Abstract
Objective
This systematic review and meta-analysis aimed to evaluate the impact of cannabinoid (CB) receptor modulation on infarct volume and behavioral deficits in animal models of focal ischemic stroke, with a primary focus on infarct outcomes.
Method
A comprehensive literature search was conducted following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, yielding 29 eligible studies for inclusion.
Results
The analysis revealed significant reductions in infarct volume with CB agonists, particularly CB1 and CB2 agonists, indicating their potential as neuroprotective agents. Subgroup analyses further highlighted specific agonists, such as ACEA and KN38-72717, as particularly effective in reducing infarct volume. Additionally, CB antagonists, particularly CB1 antagonists, such as SR141716, showed promising results in reducing infarct volume. Although improvements in neurological scores were observed with both agonists and antagonists, statistical significance was not reached, indicating the need for further investigation.
Conclusion
These results highlight the potential of CB receptor modulation as a neuroprotective strategy in ischemic strokes and underscore the need for further research to elucidate the underlying mechanisms and optimize therapeutic approaches.
Keywords: Ischemic stroke, Cannabinoid receptors, Neuroprotection, Meta-analysis, Therapeutic potential
Highlights of the Study
Cannabinoid (CB) agonists, particularly CB1 and CB2 agonists, significantly reduced infarct volume, with standardized mean differences indicating robust neuroprotective effects.
Among CB antagonists, CB1 antagonists like SR141716 significantly reduced infarct volume (SMD −2.93; p < 0.0001), while CB2 antagonists did not show similar effects, highlighting a differential role for CB1 versus CB2 receptors in mitigating ischemic injury.
Both CB2 agonists and non-selective CB1/CB2 agonists demonstrated significant reductions in infarct volume, with SMDs of −3.69 and −3.60, respectively, suggesting a broader neuroprotective role for CB receptor activation across different subtypes.
Introduction
Stroke remains a predominant global health challenge, consistently escalating in prevalence across its subtypes, yet there exists no effective treatment to counteract its detrimental impacts [1–3]. Ischemic stroke, the most prevalent form, occurs when the blood flow to brain tissue becomes inadequate, causing cerebral infarction or damage. This often leads to lasting behavioral impairments. Animal models of ischemic stroke can be classified as global or focal [4]. Focal ischemic stroke can be induced in animal models using various methods, most commonly by obstructing the middle cerebral artery (middle cerebral artery occlusion [MCAO]). Global ischemic stroke can be achieved or simulated using several techniques, including 4-vessel occlusion (4-VO) or 2-vessel occlusion (2-VO), among other approaches [5, 6].
The endocannabinoid system (ECS) is a vital communication network between cells that regulates a range of physiological processes in the body, including synaptic transmission, memory processes, and inflammation [7]. This system primarily operates through cannabinoid receptor 1 (CB1R) and cannabinoid receptor 2 (CB2R), which are widely expressed in the central nervous and immune systems [7, 8]. Cannabinoids (CBs) are mainly classified into three categories: endocannabinoids, phytocannabinoids, and synthetic CBs [9]. In addition, research has shown that components of the ECS, such as CB receptors and endocannabinoids, are modified after stroke, suggesting their role in the body’s natural response to stroke damage [10, 11]. Preclinical studies have demonstrated the neuroprotective effects of various approaches to modulate the ECS, indicating a potential therapeutic benefit of targeting the ECS in stroke treatment [10, 11].
Although the meta-analysis by England et al. [9] provided foundational insights into CB therapies in experimental stroke, its inclusion of compounds with off-target mechanisms (i.e., effects independent of CB1/CB2 receptor activation) limits the ability to isolate neuroprotective pathways specific to canonical CB receptor signaling [9]. To address this gap, our systematic review employs a targeted methodological approach, restricting analysis to pharmacological agents that directly modulate CB1 or CB2 receptors. By excluding compounds with non-receptor-mediated mechanisms, this strategy enables a rigorous evaluation of the neuroprotective efficacy and therapeutic potential of selective CB1/CB2 receptor modulation in ischemic stroke, thereby clarifying their distinct roles in mitigating neuronal injury. The objective of this study was to systematically evaluate the effects of CB receptor modulation on infarct volume and behavioral deficits in animal models of focal ischemic stroke.
Methods
Search Strategy
Our search strategy adhered to established guidelines recommended by the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A thorough examination of the existing literature was conducted, encompassing the electronic databases PubMed, Google Scholar, and ScienceDirect, from January 2000 to January 2024. The search was systematically executed utilizing Medical Subject Headings terms, specifically targeting relevant studies on stroke or ischemia in conjunction with CB-related terms, including “cannabinoid,” “endocannabinoids,” “CB1,” and “CB2.”
Study Selection
Two authors independently devised the search approach and resolved discrepancies with a third investigator. Eligible studies included original empirical research with quantitative assessment of behavioral deficits and/or infarct volume, utilizing CB1 or CB2 receptor-targeting agents for in vivo administration, focusing exclusively on brain injury outcomes, employing accepted focal ischemia induction methods, and intervention with CB receptor agonists or antagonists. The exclusion criteria were in vitro experiments, global ischemia models, neonatal stroke models, adjunctive treatments lacking CB-related mechanisms, CBs primarily acting on non-CB receptors, samples with genetic modulation of CB1 or CB2 receptors, and sample groups administered two drugs affecting the same receptor. Exclusion of global ischemia models and neonatal stroke ensured homogeneity in pathophysiology, while restricting to CB1/CB2-targeted agents isolated receptor-specific effects.
Outcome Measures and Data Extraction
The data extraction process was conducted by a team of four members, following rigorously established protocols. To ensure data accuracy, verification was performed by all four members, and any inconsistencies were resolved through consultation with two other team members. Pertinent study parameters, including study name, publication year, geographical location, sample size, drug classification, animal species, methodological approach to inducing stroke, injury duration, therapeutic regimen, frequency of administration, assessment time points, and Stroke Therapy Academic Industry Roundtable (STAIR) score, were meticulously collected. The primary outcomes under scrutiny were brain infarct volume and behavioral deficit measurements between the experimental and control cohorts.
Assessment of Risk of Bias
Studies that met our predefined criteria underwent a meticulous assessment to ensure the reliability of our findings. This evaluation utilized an eight-criterion scale adapted from the STAIR criteria and England et al. [9, 12, 13]. The key methodological aspects evaluated included the implementation of randomization to minimize selection bias, continuous temperature monitoring to control for confounding variables, blinding of outcome assessment to mitigate performance bias, evaluation at both acute and subacute/chronic phases to capture comprehensive outcomes, assessment beyond infarct size to consider broader impact measures, testing multiple doses to elucidate dose-response relationships, and repeated outcome assessments at various time points to validate findings longitudinally. By systematically addressing these criteria, we comprehensively evaluated the risk of bias within the included studies, thereby enhancing the reliability and validity of the meta-analysis. Detailed quality assessments of all included studies are shown in Table 1.
Table 1.
Quality assessment and risk of bias evaluation for each included study based on the Stroke Therapy Academic Industry Roundtable (STAIR) criteria
| Quality Assessment by STAIR criteria | |||||||||
|---|---|---|---|---|---|---|---|---|---|
| study name | A | B | C | D | E | F | G | H | total score |
| Yang et al. [14] (2020) | No | No | No | Yes | Yes | Yes | Yes | Yes | 5 |
| Bai et al. [15] (2017) | Yes | No | Yes | No | Yes | Yes | No | Yes | 5 |
| Caltana et al. [16] (2015) | No | Yes | Yes | Yes | Yes | No | Yes | No | 5 |
| Sun et al. [17] (2013) | Yes | Yes | Yes | No | Yes | Yes | Yes | Yes | 7 |
| Yokubaitis et al. [18] (2021) | No | Yes | No | No | Yes | Yes | No | Yes | 4 |
| Yang et al. [19] (2017) | Yes | Yes | Yes | No | Yes | Yes | Yes | No | 6 |
| Pottier et al. [20] (2017) | No | No | No | Yes | Yes | No | No | Yes | 3 |
| Ronca et al. [21] (2015) | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | 8 |
| Yu et al. [22] (2015) | No | Yes | Yes | Yes | Yes | Yes | No | Yes | 6 |
| Choi et al. [23] (2013) | No | Yes | No | No | Yes | Yes | No | Yes | 4 |
| Zarruk et al. [24] (2012) | Yes | Yes | Yes | No | Yes | Yes | No | Yes | 6 |
| Shirazi et al. [25] (2021) | No | No | Yes | No | Yes | Yes | Yes | Yes | 5 |
| Murakami et al. [26] (2013) | No | Yes | No | No | Yes | Yes | No | Yes | 4 |
| Suzuki et al. [27] (2012) | Yes | Yes | Yes | Yes | Yes | Yes | No | Yes | 7 |
| Schmidt et al. [28] (2012) | No | Yes | Yes | Yes | Yes | Yes | No | Yes | 6 |
| Reichenbach et al. [29] (2016) | No | Yes | No | No | Yes | No | Yes | No | 3 |
| Jalin et al. [30] (2015) | Yes | No | Yes | No | Yes | Yes | No | Yes | 5 |
| Bravo-Ferrer et al. [31] (2016) | No | Yes | No | Yes | Yes | Yes | Yes | Yes | 6 |
| Zhang et al. [32] (2012) | No | Yes | No | No | Yes | Yes | Yes | Yes | 5 |
| Zhang et al. [33] (2009) | No | Yes | No | No | Yes | No | Yes | Yes | 4 |
| Zhang et al. [34] (2007) | No | Yes | No | Yes | Yes | Yes | Yes | Yes | 6 |
| Zhang et al. [35] (2008) | Yes | Yes | Yes | Yes | Yes | Yes | Yes | Yes | 8 |
| Hu et al. [36] (2010) | Yes | Yes | Yes | No | Yes | No | No | No | 4 |
| Berger et al. [37] (2004) | Yes | Yes | No | No | Yes | No | No | No | 3 |
| Muthian et al. [38] (2004) | Yes | Yes | No | No | Yes | Yes | Yes | No | 5 |
| Hayakawa et al. [39] (2007) | No | Yes | No | No | Yes | Yes | Yes | Yes | 5 |
| Leker et al. [40] (2003) | No | Yes | Yes | No | Yes | Yes | No | Yes | 5 |
Stroke Therapy Academic Industry Roundtable (STAIR) criteria: A, implementation of randomization to minimize selection bias; B, continuous temperature monitoring to control for confounding variables; C, blinding of outcome assessment to mitigate performance bias, D, evaluation at both acute and subacute/chronic phases to capture comprehensive outcomes; E, assessment beyond infarct size to consider broader impact measures; F, testing multiple doses to elucidate dose-response relationships; G, repeated outcome assessments at various time points to validate findings longitudinally; H, therapeutic time window relationship of a particular agonist conducted.
Statistical Analysis
For the statistical analysis, Review Manager (RevMan) version 5.4 and RStudio were utilized. By employing a random-effects model, calculation of the standard mean difference was carried out with 95% confidence interval (CI). Our analysis included constructing forest plots to illustrate primary outcomes, such as overall infarct size and behavioral deficit across CB agonists and antagonists. Further exploration included the creation of scatter diagrams to investigate the correlation between specific drug responses and infarct size. To assess the impact of time of administration, grouped bar plots with error bars and CIs were developed. A visual examination of the funnel plot was performed to evaluate publication bias. Statistical significance was set at a threshold of p < 0.05. Heterogeneity was evaluated using Higgins I2, with values exceeding 50% considered statistically significant. A leave-one-out sensitivity analysis, conducted post hoc and guided by observed data patterns, assessed individual studies’ impact on overall heterogeneity. Subgroup analyses were prioritized over meta-regression due to heterogeneity in reported variables and sample size limitations.
Study Registration and Ethical Approval
This study was registered at the King Abdullah International Medical Research Center under protocol number NRR25/063/3.
Results
Literature Search
An extensive search across the databases yielded a total of 3,405 articles. After eliminating duplicates (n = 574), 2,831 unique articles remained for screening. Each of these 2,831 records underwent a thorough evaluation based on their titles and abstracts, resulting in the identification of 44 articles for further scrutiny. A subsequent detailed full-text examination led to the exclusion of 15 articles [41–55], citing reasons such as failure to meet the inclusion criteria and lack of relevant data. These reasons are documented in online supplementary Table 1 (for all online suppl. material, see https://doi.org/10.1159/000547277). Ultimately, 29 articles [14–40, 56, 57] were deemed appropriate for inclusion in the systematic review, representing a qualitative synthesis. Among these, 26 studies were eligible for incorporation into the meta-analysis, representing a quantitative synthesis. The selection process for the included studies is shown in online supplementary Figure 1.
Study Characteristics and Quality Assessment
The baseline characteristics of the included studies, detailed in Tables 2, revealed a diverse landscape spanning from 2003 to 2021 across several countries, including China, the USA, Argentina, Spain, Japan, South Korea, France, and Germany (Fig. 1–5). Predominantly employing rodent models, particularly Sprague-Dawley rats, C57Bl/6 mice, and Wistar rats, the studies investigated various drugs targeting CB receptors, such as ACEA, WIN55, 212-2, BCP, JWH-133, AM1241, TC, O-1966, O-3853, HU-210, SR141716A, KN38-72717, and SR144528. Stroke induction methods varied, including MCAO with or without reperfusion, photo thrombosis, and thromboembolic MCAO, with treatments administered either pre-stroke or post-stroke. The total number of animals included in the experimental group was 385, while the number of animals in the control or vehicle-treated group was 258. Outcome assessments were conducted at various post-stroke intervals, encompassing parameters such as infarct volume, neurological deficit scores, grip strength, ladder-rung walking, and foot-fault tests. The quality of evidence presented in each study was evaluated using the STAIR score, providing an assessment of study reliability and validity.
Table 2.
Baseline characteristics of included studies
| Author | Year | Country | Drug | Mechanism of action | Animal | Stroke method | Therapy type | Timing of administrations | Outcome | Assessment time points | Significant improvements |
|---|---|---|---|---|---|---|---|---|---|---|---|
| Yang et al. [14] | 2020 | China | ACEA | CB1 agonist | Sprague-Dawley rats | MCAO/r | Pre-treatment | 30 min before injury | Substantial reduction compared to control | Day 1 | Longa’s neurological deficit score |
| Bai et al. [15] | 2017 | China | ACEA | CB1 agonist | Sprague-Dawley rats | MCAO/r | Post-treatment | 1 h after MCAO | Significant decrease compared to control | Day 3 | Garcia JH 18-point neurological deficit score |
| Caltana et al. [16] | 2015 | Argentina | ACEA | CB1 agonist | C57Bl/6 mice | MCAO | Post-treatment | 3, 24, and 48 h after injury | Significant decrease compared to control | Day 28 | Neurological score |
| Sun et al. [17] | 2013 | China | WIN55,212-2 | CB1 agonist | Sprague-Dawley rats | MCAO | Post-treatment | 2 h after injury | Significant decrease compared to vehicle treated | Day 1 | Neurological score |
| Yokubaitis et al. [18] | 2021 | USA | BCP | CB2 agonist | C57Bl/6 mice | Photo thrombosis | Pre-/post-treatment | 1 h prior to injury and 24 h post-injury | Significant decrease compared to vehicle treated | Day 3 | Grip strength test |
| Yang et al. [19] | 2017 | China | BCP | CB2 agonist | C57BL/6 mice | MCAO/r | Pre-/post-treatment | 3 days before injury and 2 h post-injury | Significant decrease compared to control | Day 2 | Longa’s neurological deficit score |
| Pottier et al. [20] | 2017 | France | JWH-133 | CB2 agonist | Sprague-Dawley rats | MCAO/r | Post-treatment | Daily from 1 h after MCAO to day 7 | Significant improvement compared to non-treated samples | Day 7 | N/A |
| Ronca et al. [21] | 2015 | USA | O-1966 | CB2 agonist | C57B/6 mice | Photo thrombosis | Pre-/post-treatment | 1 h prior to injury and 2.5 days following or 1 h prior to injury and 2 days following | Significant decrease compared to control | Day 1, 3, and 7 | Novel object recognition task, food-motivated operant task |
| Yu et al. [22] | 2015 | USA | AM1241 | CB2 agonist | Sprague-Dawley rats | MCAO/r | Pre-/post-treatment | 5 min before injury or from day 2–5 following | Significant decrease compared to vehicle-treated (pre-treatment), non-significant improvement (post-treatment) | Day 2 and 6 | Bederson’s score |
| Choi et al. [23] | 2013 | South Korea | TC | CB2 agonist | Sprague-Dawley rats | MCAO/r | Post-treatment | 3 h after injury | Significant decrease compared to control | Day 1 | N/A |
| Zarruk et al. [24] | 2012 | Spain | JWH-133 | CB2 agonist | Swiss mice | MCAO | Post-treatment | 10 min or 3 h post-injury | Significant decrease compared to vehicle treated in both groups of treatment | Day 2 | mNSS |
| Shirazi et al. [25] | 2021 | New Zealand | Novel N-acylethanolamine derivatives | CB1/CB2 agonist | C57BL/6J mice | Photo thrombosis | Post-treatment | 1 h post-injury | Significant decrease compared to vehicle treated (NAE-18:4n-6, NAE-18:5n-3); non-significant improvement compared to vehicle treated (NAE-22:5n-6, NAE-22:6n-3, NAE-20:5n-3, NAE-20:4n-6) | Day 7 | NAE-18:4n-6, NAE-18:5n-3 |
| Murakami et al. [26] | 2013 | Japan | TAK-937 | CB1/CB2 agonist | Sprague-Dawley rats | Photo thrombosis | Post-treatment | Continuous administration from 3 h, 5 h, or 8 h–24 h | Significant decrease compared to vehicle treated (3–24 h and 5–24 h); no improvement (8–24 h) | Day 2 | N/A |
| Suzuki et al. [27] | 2012 | Japan | TAK-937 | CB1/CB2 agonist | Sprague-Dawley rats or cynomolgus monkeys | MCAO/r or Thromboembolic MCAO | Post-treatment | Continuous infusion for 22 h following perfusion (rats), continuous infusion for 23.5 h following clot injection (monkeys) | Significant decrease compared to vehicle-treated (rats), non-significant improvement compared to vehicle treated (monkeys) | Day 1 | Foot-fault test, Bederson’s score |
| Schmidt et al. [28] | 2012 | Germany | KN38-72717 | CB1/CB2 agonists | Sprague-Dawley rats | mMCAO | Pre-/post-treatment | 2 h before injury, during eMCAO, 30 min, 4 h, 3 days, 6 days after injury | Significant decrease compared to vehicle treated in multiple timepoints | Day 7 and 21 | Ladder-rung walking test |
| Reichenbach et al. [29] | 2016 | USA | SR141716A | CB1 antagonist | C57Bl/6 mice | Photo thrombosis | Pre-/post-treatment | 1 h before or after injury | Significant decrease compared to vehicle treated (pre-treatment); non-significant improvement (post-treatment) | Day 1 | Hata et al. five point neurological score |
| Jalin et al. [30] | 2015 | Spain | Hinokiresinols | CB1/CB2 antagonist | Sprague-Dawley rats | MCAO/r | Post-treatment | 2 h and 7 h after injury | Significant decrease compared to vehicle treated | Day 1 | N/A |
| Bravo-Ferrer et al. [31] | 2016 | Spain | JWH-133 or SR144528 | CB2 agonist or CB2R antagonist (respectively) | C57BL/6 mice | MCAO | Post-treatment | 2 days following injury till day 6 | No improvement compared to vehicle treated (both drugs) | Day 14 and 28 | NR |
| Zhang et al. [32] | 2012 | USA | O-1966, SR141716A, both, or SR144528 | CB2 agonist, CB1 antagonist, CB1 antagonist/CB2 agonist, or CB2 antagonist (respectively) | C57BL/6 mice | MCAO/r | Pre-treatment | 1 h before injury | O-1966: significant decrease compared to control; SR141716A: significant decrease compared to control; O-1966 and SR141716A: significant decrease compared to control; SR144528: no improvement | Day 1 | N/A |
| Zhang et al. [33] | 2009 | USA | O-1966 | CB2 agonist | C57BL/6 mice | MCAO/r | Pre-/post-treatment | 1 h before injury | Significant decrease with 5 mg dose only compared to vehicle treated | Day 1 | Significant improvement compared with vehicle group |
| Zhang et al. [34] | 2007 | USA | O-3853, O-1966 | CB2 agonist | C57BL/6 mice | MCAO/r | Pre-/post-treatment | 1 h before and after the injury | Significant decrease in size with both pre- and post-treatment | Day 1 | Significant improvement with both pre- and post-treatment group |
| Zhang et al. [33] | 2009 | USA | O-1966, SR141716A, SR144528 | CB1 and CB2 antagonist | C57BL/6 mice | MCAO/r | Pre-treatment | 1 h before injury | CB2 receptor activation and CB1 receptor inhibition were neuroprotective, with the greatest protection achieved by combining a CB2 agonist and CB1 antagonist | Day 1 | CB2 receptor activation and CB1 receptor inhibition were neuroprotective, with the greatest protection achieved by combining a CB2 agonist and CB1 antagonist |
| Hu et al. [36] | 2010 | China | WIN 55,212-2, U0126 | CB agonist, CB antagonist | Sprague-Dawley rats | MCAO/r | Pre-treatment | 1 day before the injury | CB agonist has neuroprotective function while the antagonist proves the opposite effect | Day 1, 3, 6 | Pretreatment with CB agonist has protective function |
| Berger et al. [37] | 2004 | Germany | SR141716A | CB1 antagonist | Wistar rats | MCAO/r | Post-treatment | 30 min after injury | CB1 antagonist reduces infarct size significantly | Day 1 | NR |
| Muthian et al. [38] | 2004 | USA | WIN 55212-2, SR141716, LY320135 | CB1 antagonist | Wistar rats | MCAO/r | Pre-treatment | 5 min pre-onset | CB1 antagonist reduces infarct size significantly, while no effect with CB1 agonist | Day 1 | Significantly improved neurological function with CB1 receptor antagonist |
| Hayakawa et al. [39] | 2007 | Japan | Delta(9)-THC, SR141716 | CB agonist, CB antagonist | ddY mice | MCAO/r | Pre-treatment | Pre-onset | Delta 9 THC significantly decreased the infarct volume | Day 1 | NR |
| Leker et al. [40] | 2003 | Jerusalem | HU-210 | CB antagonist | Sprague-Dawley rats | pMCAO | Post-treatment | 1, 2, 4, or 6 h | Motor disability and infarct volumes were significantly reduced in animals treated with HU-210 | Day 3 | Motor disability and infarct volumes were significantly reduced in animals treated with HU-210 |
Pre-treatment, treatment administered before inducing stroke; post-treatment, treatment administered after inducing stroke; CB, cannabinoid; CB1, cannabinoid receptor type 1; CB2, cannabinoid receptor type 2; MCAO/r, middle cerebral artery occlusion with reperfusion; MCAO, middle cerebral artery occlusion; NR, not reported; N/A, not applicable; STAIR, Stroke Therapy Academic Industry Roundtable; h, hours; min, minutes.
Fig. 1.
PRISMA flowchart illustrating the systematic process of identification, screening, eligibility, and inclusion of studies in a systematic review and meta-analysis [58].
Fig. 5.
Forest plot based on specific CB antagonist drug used vs. control group, showing the standard mean difference in infarct volume.
Fig. 2.
Forest plot illustrating the standard mean difference in brain infarct volume between the experimental and control groups, exclusively utilizing CB agonist drugs.
Fig. 3.
Subgroup analysis based on specific CB agonist drug used vs. control group, showing the standard mean difference in infarct volume.
Fig. 4.
Subgroup analysis based on drugs targeting specific CB ligands, illustrating the standard mean difference in infarct volume.
Infarct Volume with CB Agonist
The analysis of infarct volume in response to CB agonist drugs showed a significant reduction in infarct volume within the experimental group compared to controls. The combined data from various agonist drugs revealed a substantial decrease, with a standardized mean difference (SMD) of −3.61 (95% CI: −4.41 to −2.81; p < 0.00001; I2 83%), as illustrated in online supplementary Figure 2. This suggests a significant effect of CB agonists in reducing the infarct volume. Due to heterogeneity, a leave-one-out analysis was conducted, and a forest plot is included in online supplementary Figure 2.
Further investigation through subgroup analysis categorized by specific drug types revealed varying levels of efficacy among different agonists. Notably, both ACEA and KN38-72717 showed a significant reduction in infarct volume, with a SMD of −5.40 (95% CI: −7.07, −3.74; p < 0.00001; I2 15%) for ACEA and −13.12 (95% CI: −17.86, −8.38; p < 0.00001; I2 83%) for KN38-72717, as depicted in online supplementary Figure 3. Additionally, TAK-937 (−8.50) and O-1966 (−5.35) also demonstrated significant reductions in infarct volume, highlighting their potential as therapeutic agents for ischemic events. Other compounds such as WIN 55212-2 (−2.22), NAE-18:4n-6 (−4.69), BCP (−3.46), NAE-18:5n-3 (−1.33), NAE-22:5n-6 (−0.24), JWH-133 (−1.86), and NAE-20:5n-3 (−1.35) have also exhibited a reduction in infarct volume but were not as significant as ACEA and KN38-72717.
Subgroup analysis based on ligand type also provided insight into the efficacy of CB agonists in reducing infarct volume. CB1 agonists emerged as particularly effective, producing a substantial reduction with an SMD of −4.55 (95% CI: −5.54, −3.57; p < 0.00001; I2 10%), as illustrated in online supplementary Figure 4. This highlights the specific targeting of CB1 receptors as a promising strategy for mitigating ischemic damage. Furthermore, CB2 agonists and non-selective agonists (CB1/CB2) also displayed significant effects, with SMDs of −3.69 (95% CI: −4.71, −2.67; p < 0.00001) and −3.60 (95% CI: −5.83, −1.36; p = 0.002), respectively.
Infarct Volume with CB Antagonist
Analysis of infarct volume in response to CB antagonists revealed a significant overall effect between the experimental and control groups, with a SMD of −2.12 (95% CI: −3.20, −1.04; p = 0.0004). A leave-one-out analysis was conducted, and a forest plot is included in the supplementary file. Subgroup analysis provided further insights into the efficacy of specific CB antagonists. SR141716, a CB1 antagonist, demonstrated a significant reduction in infarct volume with an SMD of −2.93 (95% CI: −4.37, −1.49; p < 0.0001), while LY320135, another CB1 antagonist, also showed a notable effect with an SMD of −2.55 (95% CI: −4.42, −0.67; p = 0.008). However, SR144528, a CB2 antagonist, did not exhibit a significant effect on infarct volume, with an SMD of 0.96 (95% CI: −0.32, 2.24; p = 0.14). In contrast, U0126, a CB antagonist, demonstrated a significant reduction in infarct volume, with an SMD of −1.23 (95% CI: −2.21, −0.26; p = 0.01). These findings highlight the varying efficacies of CB antagonists in reducing infarct volume, with CB1 antagonists showing more prominent effects than CB2 antagonists (online suppl. Fig. 5).
Neurological Scores
Neurological scores for both CB agonists and antagonists were evaluated across the experimental and control groups. The SMD for neurological scores with CB agonists was −0.63 (95% CI: −1.57, 0.31; p = 0.19), suggesting a potential improvement trend, although statistical significance was not reached. Similarly, CB antagonists showed an SMD of −0.84 (95% CI: −1.81, 0.13; p = 0.09), indicating a possible enhancement in neurological function, but not statistically significant (online suppl. Fig. 5). These findings suggest a promising yet inconclusive pattern towards enhanced neurological scores with both CB agonists and antagonists. Further investigations with larger cohorts may be necessary to unveil the precise impact of CB modulation on neurological outcomes. Such insights could pave the way for potential therapeutic interventions targeting conditions that affect the central nervous system.
Summary of Effects of CB Agonist and Antagonist Drugs on Infarct Volume
This study examined the impact of CB agonist and antagonist drugs on infarct volume using SMD analysis, along with 95% CI. Among CB agonists, drugs such as ACEA and WIN 55212-2 demonstrated negative SMD values, suggesting a potential decrease in infarct volume. Conversely, some agonists, such as KN38-72717 and O-1966, displayed substantial negative SMD values, indicating a more pronounced effect on reducing infarct volume (online suppl. Fig. 6a) For the antagonist drugs, SR141716 and LY320135 exhibited negative SMD values, indicating a potential reduction in infarct volume, whereas SR144528 showed a positive SMD value, suggesting a potential increase. U0126 also displayed a negative SMD value, implying a potential reduction in the infarct volume (online suppl. Fig. 6b) These findings underscore the complex and varied effects of CB agonists and antagonists on infarct volume, highlighting the importance of considering both the central tendency and uncertainty in interpreting the results.
Effect of CB Agonist and Antagonist Drugs on Experimental Infarct Volume over Time
The grouped bar plot illustrates the effect of the time of administration on the experimental infarct volume, categorized by CB agonist and antagonist drug classes. Overall, it can be observed that the SMD in infarct volume varies across different time points for both agonist and antagonist drug classes. For the CB agonist group (online suppl. Fig. 7a), the SMD tends to decrease over time, with the most substantial reduction observed between 2–3 h and 3–4 h post-administration. Conversely, in the CB antagonist group (online suppl. Fig. 7b), SMD fluctuated across time points, with no clear trend observed. Notably, the error bars representing the 95% CIs indicate the uncertainty associated with the SMD estimates, and values crossing zero are deemed statistically insignificant.
Publication Bias
Publication bias was assessed through the examination of funnel plots. Notably, the funnel plot representing the overall CB agonist data exhibited slight asymmetry. Subsequently, an Egger test was conducted, yielding an intercept value of −7.05, a standard error of 1.22, a t value of 5.79, and a p value of 0.000001. To further validate our findings, a Duval and Tweedie trim-and-fill test was performed, revealing no trimmed studies and minimal impact on the results, thus enhancing the accuracy of our findings. Similarly, a funnel plot was generated for the CB antagonist data, with an Egger intercept value of −5.04, a standard error of 1.51, a t value of 3.33, and a p value of 0.003. Once again, a Duval and Tweedie trim-and-fill analysis was undertaken, resulting in no trimmed studies and negligible alteration to the findings. All funnel plots have been included (online suppl. Fig. 2, 4).
Discussion
CBs have demonstrated therapeutic and neuroprotective effects in various models of stroke [59]. In particular, the impact of CBs on the outcomes of focal ischemic stroke has been a subject of debate for over 2 decades [60]. A systematic review and meta-analysis conducted by England et al. [9] not only revealed the significant influence of CBs on focal ischemic strokes but also the outcomes associated with drug dose and therapeutic time window [9]. It has been demonstrated that multiple CB drugs, including oral cannabidiol (CBD), oleoylethanolamide (OEA), and palmitoylethanolamide (PEA), exert their effects through receptors other than CB receptors [9, 54, 61–63]. Given the diverse actions of different CBs, further investigation of the role of CB receptors in focal ischemic strokes is essential to establish a comprehensive understanding of their involvement in the therapeutic cascade. This study aimed to investigate the function of CB receptor-related medications in the management of focal stroke.
This comprehensive meta-analysis has demonstrated that CB receptor agonists, such as CB1 and CB2 agonists, significantly decrease infarct volume in both transient and permanent ischemic models and enhance both early and late functional outcomes. Our meta-analysis demonstrated significant outcomes in terms of diminishing infarct volume with either CB1 or CB2 agonist, as illustrated in online supplementary Figure 2. Moreover, by conducting a subgroup analysis categorized by specific drug types, we discovered varying levels of efficacy among the different agonists. Importantly, ACEA, KN38-72717, and TAK-937 were found to be the most effective compounds, showing a considerable decrease in infarct volume with a SMD [14, 15, 64]. CB1 agonists alone reduce infarct volume more than CB2 agonists and non-selective CB receptor agonists [16, 65]. The role of the CB receptor is not well understood as significant results were also obtained for reducing infarct volume with a CB1 antagonist [29, 31]. The role of CB receptors is very well established, but it is not completely understood and is far from being clear. Therefore, future studies are of utmost importance to describe these discrepancies and their exact mechanisms and roles. Neurological scores were evaluated across experimental studies and for all the CB agonists and antagonists they showed a trend towards improvement but still not significant [5, 29, 57, 64, 66, 67].
The ECS has proved substantial immunomodulatory and protective effects on glial cells, and the pharmacological modulation of this signaling pathway has demonstrated promising neuroprotective effects in various neurological diseases [68–70]. Research suggests that the ECS plays a crucial role in regulating glial cell function, not only by influencing cell reactivity or phenotype polarization but also by promoting cell survival and preventing functional impairment [11]. Research on the ECS and CB2 receptors, particularly, has been extensive owing to their significant role in neuroinflammation and their presence in microglial cells [71]. This area of study has garnered much attention from researchers owing to its potential implications in treating various neurological disorders [71]. Multiple investigations have demonstrated encouraging results related to the activation of the CB2 receptor in reducing protein aggregation-related pathologies as well as in mitigating inflammation and several dementia-related symptoms [71, 72].
The neuroprotective effects of CBs may also be attributed to other receptor targets, such as the serotonin 1A receptor (5HT1A) [61]. For instance, CBD’s effects, which are not influenced by CB1, CB2, the transient receptor potential cation channel subfamily V member 1 (TRPV1) antagonism, are partially mediated through 5HT1A, leading to decreased infarct volume and improved cerebral blood flow [54, 62, 63]. Additionally, anti-inflammatory effects have yet to be linked to specific target sites, and further investigation is needed for known CB target sites such as TRPs and the peroxisome proliferator-activated receptors PPARs [73]. Endocannabinoids PEA and OEA have been associated with reduced cell death, edema, and inflammation [74]. OEA’s infarct-reducing effects are believed to be mediated through PPARα, as demonstrated by their absence in PPARα−/− mice and inhibition by a PPARα antagonist [67, 75, 76].
The neuroprotective effects of CBs have been also linked to their role in maintaining cerebral blood flow and preserving blood-brain barrier (BBB) integrity [51, 77]. For instance, a recent study revealed a notable elevation in IgG levels within the ipsilateral cortex of mice subjected to MCAO compared to sham-control mice [44]. Interestingly, the administration of VCE-004.8 effectively mitigated this increase, indicating a potential protective effect on the BBB. Specifically, the levels of IgG were significantly lower in the VCE-004.8 treated group compared to the vehicle group. This suggests that VCE-004.8 treatment may prevent the extravasation of immunoglobulins, thus safeguarding the integrity of the BBB. Furthermore, the study observed that VCE-004.8 treatment led to a reduction in active-matrix metalloproteinase-9 (MMP-9) levels in the brain [44]. MMP-9 is recognized for its involvement in ischemic stroke models, where it plays a crucial role in the degradation of tight junction proteins and extracellular matrix proteins, contributing to BBB breakdown and edema formation [78]. This finding underscores the potential of VCE-004.8 in modulating MMP-9 activity, thereby potentially mitigating BBB disruption associated with ischemic stroke.
While England et al. [9] identified neuroprotective effects of CBs like CBD and PEA (which act via non-CB receptors), our analysis isolated CB1/CB2 agonists (e.g., ACEA, KN38-72717) as uniquely effective in reducing infarct volume. Notably, CB1 antagonists (e.g., SR141716) also showed efficacy, a finding not emphasized in prior work, underscoring the complexity of CB receptor signaling [9].
Clinical research investigating CB1 and CB2 modulation for ischemic stroke remains limited and predominantly preclinical in scope. While preclinical studies highlight neuroprotective mechanisms such as reduced neuroinflammation and oxidative stress, few compounds have advanced to human trials. Dexanabinol, a synthetic intravenous CB, progressed to Phase III trials for traumatic brain injury but failed to demonstrate efficacy in improving neurological outcomes [79]. In contrast, EPIDIOLEX (CBD) received FDA approval in 2018 for two severe forms of seizers (Lennox-Gastaut and Dravet syndromes), validating CB modulation in neurological disorders [80]. Based on England et al. [9] and the current meta-analysis, taking a step in clinical ischemic stroke research is warranted. Trials must address critical barriers, including optimal therapeutic time windows, receptor subtype specificity and compatibility with reperfusion therapies like thrombectomy.
This systematic review and meta-analysis have significantly advanced our understanding of the role of CB in managing stroke patients. Our study’s specific inclusion criteria, rigorous analysis, and clear presentation of results represent a significant milestone in this area of research. Unlike previous systematic reviews, such as those by England et al. [9], our study focused exclusively on CB receptor-related drugs, enhancing the precision and relevance of our findings. Our extensive evaluation revealed several inadequacies in the existing literature, highlighting the need for further investigation, particularly in CB2 antagonists against mixed CB1/CB2 agonists, which could offer significant therapeutic benefits. Moreover, it is worth mentioning that the expression of CB2 receptors experiences a decline in the first 3 h post-MCAO, followed by a gradual rise within the 24-h timeframe [35].
Our findings face several limitations that may impact the way our findings are interpreted. Initially, there was substantial heterogeneity owing to the variety in the design of the individual studies. To address this, we used a random-effects model for analysis. Moreover, we conducted a subgroup analysis to further examine the studies and pinpoint those causing the heterogeneity. Additionally, we implemented a leave-one-out analysis to adjust for the heterogeneity, enhancing the robustness of our findings. Several studies exhibit relatively small sample sizes, emphasizing the significance of future research initiatives with larger participant cohorts. Undertaking studies with substantial sample sizes is essential to mitigate potential biases, increase statistical power, and ensure the overall quality of the research outcomes. Furthermore, the limited size of the study groups resulted in imprecise estimates of variance and, subsequently, the SMD. It is worth mentioning that we opted for the use of SMD rather than weighted mean differences to combine diverse scales to assess the same parameter. Although interpreting SMD may be less intuitive, this choice has enabled the inclusion of a considerably greater number of studies in this analysis. A limitation of our study is the absence of human-level investigations. We solely relied on animal-level studies due to the extensive body of research conducted at this level, whereas human-level studies were not included in our analysis.
Looking ahead, the implications of our findings hold promise for the development of novel therapeutic interventions targeting CB receptors in the management of ischemic stroke. The observed reduction in infarct volume with CB agonists, particularly CB agonists such as ACEA, KN38-72717, and TAK-937, suggests a potential avenue for the development of neuroprotective agents. Further investigation of the specific mechanisms underlying the neuroprotective effects of CBs could lead to the development of targeted therapies aimed at preserving neural tissue integrity and improving functional outcomes following stroke. Additionally, the varying efficacy of CB antagonists in reducing infarct volume highlights the complexity of CB receptor signaling and warrants further exploration of their therapeutic potential. Future research should focus on elucidating the precise mechanisms of CB receptor modulation and optimizing therapeutic strategies to enhance their clinical involvement in practice. Further studies assessing behavioral outcomes are warranted to have a broader understanding to the effects of CB receptors modulation in ischemic brain injury. Ultimately, the translation of these preclinical findings into clinical practice has the potential to revolutionize stroke management and improve patient outcomes.
Conclusion
This systematic review and meta-analysis underscore the significant potential of CB receptor modulation in mitigating infarct volume in animal models of focal ischemic stroke. Our findings revealed a consistent reduction in infarct volume with CB agonists, particularly CB agonists such as ACEA, KN38-72717, and TAK-937, suggesting their promising therapeutic efficacy. The subgroup analyses provided further insight into the varying levels of efficacy among different agonists and emphasized the need for continued exploration of their specific mechanisms of action. Moreover, CB antagonists, particularly CB1 antagonists such as SR141716, showed significant reductions in infarct volume but with varying degrees of effectiveness. Although promising trends were observed, the impact on neurological scores remained inconclusive, indicating the need for more extensive investigation with larger cohorts. Our study not only contributes to advancing the understanding of the role of CB receptors in stroke management but also underscores the need for future research to elucidate the underlying mechanisms and optimize therapeutic strategies.
Statement of Ethics
No interventions involving animals or humans were conducted, and no confidential data were utilized.
Conflict of Interest Statement
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this article.
Funding Sources
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Author Contributions
Hussain Al Dera contributed to conception and design; supervised the study; approved the final version of the manuscript on behalf of all authors; reviewed submitted version of manuscript; critically revised the article; drafted the article, and performed acquisition, analysis, and interpretation of data. Abdulrahman M. Khojah contributed to conception and design; supervised the study; reviewed submitted version of manuscript; critically revised the article; drafted the article; and performed acquisition, analysis, and interpretation of data. Abdulrazak Sakhakhni reviewed submitted version of manuscript, drafted the article, and contributed to acquisition of data. Syed Aga and Faisal Alamri reviewed submitted version of manuscript and critically revised the article. Abdulaziz Alqahtani drafted the article and contributed to acquisition of data. Fahad Alsharef and Shadell Alghamdi contributed to administrative/technical/material support and acquisition of data.
Funding Statement
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Data Availability Statement
The data used during the current study are available from the corresponding author upon reasonable request.
Supplementary Material.
References
- 1. Feske SK. Ischemic stroke. Am J Med. 2021;134(12):1457–64. [DOI] [PubMed] [Google Scholar]
- 2. Shafique MA, Shahid A, Sajjad L, Jaber MH, Haseeb A. The Nexus of malnutrition and stroke associated pneumonia: insights and strategies for improved outcomes. J Med Surg Public Health. 2024;2:100045. [Google Scholar]
- 3. Jolugbo P, Ariëns RA. Thrombus composition and efficacy of thrombolysis and thrombectomy in acute ischemic stroke. Stroke. 2021;52(3):1131–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Li Y, Zhang J. Animal models of stroke. Anim Model Exp Med. 2021b;4(3):204–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. McBean DE, Kelly PA. Rodent models of global cerebral ischemia: a comparison of two-vessel occlusion and four-vessel occlusion. Gen Pharmacol. 1998;30(4):431–4. [DOI] [PubMed] [Google Scholar]
- 6. Mhairi MI. New models of focal cerebral ischaemia. Br J Clin Pharmacol. 1992;34(4):302–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Lu HC, Mackie K. Review of the endocannabinoid system. Cogn Neurosci Neuroimaging. 2021;6:607–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Karimian Azari E, Kerrigan A, O’Connor A. Naturally occurring cannabinoids and their role in modulation of cardiovascular health. J Diet Suppl. 2020;17(5):625–50. [DOI] [PubMed] [Google Scholar]
- 9. England TJ, Hind WH, Rasid NA, O’Sullivan SE. Cannabinoids in experimental stroke: a systematic review and meta-analysis. J Cereb Blood Flow Metab. 2015;35(3):348–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Cristino L, Bisogno T, Di Marzo V. Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat Rev Neurol. 2020;16(1):9–29. [DOI] [PubMed] [Google Scholar]
- 11. Vicente-Acosta A, Ceprian M, Sobrino P, Pazos MR, Loría F. Cannabinoids as glial cell modulators in ischemic stroke: implications for neuroprotection. Front Pharmacol. 2022;13:888222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Stroke Therapy Academic Industry Roundtable STAIR . Recommendations for standards regarding preclinical neuroprotective and restorative drug development. Stroke. 1999;30(12):2752–8. [DOI] [PubMed] [Google Scholar]
- 13. Gibson CL, Gray LJ, Bath PMW, Murphy SP. Progesterone for the treatment of experimental brain injury; a systematic review. Brain. 2008;131(Pt 2):318–28. [DOI] [PubMed] [Google Scholar]
- 14. Yang S, Hu B, Wang Z, Zhang C, Jiao H, Mao Z, et al. Cannabinoid CB1 receptor agonist ACEA alleviates brain ischemia/reperfusion injury via CB1-Drp1 pathway. Cell Death Discov. 2020;6:102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Bai F, Guo F, Jiang T, Wei H, Zhou H, Yin H, et al. Arachidonyl-2-Chloroethylamide alleviates cerebral ischemia injury through glycogen synthase kinase-3β-mediated mitochondrial biogenesis and functional improvement. Mol Neurobiol. 2017;54(2):1240–53. [DOI] [PubMed] [Google Scholar]
- 16. Caltana L, Saez TM, Aronne MP, Brusco A. Cannabinoid receptor type 1 agonist ACEA improves motor recovery and protects neurons in ischemic stroke in mice. J Neurochem. 2015;135(3):616–29. [DOI] [PubMed] [Google Scholar]
- 17. Sun J, Fang YQ, Ren H, Chen T, Guo JJ, Yan J, et al. WIN55,212-2 protects oligodendrocyte precursor cells in stroke penumbra following permanent focal cerebral ischemia in rats. Acta Pharmacol Sin. 2013;34(1):119–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Yokubaitis CG, Jessani HN, Li H, Amodea AK, Ward SJ. Effects of cannabidiol and beta-caryophyllene alone or in combination in a mouse model of permanent ischemia. Int J Mol Sci. 2021;22(6):2866. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Yang M, Lv Y, Tian X, Lou J, An R, Zhang Q, et al. Neuroprotective effect of β-caryophyllene on cerebral ischemia-reperfusion injury via regulation of necroptotic neuronal death and inflammation: in vivo and in vitro. Front Neurosci. 2017;11:583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Pottier G, Gómez-Vallejo V, Padro D, Boisgard R, Dollé F, Llop J. PET imaging of cannabinoid type 2 receptors with [11C]A-836339 did not evidence changes following neuroinflammation in rats. J Cerebr Blood Flow Metabol. 2017;37:1163–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ronca RD, Myers AM, Ganea D, Tuma RF, Walker EA, Ward SJ. A selective cannabinoid CB2 agonist attenuates damage and improves memory retention following stroke in mice. Life Sci. 2015;138:72–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Yu SJ, Reiner D, Shen H, Wu KJ, Liu QR, Wang Y. Time-dependent protection of CB2 receptor agonist in stroke. PLoS One. 2015;10(7):e0132487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Choi IY, Ju C, Anthony Jalin AMA, Lee DI, Prather PL, Kim WK. Activation of cannabinoid CB2 receptor-mediated AMPK/CREB pathway reduces cerebral ischemic injury. Am J Pathol. 2013;182(3):928–39. [DOI] [PubMed] [Google Scholar]
- 24. Zarruk JG, Fernández-López D, García-Yébenes I, García-Gutiérrez MS, Vivancos J, Nombela F, et al. Cannabinoid type 2 receptor activation downregulates stroke-induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke. 2012;43(1):211–9. [DOI] [PubMed] [Google Scholar]
- 25. Shirazi RS, Vyssotski M, Lagutin K, Thompson D, MacDonald C, Luscombe V, et al. Neuroprotective activity of new Δ3‐N‐acylethanolamines in a focal ischemia stroke model. Lipids. 2022;57(1):17–31. [DOI] [PubMed] [Google Scholar]
- 26. Murakami K, Suzuki M, Suzuki N, Hamajo K, Tsukamoto T, Shimojo M. Cerebroprotective effects of TAK-937, a novel cannabinoid receptor agonist, in permanent and thrombotic focal cerebral ischemia in rats: therapeutic time window, combination with t-PA and efficacy in aged rats. Brain Res. 2013;1526:84–93. [DOI] [PubMed] [Google Scholar]
- 27. Suzuki N, Suzuki M, Murakami K, Hamajo K, Tsukamoto T, Shimojo M. Cerebroprotective effects of TAK-937, a cannabinoid receptor agonist, on ischemic brain damage in middle cerebral artery occluded rats and non-human primates. Brain Res. 2012;1430:93–100. [DOI] [PubMed] [Google Scholar]
- 28. Schmidt W, Schäfer F, Striggow V, Fröhlich K, Striggow F. Cannabinoid receptor subtypes 1 and 2 mediate long-lasting neuroprotection and improve motor behavior deficits after transient focal cerebral ischemia. Neuroscience. 2012;227:313–26. [DOI] [PubMed] [Google Scholar]
- 29. Reichenbach ZW, Li H, Ward SJ, Tuma RF. The CB1 antagonist, SR141716A, is protective in permanent photothrombotic cerebral ischemia. Neurosci Lett. 2016;630:9–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Jalin AMA, Rajasekaran M, Prather PL, Kwon JS, Gajulapati V, Choi Y, et al. Non-selective cannabinoid receptor antagonists, hinokiresinols reduce infiltration of microglia/macrophages into ischemic brain lesions in rat via modulating 2-arachidonolyglycerol-induced migration and mitochondrial activity. PLoS One. 2015;10:e0141600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Bravo-Ferrer I, Cuartero MI, Zarruk JG, Pradillo JM, Hurtado O, Romera VG, et al. Cannabinoid type-2 receptor drives neurogenesis and improves functional outcome after stroke. Stroke. 2017;48(1):204–12. [DOI] [PubMed] [Google Scholar]
- 32. Zhang M, Mahadevan A, Amere M, Li H, Ganea D, Tuma RF. Unique effects of compounds active at both cannabinoid and serotonin receptors during stroke. Transl Stroke Res. 2012;3:348–56. [DOI] [PubMed] [Google Scholar]
- 33. Zhang M, Adler MW, Abood ME, Ganea D, Jallo J, Tuma RF. CB2 receptor activation attenuates microcirculatory dysfunction during cerebral ischemic/reperfusion injury. Microvasc Res. 2009;78(1):86–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Zhang M, Martin BR, Adler MW, Razdan RK, Jallo JI, Tuma RF. Cannabinoid CB2 receptor activation decreases cerebral infarction in a mouse focal ischemia/reperfusion model. J Cereb Blood Flow Metab. 2007;27(7):1387–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Zhang M, Martin B, Adler M, Razdan R, Ganea D, Tuma R. Modulation of the balance between cannabinoid CB1 and CB2 receptor activation during cerebral ischemic/reperfusion injury. Neuroscience. 2008;152(3):753–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Hu B, Wang Q, Chen Y, Du J, Zhu X, Lu Y, et al. Neuroprotective effect of WIN 55,212-2 pretreatment against focal cerebral ischemia through activation of extracellular signal-regulated kinases in rats. Eur J Pharmacol. 2010;645(1–3):102–7. [DOI] [PubMed] [Google Scholar]
- 37. Berger C, Schmid PC, Schabitz WR, Wolf M, Schwab S, Schmid HHO. Massive accumulation of N‐acylethanolamines after stroke. Cell signaling in acute cerebral ischemia? J Neurochem. 2004;88(5):1159–67. [DOI] [PubMed] [Google Scholar]
- 38. Muthian S, Rademacher D, Roelke C, Gross G, Hillard C. Anandamide content is increased and CB1 cannabinoid receptor blockade is protective during transient, focal cerebral ischemia. Neuroscience. 2004;129(3):743–50. [DOI] [PubMed] [Google Scholar]
- 39. Hayakawa K, Mishima K, Nozako M, Hazekawa M, Ogata A, Fujioka M, et al. Delta9-tetrahydrocannabinol (Delta9-THC) prevents cerebral infarction via hypothalamic-independent hypothermia. Life Sci. 2007;80(16):1466–71. [DOI] [PubMed] [Google Scholar]
- 40. Leker RR, Gai N, Mechoulam R, Ovadia H. Drug-induced hypothermia reduces ischemic damage: effects of the cannabinoid HU-210. Stroke. 2003;34(8):2000–6. [DOI] [PubMed] [Google Scholar]
- 41. Landucci E, Scartabelli T, Gerace E, Moroni F, Pellegrini-Giampietro DE. CB1 receptors and post-ischemic brain damage: studies on the toxic and neuroprotective effects of cannabinoids in rat organotypic hippocampal slices. Neuropharmacology. 2011;60(4):674–82. [DOI] [PubMed] [Google Scholar]
- 42. Xu BT, Li MF, Chen KC, Li X, Cai NB, Xu JP, et al. Mitofusin-2 mediates cannabidiol-induced neuroprotection against cerebral ischemia in rats. Acta Pharmacol Sin. 2023;44(3):499–512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Wang S, Sun H, Liu S, Wang T, Guan J, Jia J. Role of hypothalamic cannabinoid receptors in post-stroke depression in rats. Brain Res Bull. 2016;121:91–7. [DOI] [PubMed] [Google Scholar]
- 44. Lavayen BP, Yang C, Larochelle J, Liu L, Tishko RJ, De Oliveira ACP, et al. Neuroprotection by the cannabidiol aminoquinone VCE-004.8 in experimental ischemic stroke in mice. Neurochem Int. 2023;165:105508. [DOI] [PubMed] [Google Scholar]
- 45. Hosoya T, Fukumoto D, Kakiuchi T, Nishiyama S, Yamamoto S, Ohba H, et al. In vivo TSPO and cannabinoid receptor type 2 availability early in post-stroke neuroinflammation in rats: a positron emission tomography study. J Neuroinflammation. 2017;14(1):69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Karwad M, Couch D, Wright K, Tufarelli C, Larvin M, Lund J, et al. Endocannabinoids and endocannabinoid-like compounds modulate hypoxia-induced permeability in CaCo-2 cells via CB1, TRPV1, and PPARα. Biochem Pharmacol. 2019b;168:465–72. [DOI] [PubMed] [Google Scholar]
- 47. Abdel Mageed SS, Ammar RM, Nassar NN, Moawad H, Kamel AS. Role of PI3K/Akt axis in mitigating hippocampal ischemia-reperfusion injury via CB1 receptor stimulation by paracetamol and FAAH inhibitor in rat. Neuropharmacology. 2022b;207:108935. [DOI] [PubMed] [Google Scholar]
- 48. Mori MA, Meyer E, Da Silva FF, Milani H, Guimarães FS, Oliveira RMW. Differential contribution of CB1, CB2, 5‐HT1A, and PPAR‐γ receptors to cannabidiol effects on ischemia‐induced emotional and cognitive impairments. Eur J Neurosci. 2021;53(6):1738–51. [DOI] [PubMed] [Google Scholar]
- 49. Ward SJ, Castelli F, Reichenbach ZW, Tuma RF. Surprising outcomes in cannabinoid CB1/CB2 receptor double knockout mice in two models of ischemia. Life Sci. 2018;195:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Zani A, Braida D, Capurro V, Sala M. Delta9-tetrahydrocannabinol (THC) and AM 404 protect against cerebral ischaemia in gerbils through a mechanism involving cannabinoid and opioid receptors. Br J Pharmacol. 2007;152(8):1301–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Chi OZ, Barsoum S, Grayson J, Hunter C, Liu X, Weiss HR. Effects of cannabinoid receptor agonist WIN 55,212-2 on blood-brain barrier disruption in focal cerebral ischemia in rats. Pharmacology. 2012;89(5–6):333–8. [DOI] [PubMed] [Google Scholar]
- 52. Tang J, Tao Y, Tan L, Yang L, Niu Y, Chen Q, et al. Cannabinoid receptor 2 attenuates microglial accumulation and brain injury following germinal matrix hemorrhage via ERK dephosphorylation in vivo and in vitro. Neuropharmacology. 2015;95:424–33. [DOI] [PubMed] [Google Scholar]
- 53. Suzuki N, Suzuki M, Hamajo K, Murakami K, Tsukamoto T, Shimojo M. Contribution of hypothermia and CB1 receptor activation to protective effects of TAK-937, a cannabinoid receptor agonist, in rat transient MCAO model. PLoS One. 2012;7:e40889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Mishima K, Hayakawa K, Abe K, Ikeda T, Egashira N, Iwasaki K, et al. Cannabidiol prevents cerebral infarction via a serotonergic 5-hydroxytryptamine 1A receptor–dependent mechanism. Stroke. 2005;36(5):1077–82. [DOI] [PubMed] [Google Scholar]
- 55. Teichner A, Ovadia H, Lavie G, Leker R. Combination of dexanabinol and tempol in focal cerebral ischemia: is there a ceiling effect? Exp Neurol. 2003;182(2):353–60. [DOI] [PubMed] [Google Scholar]
- 56. Bonfils PK, Reith J, Hasseldam H, Johansen FF. Estimation of the hypothermic component in neuroprotection provided by cannabinoids following cerebral ischemia. Neurochem Int. 2006;49(5):508–18. [DOI] [PubMed] [Google Scholar]
- 57. Murikinati S, Jüttler E, Keinert T, Ridder DA, Muhammad S, Waibler Z, et al. Activation of cannabinoid 2 receptors protects against cerebral ischemia by inhibiting neutrophil recruitment. FASEB J. 2010;24(3):788–98. [DOI] [PubMed] [Google Scholar]
- 58. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. BMJ. 2009:339:b2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Greco R, Demartini C, Zanaboni A, Tumelero E, Elisa C, Persico A, et al. Characterization of CB2 receptor expression in peripheral blood monocytes of acute ischemic stroke patients. Transl Stroke Res. 2021;12(4):550–8. [DOI] [PubMed] [Google Scholar]
- 60. Fernández-Ruiz J, Moro MA, Martínez-Orgado J. Cannabinoids in neurodegenerative disorders and stroke/brain trauma: from preclinical models to clinical applications. Neurotherapeutics. 2015;12(4):793–806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Hayakawa K, Mishima K, Abe K, Hasebe N, Takamatsu F, Yasuda H, et al. Cannabidiol prevents infarction via the non-CB1 cannabinoid receptor mechanism. Neuroreport. 2004;15:2381–5. [DOI] [PubMed] [Google Scholar]
- 62. Hayakawa K, Mishima K, Nozako M, Hazekawa M, Irie K, Fujioka M, et al. Delayed treatment with cannabidiol has a cerebroprotective action via a cannabinoid receptor‐independent myeloperoxidase‐inhibiting mechanism. J Neurochem. 2007;102(5):1488–96. [DOI] [PubMed] [Google Scholar]
- 63. Hayakawa K, Mishima K, Nozako M, Ogata A, Hazekawa M, Liu AX, et al. Repeated treatment with cannabidiol but not Delta9-tetrahydrocannabinol has a neuroprotective effect without the development of tolerance. Neuropharmacology. 2007;52(4):1079–87. [DOI] [PubMed] [Google Scholar]
- 64. Ma L, Jia J, Niu W, Jiang T, Zhai Q, Yang L, et al. Mitochondrial CB1 receptor is involved in ACEA-induced protective effects on neurons and mitochondrial functions. Sci Rep. 2015;5:12440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Zhuang Q, Dai C, Yang L, Wen H, Wang H, Jiang X, et al. Stimulated CB1 cannabinoid receptor inducing ischemic tolerance and protecting neuron from cerebral ischemia. Cent Nerv Syst Agents Med Chem. 2017;17(2):141–50. [DOI] [PubMed] [Google Scholar]
- 66. Kiran S, Rakib A, Moore BM, Singh UP. Cannabinoid receptor 2 (CB2) inverse agonist SMM-189 induces expression of endogenous CB2 and protein kinase a that differentially modulates the immune response and suppresses experimental colitis. Pharmaceutics. 2022;14(5):936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Romano A, Friuli M, Eramo B, Gallelli CA, Koczwara JB, Azari EK, et al. “To brain or not to brain”: evaluating the possible direct effects of the satiety factor oleoylethanolamide in the central nervous system. Front Endocrinol. 2023;14:1158287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Egaña-Huguet J, Soria-Gómez E, Grandes P. The endocannabinoid system in glial cells and their profitable interactions to treat epilepsy: evidence from animal models. Int J Mol Sci. 2021;22(24):13231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. De Almeida V, Martins-De-Souza D. Cannabinoids and glial cells: possible mechanism to understand schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2018;268(7):727–37. [DOI] [PubMed] [Google Scholar]
- 70. Luongo L, Maione S, Di Marzo V. Endocannabinoids and neuropathic pain: focus on neuron–glia and endocannabinoid–neurotrophin interactions. Eur J Neurosci. 2014;39(3):401–8. [DOI] [PubMed] [Google Scholar]
- 71. Vuic B, Milos T, Tudor L, Konjevod M, Nikolac Perkovic M, Jazvinscak Jembrek M, et al. Cannabinoid CB2 receptors in neurodegenerative proteinopathies: new insights and therapeutic potential. Biomedicines. 2022;10(12):3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Mukadam AS, Miller LVC, Smith AE, Vaysburd M, Sakya SA, Sanford S, et al. Cytosolic antibody receptor TRIM21 is required for effective tau immunotherapy in mouse models. Science. 2023;379(6639):1336–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Lillo J, Raïch I, Silva L, Zafra DA, Lillo A, Ferreiro-Vera C, et al. Regulation of expression of cannabinoid CB2 and serotonin 5HT1A receptor complexes by cannabinoids in animal models of hypoxia and in oxygen/glucose-deprived neurons. Int J Mol Sci. 2022;23(17):9695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Ahmad A, Genovese T, Impellizzeri D, Crupi R, Velardi E, Marino A, et al. Reduction of ischemic brain injury by administration of palmitoylethanolamide after transient middle cerebral artery occlusion in rats. Brain Res. 2012;1477:45–58. [DOI] [PubMed] [Google Scholar]
- 75. Sun Y, Alexander SPH, Garle MJ, Gibson CL, Hewitt K, Murphy SP, et al. Cannabinoid activation of PPAR alpha; a novel neuroprotective mechanism. Br J Pharmacol. 2007;152(5):734–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Zhou Y, Yang L, Ma A, Zhang X, Li W, Yang W, et al. Orally administered oleoyl ethanolamide protects mice from focal cerebral ischemic injury by activating peroxisome proliferator-activated receptor α. Neuropharmacology. 2012;63(2):242–9. [DOI] [PubMed] [Google Scholar]
- 77. Hagan K, Varelas P, Zheng H. Endocannabinoid system of the blood–brain barrier: current understandings and therapeutic potentials. Cannabis Cannabinoid Res. 2022;7(5):561–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Yang C, Hawkins KE, Doré S, Candelario-Jalil E. Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am J Physiol Cel Physiol. 2019;316(2):C135–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Maas AI, Murray G, Henney H, Kassem N, Legrand V, Mangelus M, et al. Efficacy and safety of dexanabinol in severe traumatic brain injury: results of a phase III randomised, placebo-controlled, clinical trial. Lancet Neurol. 2006;5(1):38–45. [DOI] [PubMed] [Google Scholar]
- 80. US Food and Drug Administration (FDA) . FDA approves first drug comprised of an active ingredient derived from marijuana to treat rare, severe forms of epilepsy [Press release]. 2018. Available from: https://www.fda.gov/news-events/press-announcements/fda-approves-first-drug-comprised-active-ingredient-derived-marijuana-treat-rare-severe-forms [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data used during the current study are available from the corresponding author upon reasonable request.





