it undergoes further randomized clinical trials (Alexandri et al. 2023).

Investment in studies aimed at understanding the diverse effects of CBD on aspects of ad has provided a wide range of information on the mechanisms related to this therapeutic approach. A synthesis of these findings can be seen in Figure 3, which demonstrates the multiple mechanisms of action of this compound, offering several possibilities for the investigation of new treatment methods based on CBD.

FIGURE 3.

FIGURE 3

Mechanisms of action of cannabidiol in Alzheimer's disease. Schematic representation of the effects of CBD associated with AD found in the literature review.

3.2. In Silico Identification of Key Therapeutic Pathways Modulated by CBD

In ORA, five KEGG pathways were identified: AD (H00056 code in the database), lipid and atherosclerosis pathways (map 05417), neurotrophin signaling pathway (HSA04722), pathways of neurodegeneration—multiple diseases (HSA05022), and Shigellosis (HSA05131). The relationships between genes and pathways can be seen in Figure 4.

FIGURE 4.

FIGURE 4

Enrichment network of KEGG pathways related to the Alzheimer disease. Obtained through the analysis of a set of 64 genes modulated by CBD treatment, obtained from the literature, in an Over‐Representation Analysis (ORA) using the clusterProfiler package in R and the KEGG gene set. The defined parameters were as follows: pvalueCutoff = 0.05, qvalueCutoff = 0.05, universe = org. Hs.eg.db, minGSSize = 10, maxGSSize = 500.

3.2.1. CBD Modulates Key Genes in Aβ Formation and Neurodegeneration Pathways

The AD category is related to the mechanisms of pathology development, including genes and variants associated with processes of apoptosis, autophagy, impaired neurotransmission, mitochondrial abnormalities, impaired synaptic plasticity, and neurodegeneration. Some of these genes are associated with the production of Aβ, such as PSEN1 and PSEN2, genes strongly associated with early‐onset ad (Do et al. 2023). These genes encode presenilins, proteins that constitute the γ‐secretase protein complex, which processes type I transmembrane proteins, such as the Aβ precursor protein (Hutton 1997; Fraser et al. 2000). Thus, as indicated in the work of Libro et al. (2016), the decrease in the expression of these genes and others related to the formation of the γ‐secretase complex—such as aph‐1 homologue A (APH1A), presenilin enhancer (PSENEN), nicastrin (NCSTN), as well as beta‐secretase 1 (BACE1), which encodes the β‐secretase enzyme also related to the production of Aβ—indicates that treatment with CBD may lead to a decrease in the formation of Aβ plaques and have a beneficial effect in the AD therapeutics (Fraser et al. 2000; Kitazume et al. 2001; Libro et al. 2016).

Genes of the AD pathway linked to the neurodegeneration pathway were identified as well. This category comprises processes that include abnormal protein dynamics due to factors such as deficiency in the ubiquitin‐proteasome‐autophagy system; oxidative stress and free radical formation; endoplasmic reticulum stress; mitochondrial dysfunction; and axonal transport interruptions. In addition, other gene families can be cited as a common point between these pathways, such as the class of protein subunits encoded by the PSMB genes. They take part in the formation of the proteasome complex, which has dysfunctions related to oxidative stress and the development of AD (Bonet‐Costa et al. 2016; Moya‐Alvarado et al. 2016).

Other genes modulated by CBD and related to both pathways are the calpain 1 (CAPN1) and 2 (CAPN2) genes, which encode calcium‐activated intracellular proteins whose increased activity is related to cognitive disorders, as in the case of AD, and the cyclin dependent kinase 5 (CDK5) and cyclin dependent kinase 5 regulatory subunit 1 (CDK5R1) genes (Saito et al. 1993; Ferreira and Bigio 2011). Unlike other genes in the cyclin dependent kinase (CDK) family, which are related to cell cycle regulatory proteins, the CDK5 gene encodes a cyclin‐dependent protein kinase that is highly expressed in neurons of the CNS. Its main function is to act on synaptic plasticity and neuronal migration through the synaptic phosphorylation of proteins needed for the organization of the cytoskeleton, endocytosis, exocytosis, and apoptosis (Liu et al. 2016). A study investigating the modulation of CDK proteins in cases of AD shows increased expression of these proteins in the disease condition (Lim et al. 2021). Therefore, since CBD decreases the expression of these proteins, it may indicate a protective effect in the AD context.

3.2.2. CBD's Impact on Lipid‐Related and Chaperone‐Mediated Pathways

The lipids and atherosclerosis category is characterized by processes related to lipid metabolism and atherosclerosis, a chronic inflammatory disease caused by the accumulation of lipid plaques on artery walls, leading to complications involved in cardiovascular diseases. Among the genes related to this pathway, some demonstrate links with a greater number of pathways and consequent cumulative action in ad, others present a punctual action. This is the case of genes from the CAMK2 family and genes related to the activity of heat shock proteins (HSPs). These proteins are part of a group of chaperones that regulate the folding, signaling, monitoring, and maintenance of cellular proteins, conferring a protective effect on the cells of the nervous system in cases of neuropathology (Macario and De Conway Macario 2007; Marino Gammazza et al. 2016). Studies have shown an important role for HSPs in inhibiting Aβ aggregation, in binding to ubiquitin, and in protein degradation, including that of Aβ and tau (Walls et al. 2012; Mateju et al. 2017; Meriin et al. 2018). Thus, the findings related to the upregulation of these genes with CBD treatment may indicate an interesting strategy to regulate pathways associated with ad development (Libro et al. 2016).

Moreover, the CAMK2 gene family was identified in this pathway, which includes some of the protein kinases related to the phosphorylation of tau protein in ad. In the context of atherosclerosis, CAMK2 members act in the regulation of inflammatory processes, since they promote the release of pro‐inflammatory proteins (Liu et al. 2008), which is a hallmark condition in the pathology of atherosclerosis. One of the induction pathways for inflammatory reactions occurs through the triggering of a metabolic cascade that results in the release of interleukins involving the activation of mitogen‐activated protein kinase (MAPK) (Zhang et al. 2011). Also named extracellular signal‐regulated kinases (ERKs), MAPKs play a fundamental role in the communication and integration of various biochemical signals within cells. Their action extends to a wide range of cellular processes, including proliferation, differentiation, transcription regulation, and development. After activation, MAPKs migrate to the cell nucleus, where they exert their function by phosphorylating various nuclear targets. This phosphorylation triggers metabolic cascades regulated by the action of these proteins (Blum and Dash 2009; Cargnello and Roux 2011). This family of proteins is encoded by numerous genes in which CBD has a modulating function, as well as playing a role in various pathways related to the development of ad. One such example is MAP 2K2, which is associated with the development of AD, pathways of neurodegeneration, and the neurotrophin signaling pathway (Aso et al. 2014).

3.2.3. CBD Regulation of Neurotrophin, Ubiquitination, and Kinase Signaling Pathways

The neurotrophin signaling pathway, despite being related to the fewest genes compared to the others, was genes closely linked to at least two other pathways in the analysis carried out. This pathway includes mechanisms related to neurotrophins, a family of trophic factors involved in the differentiation and maintenance of neuronal cells. The regulation of these mechanisms is related to various regulatory cascades, which include signaling pathways mediated by kinases. Among the genes that represent the pathway are those encoding presenilins, mentioned previously, and proteins of the MAPK family, which are negatively modulated by treatments with CBD, as well as other related genes encoding other kinases such as phosphatidylinositol 3 (PIK3C) and human serine–threonine kinase (AKT) (Aso et al. 2014; Libro et al. 2016).

In addition to MAP 2K2, other genes from the MAPK family are modulated by the effect of CBD and are related to ad. The MAPK12 and MAPK14 genes have been linked to pathways involved in neurodegeneration, neurotrophin signaling, lipids, and atherosclerosis and the pathway related to Shigellosis. Shigellosis, or bacillary dysentery, is an intestinal infection caused by Shigella, a genus of enterobacteria. Shigella are potential food‐borne pathogens that are able to colonize the intestinal epithelium, exploiting the functions of epithelial cells and bypassing the host's innate immune response (Echeverria et al. 1991). The enrichment of this pathway in our functional analysis was not expected in the context of ad. This finding could be explained by the effect of CBD on genes related to ubiquitination processes, which correspond to some of the genes associated with the Shigellosis pathway. Ubiquitination consists of a post‐transcriptional modification mediated by proteins that insert an ubiquitin into a target protein, which leads to different processes, such as protein degradation, subcellular localization, and kinase activation (Callis 2014). The ubiquitination mechanism is crucial in the process of Shigella infection, as the bacterium exploits the host's ubiquitin‐proteasome system via bacterial ubiquitin ligases. This facilitates the targeted degradation of proteins involved in the host's immune defense, thereby aiding in the establishment and progression of the infection (Tanner et al. 2015; Wandel et al. 2017).

Since ad involves the abnormal accumulation of Aβ proteins and a large part of the protein clearance mechanisms depends on ubiquitination, this pathway plays an important role (Glickman and Ciechanover 2002; McKinnon and Tabrizi 2014; Tramutola et al. 2016; Zhang et al. 2017). Based on this mechanism, a study proposes a three‐dimensional model for studying ad in human neural cell cultures induced by alterations in the ubiquitin signaling pathway (Maniv et al. 2023). In this case, treatments that result in the positive modulation of genes related to the ubiquitin signaling pathway, like CBD, may reduce the protein accumulation in affected brains.

The regulation of signaling hubs seems to be a good target for understanding and treating the pathology involved in ad and CBD has shown promising effects in this regard. Regulatory mechanisms associated with the action of neurotrophins, ubiquitins, and signaling related to protein kinases were studied. This must be highlighted since the genes related to these mechanisms interact with all the addressed pathways found in the analysis. As an example, we have the MAPK1 gene, which plays a role in all the pathways shown in Figure 2.

The PIK3CA and PIK3CB genes are related to the expression of the PIK3 protein. The activity of this protein, together with the action of the protein kinase 3 (AKT), one of whose related genes is AKT1, participates in the important PIK3/AKT signaling axis with a wide range of functions in the brain, regulating cell survival, proliferation, growth, differentiation, motility, intracellular traffic, and the extension of neurites (dendrites and axons) (Vanhaesebroeck et al. 2010; Ye et al. 2019). In the AD brain, accumulated Aβ prevents the propagation of the PIK3/Akt signaling axis and increases the activity of this protein kinase in neurons, eliminating the suppressive effect of this pathway on GSK‐3. In addition to the indirect impact on GSK‐3, Aβ oligomers could also directly stimulate this kinase in neurons and neuron stem cells. In turn, activated GSK‐3 could increase apoptotic signals and decrease cell survival capacity. This enzyme also plays a profound role in regulating tau hyperphosphorylation. In fact, activation of the PI3K/Akt signaling axis in neurons could reduce tau hyperphosphorylation by suppressing GSK‐3 activity (Ariga et al. 2008; Choi and Ho Koh 2016; Ryu et al. 2016; Ren et al. 2018). In addition to this mechanism, the PIK3/Akt signaling axis plays a regulatory role in pathways related to glucose and insulin metabolism, changes in autophagy processes, oxidative stress, and neuroinflammation. A review by Razani et al. (2021) provides a robust and detailed summary of the molecular mechanisms that drive the effects of this pathway in AD brains (Razani et al. 2021).

Even though more studies are needed to reveal CBD's modulating effect at a systemic level, therapeutics based on CBD show interesting relevance due to its potential to modulate genes related to important signaling pathways, such as those of the PIK3 family, AKT1, and GSK3β (Aso et al. 2014; Libro et al. 2016).

In silico analysis identified important pathways involved in ad and related mechanisms, such as neurodegeneration, inflammatory processes, and neurotrophic signaling. The genes regulated by CBD treatment are related to pathological mechanisms, such as the accumulation of Aβ plaques, mitochondrial dysfunction, and oxidative stress, reinforcing the beneficial effect of this cannabinoid at the molecular level. In addition, CBD treatment demonstrated promising effects by regulating the expression of crucial genes, such as those linked to the γ‐secretase complex and protein kinase signaling, suggesting a possible reduction in Aβ plaque formation and tau phosphorylation. Thus, CBD may play a protective role by regulating pathways such as ubiquitination, PIK3/AKT signaling, and lipid metabolism, reinforcing its therapeutic potential for ad. Although further studies are needed, CBD presents itself as a relevant therapeutic alternative due to its ability to genetically modulate key ad processes.

3.3. CBD as a Complementary Method for the Treatment of ad

With the data presented in this study, the beneficial effects of CBD on key pathological pathways of ad are evident. However, when considering the implementation of a new therapeutic approach, special attention should be paid to adverse effects, optimal dosing strategies, and long‐term safety in clinical settings.

In this sense, despite promising preclinical findings, current clinical evidence for CBD remains limited. Most studies to date have used heterogeneous formulations and small sample sizes, often combining CBD with other cannabinoids such as THC, which makes interpretation of results difficult (Singh et al. 2023; Martiniano et al. 2024). The lack of randomized phase III clinical trials specifically focused on ad represents a significant barrier to clinical translation.

In addition, although CBD is generally considered well tolerated, safety concerns have been raised, particularly at high doses or in combination with other medications. A meta‐analysis demonstrated a dose‐dependent increase in the risk of elevated liver enzymes and drug‐induced liver injury (Lo et al. 2023). Furthermore, CBD inhibits several cytochrome P450 enzymes, increasing the potential for significant pharmacokinetic interactions, especially in elderly patients on polypharmacy regimens (Huestis et al. 2019).

In contrast, conventional therapies such as acetylcholinesterase inhibitors are supported by a solid clinical evidence base. Studies have confirmed their modest but consistent cognitive benefits (Ritchie 2004; Moreta et al. 2021). However, they are often associated with adverse effects, particularly gastrointestinal and cardiovascular complications, including bradycardia, atrioventricular block, and increased risk of hospitalization, especially in the first months of use (Gauthier 2001; Ruangritchankul et al. 2021). In this context, CBD may be better positioned as a complementary therapy rather than a replacement for established treatments. Its multimodal mechanisms—anti‐inflammatory, antioxidant, and receptor modulation—combined with a favorable safety profile at therapeutic doses, suggest its potential for integration into combination regimens (Fernández‐Ruiz et al. 2013; Noreen et al. 2018). This may be particularly beneficial for patients who are intolerant to cholinesterase inhibitors or who have neuropsychiatric symptoms not well controlled by current medications. To expand this potential, more clinical research is needed to define optimal dosing regimens, evaluate long‐term outcomes, and establish safety parameters in elderly and comorbid populations. Regulatory challenges and standardization of formulations must also be addressed to enable safe and effective clinical use of CBD in ad.

4. Conclusion

To the authors' knowledge, this is the first work to collectively address the key findings about the role of CBD in ad, adding in silico enrichment analysis to provide a comprehensive overview of the genes whose expression levels are modulated by CBD treatment.

The bibliographic analysis presented a rich understanding of CBD's action mechanisms. The results point to a broad action of CBD, with effects that can be positive in several aspects of ad, such as reducing the production and aggregation of amyloid beta protein, as well as decreasing the hyperphosphorylation of tau protein. CBD is also capable of decreasing the effect of AChE and BChE, a mechanism used to decrease the progression of the disease through other anticholinesterases, as well as regulating processes associated with glucose and lipid metabolism, also decreasing the tissue inflammatory response and oxidative stress. The combination of these effects related to CBD treatment may explain the behavioral improvement observed in in vivo studies and the beneficial physiological changes reported in the literature. Therefore, even though most of the explored studies are in a pre‐clinical scope, these data support a promising future regarding CBD‐based treatments for patients with AD.

The in silico analysis performed corroborates the idea of the positive effect of CBD for the treatment of ad. The identification of key ad pathways, such as inflammatory processes, signaling mechanisms, processing of Aβ and tau proteins, and ubiquitination pathways based on genes regulated by the treatment, provides a greater understanding of the molecular bases that act on the therapeutic effect of CBD observed in preclinical studies. Furthermore, this analysis identified genes that are central to ad‐associated pathways. These genes are of great interest since they may act as targets for the regulation of several mechanisms that promote ad‐associated dementia and may also experience a cumulative positive effect of CBD treatment, as they are involved in multiple pathways influenced by it. In this sense, great effort is recommended to analyze the mechanisms and effects related to their modulation.

As limitations, the literature review aimed to survey the results of CBD treatments already associated with ad; thus, other studies that demonstrate the CBD effects on mechanisms associated but without directly relating it to the disease were left out. Additionally, no studies were identified that examine the potential adverse effects or toxicity associated with CBD treatments in the search performed.

Another limitation is the limited amount of data on the effect of CBD on the expression of genes associated with ad. Studies involving candidate genes were not used for this analysis, in order to avoid bias in the identification of the main affected pathways. Thus, this analysis can be further enriched as more studies evaluating gene expression after CBD treatment are performed. In this sense, research based on hypothesis‐free analysis methodologies that provide a broader view of the effect of CBD on ad is encouraged.

In summary, the combination of literature review and in silico analysis brings together classical and contemporary data analysis methods, promoting a rich understanding of a complex disease such as ad. This combination of methodologies, capable of bringing together the effects and pathways of action associated with genetic modulation promoted by CBD treatment, demonstrates the enabling potential of this cannabinoid for the development of a complementary therapeutic method for ad.

Author Contributions

João V. Mello‐Hortega: conceptualization, investigation, methodology, writing – original draft. Carolina S. de Oliveira: writing – review and editing. Vitoria S. de Araujo: software. Lupe Furtado‐Alle: writing – review and editing. Luciane V. Tureck: funding acquisition, writing – review and editing. Ricardo L. R. Souza: conceptualization, project administration, supervision, writing – review and editing.

Conflicts of Interest

The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Joao V M Hortega reports financial support was provided by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior; Carolina S. de Oliveira reports financial support was provided by Conselho Nacional de Desenvolvimento Científico e Tecnológico.

Peer Review

The peer review history for this article is available at https://www.webofscience.com/api/gateway/wos/peer‐review/10.1111/ejn.70229.

Supporting information

Table S1: Supporting Information.

EJN-62-0-s002.docx (72.3KB, docx)

Table S2: Supporting Information.

EJN-62-0-s001.docx (26.3KB, docx)

Acknowledgements

The Article Processing Charge for the publication of this research was funded by the Coordenação de Aperfeiçoamento de Pessoal de Nível Superior – Brasil (CAPES) (ROR identifier: 00x0ma614).

Mello‐Hortega, J. , de Oliveira C., de Araujo V., Furtado‐Alle L., Tureck L., and Souza R.. 2025. “Cannabidiol and Alzheimer Disease: A Comprehensive Review and In Silico Insights Into Molecular Interactions.” European Journal of Neuroscience 62, no. 4: e70229. 10.1111/ejn.70229.

Associate Editor: Yan Zhang

Funding: This research was supported by the Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior for scholarships.

References

  1. 2022 Alzheimer's Disease Facts and Figures. 2022. Alzheimer's & Dementia 18: 700–789. 10.1002/alz.12638. [DOI] [PubMed] [Google Scholar]
  2. Abreu, I. D. D. , Forlenza O. V., and Barros H. L. D.. 2005. “Demência de Alzheimer: correlação entre memória e autonomia.” Revista de Psiquiatria Clinica 32: 131–136. [Google Scholar]
  3. Alali, S. , Riazi G., Ashrafi‐Kooshk M. R., et al. 2021. “Cannabidiol Inhibits Tau Aggregation In Vitro.” Cells 10: 3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Alexandri, F. , Papadopoulou L., Tsolaki A., Papantoniou G., Athanasiadis L., and Tsolaki M.. 2023. “The Effect of Cannabidiol 3% on Neuropsychiatric Symptoms in Dementia—Six‐Month Follow‐Up.” Clinical Gerontologist 47: 1–8. [DOI] [PubMed] [Google Scholar]
  5. Amini, M. , and Abdolmaleki Z.. 2022. “The Effect of Cannabidiol Coated by Nano‐Chitosan on Learning and Memory, Hippocampal CB1 and CB2 Levels, and Amyloid Plaques in an Alzheimer's Disease Rat Model.” Neuropsychobiology 81: 171–183. [DOI] [PubMed] [Google Scholar]
  6. Ariga, T. , McDonald M. P., and Yu R. K.. 2008. “Thematic Review Series: Sphingolipids. Role of Ganglioside Metabolism in the Pathogenesis of Alzheimer's Disease—A Review.” Journal of Lipid Research 49: 1157–1175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Aso, E. , Sánchez‐Pla A., Vegas‐Lozano E., Maldonado R., and Ferrer I.. 2014. “Cannabis‐Based Medicine Reduces Multiple Pathological Processes in AβPP/PS1 Mice.” JAD 43: 977–991. [DOI] [PubMed] [Google Scholar]
  8. Aso, E. , Andrés‐Benito P., and Ferrer I.. 2016. “Delineating the Efficacy of a Cannabis‐Based Medicine at Advanced Stages of Dementia in a Murine Model.” Journal of Alzheimer's Disease 54: 903–912. [DOI] [PubMed] [Google Scholar]
  9. Bliss, T. V. P. , and Collingridge G. L.. 1993. “A Synaptic Model of Memory: Long‐Term Potentiation in the Hippocampus.” Nature 361: 31–39. [DOI] [PubMed] [Google Scholar]
  10. Bloomfield, M. A. P. , Green S. F., Hindocha C., et al. 2020. “The Effects of Acute Cannabidiol on Cerebral Blood Flow and Its Relationship to Memory: An Arterial Spin Labelling Magnetic Resonance Imaging Study.” Journal of Psychopharmacology 34: 981–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Blum, S. , and Dash P. K.. 2009. “MAP Kinase Signaling in Learning and Memory.” In Encyclopedia of Neuroscience, 657–662. Elsevier. [Google Scholar]
  12. Bondan, E. F. , Cardoso C. V., Martins M. D. F. M., and Otton R.. 2019. “Memory Impairments and Increased GFAP Expression in Hippocampal Astrocytes Following Hypercaloric Diet in Rats.” Arquivos de Neuro‐Psiquiatria 77: 601–608. [DOI] [PubMed] [Google Scholar]
  13. Bonet‐Costa, V. , Pomatto L. C.‐D., and Davies K. J. A.. 2016. “The Proteasome and Oxidative Stress in Alzheimer's Disease.” Antioxidants & Redox Signaling 25: 886–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Callis, J. 2014. “The Ubiquitination Machinery of the Ubiquitin System.” Arabidopsis Book 12: e0174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Cargnello, M. , and Roux P. P.. 2011. “Activation and Function of the MAPKs and Their Substrates, the MAPK‐Activated Protein Kinases.” Microbiology and Molecular Biology Reviews 75: 50–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Casarejos, M. J. , Perucho J., Gomez A., et al. 2013. “Natural Cannabinoids Improve Dopamine Neurotransmission and Tau and Amyloid Pathology in a Mouse Model of Tauopathy.” Journal of Alzheimer's Disease 35: 525–539. [DOI] [PubMed] [Google Scholar]
  17. Casares, L. , Unciti‐Broceta J. D., Prados M. E., et al. 2020. “Isomeric O‐Methyl Cannabidiolquinones with Dual BACH1/NRF2 Activity.” Redox Biology 37: 101689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Charytoniuk, T. , Sztolsztener K., Harasim‐Symbor E., Berk K., Chabowski A., and Konstantynowicz‐Nowicka K.. 2021. “Cannabidiol—A Phytocannabinoid That Widely Affects Sphingolipid Metabolism Under Conditions of Brain Insulin Resistance.” Biomedicine & Pharmacotherapy 142: 112057. [DOI] [PubMed] [Google Scholar]
  19. Cheng, D. , Low J. K., Logge W., Garner B., and Karl T.. 2014. “Chronic Cannabidiol Treatment Improves Social and Object Recognition in Double Transgenic APPswe/PS1∆E9 Mice.” Psychopharmacology 231: 3009–3017. [DOI] [PubMed] [Google Scholar]
  20. Cheng, D. , Spiro A. S., Jenner A. M., Garner B., and Karl T.. 2014. “Long‐Term Cannabidiol Treatment Prevents the Development of Social Recognition Memory Deficits in Alzheimer's Disease Transgenic Mice.” Journal of Alzheimer's Disease 42: 1383–1396. [DOI] [PubMed] [Google Scholar]
  21. Cherry, J. D. , Olschowka J. A., and O'Banion M. K.. 2014. “Neuroinflammation and M2 Microglia: The Good, the Bad, and the Inflamed.” Journal of Neuroinflammation 11: 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chesworth, R. , Cheng D., Staub C., and Karl T.. 2022. “Effect of Long‐Term Cannabidiol on Learning and Anxiety in a Female Alzheimer's Disease Mouse Model.” Frontiers in Pharmacology 13: 931384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Choi, H. , and Ho Koh S.. 2016. “Interaction Between Amyloid Beta Toxicity and the PI3K Pathway in Alzheimer's Disease.” Journal of Alzheimers Disease & Parkinsonism 6: 269–271. [Google Scholar]
  24. Chrobak, W. , Pacut D. W., Blomgren F., Rodin A., Swenson J., and Ermilova I.. 2021. “Component of Cannabis, Cannabidiol, as a Possible Drug against the Cytotoxicity of Aβ(31‐35) and Aβ(25‐35) Peptides: An Investigation by Molecular Dynamics and Well‐Tempered Metadynamics Simulations.” ACS Chemical Neuroscience 12: 660–674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Coles, M. , Steiner‐Lim G. Z., and Karl T.. 2022. “Therapeutic Properties of Multi‐cannabinoid Treatment Strategies for Alzheimer's Disease.” Frontiers in Neuroscience 16: 962922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cooke, S. F. 2006. “Plasticity in the Human Central Nervous System.” Brain 129: 1659–1673. [DOI] [PubMed] [Google Scholar]
  27. Corey‐Bloom, J. , Thal L. J., Galasko D., et al. 1995. “Diagnosis and Evaluation of Dementia*.” Neurology 45: 211–218. [DOI] [PubMed] [Google Scholar]
  28. Cserép, C. , Pósfai B., Lénárt N., et al. 2020. “Microglia Monitor and Protect Neuronal Function Through Specialized Somatic Purinergic Junctions.” Science 367: 528–537. [DOI] [PubMed] [Google Scholar]
  29. Cummings, J. L. , and Back C.. 1998. “The Cholinergic Hypothesis of Neuropsychiatric Symptoms in Alzheimer's Disease.” American Journal of Geriatric Psychiatry 6: S64–S78. [DOI] [PubMed] [Google Scholar]
  30. De Felice, F. G. , Lourenco M. V., and Ferreira S. T.. 2014. “How Does Brain Insulin Resistance Develop in Alzheimer's Disease?” Alzheimer's & Dementia 10: S26–S32. [DOI] [PubMed] [Google Scholar]
  31. De Strooper, B. , Vassar R., and Golde T.. 2010. “The Secretases: Enzymes with Therapeutic Potential in Alzheimer Disease.” Nature Reviews. Neurology 6: 99–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Do, H. N. , Devkota S., Bhattarai A., Wolfe M. S., and Miao Y.. 2023. “Effects of Presenilin‐1 Familial Alzheimer's Disease Mutations on γ‐Secretase Activation for Cleavage of Amyloid Precursor Protein.” Communications Biology 6: 174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Duan, F. , Ju T., Song C., et al. 2023. “Synergetic Effect of β‐Asarone and Cannabidiol Against Aβ Aggregation In Vitro and In Vivo.” Computational and Structural Biotechnology Journal 21: 3875–3884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Echeverria, P. , Sethabutr O., and Pitarangsi C.. 1991. “Microbiology and Diagnosis of Infections with Shigella and Enteroinvasive Escherichia coli .” Clinical Infectious Diseases 13: S220–S225. [DOI] [PubMed] [Google Scholar]
  35. Esposito, G. , De Filippis D., Carnuccio R., Izzo A. A., and Iuvone T.. 2006. “The Marijuana Component Cannabidiol Inhibits Beta‐Amyloid‐Induced Tau Protein Hyperphosphorylation Through Wnt/Beta‐Catenin Pathway Rescue in PC12 Cells.” Journal of Molecular Medicine (Berlin, Germany) 84: 253–258. [DOI] [PubMed] [Google Scholar]
  36. Esposito, G. , De Filippis D., Maiuri M. C., De Stefano D., Carnuccio R., and Iuvone T.. 2006. “Cannabidiol Inhibits Inducible Nitric Oxide Synthase Protein Expression and Nitric Oxide Production in Beta‐Amyloid Stimulated PC12 Neurons Through p38 MAP Kinase and NF‐kappaB Involvement.” Neuroscience Letters 399: 91–95. [DOI] [PubMed] [Google Scholar]
  37. Esposito, G. , Scuderi C., Savani C., et al. 2007. “Cannabidiol in vivo Blunts β‐Amyloid Induced Neuroinflammation by Suppressing IL‐1β and iNOS Expression: CBD Blunts A β Induced Neuroinflammation in vivo .” British Journal of Pharmacology 151: 1272–1279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Esposito, G. , Scuderi C., Valenza M., et al. 2011. “Cannabidiol Reduces Aβ‐Induced Neuroinflammation and Promotes Hippocampal Neurogenesis Through PPARγ Involvement.” PLoS ONE 6: e28668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Fernández‐Ruiz, J. , Sagredo O., Pazos M. R., et al. 2013. “Cannabidiol for Neurodegenerative Disorders: Important New Clinical Applications for This Phytocannabinoid?” British Journal of Clinical Pharmacology 75: 323–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Ferreira, A. , and Bigio E. H.. 2011. “Calpain‐Mediated Tau Cleavage: A Mechanism Leading to Neurodegeneration Shared by Multiple Tauopathies.” Molecular Medicine 17: 676–685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Frandsen, J. , and Narayanasamy P.. 2022. “Effect of Cannabidiol on the Neural Glyoxalase Pathway Function and Longevity of Several C. elegans Strains Including a C. elegans Alzheimer's Disease Model.” ACS Chemical Neuroscience 13: 1165–1177. [DOI] [PubMed] [Google Scholar]
  42. Fraser, P. E. , Yang D.‐S., Yu G., et al. 2000. “Presenilin Structure, Function and Role in Alzheimer Disease.” Biochimica et Biophysica Acta (BBA) ‐ Molecular Basis of Disease 1502: 1–15. [DOI] [PubMed] [Google Scholar]
  43. Gauthier, S. 2001. “Cholinergic Adverse Effects of Cholinesterase Inhibitors in Alzheimer's Disease: Epidemiology and Management.” Drugs & Aging 18: 853–862. [DOI] [PubMed] [Google Scholar]
  44. Glickman, M. H. , and Ciechanover A.. 2002. “The Ubiquitin‐Proteasome Proteolytic Pathway: Destruction for the Sake of Construction.” Physiological Reviews 82: 373–428. [DOI] [PubMed] [Google Scholar]
  45. Halliwell, B. , and Gutteridge J. M. C.. 2015. Free Radicals in Biology and Medicine. Fifth ed. Oxford University Press; Oxford. [Google Scholar]
  46. Hampel, H. , Mesulam M.‐M., Cuello A. C., et al. 2018. “The Cholinergic System in the Pathophysiology and Treatment of Alzheimer's Disease.” Brain 141: 1917–1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Hampson, A. J. , Grimaldi M., Axelrod J., and Wink D.. 1998. “Cannabidiol and (−)Delta9‐tetrahydrocannabinol are Neuroprotective Antioxidants.” Proceedings of the National Academy of Sciences of the United States of America 95: 8268–8273. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Han, Y. , Ma Y., Liu Y., et al. 2019. “Plasma Cholinesterase is Associated with Chinese Adolescent Overweight or Obesity and Metabolic Syndrome Prediction.” Diabetes, Metabolic Syndrome and Obesity: Targets and Therapy 12: 685–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Hao, F. , and Feng Y.. 2021. “Cannabidiol (CBD) Enhanced the Hippocampal Immune Response and Autophagy of APP/PS1 Alzheimer's Mice Uncovered by RNA‐seq.” Life Sciences 264: 118624. [DOI] [PubMed] [Google Scholar]
  50. Harvey, B. S. , Ohlsson K. S., Mååg J. L. V., Musgrave I. F., and Smid S. D.. 2012. “Contrasting Protective Effects of Cannabinoids Against Oxidative Stress and Amyloid‐β Evoked Neurotoxicity in Vitro.” Neurotoxicology 33: 138–146. [DOI] [PubMed] [Google Scholar]
  51. Hashioka, S. , Wu Z., and Klegeris A.. 2021. “Glia‐Driven Neuroinflammation and Systemic Inflammation in Alzheimer's Disease.” Current Neuropharmacology 19: 908–924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Hernandez, F. , Lucas J. J., and Avila J.. 2012. “GSK3 and Tau: Two Convergence Points in Alzheimer's Disease.” JAD 33: S141–S144. [DOI] [PubMed] [Google Scholar]
  53. Hu, X. , Leak R. K., Shi Y., et al. 2015. “Microglial and Macrophage Polarization—New Prospects for Brain Repair.” Nature Reviews. Neurology 11: 56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Huestis, M. A. , Solimini R., Pichini S., Pacifici R., Carlier J., and Busardò F. P.. 2019. “Cannabidiol Adverse Effects and Toxicity.” Current Neuropharmacology 17: 974–989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Hughes, B. , and Herron C. E.. 2019. “Cannabidiol Reverses Deficits in Hippocampal LTP in a Model of Alzheimer's Disease.” Neurochemical Research 44: 703–713. [DOI] [PubMed] [Google Scholar]
  56. Hutton, M. 1997. “The Presenilins and Alzheimer's Disease.” Human Molecular Genetics 6: 1639–1646. [DOI] [PubMed] [Google Scholar]
  57. Iuvone, T. , Esposito G., Esposito R., Santamaria R., Di Rosa M., and Izzo A. A.. 2004. “Neuroprotective Effect of Cannabidiol, A Non‐Psychoactive Component From Cannabis sativa, on Beta‐Amyloid‐Induced Toxicity in PC12 Cells.” Journal of Neurochemistry 89: 134–141. [DOI] [PubMed] [Google Scholar]
  58. Jiang, X. , Zhang Z., Zuo J., et al. 2021. “Novel Cannabidiol‐Carbamate Hybrids as Selective BuChE Inhibitors: Docking‐Based Fragment Reassembly for the Development of Potential Therapeutic Agents Against Alzheimer's Disease.” European Journal of Medicinal Chemistry 223: 113735. [DOI] [PubMed] [Google Scholar]
  59. Kandimalla, R. , Thirumala V., and Reddy P. H.. 2017. “Is Alzheimer's Disease a Type 3 Diabetes? A Critical Appraisal.” Biochimica et Biophysica Acta (BBA) ‐ Molecular Basis of Disease 1863: 1078–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Kettenmann, H. , Hanisch U.‐K., Noda M., and Verkhratsky A.. 2011. “Physiology of Microglia.” Physiological Reviews 91: 461–553. [DOI] [PubMed] [Google Scholar]
  61. Khodadadi, H. , Salles É. L., Jarrahi A., et al. 2021. “Cannabidiol Ameliorates Cognitive Function via Regulation of IL‐33 and TREM2 Upregulation in a Murine Model of Alzheimer's Disease.” Journal of Alzheimer's Disease 80: 973–977. [DOI] [PubMed] [Google Scholar]
  62. Kitazume, S. , Tachida Y., Oka R., Shirotani K., Saido T. C., and Hashimoto Y.. 2001. “Alzheimer's β‐Secretase, β‐Site Amyloid Precursor Protein‐Cleaving Enzyme, is Responsible for Cleavage Secretion of a Golgi‐Resident Sialyltransferase.” Proceedings of the National Academy of Sciences of the United States of America 98: 13554–13559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Klegeris, A. , Walker D. G., and Mcgeer P. L.. 1994. “Activation of Macrophages by Alzheimer β Amyloid Peptide.” Biochemical and Biophysical Research Communications 199: 984–991. [DOI] [PubMed] [Google Scholar]
  64. Kreilaus, F. , Przybyla M., Ittner L., and Karl T.. 2022. “Cannabidiol (CBD) Treatment Improves Spatial Memory in 14‐Month‐Old Female TAU58/2 Transgenic Mice.” Behavioural Brain Research 425: 113812. [DOI] [PubMed] [Google Scholar]
  65. Landreth, G. , Jiang Q., Mandrekar S., and Heneka M.. 2008. “PPARγ Agonists as Therapeutics for the Treatment of Alzheimer's Disease.” Neurotherapeutics 5: 481–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Landucci, E. , Mazzantini C., Lana D., et al. 2022. “Cannabidiol Inhibits Microglia Activation and Mitigates Neuronal Damage Induced by Kainate in an In‐Vitro Seizure Model.” Neurobiology of Disease 174: 105895. [DOI] [PubMed] [Google Scholar]
  67. Lee, M. W. , Lee E. Y., Lai G. H., et al. 2017. “Molecular Motor Dnm1 Synergistically Induces Membrane Curvature To Facilitate Mitochondrial Fission.” ACS Central Science 3: 1156–1167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Li, S. , Huang Y., Yu L., Ji X., and Wu J.. 2023. “Impact of the Cannabinoid System in Alzheimer's Disease.” Current Neuropharmacology 21: 715–726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Libro, R. , Diomede F., Scionti D., et al. 2016. “Cannabidiol Modulates the Expression of Alzheimer's Disease‐Related Genes in Mesenchymal Stem Cells.” International Journal of Molecular Sciences 18: 26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Lim, K.‐H. , Kim S.‐H., Yang S., Chun S., and Joo J.‐Y.. 2021. “Advances in Multiplex PCR for Alzheimer's Disease Diagnostics Targeting CDK Genes.” Neuroscience Letters 749: 135715. [DOI] [PubMed] [Google Scholar]
  71. Liu, X. , Yao M., Li N., Wang C., Zheng Y., and Cao X.. 2008. “CaMKII Promotes TLR‐Triggered Proinflammatory Cytokine and Type I Interferon Production by Directly Binding and Activating TAK1 and IRF3 in Macrophages.” Blood 112: 4961–4970. [DOI] [PubMed] [Google Scholar]
  72. Liu, S.‐L. , Wang C., Jiang T., Tan L., Xing A., and Yu J.‐T.. 2016. “The Role of Cdk5 in Alzheimer's Disease.” Molecular Neurobiology 53: 4328–4342. [DOI] [PubMed] [Google Scholar]
  73. Lo, L. A. , Christiansen A., Eadie L., et al. 2023. “Cannabidiol‐Associated Hepatotoxicity: A Systematic Review and Meta‐Analysis.” Journal of Internal Medicine 293: 724–752. [DOI] [PubMed] [Google Scholar]
  74. Macario, A. J. L. , and De Conway Macario E.. 2007. “Chaperonopathies and Chaperonotherapy.” FEBS Letters 581: 3681–3688. [DOI] [PubMed] [Google Scholar]
  75. Maniv, I. , Sarji M., Bdarneh A., et al. 2023. “Altered Ubiquitin Signaling Induces Alzheimer's Disease‐Like Hallmarks in a Three‐Dimensional Human Neural Cell Culture Model.” Nature Communications 14: 5922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Mariani, S. M. 2004. “Amyloid, Tau, and Cell Death in Alzheimer's Disease.” MedGenMed 6: 46. [PMC free article] [PubMed] [Google Scholar]
  77. Marino Gammazza, A. , Caruso Bavisotto C., Barone R., De Conway Macario E., and Macario A. J. L.. 2016. “Alzheimer's Disease and Molecular Chaperones: Current Knowledge and the Future of Chaperonotherapy.” CPD 22: 4040–4049. [DOI] [PubMed] [Google Scholar]
  78. Marsh, D. T. , Sugiyama A., Imai Y., Kato R., and Smid S. D.. 2023. “The Structurally Diverse Phytocannabinoids Cannabichromene, Cannabigerol and Cannabinol Significantly Inhibit Amyloid β‐Evoked Neurotoxicity and Changes in Cell Morphology in PC12 Cells.” Basic & Clinical Pharmacology & Toxicology 134: 293–309. [DOI] [PubMed] [Google Scholar]
  79. Martiniano, F. S. T. , Barbosa J., Cardozo‐Batista L., et al. 2024. “Efficacy Of Cannabidiol (CBD) in the Treatment Of Alzheimer's Disease: A Critical Review Of Evidence and Clinical Implications.” Revft 29: 28–29. [Google Scholar]
  80. Martín‐Moreno, A. M. , Reigada D., Ramírez B. G., et al. 2011. “Cannabidiol and Other Cannabinoids Reduce Microglial Activation In Vitro and In Vivo: Relevance to Alzheimer's Disease.” Molecular Pharmacology 79: 964–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Mateju, D. , Franzmann T. M., Patel A., et al. 2017. “An Aberrant Phase Transition of Stress Granules Triggered by Misfolded Protein and Prevented by Chaperone Function.” EMBO Journal 36: 1669–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. McKinnon, C. , and Tabrizi S. J.. 2014. “The Ubiquitin‐Proteasome System in Neurodegeneration.” Antioxidants & Redox Signaling 21: 2302–2321. [DOI] [PubMed] [Google Scholar]
  83. Meda, L. , Cassatella M. A., Szendrei G. I., et al. 1995. “Activation of Microglial Cells by β‐Amyloid Protein and Interferon‐γ.” Nature 374: 647–650. [DOI] [PubMed] [Google Scholar]
  84. Meriin, A. B. , Narayanan A., Meng L., et al. 2018. “Hsp70–Bag3 Complex is a Hub for Proteotoxicity‐Induced Signaling That Controls Protein Aggregation.” Proceedings of the National Academy of Sciences of the United States of America 115: E7043–E7052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Mooko, T. , Bala A., Tripathy S., et al. 2022. “Cannabis Sativa L. Flower and Bud Extracts Inhibited In Vitro Cholinesterases and β‐Secretase Enzymes Activities: Possible Mechanisms of Cannabis Use in Alzheimer Disease.” Endocrine, Metabolic & Immune Disorders Drug Targets 22: 297–309. [DOI] [PubMed] [Google Scholar]
  86. Moreta, M. P.‐G. , Burgos‐Alonso N., Torrecilla M., Marco‐Contelles J., and Bruzos‐Cidón C.. 2021. “Efficacy of Acetylcholinesterase Inhibitors on Cognitive Function in Alzheimer's Disease. Review of Reviews.” Biomedicine 9: 1689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Moya‐Alvarado, G. , Gershoni‐Emek N., Perlson E., and Bronfman F. C.. 2016. “Neurodegeneration and Alzheimer's Disease (AD). What Can Proteomics Tell Us About the Alzheimer's Brain?” Molecular & Cellular Proteomics 15: 409–425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Noreen, N. , Muhammad F., Akhtar B., Azam F., and Anwar M. I.. 2018. “Is Cannabidiol a Promising Substance for New Drug Development? A Review of its Potential Therapeutic Applications.” Critical Reviews in Eukaryotic Gene Expression 28: 73–86. [DOI] [PubMed] [Google Scholar]
  89. Orihuela, R. , McPherson C. A., and Harry G. J.. 2016. “Microglial M1/M2 Polarization and Metabolic States.” British Journal of Pharmacology 173: 649–665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Patil, N. , Chandel V., Rana A., Jain M., and Kaushik P.. 2023. “Investigation of Cannabis sativa Phytochemicals as Anti‐Alzheimer's Agents: An In Silico Study.” Plants (Basel) 12: 510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. de Paula Faria, D. , Estessi de Souza L., Duran F. L. S., et al. 2022. “Cannabidiol Treatment Improves Glucose Metabolism and Memory in Streptozotocin‐Induced Alzheimer's Disease Rat Model: A Proof‐of‐Concept Study.” International Journal of Molecular Sciences 23: 1076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Pereira, M. G. , Gonçalves L. C., Da Silva P. A. N., et al. 2021. “Association Between Alzheimer's Disease and Apoe Gene Genotypic Polymorphisms: Systematic Review and Meta‐Analysis/Associação entre doença de alzheimer e polimorfismos genotipicos do gene apoe: revisão sistemática e metanálise.” BJDV 7: 59825–59843. [Google Scholar]
  93. Perry, E. K. , Tomlinson B. E., Blessed G., Bergmann K., Gibson P. H., and Perry R. H.. 1978. “Correlation of Cholinergic Abnormalities With Senile Plaques and Mental Test Scores in Senile Dementia.” BMJ 2: 1457–1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Prince, M. J. 2015. “S3‐01‐01: Prevention Report from Alzheimer's Disease International (ADI).” Alzheimer's & Dementia 11: 210. [Google Scholar]
  95. Prinz, M. , Jung S., and Priller J.. 2019. “Microglia Biology: One Century of Evolving Concepts.” Cell 179: 292–311. [DOI] [PubMed] [Google Scholar]
  96. Rajan, T. S. , Scionti D., Diomede F., et al. 2017. “Gingival Stromal Cells as an In Vitro Model: Cannabidiol Modulates Genes Linked with Amyotrophic Lateral Sclerosis: C ANNABIDIOL A LTERS ALS L INKED G ENES in hGMSCs.” Journal of Cellular Biochemistry 118: 819–828. [DOI] [PubMed] [Google Scholar]
  97. Razani, E. , Pourbagheri‐Sigaroodi A., Safaroghli‐Azar A., Zoghi A., Shanaki‐Bavarsad M., and Bashash D.. 2021. “The PI3K/Akt Signaling Axis in Alzheimer's Disease: A Valuable Target to Stimulate or Suppress?” Cell Stress and Chaperones 26: 871–887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Reitz, C. , Brayne C., and Mayeux R.. 2011. “Epidemiology of Alzheimer Disease.” Nature Reviews. Neurology 7: 137–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Ren, Z. , Yang M., Guan Z., and Yu W.. 2018. “Astrocytic α7 Nicotinic Receptor Activation Inhibits Amyloid‐β Aggregation by Upregulating Endogenous αB‐crystallin Through the PI3K/Akt Signaling Pathway.” Carcinogenesis 16: 39–48. [DOI] [PubMed] [Google Scholar]
  100. Ritchie, C. W. 2004. “Meta‐analysis of Randomized Trials of the Efficacy and Safety of Donepezil, Galantamine, and Rivastigmine for the Treatment of Alzheimer Disease.” American Journal of Geriatric Psychiatry 12: 358–369. [DOI] [PubMed] [Google Scholar]
  101. Ruangritchankul, S. , Chantharit P., Srisuma S., and Gray L. C.. 2021. “Adverse Drug Reactions of Acetylcholinesterase Inhibitors in Older People Living with Dementia: A Comprehensive Literature Review.” Therapeutics and Clinical Risk Management 17: 927–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Ryu, J. , Hong B.‐H., Kim Y.‐J., et al. 2016. “Neuregulin‐1 Attenuates Cognitive Function Impairments in a Transgenic Mouse Model of Alzheimer's Disease.” Cell Death & Disease 7: e2117–e2117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Saito, K. , Elce J. S., Hamos J. E., and Nixon R. A.. 1993. “Widespread Activation of Calcium‐Activated Neutral Proteinase (Calpain) in the Brain in Alzheimer Disease: A Potential Molecular Basis for Neuronal Degeneration.” Proceedings of the National Academy of Sciences of the United States of America 90: 2628–2632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Scuderi, C. , Steardo L., and Esposito G.. 2014. “Cannabidiol Promotes Amyloid Precursor Protein Ubiquitination and Reduction of Beta Amyloid Expression in SHSY5YAPP+ Cells Through PPARγ Involvement.” Phytotherapy Research 28: 1007–1013. [DOI] [PubMed] [Google Scholar]
  105. da Silva, V. K. , de Freitas B. S., da Silva Dornelles A., et al. 2014. “Cannabidiol Normalizes Caspase 3, Synaptophysin, and Mitochondrial Fission Protein DNM1L Expression Levels in Rats with Brain Iron Overload: Implications for Neuroprotection.” Molecular Neurobiology 49: 222–233. [DOI] [PubMed] [Google Scholar]
  106. Silva, M. V. F. , Loures C. D. M. G., Alves L. C. V., De Souza L. C., Borges K. B. G., and Carvalho M. D. G.. 2019. “Alzheimer's Disease: Risk Factors and Potentially Protective Measures.” Journal of Biomedical Science 26: 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Silva, S. S. L. , Tureck L. V., Souza L. C., et al. 2023. “Animal Model of Alzheimer's Disease Induced by Streptozotocin: New Insights About Cholinergic Pathway.” Brain Research 1799: 148175. [DOI] [PubMed] [Google Scholar]
  108. Simon, D. W. , McGeachy M. J., Bayır H., Clark R. S. B., Loane D. J., and Kochanek P. M.. 2017. “The Far‐Reaching Scope of Neuroinflammation After Traumatic Brain Injury.” Nature Reviews. Neurology 13: 171–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Singh, K. , Bhushan B., Chanchal D. K., et al. 2023. “Emerging Therapeutic Potential of Cannabidiol (CBD) in Neurological Disorders: A Comprehensive Review.” Behavioural Neurology 2023: 8825358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Smith, B. , and Ownby R. L.. 2024. “Disease‐Modifying Treatments and Their Future in Alzheimer's Disease Management.” Cureus 16: e56105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Summers, W. K. , Majovski L. V., Marsh G. M., Tachiki K., and Kling A.. 1986. “Oral Tetrahydroaminoacridine in Long‐Term Treatment of Senile Dementia, Alzheimer Type.” New England Journal of Medicine 315: 1241–1245. [DOI] [PubMed] [Google Scholar]
  112. Sze, C.‐I. , Troncoso J. C., Kawas C., Moution P., Price D. L., and Martin L. J.. 1997. “Loss of the Presynaptic Vesicle Protein Synaptophysin in Hippocampus Correlates with Cognitive Decline in Alzheimer Disease.” Journal of Neuropathology and Experimental Neurology 56: 933–944. [DOI] [PubMed] [Google Scholar]
  113. Tanner, K. , Brzovic P., and Rohde J. R.. 2015. “The Bacterial Pathogen‐Ubiquitin Interface: Lessons Learned from S higella: The Bacterial Pathogen‐Ubiquitin Interface.” Cellular Microbiology 17: 35–44. [DOI] [PubMed] [Google Scholar]
  114. Tanzi, R. E. 2012. “The Genetics of Alzheimer Disease.” Cold Spring Harbor Perspectives in Medicine 2: a006296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Tramutola, A. , Di Domenico F., Barone E., Perluigi M., and Butterfield D. A.. 2016. “It Is All about (U)biquitin: Role of Altered Ubiquitin‐Proteasome System and UCHL1 in Alzheimer Disease.” Oxidative Medicine and Cellular Longevity 2016: 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Vallée, A. , Lecarpentier Y., Guillevin R., and Vallée J.‐N.. 2017. “Effects of Cannabidiol Interactions with Wnt/β‐Catenin Pathway and PPARγ on Oxidative Stress and Neuroinflammation in Alzheimer's Disease.” Acta Biochimica et Biophysica Sinica 49: 853–866. [DOI] [PubMed] [Google Scholar]
  117. Vanhaesebroeck, B. , Guillermet‐Guibert J., Graupera M., and Bilanges B.. 2010. “The Emerging Mechanisms of Isoform‐Specific PI3K Signalling.” Nature Reviews. Molecular Cell Biology 11: 329–341. [DOI] [PubMed] [Google Scholar]
  118. Vanin, A. P. , Tamagno W. A., Alves C., et al. 2022. “Neuroprotective Potential of Cannabis sativa‐Based Oils in Caenorhabditis elegans .” Scientific Reports 12: 15376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Walls, K. C. , Coskun P., Gallegos‐Perez J. L., et al. 2012. “Swedish Alzheimer Mutation Induces Mitochondrial Dysfunction Mediated by HSP60 Mislocalization of Amyloid Precursor Protein (APP) and Beta‐Amyloid.” Journal of Biological Chemistry 287: 30317–30327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Wandel, M. P. , Pathe C., Werner E. I., et al. 2017. “GBPs Inhibit Motility of Shigella flexneri but Are Targeted for Degradation by the Bacterial Ubiquitin Ligase IpaH9.8.” Cell Host & Microbe 22: 507–518.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Wang, Z. , Zheng P., Xie Y., et al. 2021. “Cannabidiol Regulates CB1‐pSTAT3 Signaling for Neurite Outgrowth, Prolongs Lifespan, and Improves Health Span in Caenorhabditis elegans of Aβ Pathology Models.” FASEB Journal 35: e21537. [DOI] [PubMed] [Google Scholar]
  122. Wang, Z. , Zheng P., Nagaratnam N., Solowij N., and Huang X.‐F.. 2023. “Parkin Mediates Cannabidiol Prevention of Amyloid‐Beta‐Induced Senescence in Human Astrocytes.” Cannabis and Cannabinoid Research 8: 309–320. [DOI] [PubMed] [Google Scholar]
  123. Watt, G. , Shang K., Zieba J., et al. 2020. “Chronic Treatment with 50 mg/kg Cannabidiol Improves Cognition and Moderately Reduces Aβ40 Levels in 12‐Month‐Old Male AβPPswe/PS1ΔE9 Transgenic Mice.” JAD 74: 937–950. [DOI] [PubMed] [Google Scholar]
  124. Weinreb, O. , Mandel S., Bar‐Am O., et al. 2009. “Multifunctional Neuroprotective Derivatives of Rasagiline as Anti‐Alzheimer's Disease Drugs.” Neurotherapeutics 6: 163–174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Williams, J. W. , Plassman B. L., Burke J., and Benjamin S.. 2010. “Preventing Alzheimer's Disease and Cognitive Decline.” Evidence Report Technology Assessment (Full Rep) 193: 1–727. [PMC free article] [PubMed] [Google Scholar]
  126. Woelfl, T. , Rohleder C., Mueller J. K., et al. 2020. “Effects of Cannabidiol and Delta‐9‐Tetrahydrocannabinol on Emotion, Cognition, and Attention: A Double‐Blind, Placebo‐Controlled, Randomized Experimental Trial in Healthy Volunteers.” Frontiers in Psychiatry 11: 576877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Wu, D. , and Pan W.. 2010. “GSK3: A Multifaceted Kinase in Wnt Signaling.” Trends in Biochemical Sciences 35: 161–168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Xu, H. , Garcia‐Ptacek S., Jönsson L., Wimo A., Nordström P., and Eriksdotter M.. 2021. “Long‐term Effects of Cholinesterase Inhibitors on Cognitive Decline and Mortality.” Neurology 96: e2220–e2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Yang, S. , Du Y., Zhao X., et al. 2022. “Cannabidiol Enhances Microglial Beta‐Amyloid Peptide Phagocytosis and Clearance via Vanilloid Family Type 2 Channel Activation.” International Journal of Molecular Sciences 23: 5367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Ye, L. , Wang X., Cai C., et al. 2019. “FGF21 Promotes Functional Recovery After Hypoxic‐Ischemic Brain Injury in Neonatal Rats by Activating the PI3K/Akt Signaling Pathway via FGFR1/β‐klotho.” Experimental Neurology 317: 34–50. [DOI] [PubMed] [Google Scholar]
  131. Yu, G. , Wang L.‐G., Han Y., and He Q.‐Y.. 2012. “ClusterProfiler: An R Package for Comparing Biological Themes Among Gene Clusters.” OMICS: A Journal of Integrative Biology 16: 284–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Zhang, X. , Guo L., Collage R. D., et al. 2011. “Calcium/calmodulin‐dependent Protein Kinase (CaMK) Iα Mediates the Macrophage Inflammatory Response to Sepsis.” Journal of Leukocyte Biology 90: 249–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Zhang, B. , Gaiteri C., Bodea L.‐G., et al. 2013. “Integrated Systems Approach Identifies Genetic Nodes and Networks in Late‐Onset Alzheimer's Disease.” Cell 153: 707–720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Zhang, Y. , Chen X., Zhao Y., Ponnusamy M., and Liu Y.. 2017. “The Role of Ubiquitin Proteasomal System and Autophagy‐Lysosome Pathway in Alzheimer's Disease.” Reviews in the Neurosciences 28: 861–868. [DOI] [PubMed] [Google Scholar]
  135. Zhang, X.‐X. , Tian Y., Wang Z.‐T., Ma Y.‐H., Tan L., and Yu J.‐T.. 2021. “The Epidemiology of Alzheimer's Disease Modifiable Risk Factors and Prevention.” Journal of Prevention of Alzheimer's Disease 8: 313–321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Zhang, Y. , Li H., Jin S., et al. 2022. “Cannabidiol Protects against Alzheimer's Disease in C. elegans via ROS Scavenging Activity of Its Phenolic Hydroxyl Groups.” European Journal of Pharmacology 919: 174829. [DOI] [PubMed] [Google Scholar]
  137. Zhang, J. , Zhang Y., Wang J., Xia Y., Zhang J., and Chen L.. 2024. “Recent Advances in Alzheimer's Disease: Mechanisms, Clinical Trials and New Drug Development Strategies.” Signal Transduction and Targeted Therapy 9: 211. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Table S1: Supporting Information.

EJN-62-0-s002.docx (72.3KB, docx)

Table S2: Supporting Information.

EJN-62-0-s001.docx (26.3KB, docx)

Articles from The European Journal of Neuroscience are provided here courtesy of Wiley

RESOURCES