Previous Article in Journal
Sepsis Biomarkers: What Surgeons Need to Know
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endocannabinoid System in Sepsis: A Scoping Review

by
Brandon Thai
1,
Hideaki Yamamoto
2,
Aristides Koutrouvelis
3 and
Satoshi Yamamoto
3,4,*
1
School of Medicine, University of Texas Medical Branch, Galveston, TX 77555, USA
2
Division of Biological Science, University of California San Diego, La Jolla, CA 92093, USA
3
Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
4
Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, CA 94143, USA
*
Author to whom correspondence should be addressed.
Anesth. Res. 2025, 2(4), 24; https://doi.org/10.3390/anesthres2040024
Submission received: 5 August 2025 / Revised: 19 September 2025 / Accepted: 15 October 2025 / Published: 24 October 2025

Abstract

Sepsis is a life-threatening syndrome marked by a dysregulated host response to infection, resulting in systemic inflammation, organ dysfunction, and high mortality globally. Despite advancements in supportive care, effective immunomodulatory therapies remain elusive, necessitating exploration of novel biological pathways and subsequent therapeutic development. The endocannabinoid system (ECS), which regulates immune function and homeostasis, has emerged as a key modulator of immunological and metabolic pathways central to sepsis pathophysiology. The ECS mediates its effects through endogenous ligands, G-protein-coupled cannabinoid receptors (CB1 and CB1), and regulatory enzymes that control its synthesis and degradation. Following PRISMA-ScR guidelines, this scoping review synthesizes current evidence on the mechanistic roles of ECS components in experimental and clinical models of sepsis, identifies knowledge gaps, and delineates future areas of work. A comprehensive literature search across multiple databases without restrictions on date or publication type was executed to ensure broad coverage of original studies investigating ECS mechanisms and their intersection with sepsis and septic shock. Across 53 studies, CB2 receptor activation was consistently associated with anti-inflammatory process, organ-protective outcomes, and increased survival rates against septic challenges in preclinical rodent models. CB1 receptor activation trends, however, showed context dependent outcomes. Central antagonism improved hemodynamics and survival rate, but peripheral effects varied with cell type and timing. Non-canonical ECS components (TRPV1, GPR55, PPAR-α, FAAH, MAGL) also contributed to neuroimmune and metabolic regulation. Limited clinical data linked ECS lipid profiles and gene expression with sepsis severity and outcomes. Collectively, ECS modulation, particularly CB2 agonism, TRPV1 activation, and FAAH/MAGL inhibition, shows promise in mitigating sepsis-induced inflammation and organ dysfunction. However, complex, context-dependent effects, especially involving CB1, highlight the need for precision-targeted therapeutic approaches. Further preclinical research is needed to expand generalizable trends to allow translational research to refine ECS-based interventions for sepsis management.

Graphical Abstract

1. Introduction

Sepsis continues to be recognized as one of the foremost contributors to global morbidity and mortality rates, characterized as a dysregulated response from the host to infectious agents that leads to an extensive and often deleterious cascade of inflammation, subsequent organ dysfunction, and outcomes that can frequently result in death. Current epidemiological estimates indicate that there are approximately 49 million cases of sepsis reported annually, with around 11 million fatalities attributed to this condition, which collectively accounts for nearly 20% of the total mortality observed worldwide [1]. In addition to the significant clinical implications of sepsis, it also imposes a considerable economic burden on healthcare systems. This condition is consistently identified as the most financially burdensome ailment encountered within hospital settings, a status that is largely driven by the necessity for prolonged stays in intensive care units (ICUs), the execution of invasive medical procedures, and the alarming rates of patient readmission that are often associated with sepsis. In the context of the United States, it has been reported that the annual financial expenditures related to the care and management of sepsis surpassed $38 billion as of the year 2017 [2].
Individuals who survive an episode of sepsis frequently confront a range of long-term health complications, which may include, but are not limited to, cognitive dysfunction, chronic kidney disease, and various cardiovascular issues [3]. Despite significant advancements in the provision of supportive medical care, there remains a notable absence of immunomodulatory therapies that have consistently proven to offer a tangible survival advantage to patients suffering from sepsis. The inherent clinical variability of sepsis—spanning from initial hyperinflammatory responses characterized by cytokine storms to the late-phase phenomenon of immunoparalysis—has introduced considerable challenges in the formulation of effective targeted therapeutic interventions. In light of its profound global implications, the World Health Organization has formally classified sepsis as a crucial public health concern and has identified it as the leading cause of mortality among children around the globe [4]. The repeated shortcomings observed in various immunomodulatory therapeutic strategies, which include corticosteroids, anti–TNF-α agents, and immune checkpoint inhibitors, highlight the pressing necessity for the development of innovative mechanistic understandings and novel therapeutic modalities [5,6,7,8].
The endocannabinoid system (ECS), which is a complex lipid-based signaling network that consists of endogenous ligands, G-protein-coupled receptors—primarily CB1 and CB2—and associated metabolic enzymes, has garnered attention as a significant regulator of immune homeostasis and inflammation within the body [9]. Initially identified for its involvement in neurobehavioral processes, the activity of the ECS is now recognized to exert influence over a broad spectrum of physiological functions, which encompass vascular tone regulation, cellular metabolic processes, and the differentiation of immune cells [10]. Considering the perturbations of these critical pathways that occur during sepsis, the modulation of the ECS represents a particularly compelling therapeutic opportunity for intervention.
Preclinical studies spanning murine and rat models of polymicrobial sepsis, endotoxemia, acute respiratory distress syndrome (ARDS), and septic encephalopathy have not revealed distinct ECS-mediated mechanisms [11,12,13,14,15]. CB2 receptor activation consistently exerts anti-inflammatory effects by tempering neutrophil influx, repressing pyroptosis-related pathways (e.g., NLRP3–caspase-1–GSDMD), and promoting immunoregulatory phenotypes such as M2 macrophages and FOXP3+ T cells [16,17,18,19]. Conversely, the signaling of the CB1 receptor demonstrates a dichotomous influence, whereby central antagonism may mitigate septic hypotension and cognitive deterioration, whereas peripheral modulation of CB1 may affect metabolic and immunological functions [20,21].
Emerging data also highlights the role of non-canonical ECS actors—such as TRPV1, GPR55, and PPAR-α—and enzymatic regulators like FAAH and MAGL, revealing intricate layers of neuroimmune crosstalk, metabolic reprogramming, and organ-specific protection [22,23,24,25,26]. Clinical observations, albeit limited, lend further credibility to ECS relevance, with circulating ligand profiles and transcriptomic signatures correlating with sepsis outcomes [27,28,29,30].
Given the breadth and heterogeneity of ECS research in sepsis, including variations in animal models, ligands, dosing, strategies, and outcomes, a scoping review is the most appropriate direction to map existing evidence. This scoping review aims to synthesize the mechanistic landscape of ECS in sepsis by cataloging experimental models, receptor-specific pathways, and immunological outcomes, thereby identifying knowledge gaps and guiding future translational efforts. This methodology allows for comprehensive data review while acknowledging the limitations of comparability across studies. Through critical evaluation of the literature, we explore whether ECS modulation can fulfill the long-standing therapeutic void in sepsis management and pave the way toward precision-targeted interventions.

2. Methods

This scoping review was guided by the framework developed by Arksey and O’Malley (2005) [31]. The purpose of this review is to examine the extent and breadth of available original literature for the ECS and its involvement with sepsis. As such, critical appraisal of individual sources is not required. To ensure adherence to structured, systematic reporting, this scoping review followed the PRISMA-ScR (Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews) [32] methodology and checklist.

2.1. Protocol and Registration

A review protocol was developed and registered on the Open Science Framework (OSF) [33]. The methods were established a priori and adhered to throughout the review process. While scoping review protocol registrations are not required, this registration ensures transparency and further aligns with PRISMA-ScR recommendations.

2.2. Information Sources

Following consultation with a qualified medical librarian, a comprehensive literature search was conducted across several medical and scientific databases, including Ovid (Medline), Central, Scopus, and CINAHL, all of which were executed on 13 June 2025. The objective was to identify studies investigating the mechanisms of the endocannabinoid system (ECS) during sepsis and septic shock. The search used controlled vocabulary to increase coverage such as: MeSH terms (e.g., “Sepsis”, “Endocannabinoids”), keywords (e.g., “bloodstream infection”) and Boolean operators (AND/OR). A key challenge in searching this field was a lack of standardized terminology and indexing. Relevant papers occasionally described individual components (e.g., CB1, CB2) without explicitly using “endocannabinoid system” as a keyword or may not have been indexed under our searched terms. Additionally, given the relatively niche research intersection between sepsis and ECS, there were no constraints regarding animal model, publication year, subject type, or publication type. These factors may have increased the initial pool of records, but ensure broad coverage of relevant terms and their subcategories. (for full search strategy see Appendix A Table A1)

2.3. Eligibility Criteria and Study Selection

This scoping review focuses on studies that investigate mechanisms of the ECS as it relates to sepsis and septic shock. Specifically, studies were included if they: (i) contain original experimental data with a mechanistic focus of the ECS; (ii) have direct experimental modulation of at least one of the ECS’s components (e.g., receptor expression, ligand modulation, enzymatic activity); (iii) utilize a recognized septic model (e.g., cecal ligation and puncture (CLP), lipopolysaccharide (LPS), bacterial infection) or experimental samples from septic patients; and (iv) were published in English. Studies were excluded if they: (i) did not contain original experimental data (reviews, editorials, etc.); (ii) did not elicit mechanistic understanding through direct modulation of ECS components (e.g., only clinical/observational associations); (iii) lacked an established sepsis model; or (iv) were non-English. Using these criteria, the original 253 results were screened independently by 2 authors (B.T. and H.Y.) for a final selection of 53 included studies. Any discrepancy was resolved through consensus among the authors. This process was facilitated using Rayyan (Rayyan Systems Inc.; https://www.rayyan.ai), an online systematic review management platform accessed in June 2025.
Although we did not perform a formal risk-of-bias assessment in line with scoping review methodology, it is notable that many included studies incorporated internal validity checks. For example, several studies confirmed receptor specificity by combining agonist administration with receptor antagonism or by comparing wild type to receptor knockout animals. These “opposite-control” designs strengthen the causality, ensuring that observed effects are attributable to modulation of the ECS and not due to unknown off-target interactions [34].

2.4. Data Charting

Data extraction was independently performed by the screeners using Microsoft Excel, with disputes resolved by author consensus. After several revisions, the charted data was finalized as: first author, year of publication, animal model system (e.g., C57BL/6 mice), sepsis model (e.g., CLP, LPS), experimental intervention, key mechanistic findings, outcomes, and summary. Study interventions not relevant to the scope of the review or our understanding of the study were excluded (see Appendix A for full detailed data chart). The extracted data was summarized by grouping individual studies based on their investigated ECS component(s) (e.g., CB1, CB2, enzyme, etc.) along with calculating percentages of data columns to show trends in study design.
As this was a scoping review, no formal statistical analyses or meta-analyses were performed. Instead, results were synthesized descriptively. Frequencies and proportions were calculated by dividing the number of studies reporting a given characteristic (e.g., model type) by the total number of studies included. Percentages are displayed to summarize trends across the various study categories (Table 1 and Table 2).

3. Results

3.1. Study Identification and Inclusion

In the initial search conducted by the librarian, we identified 253 studies across 4 databases. Using an automated Rayyan program, 73 duplicate studies were removed following manual approval from the authors. 180 records remaining for screening. Of the 180, 106 records were excluded based on title and abstract due to lack of relevance or adherence to the stated inclusion criteria. The remaining 74 records were sought for retrieval, with 5 being unavailable. Thus, 69 studies were available for full-text eligibility screening, of which 16 were excluded due to failure to meet criteria as listed in the PRIMSA flow diagram (Figure 1). A final total of 53 studies remained for data extraction and further analysis. This methodology ensures a more comprehensive review that should reflect the rigor as defined by PRISMA-ScR guidelines [32].
Each stage of the study selection is listed, with showcased reasons for narrowing of the article pool.
PRISMA, Preferred Reporting Items for Systematic Review and Meta-Analyses; ECS, endocannabinoid system

3.2. Characteristics of Sources

A comprehensive overview of the proportions pertaining to the animal models utilized in the included studies, the sepsis models employed, the ECS targets investigated, the organ tissues examined, and the respective publication years are presented in Table 1. An elaborated account of the extracted data is provided in Appendix A.
Among the 53 studies incorporated in this analysis, a notable augmentation in publication frequency was observed post-2020 (36%), potentially associated with advancements in technology and heightened focus on immune dysregulation in the context of the COVID-19 pandemic (2020+) (Figure 2). A majority of the investigations concentrated on murine models (58%) employing lipopolysaccharide (LPS) (70%) to simulate a septic condition. LPS constitutes the predominant percentage within its corresponding study characteristic across all classifications, yielding findings that support the involvement of Gram-negative bacteria. Additionally, lung tissue and the cannabinoid receptor type 2 (CB2R) were subjects of substantial inquiry, attributed to the extensive distribution of CB2R among immune cells and the prevalent pulmonary manifestations associated with sepsis.
This plots the number of publications for the included studies over time. It is noted that 3 of the 5 highest years for publications are after 2020, indicating growing interest in ECS immunoregulatory effects.

3.3. Study Results and Outcomes

A total of fifty-three scholarly investigations were incorporated into the conclusive analysis and categorized thematically according to their principal endocannabinoid system (ECS) target of action. The majority of these studies, to varying extents, examined the cannabinoid receptor type 2 (CB2R) (42%), revealing two predominant patterns: the activation of CB2R is associated with the promotion of anti-inflammatory responses and the enhancement of survival via organ-protective mechanisms (e.g., amelioration of lung injury [16,35,36,37,38,39,40,41] and attenuation of neuroinflammation [42,43]) (Table 2). In contrast, it was frequently demonstrated that the inhibition of cannabinoid receptor type 1 (CB1R) mitigates the severity of sepsis in 13 out of the 23 studies that predominantly concentrated on CB1R. A recurring theme was the stabilization of hemodynamics through either the reduction in hypotensive/dilatory effects [44,45,46] or the enhancement of activity/sensitivity to vasopressor modulation (including norepinephrine and arginine vasopressin) [47,48,49,50].
Across 53 preclinical and clinical studies, the endocannabinoid system (ECS) emerged as a multifaceted modulator of sepsis pathophysiology. Of the 53 studies, 18 rodent models sought to illicit the mechanistic insights regarding CB2 activation in sepsis. The majority (10/18) reported reductions in pro-inflammatory cytokines, most notably: IL-6, IL-1β, and TNF-α. In eight studies, CB2 agonists such as HU-308, JWH-133, and AM-1241, attenuated organ-damage during septic challenge. This organ protection was most frequently observed in lung (6 studies), followed by heart (1 study) and brain (1 study) tissue. One study using a CLP sepsis model, found that JWH-133 agonism of CB2 shielded lung, liver, brain, and heart tissue in rats. Histopathological scores were dose-dependent, dropping to below 50% at the max 5 mg/kg dosage [36]. Many of the studies presenting decreased organ injury also had corresponding attenuation of pyroptosis markers (4 studies) (e.g., NLRP3, GSDMD) or NF-κB (2 studies). Reduction in leukocyte adhesion was also noted (6 studies), mainly in lung or intestinal tissue. Survival improvement was reported in 4 studies, with the longest duration being a decrease in 7-day mortality rates [39]. Mechanistically, CB2 signaling promoted anti-inflammatory polarization of microglia and macrophages [35,42], repressed dendritic cell maturation [40], and supported autophagy flux [38,51,52]. Interestingly, 1 study found that CB2 activation of CD4-T-cells resulted in decreased IL-10 production, increased lung injury, and decreased survival [53]. CB2 inhibition/knockout presented with a comparably more varied septic outcome. Of the 4 studies with CB2 inhibition as the primary investigation, 2 displayed an increase in leukocyte adhesion, either through AM630 antagonism or genetic deletion [17,54]. Two additional studies where CB2 inhibition was a secondary experiment also showed an increase in or maintenance of pro-inflammatory leukocyte influx [55,56]. Other studies suggested that CB2 signaling impairs antibacterial defense, with its downregulation resulting in less lymphoid apoptosis and aversion of gut stasis (Appendix A Table A2).
Following CB2 modulation, the next most frequently investigated ECS component was CB1 receptor inhibition. Thirteen of the 23 studies assessing CB1 inhibition reported attenuation of sepsis severity. Eight of the 13 studies demonstrated that application of CB1 antagonist (e.g., Rimonabant, AM281) to rodent models reduced sepsis associated hypotension by stabilizing vasoplegia or increasing vascular response to vasopressors (e.g., vasopressin). Three studies employing central blockage of CB1 signaling reported attenuation of late septic hypotension and increasing survival; in 1 study, 7-day survival rate significantly improved by 39% in a CLP rat model [47]. This phenomenon of increased survival benefit was elicited in 4 other CB1 inhibitor studies. Improved neurological outcomes were seen in 2 studies by attenuation of pro-inflammatory mechanisms, such as caspase-3 activation [46]. Collectively, 4 studies showcased a reduction in pro-inflammatory cytokines, of which all were documented with a decrease in TNF-α. In contrast, CB1 activation produced mixed results. Among the 10 studies investigating CB1 activation, 6 reported improved sepsis outcomes through a reduction in pro-inflammatory cytokine levels, most notably IL-6 and TNF-α, with 3 studies concurrently displaying an upregulation of IL-10. Conversely, other studies showed that CB1 activation translated to worsened septic outcomes with hypotension, hypothermia, nitric oxide mediated organ damage, and reduced intestinal motility [57,58] (Appendix A Table A2).
Beyond the canonical ECS, several ECS-adjacent targets we included in the 53 selected studies. TRPV1 activation triggered IL-10–centered neuroimmune reflexes that alleviated lung and brain injury. This was achieved through central activation but required peripheral macrophages independent of TRPV1 to convey their protective effect. Inhibition of GPR55 reduced renal apoptosis and inflammation in acute kidney injury models. Additionally, pharmacologic modulation of ECS-metabolizing enzymes like FAAH and MAGL enhanced endogenous cannabinoid tone, supporting vascular and immunologic recovery [54,56,59,60,61].
Human observational studies demonstrated correlations between ECS lipid levels (AEA, 2-AG) and clinical outcomes such as invasive mechanical ventilation and ICU stay length [62]. Transcriptomic profiling further identified ECS-linked gene signatures and hub targets (e.g., PTEN, HIST2H2BE) with diagnostic potential [63].
Collectively, these results indicate that ECS modulation—particularly via CB2 agonism and TRPV1 signaling—confers organ-specific and systemic benefits in sepsis. However, context-dependent findings, particularly regarding CB1 and IL-10 dynamics, reveal a complex landscape requiring precise therapeutic targeting (Figure 3).
This figure displays the general mechanistic trends of the three most studied endocannabinoid receptors: CB1, CB2, and TRPV1. Activation of CB1 shows the most nuanced and context dependent effects, often resulting in hypotension, loss of vasopressin tone, and anti-inflammatory release. CB2 and TRPV1 were more consistent, leading to an increase in systemic anti-inflammatory cytokines like IL-10. CB2 activation additionally demonstrated attenuation of leukocyte adhesion, pyroptosis, and tissue injury.

4. Discussion

This scoping review elucidates the intricate and contextually dependent function of the endocannabinoid system (ECS) within the pathophysiology of sepsis, synthesizing molecular, cellular, and systemic perspectives derived from both preclinical and clinical investigations. The role of the CB2 receptor has repeatedly been interpreted as a safeguard marker within the ECS components in numerous testing environments. Selective agonists of the CB2 receptor—including JWH-133, HU-308, and AM-1241—exhibited significant mitigation of inflammation, immune dysregulation, and pyroptosis, thereby establishing CB2 signaling as a pivotal regulator of immune homeostasis [16,40,52,64,65,66,67,68,69].
In models of endotoxemia and cecal ligation and puncture (CLP), the activation of CB2 receptors has been observed to mitigate neutrophil infiltration, oxidative stress, and the expression of pro-inflammatory cytokines (such as TNF-α, IL-1β, IL-6), whilst concurrently augmenting IL-10 production and promoting autophagic clearance [51,53,55,69,70]. Additionally, CB2 signaling has been implicated in the promotion of M2 macrophage polarization, the inhibition of dendritic cell maturation [16,53,55], and the facilitation of microglial reprogramming from the pro-inflammatory M1 phenotype to the reparative M2 phenotype—thereby ameliorating cognitive impairments associated with septic encephalopathy [43,54,59,71]. Nevertheless, the ramifications of CB2 activation were not entirely advantageous. Within CD4+ T cells, the activation of CB2 paradoxically resulted in the inhibition of IL-10 production and exacerbated lung injury and mortality, thereby highlighting the necessity for therapeutic strategies that are tailored to specific cell types and tissue contexts [16]. These findings underscore the risk that indiscriminate CB2 agonism could potentially worsen immunosuppression in patients already progressing toward sepsis-associated immune paralysis. Translational strategies will therefore require precise approaches guided by biomarkers to identify patient subsets that would most likely benefit. Circulating cytokine ratios such as IL-6/IL-10, or immune cell phenotyping may help stratify patients in either hyperinflammatory or immunosuppressed states, guiding clinical evaluation and guarding against indiscriminate use [72].
In a different light, the interaction with CB1 receptors illustrated an increased complexity and range of variability. Central antagonism of CB1 was correlated with enhanced survival rates and hemodynamic stability in multiple rodent models [46,56,73,74], with one investigation documenting a 39% increase in survival among CLP rats. Also, blocking CB1 led to reduced caspase-3 activation and neuroinflammation, which points towards possible neuroprotective roles within the scenario of septic encephalopathy [75]. Conversely, the triggering of CB1 generated effects that changed substantially in relation to the anatomical and cellular frameworks. Specifically, central CB1 activation intensified the severity of sepsis, whereas peripheral CB1 signaling—especially in monocytic myeloid-derived suppressor cells—facilitated the release of IL-10 and provided immunosuppressive advantages [50,76].
Conversely, the overactivation of CB1 receptors has been associated with gastrointestinal dysmotility [77], diminished vascular responsiveness [49,75,78], and an exacerbation of inflammatory processes [35,58]. The application of enzymatic modulation through the use of FAAH and MAGL inhibitors has proven effective in restoring the equilibrium of the endocannabinoid system (ECS), thereby alleviating these detrimental consequences [51,69,70]. Collectively, these findings indicate that CB1 signaling operates with dual, context-dependent roles in the pathophysiology of sepsis, with central blockade and selective peripheral activation presenting unique therapeutic opportunities. Despite this, the unregulated activation of CB1 may cause hypotensive incidents, collapse of the circulatory system, and organ malfunction—underscoring the need for accurate approaches that explicitly separate the central and peripheral CB1 signaling routes.
It is noteworthy that divergent thermoregulatory outcomes were documented in investigations utilizing Rimonabant for the antagonism of CB1 receptors. Leite-Avalca et al. (2016) observed hypothermia subsequent to CB1 blockade, whereas Steiner et al. (2011) ascribed hypothermia to the activation of CB1 receptors [47,58]. These inconsistencies are likely attributable to variations in the route of administration (intracerebroventricular versus oral) and the timing of intervention (pretreatment versus post-sepsis induction), thereby underscoring the necessity for mechanistic studies to clarify the temporal and spatial dynamics associated with the modulation of the ECS.
Beyond cannabinoid receptors, the activation of TRPV1 by both endogenous and synthetic ligands (for instance, OLDA and NADA) initiated IL-10-mediated neuroimmune reflexes, which contributed to the attenuation of inflammation within the pulmonary and central nervous systems [61,67,79]. The inhibition of GPR55 resulted in a reduction in renal injury through the downregulation of the RhoA/ROCK signaling pathway and the induction of apoptotic processes [60], whereas the modulation of GPR18 and PPARγ introduced novel pathways of endocannabinoid system-driven resolution signaling [38,41,42].
The enzymatic modulation of the endocannabinoid system (ECS) tone through the inhibition of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) markedly enhanced the protective mechanisms of autophagy, augmented vascular perfusion, and preserved intestinal integrity [37,51,70,80]. These observed phenomena were particularly pronounced within the endothelial and gastrointestinal systems, wherein lipids derived from the ECS influenced permeability and alleviated oxidative stress [34,49,57]. Moreover, ECS-mediated augmentation of autophagy significantly mitigated systemic cytokine storms through the reduction in high mobility group box 1 (HMGB1) and damage-associated molecular patterns (DAMPs), thus promoting the resolution process [69,70].
Clinically, the levels of circulating endocannabinoid system (ECS) lipids, such as anandamide (AEA) and 2-arachidonoylglycerol (2-AG), exhibited an inverse correlation with the duration of mechanical ventilation, the length of stay in the intensive care unit (ICU), and the severity of sepsis [17,36,45,60]. Transcriptomic and metabolomic investigations have elucidated ECS-associated enzymes and pivotal genes, including phosphatase and tensin homolog (PTEN) and HIST2H2BE, which may possess significant diagnostic and therapeutic implications [35,36,81].
Notwithstanding these encouraging observations, several vulnerabilities associated with the ECS necessitate prudent consideration. Reports have indicated the occurrence of CB1 receptor-induced vasoplegia, maladaptive responses of transient receptor potential vanilloid 1 (TRPV1), and timing-dependent immunosuppressive effects stemming from monoacylglycerol lipase (MAGL) inhibition [47,82,83]. This highlights the necessity for a more profound mechanistic comprehension of the interactions involving the endocannabinoid system across the immune, vascular, and neural systems.
In synthesis, the modulation of the endocannabinoid system—particularly through the mechanisms of CB2 receptor agonism, TRPV1 receptor activation, and the inhibition of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL)—provides a complex therapeutic framework in the management of sepsis. Nonetheless, the variable outcomes associated with CB1 receptor activation and the interactions between various receptors necessitate the implementation of precision medicine strategies that consider receptor subtype specificity, cellular lineage, temporal dynamics, and organ-specific environments. Lastly, examining interventions associated with the endocannabinoid system should be done concurrently with existing immunomodulatory frameworks. Corticosteroids, though widely adopted, are tied to the possibility of indiscriminate immunosuppression along with extended recovery intervals. Innovative treatment strategies like cytokine adsorption and immune checkpoint inhibitors are still in early development phases and show variable effectiveness [84].
In contrast to these considerations, endocannabinoid system (ECS) modulation—specifically through the agonism of CB2 receptors—may distinctly mitigate hyperinflammation whilst fostering reparative immune phenotypes [85]. Nevertheless, challenges associated with translation into clinical practice persist. The cardiovascular safety profile and the immunosuppressive implications, particularly in relation to CB2 receptor activation, are yet to be thoroughly delineated. The specificity inherent in ECS modulation, although therapeutically beneficial, introduces complexities regarding predictability. As an illustration, equivalent pharmacological substances can have divergent impacts due to differing model types, administration routes, and intervention timings. Furthermore, existing sepsis models (such as lipopolysaccharide (LPS), cecal ligation and puncture (CLP), and caspase-1 activation (CASP)) inadequately represent the heterogeneity observed in human sepsis, which frequently results from concurrent infections in older populations with multiple comorbidities. Additionally, the reliance on the LPS sepsis model, which was present in 70% of selected studies, introduces a Gram-negative bacterial bias to mechanistic insights, overshadowing non-Gram-negative infections that make up approximately 42% of sepsis cases [86].
The unification of preliminary research models along with the segmentation of patient groups is key to increasing the broader applicability and clinical importance of our findings. Additionally, neurobehavioral side effects warrant careful consideration; however, the modulation of the endocannabinoid system (ECS) through the inhibition of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) may potentially reduce the risks associated with cannabinoid-related adverse events [87,88]. The limited availability of human ECS data further constrains the comprehension of interindividual variability in ECS functionality throughout various stages of sepsis and the presence of comorbid conditions. Future translational investigations should emphasize the importance of stratified patient populations in order to exploit ECS signaling mechanisms for the maintenance of immunologic homeostasis and recovery in the context of human sepsis.
From a translational perspective, several limited clinical studies investigating ECS-targeting compounds that have moved beyond preclinical models. Proof-of-pharmacology clinical trials of FAAH inhibitor PF-04457845 sought to establish bioavailability and dosing parameters, while CB1 antagonist ANEB-001 has demonstrated penetration of CNS blood–brain barrier in early human studies [89,90]. Off-label cannabinoid drugs such as Sativex and Nabilone, investigated in related neuroinflammatory conditions such as Huntington’s or Parkinson’s disease, could shine light on potential applications regarding neuroinflammation found in sepsis [91,92]. Although selective CB2 agonists (e.g., HU-308, JWH-133) remain in preclinical development, their consistent anti-inflammatory effects highlight promising pharmacological direction. While to date there are no clinical studies directly investigating ECS compounds in the context of human sepsis, the growing clinical experience with ECS modulation in other indications suggests the feasibility for future translational studies.
This scoping review is subject to several constraints. First, the exclusion of articles published in languages other than English may have resulted in the inadvertent omission of pertinent studies, particularly from regions such as China, where research pertaining to ECS-related sepsis is notably vigorous. This judgment reflects practical concerns, consequently narrowing the scope and impact of our perspectives. Subsequent reviews may derive advantages from implementing multilingual search methodologies or fostering international cooperative efforts. Aligned with the standards described in scoping review methodology, we resolved not to investigate the methodological robustness of the studies highlighted in our evaluation. As a result, it is recommended that individuals carefully assess the conclusions, recognizing that the strength of the data might vary among the different research works.

5. Conclusions

In conclusion, the endocannabinoid system constitutes a sophisticated yet promising focal point for sepsis intervention, with empirical evidence corroborating the beneficial effects of CB2 agonism, TRPV1 activation, and FAAH/MAGL inhibition on immune modulation, organ safeguarding, and the resolution of inflammatory responses. Although no ECS-directed therapies have been tested clinically, the preclinical outcomes provide hypothetical extrapolations to frame clinical translation. CB2 agonism associated with improved survival in rodent sepsis, while TRPV1 activation attenuated lung and brain injury, suggest potential to shorten duration of mechanical ventilation and reduce encephalopathy. Similarly, FAAH and MAGL modulation showed improvement to vascular and intestinal microcirculation, showcasing potential relevance for attenuation of multi-organ dysfunction and mortality. Nevertheless, the ambivalent roles of CB1 signaling and context-dependent receptor interactions underscore the imperative for precise, cell- and organ-specific therapeutic modalities. While preclinical and preliminary clinical investigations emphasize ECS modulation as a multifaceted approach for reinstating immune equilibrium and enhancing clinical outcomes, additional translational research is critically required to refine treatment timing, selectivity, and safety in the context of human sepsis. Customized ECS-targeted interventions may improve the management of this complex syndrome, provided that these strategies are guided by a thorough comprehension of molecular, cellular, and systemic principles.

Author Contributions

Conceptualization, B.T., H.Y., A.K. and S.Y.; Methodology, B.T., H.Y., A.K. and S.Y.; Software, B.T., H.Y., A.K. and S.Y.; Validation, B.T., H.Y., A.K. and S.Y.; Formal Analysis, B.T., H.Y., A.K. and S.Y.; Investigation, B.T., H.Y., A.K. and S.Y.; Resources, B.T., H.Y., A.K. and S.Y.; Data Curation, B.T., H.Y., A.K. and S.Y.; Writing—Original Draft Preparation, B.T., H.Y.; Writing—Review and Editing, S.Y.; Visualization, S.Y.; Supervision, S.Y.; Project Administration, S.Y.; Funding Acquisition, NA. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article. The data that support the findings of this study are available from the corresponding author upon reasonable request.

Conflicts of Interest

The authors declare no conflicts of interest.

Appendix A

Table A1. Full Literature Database Search Strategy. * Identifies variations in the root word, “exp” and “mh” is a search field tag for “Exploded MeSH” which will include all related subcategories with functionality similar to “INDEXTERMS”, “NEXT” requires the search to have the two terms in immediate proximity, “.mp.” is a multi-purpose field search similar to “:ti,ab,kw” to search for text in the title, abstract, and keywords.
Table A1. Full Literature Database Search Strategy. * Identifies variations in the root word, “exp” and “mh” is a search field tag for “Exploded MeSH” which will include all related subcategories with functionality similar to “INDEXTERMS”, “NEXT” requires the search to have the two terms in immediate proximity, “.mp.” is a multi-purpose field search similar to “:ti,ab,kw” to search for text in the title, abstract, and keywords.
Ovid(Medline): Search ran on 6/13/25 and produced 134 results.
1. exp Sepsis/or (sepsis or “bloodstream infection *” or septicemia * or “blood poisoning *” or pyemia * or pyaemia * or pyohemia * or septic *).mp.
2. exp Endocannabinoids/or (endocannabinoid * or endo-cannabinoid *).mp.
3. exp Receptors, Cannabinoid/or “cannabinoid receptor *”.mp.
4. 2 or 3
5. 1 and 4
Cochrane Trials (CENTRAL): Search ran on 6/13/25 and produced 4 results.
1. [mh Sepsis] OR (sepsis:ti,ab,kw OR (“bloodstream” NEXT infection *):ti,ab,kw OR septicemia *:ti,ab,kw OR (“blood” NEXT poisoning *):ti,ab,kw OR pyemia *:ti,ab,kw OR pyaemia *:ti,ab,kw OR pyohemia *:ti,ab,kw OR septic *:ti,ab,kw)
2. [mh Endocannabinoids] OR (endocannabinoid*:ti,ab,kw OR endo-cannabinoid *:ti,ab,kw)
3. [mh “Receptors, Cannabinoid”] OR (“cannabinoid” NEXT receptor *):ti,ab,kw
4. #2 OR #3
5. #1 AND #4
Scopus: Search ran on 6/13/25 and produced 164 results.
(INDEXTERMS (sepsis) OR TITLE-ABS-KEY (sepsis OR “bloodstream infection *” OR septicemia * OR “blood poisoning *” OR pyemia * OR pyaemia * OR pyohemia * OR septic *) AND (INDEXTERMS (endocannabinoids) OR TITLE-ABS-KEY (endocannabinoid * OR endo-cannabinoid *) OR INDEXTERMS (“Receptors, Cannabinoid”) OR TITLE-ABS-KEY (“cannabinoid receptor *”)))
CINAHL: Search ran on 6/13/25 and produced 11 results.
(MH “Sepsis+” OR (sepsis OR “bloodstream infection *” OR septicemia * OR “blood poisoning *” OR pyemia * OR pyaemia * OR pyohemia * OR septic *)) AND ((endocannabinoid * OR endo-cannabinoid * OR “cannabinoid receptor *”))
Table A2. Full Detailed Data Collection Chart. This table lists the entirety of the data collected from the included studies. Arrows denote regulatory direction: ↑ indicates upregulation/increase, ↓ indicates downregulation/decrease, and → indicates a causal relationship.
Table A2. Full Detailed Data Collection Chart. This table lists the entirety of the data collected from the included studies. Arrows denote regulatory direction: ↑ indicates upregulation/increase, ↓ indicates downregulation/decrease, and → indicates a causal relationship.
Study (1st Author, Year)Model SystemECS ComponentIntervention/ComparisonKey Mechanistic Finding(s)Outcome(s)Quick Summary
Caraceni, 2009 [83]Sprague-Dawley rats; partial hepatic ischemia–reperfusion (IR) + LPS; in vivoCB1RCB1 antagonist (Rimonabant) vs. vehicle at 3 and 10 mg/kgCB1 antagonism reduced neutrophil infiltration, modulated cytokines (↓ TNF-α, ↑ IL-6, IFN-γ), and improved STAT3 signalingReduced liver injury (↓ ALT, necrosis), improved hemodynamics, reduced oxidative stressRimonabant mitigated LPS-enhanced hepatic IR injury via CB1 antagonism, modulating inflammation and oxidative stress.
Gardiner et al., 2005 [78]Sprague-Dawley rats; 24 h LPS infusion (150 µg kg−1 h−1); consciousCB1RCB1 antagonist AM251 (3 mg kg−1) pretreatmentAM251 inhibited LPS-induced tachycardia & hind-quarter vasodilation, an effect mimicked by β-adrenoceptor blockadePartial normalization of regional vascular conductanceSuggests ECS amplifies sympathetic beta-adrenergic vasodilator drive during sustained endotoxemia.
Joffre 2020 [64]C57BL/6 mice, LPS endotoxemia, in vivoCB1RΔ9-THC (agonist) ± SR141716 (CB1 inverse agonist)CB1 activation sharply increased IL-10 from Mo-MDSCs and suppressed IL-6/CCL-2 and NF-κB activationLower clinical sepsis score, reduced lung & spleen injuryCannabinoid agonism can be protective when it drives early IL-10 via CB1R.
Kadoi et al., 2005 [46]Male Wistar rats; bolus LPS (10 mg kg−1 i.v.); in vivoCB1RAM281 antagonist (1 mg kg−1 i.v.) given with LPSPrevented systemic hypotension and carotid blood-flow fall; moderated TNF-α/IL-1β riseImproved hemodynamics & halved 12 h mortalityCB1 blockade stabilized circulation and enhanced short-term survival during endotoxic shock.
Kadoi, 2005 [75]Male Wistar rats; cecal ligation & puncture (CLP);CB1RCB1 antagonist AM281 (1 mg kg−1 i.p.) vs. vehicleAM281 blunted caspase-3 activation in hippocampus and preserved brainstem reflexes↓ Neurologic dysfunction & mortality after CLPBlocking CB1 improved survival and neurological outcomes in septic rats.
Kadoi, 2008 [44]Streptozotocin-induced diabetic and non-diabetic rats; LPS-induced endotoxemia; in vivoCB1RCB1 antagonist (AM281) vs. vehicleAM281 reduced hypotension in diabetic rats more effectively; affected survival ratesImproved blood pressure and survival in diabetic ratsCB1 antagonism by AM281 improved hemodynamics and survival in diabetic endotoxemia model.
Kianian 2014 [93]Male Lewis rats, LPS-induced endotoxemia—in vivoCB1RAM281 (CB1 antagonist) ± ACEA (CB1 agonist)CB1 blockade lowered leukocyte adhesion in intestinal venules and restored functional capillary density (FCD)Improved gut microcirculation; no hypotension reportedBlocking CB1 during early endotoxemia rapidly normalized intestinal microvascular perfusion, suggesting CB1 drives microvascular inflammation.
Leite-Avalca 2016 [47]Male Wistar rats, CLP (severe, 3 punctures); central i.c.v. cannula (in vivo)CB1RCB1 antagonist Rimonabant (oral or i.c.v.) vs. vehicleCB1 blockade boosted plasma vasopressin and dropped core temperature without altering peritoneal bacterial load↑ 12-h and 5-day survival after CLPInhibiting CB1 increases AVP release and improves survival in late septic shock.
Leite-Avalca 2019 [48]Wistar male rats, CLP (3 × 16 G punctures), in vivoCB1RRimonabant 10 mg kg−1 p.o. at 4 h post-CLP (antagonist) vs. vehicleBlockade did not change NOx but normalized aortic AVP reactivity and tempered AVP hyper-responsiveness↓ plasma lactate, LDH, CK-MB; attenuated tail-artery vasoconstriction; improved organ-dysfunction panelLate-phase CB1 antagonism limits metabolic/vascular dysfunction in sepsis.
Varga 1998 [57]Sprague-Dawley rats, LPS 15 mg kg−1 i.v.; plus adoptive transfer of LPS-stimulated macrophages/platelets; in vivoCB1RLPS or transfer ± CB1 antagonist SR141716A 3 mg kg−1 i.v.LPS ↑ 2-AG in platelets & anandamide in macrophages → CB1-mediated vasodilation; SR141716A blocks hypotensionSR141716A prevents hypotension & improves survival after LPSMacrophage anandamide + platelet 2-AG paracrine drive septic hypotension via CB1; antagonist rescues BP and survival.
Vercelli 2009 [94]Pregnant mouse uterine explants; LPS challenge; in vitroCB1RCB1 antagonist AM-251 vs. vehicle (±exogenous AEA)LPS raises uterine AEA (↓ FAAH) and drives iNOS-derived NO; blockade of CB1 abolishes NO spike and tissue injuryAEA–CB1 promotes NO-mediated uterine damage in septic abortion modelEndotoxin uses local ECS tone (AEA/CB1) to amplify inflammatory NO production
Villanueva 2009 [45]Sprague-Dawley rats; i.v. LPS endotoxemia; in vivoCB1RI.c.v. rimonabant (CB1 antagonist) vs. vehicleCB1 blockade prevents early & late hypotension and blunts LPS-evoked NE overflow in pre-optic anterior hypothalamus, plus lowers plasma TNF-αMaintained mean arterial pressure; ↓ systemic TNF-αCentral CB1 activation is required for endotoxic hypotension
Çakır 2020 [36]Sprague-Dawley rats + CLP sepsisCB2RJWH-133 (agonist) 0.2–5 mg kg−1CB2 activation lowers NF-κB, caspase-3 and pro-inflammatory cytokines in brain, lung, liver & heart↓ Multi-organ histopathology; ↑ IL-10Systemic CB2 agonism shields multiple organs from CLP-induced damage.
Chen 2022 [53]C57BL/6 mice; CLP sepsis; in vivo lung (plus CD4-specific Cnr2 knock-out)CB2RHU-308 (CB2 agonist) vs. vehicle; CD4-Cre Cnr2^fl/fl vs. WT;CD4-T-cell CB2R signaling suppresses IL-10; loss of CB2R switches the effect↓ Survival, ↑ lung injury with HU-308; protection in CD4-CB2-KOCB2 activation on CD4 T cells hampers IL-10 and worsens CLP sepsis.
Chen 2025 [42]C57BL/6 mice, CLP sepsis; BV-2 microglia in vitro, brainCB2RHU-308 agonist ± AM-630 inverse agonist; Nogo-B over-expressionCB2R activation ↓ Nogo-B, shifts microglia M1→M2, ↓ IL-1β/IL-6/TNF-α; neuroprotection↑ Cognitive scores; Nogo-B over-expression blunted benefitsCB2 activation represses Nogo-B, steering microglia to a pro-repair M2 state and easing septic encephalopathy.
Csóka 2009 [82]CB2-KO vs. WT mice; CLP polymicrobial sepsis; in vivoCB2RGenetic knockout of CB2Loss of CB2 → ↓ NF-κB activation, ↓ IL-10/IL-6/MIP-2, less lymphoid apoptosis, more immune cellsLower mortality, ↓ bacteremia & organ (kidney, muscle) injuryCB2 signaling impairs antibacterial defense; antagonism could be beneficial
Gui 2013 [81]C57BL/6 mice, high-dose LPS endotoxemia—in vivo & ex vivo splenocytes/macrophagesCB2RGW405833 (CB2 agonist) and CB2-/- knockoutAgonist inhibited ERK1/2, STAT3 and NF-κB activation in macrophages; CB2-/- mice showed exaggerated cytokine stormGW405833 improved 72 h survival and lowered serum TNF-α/IL-6/HMGB1Genetic and pharmacologic evidence that CB2 signaling restrains pro-inflammatory pathways and improves survival in acute LPS sepsis.
Kapellos 2017 [17]C57BL/6 WT vs. CB2/ mice, LPS 10 mg kg−1 i.p., in vivoCB2RGenetic deletionCB2 deficiency significantly increased rolling/adhesion and neutrophil influx into lung & liver (pro-recruitment)Heightened neutrophil tissue accumulation; no mortality difference reported.CB2 signaling restrains early neutrophil recruitment during endotoxemia.
Lehmann 2012 [34]Lewis rats, LPS endotoxemia & CASP peritonitis models—in vivoCB2RHU308 (CB2 agonist) or AM630 (CB2 antagonist)CB2 activation reduced intestinal leukocyte adhesion and lowered systemic cytokines in both sepsis modelsDecreased inflammatory mediator release; microvascular protectionPharmacologic CB2 stimulation dampens early hyper-inflammation in sepsis, pointing to CB2 as a therapeutic target.
Liu 2014 [37]Wistar rats, CLP sepsis; lung tissue (in vivo)CB2ROral Melilotus extract vs. vehicle (2 h pre-CLP)Extract up-regulated CB2R and impeded NF-κB activation in PBMCs/lung↓ TNF-α, ↓ IL-6, fewer BAL neutrophils, attenuated histologic lung injuryHerbal up-regulation of CB2 breaks NF-κB-driven lung injury in polymicrobial sepsis.
Liu 2020 [38]C57BL/6 mice + CLP; RAW264.7 macrophages + LPSCB2RHU-308 (agonist) ± 3-MA (autophagy blocker)HU-308 triggers protective autophagy; 3-MA abrogates the effect, linking CB2 → autophagy → anti-inflammation↓ Lung pathology, ↓ TNF-α/IL-6, ↑ cell viabilityCB2-driven autophagy is central to limiting septic lung injury.
Sardinha 2014 [56]C57BL/6 mice, i.v. LPS endotoxemia; intestinal microcirculation (in vivo)CB2RCB2 agonist (HU-308), antagonist (AM630), FAAH inhibitor (URB597), MAGL inhibitor (JZL184)HU-308, URB597 & JZL184 each lowered leukocyte rolling/adhesion; AM630 maintained adherent leukocyte levels↓ Leukocyte endothelial interactions; preserved capillary densityMultiple pharmacologic routes that raise CB2 activation reduce gut microvascular inflammation in sepsis.
Souza 2023 [39]Swiss mice (♂/♀), K. pneumoniae pneumonia-sepsis (in vivo, lung)CB2RAM-1241 agonist (0.3/3 mg kg−1 i.p.)CB2R activation ↓ neutrophil influx, NOS-2, MPO, protein leak; plasma IL-1β ↓, IL-10 ↑↓ Lung bacterial load, injury score & 7-day mortalityPeripheral CB2 agonism tempers lung neutrophilia and systemic cytokines, boosting survival in pneumonia-sepsis.
Tschöp 2009 [55]CB2-KO & WT mice; CLP; plus WT + CB2 agonist (GP1a); in vivoCB2RKO (loss-of-function) & agonist (GP1a) (gain-of-function)CB2-KO → ↑ IL-6, bacteremia, lung injury, neutrophil influx & ↓ activation; GP1a reverses in WT, boosts p38 in neutrophilsKO: ↓ survival (22%); GP1a: longer survival, ↓ IL-6, ↓ bacteremiaHere CB2 supports antibacterial immunity; precise role is host-context dependent
Yang 2022 [43]Male C57BL/6 mice; CLP-induced sepsis-associated encephalopathy; in vivoCB2RHU-308 (agonist) vs. vehicleCB2 activation curbs microglial over-activation and neuronal pyroptosis (↓ NLRP3, GSDMD-NT)Improved cognition (OFT, NORT, MWM) & histologyCB2 agonism safeguards the brain from CLP-induced neuro-inflammation and pyroptotic damage.
Zhang 2021 [16]C57BL/6 mice + CLP (in vivo); BMDM + LPS/ATP (in vitro)CB2RHU-308 (agonist) vs. AM-630 (antagonist) and CB2-knock-downCB2 activation dampens NLRP3-caspase-1-GSDMD pyroptosis cascade↓Pro-inflammatory cytokines, ↓ lung injury, ↑survivalCB2 agonism curbs pyroptosis-driven inflammation and protects septic mice.
Zhang 2023 [65]C57BL/6 mice, CLP sepsis; heart tissueCB2RHU-308 agonist vs. CLP vehicleCB2R activation ↓ NLRP3, caspase-1, GSDMD and myocardial pyroptosis; ↓ IL-1β, LDH, CK-MBImproved myocardial histology & injury markersHU-308-driven CB2 signaling weakens cardiac pyroptosis and biochemical injury after CLP.
Zhao 2023 [40]C57BL/6 mice, LPS-induced SA-ALI; DCs in vitro/in vivoCB2RHU-308 agonist, SR144528 antagonist, DC-specific CNR2 KOCB2R signaling limits dendritic-cell maturation & pro-cytokines, mitigating lung pathology↓ Histologic lung injury & cytokines; antagonist/KO reversed protectionCB2 signaling in DCs dampens cytokine storm and rescues LPS-driven acute lung injury.
Zhou 2020 [51]Mouse peritoneal macrophages + LPS (in vitro)CB2RGW-405833 (agonist); 3-MA vs. MG-132CB2 activation enhances autophagy-lysosome flux → Cathepsin-B-dependent degradation of HMGB1↓ Extracellular HMGB1 and downstream cytokinesCB2 signaling clears danger-signal HMGB1 via autophagy, tempering inflammation.
Braile 2021 [67]Human neutrophils (PMNs) + LPS (in vitro)CB1R/CB2RACEA (CB1 agonist)/JWH-133 (CB2 agonist) ± AM-251(CB1 antagonists)/AM630(CB2 antagonist)Low-dose CB agonists selectively inhibit LPS-induced VEGF-A transcription & release without affecting CXCL8/HGF↓ Endothelial permeability & tube formation (VEGF-A-driven)Cannabinoid signaling tempers neutrophil-driven angiogenic leakage relevant to septic vasculopathy.
Godlewski et al., 2004 [49]Pithed, vagotomised Wistar rats; LPS (0.4–4 mg kg−1); in vivoCB1R/CB2RCB1 antagonist SR141716A; CB2 antagonist; VR1 & H3 antagonistsLPS suppressed neurogenic vasopressor response via presynaptic CB1—not CB2/VR1/H3—receptorsCB1 blockade restored sympathetic vasopressor tone in early septic shockEndocannabinoids acting on presynaptic CB1 dampen sympathetic vasoconstriction during sepsis.
Li 2010 [77]Rat jejunal myoelectrical activity & mouse charcoal transit; LPS septic ileus; in vivoCB1R/CB2RAgonists HU210 (CB1) & JWH133 (CB2); antagonists AM251 (CB1) & AM630 (CB2)LPS lowers spike amplitude/frequency & GI transit; CB1/CB2 agonists mimic this, while antagonists prevent motility loss and cytokine riseAntagonists restore GI transit and jejunal activityBlocking either receptor averts LPS-induced gut stasis—CB1/CB2 antagonists may treat septic ileus
Matias 2023 [66]Wistar rats, CLP (1- or 3-puncture) sepsis; CNS focusCB1R/CB2RCB1 antagonist (AM-251), CB2 antagonist (AM-630) 4 h post-CLPEarly CB1 blockade ↑ survival & prevented long-term fear-memory generalization; CB2 blockade or minocycline prevented fear generalization without ↑ survival, linking early ECS & neuro-inflam. to PTSD-like phenotypeSurvival, hyperalgesia, contextual-fear tests; hippocampal TNF-α levels ↓ with minocyclineEarly central CB1 (±CB2) antagonism raises survival and blocks long-term fear generalization via dampened neuro-inflammation.
Smith 2000 [76]BDF1 mice (±C. parvum priming) → LPS endotoxemia (E. coli O55:B5); in vivoCB1R/CB2Ri.p. agonists (0.05–0.4 mg HU-210; 3.1–50 mg WIN 55 212-2) ± SR141716A/SR144528CB1 activation ↓ TNF-α & IL-12, ↑ IL-10; effects abolished by CB1—but not CB2—antagonismProtected primed mice from lethal LPS challenge (↑ survival)Synthetic CB1 agonists blunt the cytokine storm and rescue survival in endotoxemic, C. parvum–primed mice.
Smith 2001 [68]BDF1 mice (±C. parvum priming) → LPS endotoxemia (E. coli O55:B5); in vivoCB1R/CB2RLow-dose i.c.v. agonists (≈¼ i.p. dose) ± central SR141716ACentral CB1 activation suppresses TNF-α & IL-12 and elevates IL-10 at much lower doses; blockade by SR141716A, not SR144528Demonstrated potent cytokine modulation; survival not assessedBrain CB1 receptors are a sensitive switch that controls systemic cytokine output during endotoxemia.
Steiner 2011 [58]Conscious rats, systemic LPS (25–100 µg kg−1)—in vivoCB1R/CB2RRimonabant (CB1 antagonist), SLV319, CB2/TRPV1 antagonists, ICV AEACentral CB1 blockade abolished LPS-induced hypothermia; CB2/TRPV1 antagonism had no effect; ICV AEA enhanced hypothermiaBody-temperature drop during severe sepsis is CB1-dependentEndocannabinoids acting on brain CB1 receptors drive the hypothermic phase of systemic inflammation, distinguishing CB1 from CB2/TRPV1 roles.
Bányai 2023 [71]Wistar rats, LPS (5 mg kg−1 i.v.) endotoxemia; aorta & heart ex vivoMixed CB1/CB2 (THC)Δ9-THC 10 mg kg−1 i.p.THC ↓ 4-HNE, 3-nitrotyrosine & COX-2; preserved endothelial-dependent relaxation & ventricular volumesMaintained cardiac output; mitigated vascular dysfunctionTHC safeguards cardiovascular function in endotoxemic rats by cutting oxidative-nitrative stress and COX-2.
Angelina 2022 [52]BALB/c female mice + LPS sepsis; human monocyte-DC/T-cell co-cultureWIN55,212-2 (mixed CB1/CB2 agonist)WIN55-212-2 (mixed CB1/CB2 agonist) ± autophagy, CB1 or PPAR-α blockersCB1/PPAR-α-driven autophagy reprograms DC metabolism → tolerogenic DCs → FOXP3+ Tregs; blocking autophagy or receptors reverses benefit↑ 78 h survival from 0% → 90% in lethal LPS; ↑ splenic Tregs; ↓ IL-6Cannabinoids re-educate immunity through autophagy-dependent DC-Treg axis, rescuing mice from endotoxic shock.
He 2022 [35]MH-S alveolar-macrophage line (LPS) & C57BL/6 LPS-sepsis mice; in vitro + in vivo lungWIN55,212-2 (mixed CB1/CB2 agonist)WIN (0.625–40 µM in vitro; dosing in mice) vs. LPS; miR-29b-3p/FOXO3 gain–lossWIN ↑ miR-29b-3p → ↓ FOXO3 → ↓ PFKFB3 glycolysis → shifts M1 → M2 phenotype↓ BALF protein, cells, lactate; better lung histologyWIN blocks macrophage glycolysis via a miR-29b-3p axis, easing LPS-induced acute lung injury.
Toguri 2015 [80]Lewis rats, LPS endotoxemia; (in vivo)WIN55,212-2 (mixed CB1/CB2 agonist)WIN55,212-2 ± CB1 antagonist (AM281) or CB2 antagonist (AM630)CB2 (but not CB1) activation curtailed leukocyte-endothelium adhesion and normalized micro-vascular flow↓ Adherent leukocytes; ↑ capillary perfusionSystemic CB2 stimulation rapidly rescues microcirculatory dysfunction in endotoxemia.
Wilhelmsen 2014 [79]Primary human lung microvascular ECs (HMVEC-lung) stimulated with LPS/FSL-1/TNF-α (in vitro)WIN55,212-2 (mixed CB1/CB2 agonist)NADA or WIN vs. vehicle ± CB1/CB2 antagonistsWIN and NADA dose-dependently cut IL-6/IL-8 release & neutrophil adhesion; NADA effect is CB1/CB2-dependent;↓ Endothelial cytokines & leukocyte binding; no effect on permeabilityEndothelial ECS (via NADA or WIN) is a brake on cytokine-driven vascular activation relevant to sepsis.
Singh 2018 [50]Swiss-albino mice, CLP (6 h & 20 h), ex vivo aortic rings2-AG/CB1RDAGL inhibitor (KT-109), MAGL inhibitor (JZL-184), CB1 antagonist (AM-251)Sepsis up-regulated CB1R & reduced MAGL mRNA; blocking DAGL or CB1 restored NA-induced vasoconstrictionKT-109 or AM-251 normalized vascular reactivity; improved mean arterial tone (survival not assessed).Endocannabinoid 2-AG acting on CB1 underlies septic vasoplegia.
Sultan 2021 [41]Female C57BL/6 Mice; SEB-induced ARDS (systemic super-antigen sepsis); in vivo lungAnandamide (AEA) ligandExogenous AEA 40 mg kg−1 vs. vehicleAEA down-regulates miR-23a-3p/miR-34a-5p → ↑ ARG1, TGF-β2, FOXP3 → expansion of MDSCs & Tregs; ↓ IL-2, IFN-γ, TNF↑ Lung function, ↓ cell infiltrates, cytokinesAEA re-programs miRNA networks to tilt immunity toward MDSC/Treg-driven resolution of SEB-ARDS.
Murakami 2009 [69]C57BL/6 mice sensitized with D-galactosamine + LPS (endotoxin shock, in vivo) ± RAW264.7 cells (in vitro)Anandamide (ligand)Cationic antimicrobial peptide CAP11 vs. scrambled controlCAP11 prevents LPS-induced rise in AEA, HMGB1, TNF-α & IL-6 in macrophages and in serum↓ systemic mediators, improved survival in shock model; suppresses ECS ligand generationCAP11’s protection is partly via curbing endocannabinoid (AEA) over-production
Liu, 2006 [73]RAW264.7 macrophages with/without LPS; mouse brain extracts; in vitroAEA biosynthetic enzymes (NAPE-PLD, PLC, PTPN22)LPS stimulation; siRNA knock-down; overexpression; enzyme inhibitorsLPS up-regulates a PLC–phosphatase pathway (involving PTPN22) that converts NAPE→phospho-AEA→AEA, bypassing NAPE-PLD↑ AEA synthesis under endotoxin challenge; identifies alternate biosynthetic routeDefined a novel PLC/PTPN22 route for LPS-induced anandamide production in macrophages.
De Filippis, 2008 [74]Swiss OF1 mice; LPS-induced sepsis; in vivo + intestinal muscle stripsCB1 receptor, FAAH (indirectly through cannabidiol)Cannabidiol alone and with CB1 antagonist (AM251)Sepsis upregulated CB1 and FAAH in intestine; cannabidiol reversed FAAH increase but worsened motility via CB1Cannabidiol reduced motility in septic mice; CB1 antagonist reversed this effectCannabidiol worsened sepsis-induced ileus through CB1 signaling; reversed by CB1 antagonist.
Szafran 2015 [70]Female C57BL/6 mice, single i.p. LPS (6–24 h); spleen & splenocytes (in vivo & ex vivo)Ces2g (2-AG-hydrolyzing carboxylesterase)Endotoxin vs. salineLPS selectively suppressed Ces2g activity → ↓ 2-AG hydrolysis in spleen & splenocytesPotential ↑ 2-AG levels (not directly measured)—suggests feedback dampening of inflammationEndotoxin shuts down a 2-AG catabolic enzyme, potentially prolonging anti-inflammatory eCB signaling during systemic inflammation.
Wolfson 2013 [59]Murine peripheral blood mononuclear cells (PBMCs) from non-pregnant vs. pregnant mice—in vitro/in vivo LPSFAAHProgesterone (PR agonist) ± PR antagonistLPS lowered FAAH activity in PBMCs; progesterone restored FAAH via progesterone-receptor signalingNormalized FAAH prevents excess anandamide buildup linked to pregnancy failureHormonal regulation can counter LPS-driven suppression of FAAH, highlighting cross-talk between endocrine and ECS during systemic inflammation.
Kianian 2013 [54]Lewis rats, LPS endotoxemia—in vivoFAAH & CB2URB597 (FAAH inhibitor) ± AM630 (CB2 antagonist)FAAH inhibition raised endocannabinoids, cutting leukocyte adhesion and increasing FCD; CB2 blockade reversed adhesion benefitEnhanced intestinal perfusion; reduced microvascular inflammationElevating endogenous cannabinoids via FAAH inhibition protects gut microcirculation during endotoxemia, largely through CB2.
Chen 2022 [60]C57BL/6 mice + CLP sepsis; serum from septic patients (observational)GPR55 (non-classical ECS GPCR)CID16020046 (antagonist) 20 mg kg−1GPR55 blockade suppresses ROCK1/2 signaling, inflammation and apoptosis in kidney↓ BUN/Creatinine, ↓ KIM-1/NGAL, ↓ TNF-α/IL-6, no off-target toxicityTargeting GPR55 mitigates sepsis-induced AKI via anti-inflammatory/anti-apoptotic routes.
Joffre 2022 [61]C57BL/6 mice, LPS i.v./i.t. endotoxemia; S. aureus i.t. pneumonia/sepsis; in vivo; plus BMDM, HMVEC, astrocytes in vitroTRPV1; OLDA agonistOLDA 5–10 mg/kg i.v. given at 0–2 h post-challenge vs. vehicle; WT vs. Trpv1−/−; cell-specific TRPV1 knockdowns; monocyte/macrophage depletionPan-neuronal (CNS) TRPV1 triggers an early surge in IL-10, suppresses IL-6/TNF/chemokines; reversed with knockdown, not with PNS or myeloid TRPV1 knockdown. Monocyte depletion blocks OLDA-induced IL-10.↓ Lung injury, ↑ Lung IL-10; improved mouse sepsis scores; bacterial load unchanged in S. aureus model.CNS Neuronal TRPV1 activation by ODA results in a neuro-immune pathway that drives peripheral monocyte IL-10; attenuates systemic inflammation; reduces clinical severity
Lawton 2017 [95]Mice, LPS i.v., CLP; ex vivo hematopoieticTRPV1; NADA agonistNADA 1–10 mg/kg i.v. (often 5–10 mg/kg) ± LPS/CLP; WT vs. Trpv1−/−; bone-marrow chimerasNon-hematopoietic cell TRPV1 (pointing to neuronal) reduces systemic pro-inflammatory cytokines and PAI-1; raises IL-10. NADA alters neuropeptides (↓ CGRP, ↑ substance P) during LPS.↑ Survival in endotoxemic mice; ↓ inflammatory mediator levels across models; ↑ IL-10, ↓ IL-6, CCL2, PAI-1Nonhematopoietic TRPV1 activation attenuates systemic inflammation and improves survival in endotoxemic mice
Orliac 2007 [96]Rat, i.p. endotoxemia (6 h); isolated mesenteric vascular bed; in vivo + ex vivoTRPV1; AEA agonistLPS vs. saline; ex vivo anandamide (0.01–10 μM) and capsaicin; TRPV1 antagonist capsazepine; PKC activator PMALPS increases TRPV1 and CGRP-positive nerve density in mesenteric arteries; ↑ AEA mediated CGRP and vasorelaxation (TRPV1 dependent) post-LPS; PKC activation ↑ AEA efficacyUpregulation of TRVP1 during endotoxemia implicated in septic hypotensionEarly endotoxemia upregulates TRPV1/CGRP signaling in perivascular sensory nerves, amplifying AEA-driven CGRP release and vasodilation; indicates an ECS–TRPV1 contribution to vascular changes in sepsis.

References

  1. Rudd, K.E.; Johnson, S.C.; Agesa, K.M.; Shackelford, K.A.; Tsoi, D.; Kievlan, D.R.; Colombara, D.V.; Ikuta, K.S.; Kissoon, N.; Finfer, S.; et al. Global, regional, and national sepsis incidence and mortality, 1990–2017: Analysis for the Global Burden of Disease Study. Lancet 2020, 395, 200–211. [Google Scholar] [CrossRef] [PubMed]
  2. Chechulina, V.; Sheikh, F.; Lóser, M.; Englesakis, M.; Barrett, K.; Canada, S. Healthcare costs after sepsis: A systematic review. Crit. Care 2025, 29, 381. [Google Scholar] [CrossRef] [PubMed]
  3. Meyer, N.J.; Prescott, H.C. Sepsis and Septic Shock. N. Engl. J. Med. 2024, 391, 2133–2146. [Google Scholar] [CrossRef] [PubMed]
  4. Kissoon, N.; Reinhart, K.; Daniels, R.; Machado, M.F.R.; Schachter, R.D.; Finfer, S. Sepsis in Children: Global Implications of the World Health Assembly Resolution on Sepsis. Pediatr. Crit. Care Med. 2017, 18, e625–e627. [Google Scholar] [CrossRef]
  5. Robey, R.C.; Logue, C.; Caird, C.A.; Hansel, J.; Hellyer, T.P.; Simpson, J.; Dark, P.; Mathioudakis, A.G.; Felton, T. Immunomodulatory drugs in sepsis: A systematic review and meta-analysis. Anaesthesia 2024, 79, 869–879. [Google Scholar] [CrossRef]
  6. Liu, D.; Langston, J.C.; Prabhakarpandian, B.; Kiani, M.F.; Kilpatrick, L.E. The critical role of neutrophil-endothelial cell interactions in sepsis: New synergistic approaches employing organ-on-chip, omics, immune cell phenotyping and. Front. Cell. Infect. Microbiol. 2023, 13, 1274842. [Google Scholar]
  7. Seymour, C.W.; Kennedy, J.N.; Wang, S.; Chang, C.-C.H.; Elliott, C.F.; Xu, Z.; Berry, S.; Clermont, G.; Cooper, G.; Gomez, H.; et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA 2019, 321, 2003–2017. [Google Scholar] [CrossRef]
  8. Martin-Loeches, I.; Singer, M.; Leone, M. Sepsis: Key insights, future directions, and immediate goals. A review and expert opinion. Intensiv. Care Med. 2024, 50, 2043–2049. [Google Scholar] [CrossRef]
  9. Cuddihey, H.; Macnaughton, K.W.; Sharkey, A.K. Role of the Endocannabinoid System in the Regulation of Intestinal Homeostasis. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 947–963. [Google Scholar] [CrossRef]
  10. Pandey, R.; Mousawy, K.; Nagarkatti, M.; Nagarkatti, P. Endocannabinoids and immune regulation. Pharmacol. Res. 2009, 60, 85–92. [Google Scholar] [CrossRef]
  11. Gong, F.; Zheng, X.; Xu, W.; Xie, R.; Liu, W.; Pei, L.; Zhong, M.; Shi, W.; Qu, H.; Mao, E.; et al. H3K14la drives endothelial dysfunction in sepsis-induced ARDS by promoting SLC40A1/transferrin-mediated ferroptosis. MedComm 2025, 6, e70049. [Google Scholar] [CrossRef]
  12. Joffre, J.; Hellman, J.; Ince, C.; Ait-Oufella, H. Endothelial Responses in Sepsis. Am. J. Respir. Crit. Care Med. 2020, 202, 361–370. [Google Scholar] [CrossRef] [PubMed]
  13. Xiong, S.; Hong, Z.; Huang, L.S.; Tsukasaki, Y.; Nepal, S.; Di, A.; Zhong, M.; Wu, W.; Ye, Z.; Gao, X.; et al. IL-1β suppression of VE-cadherin transcription underlies sepsis-induced inflammatory lung injury. J. Clin. Investig. 2020, 130, 3684–3698. [Google Scholar] [CrossRef] [PubMed]
  14. Xie, M.; Yu, Y.; Kang, R.; Zhu, S.; Yang, L.; Zeng, L.; Sun, X.; Yang, M.; Billiar, T.R.; Wang, H.; et al. PKM2-dependent glycolysis promotes NLRP3 and AIM2 inflammasome activation. Nat. Commun. 2016, 7, 13280. [Google Scholar] [CrossRef] [PubMed]
  15. Wang, J.; Wu, W.; Wen, T.; Zheng, G.; Qiu, G.; Qian, H.; Zhang, R.; Xia, J.; Hu, Y.; Huang, R.; et al. Extracellular vesicle-bound S100A8/A9 is differentially expressed in septic shock and prompts acute lung injury. Respir. Res. 2025, 26, 107. [Google Scholar] [CrossRef]
  16. Zhang, B.; Zheng, F.; Liu, A.; Li, Z.; Zheng, F.; Liu, Q.; Yang, L.; Chen, K.; Wang, Y.; Zhang, Z.; et al. Activation of CB2 receptor inhibits pyroptosis and subsequently ameliorates cecal ligation and puncture-induced sepsis. Int. Immunopharmacol. 2021, 99, 108038. [Google Scholar] [CrossRef]
  17. Kapellos, S.T.; Recio, C.; Greaves, R.D.; Iqbal, J.A. Cannabinoid Receptor 2 Modulates Neutrophil Recruitment in a Murine Model of Endotoxemia. Mediat. Inflamm. 2017, 2017, 4315412. [Google Scholar] [CrossRef]
  18. Tortora, C.; Di Paola, A.; Argenziano, M.; Creoli, M.; Marrapodi, M.M.; Cenni, S.; Tolone, C.; Rossi, F.; Strisciuglio, C. Effects of CB2 Receptor Modulation on Macrophage Polarization in Pediatric Celiac Disease. Biomedicines 2022, 10, 874. [Google Scholar] [CrossRef]
  19. Turcotte, C.; Blanchet, M.-R.; Laviolette, M.; Flamand, N. The CB2 receptor and its role as a regulator of inflammation. Cell. Mol. Life Sci. 2016, 73, 4449–4470. [Google Scholar] [CrossRef]
  20. Martínez-Torres, S.; Bergadà-Martínez, A.; Ortega, J.E.; Galera-López, L.; Hervera, A.; Reyes-Ramírez, L.d.L.; Ortega-Álvaro, A.; Remmers, F.; Muñoz-Moreno, E.; Soria, G.; et al. Peripheral CB1 receptor blockade acts as a memory enhancer through a noradrenergic mechanism. Neuropsychopharmacology 2023, 48, 341–350. [Google Scholar] [CrossRef]
  21. Nogueiras, R.; Veyrat-Durebex, C.; Suchanek, P.M.; Klein, M.; Tschōp, J.; Caldwell, C.; Woods, S.C.; Wittmann, G.; Watanabe, M.; Liposits, Z.; et al. Peripheral, but Not Central, CB1 Antagonism Provides Food Intake–Independent Metabolic Benefits in Diet-Induced Obese Rats. Diabetes 2008, 57, 2977–2991. [Google Scholar] [CrossRef] [PubMed]
  22. Contino, M.; Mccormick, J.P. Editorial: The Canonical and Non-Canonical Endocannabinoid System as a Target in Cancer and Acute and Chronic Pain. Front. Pharmacol. 2020, 11, 312. [Google Scholar] [CrossRef] [PubMed]
  23. Haugh, O.; Penman, J.; Irving, A.; Campbell, V. The Emerging Role of the Cannabinoid Receptor Family in Peripheral and Neuro-immune Interactions. Curr. Drug Targets 2016, 17, 1834–1840. [Google Scholar] [CrossRef] [PubMed]
  24. Kasatkina, A.L.; Rittchen, S.; Sturm, M.E. Neuroprotective and Immunomodulatory Action of the Endocannabinoid System under Neuroinflammation. Int. J. Mol. Sci. 2021, 22, 5431. [Google Scholar] [CrossRef]
  25. Jaiswal, S.; Ayyannan, R.S. Anticancer Potential of Small-Molecule Inhibitors of Fatty Acid Amide Hydrolase and Monoacylglycerol Lipase. ChemMedChem 2021, 16, 2172–2187. [Google Scholar] [CrossRef]
  26. Kouchi, Z. Physiological Role of Endocannabinoid Hydrolyzing Enzymes in Brain Development and Neurodegeneration. Biochem. Physiol. 2015, 4, 2. [Google Scholar]
  27. Antcliffe, D.B.; Burnham, K.L.; Al-Beidh, F.; Santhakumaran, S.; Brett, S.J.; Hinds, C.J.; Ashby, D.; Knight, J.C.; Gordon, A.C. Transcriptomic Signatures in Sepsis and a Differential Response to Steroids. From the VANISH Randomized Trial. Am. J. Respir. Crit. Care Med. 2019, 199, 980–986. [Google Scholar] [CrossRef]
  28. Ma, H.; Wang, W.; Tang, B.; Zhang, H.; Tan, C.; Yang, Y. Inflammatory Protein Signatures of Sepsis Risk and Mortality: A Mendelian Randomization Study. Shock 2025, 64, 148–153. [Google Scholar] [CrossRef]
  29. Reyes, M.; Filbin, R.M.; Bhattacharyya, P.R.; Billman, K.; Eisenhaure, T.; Hung, T.D.; Levy, B.D.; Baron, R.M.; Blainey, P.C.; Goldberg, M.B.; et al. An immune-cell signature of bacterial sepsis. Nat. Med. 2020, 26, 333–340. [Google Scholar] [CrossRef]
  30. Deng, H.; Li, J.; Shah, A.A.; Lin, G.; Chen, H.; Ouyang, W. Commonly expressed key transcriptomic profiles of sepsis in the human circulation and brain via integrated analysis. Int. Immunopharmacol. 2022, 104, 108518. [Google Scholar] [CrossRef]
  31. Arksey, H.; O’Malley, L. Scoping studies: Towards a methodological framework. Int. J. Soc. Res. Methodol. 2005, 8, 19–32. [Google Scholar] [CrossRef]
  32. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed]
  33. Thai, B.; Yamamoto, H.; Koutrouvelis, A.; Yamamoto, S. Endocannabinoid system in sepsis: A scoping review protocol. Open Sci. Framew. 2025. [Google Scholar] [CrossRef]
  34. Lehmann, C.; Kianian, M.; Zhou, J.; Küster, I.; Kuschnereit, R.; Whynot, S.; Hung, O.; Shukla, R.; Johnston, B.; Cerny, V.; et al. Cannabinoid receptor 2 activation reduces intestinal leukocyte recruitment and systemic inflammatory mediator release in acute experimental sepsis. Crit. Care 2012, 16, R47. [Google Scholar] [CrossRef]
  35. He, Q.; Yin, J.; Zou, B.; Guo, H. WIN55212-2 alleviates acute lung injury by inhibiting macrophage glycolysis through the miR-29b-3p/FOXO3/PFKFB3 axis. Mol. Immunol. 2022, 149, 119–128. [Google Scholar] [CrossRef]
  36. Cakir, M.; Tekin, S.; Okan, A.; Cakan, P.; Doganyigit, Z. The ameliorating effect of cannabinoid type 2 receptor activation on brain, lung, liver and heart damage in cecal ligation and puncture-induced sepsis model in rats. Int. Immunopharmacol. 2020, 78, 105978. [Google Scholar] [CrossRef]
  37. Liu, M.-W.; Su, M.-X.; Wang, Y.-H.; Wei, W.; Qin, L.-F.; Liu, X.; Tian, M.-L.; Qian, C.-Y. Effect of melilotus extract on lung injury by upregulating the expression of cannabinoid CB2 receptors in septic rats. BMC Complement. Altern. Med. 2014, 14, 94. [Google Scholar] [CrossRef]
  38. Liu, A.; Yuan, Q.; Zhang, B.; Yang, L.; He, Q.; Chen, K.; Liu, Q.; Li, Z.; Zhan, J. Cannabinoid receptor 2 activation alleviates septic lung injury by promoting autophagy via inhibition of inflammatory mediator release. Cell. Signal. 2020, 69, 109556. [Google Scholar] [CrossRef]
  39. Souza, C.F.; Borges, L.B.; Oliveira, F.R.M.B.; Silva, P.C.d.S.; Patricio, D.O.; Rosales, T.O.; Souza, N.F.; Spiller, F.; Mansur, D.S.; Assreuy, J.; et al. Cannabinoid CB2 receptor agonist reduces local and systemic inflammation associated with pneumonia-induced sepsis in mice. Eur. J. Pharmacol. 2023, 959, 176092. [Google Scholar] [CrossRef]
  40. Zhao, F.-Z.; Gu, W.-J.; Li, L.-Z.; Qu, Z.-K.; Xu, M.-Y.; Liu, K.; Zhang, F.; Liu, H.; Xu, J.; Yin, H.-Y. Cannabinoid receptor 2 alleviates sepsis-associated acute lung injury by modulating maturation of dendritic cells. Int. Immunopharmacol. 2023, 123, 110771. [Google Scholar] [CrossRef]
  41. Sultan, M.; Alghetaa, H.; Mohammed, A.; Abdulla, O.A.; Wisniewski, P.J.; Singh, N.; Nagarkatti, P.; Nagarkatti, M. The Endocannabinoid Anandamide Attenuates Acute Respiratory Distress Syndrome by Downregulating miRNA that Target Inflammatory Pathways. Front. Pharmacol. 2021, 12, 119–128. [Google Scholar] [CrossRef] [PubMed]
  42. Chen, S.; Li, Z.; Yang, L.; Xu, Z.; Liu, A.; He, Q.; Xiao, F.; Zhan, J. Cannabinoid Receptor-2 Alleviates Sepsis-Induced Neuroinflammation by Modulating Microglia M1/M2 Subset Polarization Through Inhibiting Nogo-B Expression. Mol. Neurobiol. 2025, 62, 9258–9270. [Google Scholar] [CrossRef] [PubMed]
  43. Yang, L.; Li, Z.; Xu, Z.; Zhang, B.; Liu, A.; He, Q.; Zheng, F.; Zhan, J. Protective Effects of Cannabinoid Type 2 Receptor Activation Against Microglia Overactivation and Neuronal Pyroptosis in Sepsis-Associated Encephalopathy. Neuroscience 2022, 493, 99–108. [Google Scholar] [CrossRef] [PubMed]
  44. Kadoi, Y.; Hinohara, H.; Kunimoto, F.; Saito, S. Effects of the cannabinoid antagonist AM281 on systemic hemodynamics and mortality rate in streptozotocin-induced diabetic rats with endotoxic shock: Comparison between non-diabetic and diabetic rats. Acta Anaesthesiol. Scand. 2008, 52, 664–672. [Google Scholar] [CrossRef]
  45. Villanueva, A.; Yilmaz, S.M.; Millington, W.R.; Cutrera, R.A.; Stouffer, D.G.; Parsons, L.H.; Cheer, J.F.; Feleder, C. Central cannabinoid 1 receptor antagonist administration prevents endotoxic hypotension affecting norepinephrine release in the preoptic anterior hypothalamic area. Shock 2009, 32, 614–620. [Google Scholar] [CrossRef]
  46. Kadoi, Y.; Hinohara, H.; Kunimoto, F.; Kuwano, H.; Saito, S.; Goto, F. Effects of AM281, a cannabinoid antagonist, on systemic haemodynamics, internal carotid artery blood flow and mortality in septic shock in rats. Br. J. Anaesth. 2005, 94, 563–568. [Google Scholar] [CrossRef]
  47. Leite-Avalca, C.G.M.; Lomba, A.L.; Bastos-Pereira, L.A.; Brito, O.H.; Fraga, D.; Zampronio, R.A. Involvement of Central Endothelin ETA and Cannabinoid CB1 Receptors and Arginine Vasopressin Release in Sepsis Induced by Cecal Ligation and Puncture in Rats. Shock 2016, 46, 290–296. [Google Scholar] [CrossRef]
  48. Leite-Avalca, M.C.G.; Staats, F.T.; Verona, D.; de Souza, P.; Almeida, M.C.; Silva-Santos, J.E.; Zampronio, A.R. Cannabinoid CB1 Receptor Antagonist Rimonabant Decreases Levels of Markers of Organ Dysfunction and Alters Vascular Reactivity in Aortic Vessels in Late Sepsis in Rats. Inflammation 2019, 42, 618–627. [Google Scholar] [CrossRef]
  49. Godlewski, G.; Malinowska, B.; Schlicker, E. Presynaptic cannabinoid CB1 receptors are involved in the inhibition of the neurogenic vasopressor response during septic shock in pithed rats. Br. J. Pharmacol. 2004, 142, 701–708. [Google Scholar] [CrossRef]
  50. Singh, P.; Sharma, P.; Nakade, U.P.; Sharma, A.; Gari, M.; Choudhury, S.; Shukla, A.; Garg, S.K. Endocannabinoid-mediated modulation of Gq protein-coupled receptor mediates vascular hyporeactivity to nor-adrenaline during polymicrobial sepsis. Pharmacol. Rep. 2018, 70, 1150–1157. [Google Scholar] [CrossRef]
  51. Zhou, H.; Du, R.; Li, G.; Bai, Z.; Ma, J.; Mao, C.; Wang, J.; Gui, H. Cannabinoid receptor 2 promotes the intracellular degradation of HMGB1 via the autophagy-lysosome pathway in macrophage. Int. Immunopharmacol. 2020, 78, 106007. [Google Scholar] [CrossRef]
  52. Angelina, A.; Pérez-Diego, M.; López-Abente, J.; Rückert, B.; Nombela, I.; Akdis, M.; Martín-Fontecha, M.; Akdis, C.; Palomares, O. Cannabinoids induce functional Tregs by promoting tolerogenic DCs via autophagy and metabolic reprograming. Mucosal Immunol. 2022, 15, 96–108. [Google Scholar] [CrossRef]
  53. Chen, J.; Wang, F.; Zhang, S.; Lin, Q.; Xu, H.; Zhu, T.; Peng, L.; Cen, F.; Li, F.; Wang, Z.; et al. Activation of CD4+ T Cell–Derived Cannabinoid Receptor 2 Signaling Exacerbates Sepsis via Inhibiting IL-10. J. Immunol. 2022, 208, 2515–2522. [Google Scholar] [CrossRef] [PubMed]
  54. Kianian, M.; Al-Banna, A.N.; Kelly, E.M.M.; Lehmann, C. Inhibition of endocannabinoid degradation in experimental endotoxemia reduces leukocyte adhesion and improves capillary perfusion in the gut. J. Basic Clin. Physiol. Pharmacol. 2013, 24, 27–33. [Google Scholar] [CrossRef] [PubMed]
  55. Tschöp, J.; Kasten, K.R.; Nogueiras, R.; Goetzman, H.S.; Cave, C.M.; England, L.G.; Dattilo, J.; Lentsch, A.B.; Tschöp, M.H.; Caldwell, C.C. The Cannabinoid Receptor 2 Is Critical for the Host Response to Sepsis. J. Immunol. 2009, 183, 499–505. [Google Scholar] [CrossRef] [PubMed]
  56. Sardinha, J.; Kelly, M.E.M.; Zhou, J.; Lehmann, C. Experimental Cannabinoid 2 Receptor-Mediated Immune Modulation in Sepsis. Mediat. Inflamm. 2014, 2014, 978678. [Google Scholar] [CrossRef]
  57. Varga, K.; Wagner, A.J.; Bridgen, T.D.; Kunos, G. Platelet- and macrophage-derived endogenous cannabinoids are involved in endotoxin-induced hypotension. FASEB J. 1998, 12, 1035–1044. [Google Scholar] [CrossRef]
  58. Steiner, A.A.; Molchanova, Y.A.; Dogan, D.M.; Patel, S.; Pétervári, E.; Balaskó, M.; Wanner, S.P.; Eales, J.; Oliveira, D.L.; Gavva, N.R.; et al. The hypothermic response to bacterial lipopolysaccharide critically depends on brain CB1, but not CB2 or TRPV1, receptors. J. Physiol. 2011, 589, 2415–2431. [Google Scholar] [CrossRef]
  59. Wolfson, L.M.; Aisemberg, J.; Salazar, I.A.; Rubio, D.P.A.; Vercelli, A.C.; Franchi, M.A. Progesterone reverts LPS-reduced FAAH activity in murine peripheral blood mononuclear cells by a receptor-mediated fashion. Mol. Cell. Endocrinol. 2013, 381, 97–105. [Google Scholar] [CrossRef]
  60. Chen, R.; Xu, H.; Guo, Z.; Zhang, P.; Chen, J.; Chen, Z. CID16020046, a GPR55 antagonist, attenuates sepsis-induced acute kidney injury. Mol. Med. Rep. 2022, 25, 155. [Google Scholar] [CrossRef]
  61. Joffre, J.; Wong, E.; Lawton, S.; Lloyd, E.; Nguyen, N.; Xu, F.; Sempio, C.; Kobzik, L.; Zlatanova, I.; Schumacher, M.; et al. N-Oleoyl dopamine induces IL-10 via central nervous system TRPV1 and improves endotoxemia and sepsis outcomes. J. Neuroinflamm. 2022, 19, 118. [Google Scholar] [CrossRef] [PubMed]
  62. Basu, A.; Pommerolle, L.; Arif, M.; Meliton, A.Y.; Udofia, I.; Wu, D.; Mutlu, G.M.; Gochuico, B.R.; Summer, R.; Adegunsoye, A.; et al. Elevated blood anandamide levels in acute COVID-19 pneumonia with respiratory failure. Am. J. Med. Sci. 2025, 370, 271–277. [Google Scholar] [CrossRef] [PubMed]
  63. Termine, A.; Fabrizio, C.; Gimenez, J.; Panuccio, A.; Balsamo, F.; Passarello, N.; Caioli, S.; Saba, L.; De Bardi, M.; Della Valle, F.; et al. Transcriptomic and Network Analyses Reveal Immune Modulation by Endocannabinoids in Approach/Avoidance Traits. Int. J. Mol. Sci. 2022, 23, 2538. [Google Scholar] [CrossRef] [PubMed]
  64. Joffre, J.; Yeh, C.-C.; Wong, E.; Thete, M.; Xu, F.; Zlatanova, I.; Lloyd, E.; Kobzik, L.; Legrand, M.; Hellman, J. Activation of CB1R Promotes Lipopolysaccharide-Induced IL-10 Secretion by Monocytic Myeloid-Derived Suppressive Cells and Reduces Acute Inflammation and Organ Injury. J. Immunol. 2020, 204, 3339–3350. [Google Scholar] [CrossRef]
  65. Zhang, J.; Zhu, Y.; Chen, S.; Xu, Z.; Zhang, B.; Liu, A.; He, Q.; Zhan, J. Activation of cannabinoid receptors 2 alleviates myocardial damage in cecal ligation and puncture-induced sepsis by inhibiting pyroptosis. Immunol. Lett. 2023, 264, 17–24. [Google Scholar] [CrossRef]
  66. Matias, E.M.; Radulski, R.D.; Silva, D.R.T.; Raymundi, M.A.; Stern, J.A.C.; Zampronio, R.A. Involvement of cannabinoid receptors and neuroinflammation in early sepsis: Implications for posttraumatic stress disorder. Int. Immunopharmacol. 2023, 123, 110745. [Google Scholar] [CrossRef]
  67. Braile, M.; Cristinziano, L.; Marcella, S.; Varricchi, G.; Marone, G.; Modestino, L.; Ferrara, A.L.; De Ciuceis, A.; Scala, S.; Galdiero, M.R.; et al. LPS-mediated neutrophil VEGF-A release is modulated by cannabinoid receptor activation. J. Leukoc. Biol. 2021, 109, 621–631. [Google Scholar] [CrossRef]
  68. Smith, R.S.; Terminelli, C.; Denhardt, G. Modulation of cytokine responses in Corynebacterium parvum-primed endotoxemic mice by centrally administered cannabinoid ligands. Eur. J. Pharmacol. 2001, 425, 73–83. [Google Scholar] [CrossRef]
  69. Murakami, T.; Obata, T.; Kuwahara-Arai, K.; Tamura, H.; Hiramatsu, K.; Nagaoka, I. Antimicrobial cathelicidin polypeptide CAP11 suppresses the production and release of septic mediators in D-galactosamine-sensitized endotoxin shock mice. Int. Immunol. 2009, 21, 905–912. [Google Scholar] [CrossRef]
  70. Szafran, B.; Borazjani, A.; Lee, H.J.; Ross, K.M.; Kaplan, L.F.B. Lipopolysaccharide suppresses carboxylesterase 2g activity and 2-arachidonoylglycerol hydrolysis: A possible mechanism to regulate inflammation. Prostaglandins Other Lipid Mediat. 2015, 121, 199–206. [Google Scholar] [CrossRef]
  71. Bányai, B.; Répás, C.; Miklós, Z.; Johnsen, J.; Horváth, M.E.; Benkő, R. Delta 9-tetrahydrocannabinol conserves cardiovascular functions in a rat model of endotoxemia: Involvement of endothelial molecular mechanisms and oxidative-nitrative stress. PLoS ONE 2023, 18, e0287168. [Google Scholar] [CrossRef]
  72. Antcliffe, D.B.; Mi, Y.; Santhakumaran, S.; Burnham, K.L.; Prevost, A.T.; Ward, J.K.; Marshall, T.J.; Bradley, C.; Al-Beidh, F.; Hutton, P.; et al. Patient stratification using plasma cytokines and their regulators in sepsis: Relationship to outcomes, treatment effect and leucocyte transcriptomic subphenotypes. Thorax 2024, 79, 515–523. [Google Scholar] [CrossRef]
  73. Liu, J.; Wang, L.; Harvey-White, J.; Osei-Hyiaman, D.; Razdan, R.; Gong, Q.; Chan, A.C.; Zhou, Z.; Huang, B.X.; Kim, H.Y.; et al. A biosynthetic pathway for anandamide. Proc. Natl. Acad. Sci. USA 2006, 103, 13345–13350. [Google Scholar] [CrossRef] [PubMed]
  74. De Filippis, D.; Iuvone, T.; D’Amico, A.; Esposito, G.; Steardo, L.; Herman, A.G.; Pelckmans, P.A.; De Winter, B.Y.; De Man, J.G. Effect of cannabidiol on sepsis-induced motility disturbances in mice: Involvement of CB1 receptors and fatty acid amide hydrolase. Neurogastroenterol. Motil. 2008, 20, 919–927. [Google Scholar] [CrossRef] [PubMed]
  75. Kadoi, Y.; Hinohara, H.; Kunimoto, F.; Saito, S.; Goto, F. Cannabinoid antagonist AM 281 reduces mortality rate and neurologic dysfunction after cecal ligation and puncture in rats. Crit. Care Med. 2005, 33, 2629–2636. [Google Scholar] [CrossRef] [PubMed]
  76. Smith, S.R.; Terminelli, C.; Denhardt, G. Effects of cannabinoid receptor agonist and antagonist ligands on production of inflammatory cytokines and anti-inflammatory interleukin-10 in endotoxemic mice. J. Pharmacol. Exp. Ther. 2000, 293, 136–150. [Google Scholar] [CrossRef]
  77. Li, Y.-Y.; Li, Y.-N.; Ni, J.-B.; Chen, C.-J.; Lv, S.; Chai, S.-Y.; Wu, R.-H.; Yüce, B.; Storr, M. Involvement of cannabinoid-1 and cannabinoid-2 receptors in septic ileus. Neurogastroenterol. Motil. 2010, 22, 350-e88. [Google Scholar] [CrossRef]
  78. Gardiner, M.S.; March, E.J.; Kemp, A.P.; Bennett, T. Involvement of CB1-receptors and β-adrenoceptors in the regional hemodynamic responses to lipopolysaccharide infusion in conscious rats. Am. J. Physiol.-Heart Circ. Physiol. 2005, 288, H2280–H2288. [Google Scholar] [CrossRef]
  79. Wilhelmsen, K.; Khakpour, S.; Tran, A.; Sheehan, K.; Schumacher, M.; Xu, F.; Hellman, J. The Endocannabinoid/Endovanilloid N-Arachidonoyl Dopamine (NADA) and Synthetic Cannabinoid WIN55,212-2 Abate the Inflammatory Activation of Human Endothelial Cells. J. Biol. Chem. 2014, 289, 13079–13100. [Google Scholar] [CrossRef]
  80. Toguri, J.T.; Moxsom, R.; Szczesniak, A.M.; Zhou, J.; Kelly, M.E.M.; Lehmann, C. Cannabinoid 2 receptor activation reduces leukocyte adhesion and improves capillary perfusion in the iridial microvasculature during systemic inflammation. Clin. Hemorheol. Microcirc. 2015, 61, 237–249. [Google Scholar] [CrossRef]
  81. Gui, H.; Sun, Y.; Luo, Z.-M.; Su, D.-F.; Dai, S.-M.; Liu, X. Cannabinoid Receptor 2 Protects against Acute Experimental Sepsis in Mice. Mediat. Inflamm. 2013, 2013, 741303. [Google Scholar] [CrossRef] [PubMed]
  82. Csóka, B.; Németh, Z.H.; Mukhopadhyay, P.; Spolarics, Z.; Rajesh, M.; Federici, S.; Deitch, E.A.; Bátkai, S.; Pacher, P.; Haskó, G. CB2 Cannabinoid Receptors Contribute to Bacterial Invasion and Mortality in Polymicrobial Sepsis. PLoS ONE 2009, 4, e6409. [Google Scholar] [CrossRef] [PubMed]
  83. Caraceni, P.; Pertosa, A.M.; Giannone, F.; Domenicali, M.; Grattagliano, I.; Principe, A.; Mastroleo, C.; Perrelli, M.G.; Cutrin, J.; Trevisani, F.; et al. Antagonism of the cannabinoid CB-1 receptor protects rat liver against ischaemia-reperfusion injury complicated by endotoxaemia. Gut 2009, 58, 1135–1143. [Google Scholar] [CrossRef] [PubMed]
  84. Bode, C.; Weis, S.; Sauer, A.; Wendel-Garcia, P.; David, S. Targeting the host response in sepsis: Current approaches and future evidence. Crit. Care 2023, 27, 478. [Google Scholar] [CrossRef]
  85. Leite-Avalca, M.C.G.; Zampronio, A.; Lehmann, C. Cannabinoid Receptor 1 and 2 Signaling Pathways Involved in Sepsis. Shock 2021, 56, 673–681. [Google Scholar] [CrossRef]
  86. Chun, K.; Syndergaard, C.; Damas, C.; Trubey, R.; Mukindaraj, A.; Qian, S.; Jin, X.; Breslow, S.; Niemz, A. Sepsis Pathogen Identification. J. Lab Autom. 2015, 20, 539–561. [Google Scholar] [CrossRef]
  87. Piomelli, D.; Mabou Tagne, A. Endocannabinoid-Based Therapies. Annu. Rev. Pharmacol. Toxicol. 2022, 62, 483–507. [Google Scholar] [CrossRef]
  88. Ciaramellano, F.; Fanti, F.; Scipioni, L.; Maccarrone, M.; Oddi, S. Endocannabinoid Metabolism and Transport as Drug Targets. Methods Mol. Biol. 2023, 2576, 201–211. [Google Scholar]
  89. ClinicalTrials.gov. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of CB1 Antagonist ANEB-001 in a THC Challenge Test. 2023. Available online: https://clinicaltrials.gov/study/NCT05282797 (accessed on 18 September 2025).
  90. ClinicalTrials.gov. Evaluation of the Relative Bioavailability of PF-04457845 Tablet to Solution Formulation and Food Effect in Healthy Subjects. 2009. Available online: https://clinicaltrials.gov/study/NCT00918164 (accessed on 18 September 2025).
  91. López-Sendón Moreno, J.L.; García Caldentey, J.; Trigo Cubillo, P.; Ruiz Romero, C.; García Ribas, G.; Alonso Arias, M.A.; García de Yébenes, M.J.; Tolón, R.M.; Galve-Roperh, I.; Sagredo, O.; et al. A double-blind, randomized, cross-over, placebo-controlled, pilot trial with Sativex in Huntington’s disease. J. Neurol. 2016, 263, 1390–1400. [Google Scholar] [CrossRef]
  92. Peball, M.; Werkmann, M.; Ellmerer, P.; Stolz, R.; Valent, D.; Knaus, H.-G.; Ulmer, H.; Djamshidian, A.; Poewe, W.; Seppi, K. Nabilone for non-motor symptoms of Parkinson’s disease: A randomized placebo-controlled, double-blind, parallel-group, enriched enrolment randomized withdrawal study (The NMS-Nab Study). J. Neural Transm. 2019, 126, 1061–1072. [Google Scholar] [CrossRef]
  93. Kianian, M.; Kelly, M.E.; Zhou, J.; Hung, O.; Cerny, V.; Rowden, G.; Lehmann, C. Cannabinoid receptor 1 inhibition improves the intestinal microcirculation in experimental endotoxemia. Clin. Hemorheol. Microcirc. 2014, 58, 333–342. [Google Scholar] [CrossRef]
  94. Vercelli, C.; Aisemberg, J.; Billi, S.; Cervini, M.; Ribeiro, M.; Farina, M.; Franchi, A. Anandamide regulates lipopolysaccharide-induced nitric oxide synthesis and tissue damage in the murine uterus. Reprod. Biomed. Online 2009, 18, 824–831. [Google Scholar] [CrossRef]
  95. Lawton, S.K.; Xu, F.; Tran, A.; Wong, E.; Prakash, A.; Schumacher, M.; Hellman, J.; Wilhelmsen, K. N-Arachidonoyl Dopamine Modulates Acute Systemic Inflammation via Nonhematopoietic TRPV1. J. Immunol. 2017, 199, 1465–1475. [Google Scholar] [CrossRef]
  96. Orliac, M.L.; Peroni, R.N.; Abramoff, T.; Neuman, I.; Podesta, E.J.; Adler-Graschinsky, E. Increases in vanilloid TRPV1 receptor protein and CGRP content during endotoxemia in rats. Eur. J. Pharmacol. 2007, 566, 145–152. [Google Scholar] [CrossRef]
Figure 1. PRISMA flow diagram of study screening process.
Figure 1. PRISMA flow diagram of study screening process.
Anesthres 02 00024 g001
Figure 2. Publication Date of Included Studies.
Figure 2. Publication Date of Included Studies.
Anesthres 02 00024 g002
Figure 3. Mechanistic Overview of ECS Modulation of Sepsis. Arrows (↑↓) pointing outward from the vessel walls represent vasodilation. Arrows adjacent to text represent upregulation (↑) or downregulation (↓) of their respective terms (↓ AVP Tone, ↑ IL-10, ↑ Leukocyte adhesion, ↑ Tissue injury).
Figure 3. Mechanistic Overview of ECS Modulation of Sepsis. Arrows (↑↓) pointing outward from the vessel walls represent vasodilation. Arrows adjacent to text represent upregulation (↑) or downregulation (↓) of their respective terms (↓ AVP Tone, ↑ IL-10, ↑ Leukocyte adhesion, ↑ Tissue injury).
Anesthres 02 00024 g003
Table 1. Study Characteristics. * Denotes that values are not mutually exclusive.
Table 1. Study Characteristics. * Denotes that values are not mutually exclusive.
CharacteristicCategoryn (%)
Animal Model *Mice31(58%)
Rats21 (40%)
Other5 (9%)
Sepsis Model *LPS37 (70%)
CLP17 (32%)
Bacterial Exposure2 (4%)
Primary ECS TargetCB1R12 (23%)
CB2R17 (32%)
CB1R + CB2R7 (13%)
Receptor Ligand8 (15%)
ECS Enzyme5 (9%)
Noncanonical Receptor4 (8%)
Organ/Tissue Studied *Lung26 (49%)
Heart15 (28%)
Brain10 (19%)
Kidney/Liver5 (9%)
Publication Year1998–20044 (8%)
2005–200913 (25%)
2010–201410 (19%)
2015–20197 (13%)
2020–202519 (36%)
Table 2. Themes of Septic Responses to ECS Modulation. This table lists the number of studies grouped by their ECS modulation and resulting septic response. Please note that the categories are not mutually exclusive. ↑ indicates upregulation, while ↓ denotes downregulation.
Table 2. Themes of Septic Responses to ECS Modulation. This table lists the number of studies grouped by their ECS modulation and resulting septic response. Please note that the categories are not mutually exclusive. ↑ indicates upregulation, while ↓ denotes downregulation.
ECS ComponentModulation TypeStudies (n)Direction of EffectSummary of Outcome Trends
CB1RActivation4↑ Sepsis Severity↑ Mediates hypotension/hypothermia and inflammatory damage
Activation6↓ Sepsis Severity↑ Increases Il-10 production, reduced inflammatory markers (TNF-α and Il-12), increases survival
Inhibition/Knockout0↑ Sepsis SeverityN/A
Inhibition/Knockout13↓ Sepsis Severity↓ Stabilize hemodynamics, through reduction in hypotension/dilation and increases response to vasopressors
CB2RActivation1↑ Sepsis Severity↑ Reduces IL-10 production in CD4-T cells, increasing lung injury
Activation17↓ Sepsis Severity↑ Reduces NLRP3 inflammasome related pyroptosis, leukocyte adhesion, pro-inflammatory cytokine (IL-6, TNF-α). Increases autophagy, especially in macrophages
Inhibition/Knockout2↑ Sepsis Severity↓ Results in increased leukocyte adhesion/influx
Inhibition/Knockout2↓ Sepsis Severity↓ Decreases mortality and organ damage, improves septic ileus
TRPV1Activation2↓ Sepsis Severity↑ Increases IL-10 production, reduces IL-6
FAAHModulation2Mixed Sepsis Severity↑ Reduced septic pregnancy loss, ↓Protected gut microcirculation
MAGLInhibition1↓ Sepsis SeverityReduced leukocyte adhesion and gut inflammation
AEAModulation2Mixed Sepsis Severity↑ Reduced lung injury, ↓ Reduced shock survival
Δ9-THCAdministration1↓ Sepsis SeverityReduced oxidative-nitrative cardiovascular stress
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Thai, B.; Yamamoto, H.; Koutrouvelis, A.; Yamamoto, S. Endocannabinoid System in Sepsis: A Scoping Review. Anesth. Res. 2025, 2, 24. https://doi.org/10.3390/anesthres2040024

AMA Style

Thai B, Yamamoto H, Koutrouvelis A, Yamamoto S. Endocannabinoid System in Sepsis: A Scoping Review. Anesthesia Research. 2025; 2(4):24. https://doi.org/10.3390/anesthres2040024

Chicago/Turabian Style

Thai, Brandon, Hideaki Yamamoto, Aristides Koutrouvelis, and Satoshi Yamamoto. 2025. "Endocannabinoid System in Sepsis: A Scoping Review" Anesthesia Research 2, no. 4: 24. https://doi.org/10.3390/anesthres2040024

APA Style

Thai, B., Yamamoto, H., Koutrouvelis, A., & Yamamoto, S. (2025). Endocannabinoid System in Sepsis: A Scoping Review. Anesthesia Research, 2(4), 24. https://doi.org/10.3390/anesthres2040024

Article Metrics

Back to TopTop