Abstract
The impact of the endogenous cannabinoids (AEA, 2-AG, PEA, and virodamine) on the immune cell expressed cannabinoid receptors (CB1, CB2, TRPV-1, and GPR55) and consequent regulation of immune function is an exciting area of research with potential implications in the prevention and treatment of inflammatory and autoimmune diseases. Despite significant advances in understanding the mechanisms through which cannabinoids regulate immune functions, not much is known about the role of endocannabinoids in the pathogenesis or prevention of autoimmune diseases. Inasmuch as CB2 expression on immune cells and its role has been widely reported, the importance of CB1 in immunological disorders has often been overlooked especially because it is not highly expressed on naive immune cells. Therefore, the current review aims at delineating the effect of endocannabinoids on CB1 receptors in T cell driven autoimmune diseases. This review will also highlight some autoimmune diseases in which there is evidence indicating a role for endocannabinoids in the regulation of autoimmune pathogenesis. Overall, based on the evidence presented using the endocannabinoids, specifically AEA, we propose that the peripheral CB1 receptor is involved in the regulation and amelioration of inflammation associated with autoimmune diseases.
Keywords: 2-AG, anandamide, cannabinoid receptors, cannabinoids, endocannabinoid system, T cell-mediated disease
INTRODUCTION
Human immunity has been tested time and time again against pathogens and microbes. The ability of the immune system to recognize foreign antigens and thereby protect against infections helps in the survival of the species. It is however, when the body reacts to itself that immunity can become problematic. As a form of protection, the immune system is built upon safe guards which serve to ensure that self-tolerance is maintained. It is a loss of this ability to distinguish self from nonself which underlies autoimmune diseases, ranging from mild irritations to severe life threatening symptoms [1]. Autoimmune diseases are complex immunological disorders in which it now appears that the endocannabinoid (EC) system plays an integral role [2].
The EC system was discovered while attempting to gain a more complete understanding of the mechanisms through which exogenous cannabinoids acted on the immune system [2]. Perhaps the most notable exogenous cannabinoid is Δ-9-tetrahydrocannabinol (THC), a psychoactive component of the Cannabis sativa plant, which was first described in a 1964 paper by Gaoni and Mechoulam [3]. After THC was isolated and synthesized, it was found that this cannabinoid was a ligand for two receptors termed cannabinoid receptor 1 (CB1) and cannabinoid receptor 2 (CB2), discovered in 1990 and 1992 respectively [4, 5]. Not long after the discovery of the cannabinoid receptors, endogenous ligands, which act as lipid messengers derived from cellular membranes, were found [6]. The first EC discovered, N-arachidonyl ethanolamide (AEA), was named anandamide from Sanskrit for “internal bliss” [7]. Anandamide was first found in the porcine brain in 1992 [2]. In 1995, another endogenous cannabinoid, 2-arachidonoyl glycerol (2-AG), was isolated by two independent groups [8, 9].
ECs are synthesized locally as needed rather than being stored within vesicles [10]. The synthesis of AEA can occur in two main ways: one is through N-arachidonoyl phosphatidylethanolaminephospholipase, which converts phosphatidylcholine and phosphatidylethanolamine to N-arachidonoyl phosphatidylethanolamine (NAPE), the anandamide precursor [11]. The other accepted pathway is through phospholipase C hydrolysis of NAPE and phosphoanandamide, which is then dephosphorylated [12]. However, 2-AG follows a different pathway where 2-arachidonate-containing phosphoinositols, converted to diacylglycerols (DAGs), are finally hydrolyzed by DAG lipase [13]. Upon synthesis these lipid messengers, due to the hydrophobic nature of the molecules, remain membrane bound till a high-affinity mechanism allows for cellular uptake to occur [14]. Maintenance of EC levels occurs through intercellular metabolism via the catabolic enzyme, fatty acid amide hydrolysis (FAAH), and monoacylglyceride lipase for AEA and 2-AG respectively [15–17].
While AEA and 2-AG may be the most widely studied ECs, they were only the first to be discovered. Since then, interest in palmitoylethanolamide (PEA) and O-arachidonyl ethanolamide (virodamine) has also been increasing [18, 19]. AEA has been found to be produced by neurons as well as immune cells and shows more selectivity for CB1 than CB2 [2]. AEA has also been shown to be an agonist at noncannabinoid receptors including vanilloid receptor-type 1 (TRPV-1) and GPR55 [20, 21]. 2-AG, which can stimulate through both CB1 and CB2 receptors equally, is found in higher concentrations in the immune system, plasma, and brain than AEA [22]. PEA has been found in the immune system and has been shown to interact with mast cells through the CB2 receptor [23]. PEA is also considered to have “entourage effect” where it is able to enhance the effectiveness of AEA and 2-AG [24]. Virodamine has been found to act as a partial antagonist at the CB1 receptor and as an agonist on CB2 [19].
All receptors associated with the EC system are G-protein coupled receptors (GPR) consisting of seven transmembrane helices, an extracellular N-terminus, and an intracellular C-terminus [2]. Somewhat controversial is the inclusion of the vanilloid and orphan GPR55 receptors to the EC system. The vanilloid receptor, which can be activated by voltage, protons, vanilloid compounds (such as capsaicin), as well as AEA, is found in the CNS and on immune cells [25–27]. However, as many non-cannabinoid molecules can act as activators for this receptor, it cannot be considered exclusively part of the EC system. GPR55 is present in tissues, including the adrenals, gastrointestinal tract, and central nervous system, and also affects the G13 peptide [21, 28]. The issue with including this receptor in the EC system is that while AEA, 2-AG, PEA, and virodamine have been found to activate the GPR55 in transformed human embryonic kidney cells (HEK293), these results are not always reproducible [21, 29]. The main cannabinoid receptors, CB1 and CB2, have been found to reside in numerous organ systems with CB1 having the highest rate of expression in the central nervous system and CB2, in the immune system [30, 31]. However, CB1 receptors have also been found in the immune system, in particular on activated T cells, and CB2 receptors have been found in the CNS as well as other periphery tissues [31, 32]. While CB receptors interact with multiple ligands, specificity is maintained through structural and conformation requirements. CB1 receptor binding requires 20–22 carbon fatty acid chains, no less than three homoallylic double bonds, and at least five saturated carbons at the end of the acyl chain [33]. Additionally, amino acid disparity in binding sites, due to single nucleotide polymorphisms, affects CB receptor affinity for individual ligands including the selectivity of AEA for the CB1 receptor [34–38].
There is little doubt that ECs affect the immune system. However, the discussion seems to be whether the endogenous cannabinoids are immunosuppressive or stimulatory. This is due to the differences observed in the use of invitro versus invivo models and in the concentration of ECs used. In vitro studies with exogenous cannabinoids found that a micromolar concentration elicits inhibitory effects while a nanomolar concentration causes stimulation of the immune system [39]. The conflicting nature of these results may be, in part, due to a biphasic response related to the concentration of CB receptors and affinity of the CB ligands [40, 41]. With this in mind, it appears that the role of the EC system in immune response is more complex than originally expected [41]. To better understand the conflicting nature of ECs, this review will focus on the role of the CB1 receptor in amelioration of T cell driven autoimmune diseases.
Role of CB1 in Systemic Immune Response
At first glance, the link between the EC system and autoimmune disease may appear tenuous or limited to neuroimmune disorders inasmuch as the CB1 receptor was initially understood to be CNS specific. However, in depth studies of the Jurkat T cell line has shown that CB1 transcript expression is significantly increased, 29-fold, upon CD3/CD28 activation compared to non-stimulated cells [42]. Even nonimmortalized primary human T cells, isolated form peripheral blood, exhibit an eightfold increase in CB1 transcript when activated [42]. This is of great importance in the understanding of autoimmune diseases as T cells are a major component of the adaptive immune system and are direct mediators of a number of autoimmune diseases including multiple sclerosis (MS) and autoimmune hepatitis or provide help to B cells to produce autoantibodies. Focusing first on 2-AG, it was found that its addition to anti-CD3 mAb-induced T cells inhibited proliferation, but only at low cell density [43]. Additionally, in vitro incubation with 2-AG was found to suppress IL-2 in activated Jurkat T cells at micromolar concentrations as well as in primary splenocytes [44, 45]. Looking at the mitogen-induced proliferation of T cells, a dose-dependent inhibition was observed when AEA was administered in vitro [46]. In an in vivo experiment, it was found that when AEA was added at micromolar concentrations, prior to OVA stimulation, it caused decrease in IFN-γ production in ConA-stimulated splenocytes [41].
As mentioned earlier, both AEA and 2-AG have been shown to act through the CB1 receptor. The G-proteins that CB1 receptors activate include Gi, inhibitory proteins, which are known to modulate adenylate cyclase [47]. Furthermore, the CB1 receptor upon activation has been found to increase ceramide, a bioactive lipid implicated in cellular differentiation, proliferation, and apoptosis [48]. Additionally, recent findings show that AEA (micromolar) works in a CB1 dependent manner to induce myeloid-derived suppressor cells, a regulatory cell type which has been shown to inhibit inflammatory responses to T cell activation [49]. These data support the EC system working through the CB1 receptor to elicit an anti-inflammatory response, upon activation of immune cells. Based on the fact that every autoimmune disorder represents a unique and complex pathological event, it is hard to make an overarching statement on how ECs will impact autoimmunity [2]. Nonetheless, in some autoimmune disorders it has been shown that increases in cannabinoid receptor density and EC concentration cause a reduction in clinical symptoms [50]. Below, we have highlighted the regulatory impact of ECs on certain autoimmune diseases.
Role of Endocannabinoids on the Regulation of Autoimmune Disease
Multiple sclerosis
MS is an autoimmune disease which causes chronic inflammation in the central nervous system, due to infiltration of Th17 and Th1 cells as well as macrophage activation [51]. Together, the T cell infiltration and macrophage activation can cause oligodendrocyte death, demyelination, and damage to axons [52]. The autoimmune responses to oligodendrocytes and myelin sheaths are considered to be hallmarks of MS; however, neurodegeneration, including loss of motor coordination as well as sensory deficits, occurs constituently along with neuroinflammation [6, 53]. MS can be divided into the more neuroinflammatory, relapsing-remitting (RRMS) disease which is more common in early adulthood, or the more neurodegenerative, secondary-progressive (SPMS) disease which often occurs later in life [54]. RRMS is characterized by acute relapses in disease followed by periods of recovery, however, in SPMS the disease continues to worsen without relapses [55]. Looking at both sides of MS, the EC system seems to have a good fit with cannabinoid receptors playing a part in the physiological function of both the central nervous system and the immune system [56].
The EC system has been examined to a great extent in both neuroinflammatory and neurodegenerative disease as the central nervous system is known to express both CB1 and CB2 receptors as well as the endogenous cannabinoids [4, 9]. In the beginning stages of neuroinflammation, microglia become activated and release proinflammatory cytokines including TNF-α and IL-6 which are cytotoxic and increase blood brain barrier breakdown by causing astrocyte activation [56]. As a source of suppression, it has been found that activated CB2 receptors can alter the proinflammatory tone to that of a Th2 anti-inflammatory tone, and that proinflammatory cytokines and reactive oxygen species produced by microglia or astrocytes can be modulated by AEA [57, 58]. Specifically, it has been shown that upon CB2 receptor activation the Th2 cytokine IL-4 is increased causing an induction of CB1 receptor transcripts [59]. Furthermore, neuronal degeneration often includes over activation of glutamate, which leads to generation of free radicals and secondary excitotoxicity among other pathologies [56]. However, AEA activation of CB1 receptors induces protection against excitotoxicity in murine brain [60]. As the CB1 receptor is associated with calcium channel inhibition, EC activation of this receptor helps in the restoration of calcium homeostasis [61, 62]. It has also been found that AEA signaling is involved in maintaining a healthy central nervous system as it suppresses microglial attacks on undamaged neurons [63].
Experimental autoimmune encephalomyelitis (EAE) is a murine model of MS. It is commonly induced by injection of myelin oligodendrocyte glycoprotein (MOG) peptide mixed with an adjuvant, into C57BL/6 mice, to mimic RRMS [64]. In a study by Witting et al., it was found that during EAE the levels of AEA and 2-AG did not significantly increase when compared to normal brain, most likely due to the elevated levels of IFN-γ [65]. IFN--γ is believed to be able to affect 2-AG production via its effect on monoacyglycerol lipase or phospholipase C both of which are necessary enzymatic steps in this ECs production [66]. However, a recent study found that exogenously administered 2-AG was able to ameliorate MOG-induced EAE as well as increase CB1 receptor expression in neuronal and inflammatory cells [67]. Focusing on AEA, a study by Centonze et al. observed that the endogenous level as well as the level of NAPE-PLD activity, an enzyme in AEA production, was increased in the striatum of the EAE murine brain, but not in the cortex [64]. Centonze et al. also showed that the level of FAAH activity was decreased in the striatum significantly, compared to normal controls [64].
Another murine model of MS is induced by a virus, Theiler’s murine encephalomyelitis virus, which causes demyelinating disease (TMEV-IDD) in SJL/J mice. This strain is highly susceptible to this form of encephalomyelitis and this model of murine MS mimics the progressive MS models [53]. A study that used UCM707, an EC uptake inhibitor, found that the levels of TNF-α and IL-1β were decreased in the TMEV-IDD MS model [53]. Similar results were reported by Ortega-Gutierrez et al. who administered methanandamide (Met-AEA), a known stable AEA analog, as well as used UCM707, with a suboptimal dose of AEA, in the loss of motor function seen in TMEV-IDD MS [53]. In a TMEV-IDD study, where EC levels were analyzed in spinal cord, 2-AG and PEA were found to be significantly increased [52]. Upon PEA treatment, Loria et al. found that TNF-α and IL-1β were both decreased compared to TMEV-IDD controls, and that microglia cell activation was decreased compared to the control mice [52]. While the effects of PEA treatment were anti-inflammatory, Loria et al. were unable to deduce if PEA itself was ameliorating the disease or if it was acting through possible “entourage” effect with AEA [52]. Looking at the levels of IL-12p70 and IL-23, which are involved in production of IFN-γ, a significant increase is seen in TMEV-infected spinal cords, and with AEA treatment, the levels of both IL-12p70 and IL-23 were decreased while IL-10 was increased in peripheral blood when compared to the levels in MS control [68]. Correa et al. also observed a reduction in the loss of motor function in both AEA and FAAH inhibitor treated mice [68]. An in vitro study using a mixed culture of cortical neurons and astrocytes, AEA at micromolar concentrations was able to exhibit neuroprotective effects in both NMDA and AMPA induced excitotoxicity, and that 2-AG and UCM707 were able to mimic these results in AMPA induced excitotoxicity [69]. Recent evidence was able to link not just ECs to the modulation of virus induced EAE disease symptoms, but also the importance of the CB1 receptor [70]. Rossi et al. found that loss of function at the CB1 receptor resulted in an increase of disease severity [70]. Additionally, they showed that FAAH knockout mice, with functional CB1 receptors, have decreased disease symptomology providing a possible link for AEA working through CB1 [70].
In a study of both RRMS and SPMS patients, EC levels were analyzed in plasma (nanomolar) during a time when no immunomodulatory drugs were given and the RRMS patients were in clinical remission [51]. Jean-Gilles et al. found that AEA and PEA levels were significantly increased in both RRMS and SPMS patients compared to healthy controls, and neither RRMS or SPMS patients exhibited increased levels of 2-AG in blood plasma [51]. It was also found that SPMS patients had significantly increased levels of AEA and PEA compared to RRMS patients, and also had significantly less FAAH present in whole blood samples possibly due to pro-inflammatory cytokines such as IFN-γ and IL-12 [51]. A cytokine-mediated reduction in FAAH could be affecting the EC levels seen in MS patients [71]. In a study of RRMS patients by Centonze et al. cerebrospinal fluid (CSF) showed a 6-fold increase for AEA in MS patients compared to normal controls [64]. Significantly higher levels of AEA and NAPE-PLD was found in the peripheral lymphocytes of RRMS patients when compared to healthy controls, while both FAAH activity and protein levels were decreased significantly [64]. RRMS patients with MRI gadolinium enhancing lesions were found to have higher levels of AEA, 2-AG, and virodamine in CSF than RRMS or SPMS patients without the lesions [72]. Taken together this suggests that ECs could be acting as a protective mechanism against neurological degradation and inflammation [64].
Autoimmune hepatitis
Autoimmune hepatitis (AIH) is one of many autoimmune liver diseases and can occur either in acute or chronic form [10]. One of the distinguishing hallmarks of AIH is autoantibodies against either smooth muscle actin, cytochrome P450, or soluble liver antigen [73,74]. While this immune response involves many inflammatory cytokines and cell types, it is believed to be T-cell mediated [75]. It was found that immunosuppressive treatment could greatly improve 10-year survival and the percentage of treatment failure in AIH patients with and without cirrhosis [76]. Standard treatment for this disease is based on research from nearly 30 years ago and includes prednisone alone or in combination with azathioprine, which must be reduced before 24 months to avoid severe side effects [77].
CB1 and CB2 receptors are expressed in the liver at low levels with an increase found during disease conditions, such as hepatocellular carcinoma [78]. Additionally, serum samples from cirrhotic rats as well as humans showed that AEA levels increase up to threefold when compared to normal controls [79]. Elevated AEA levels have been associated with reduced hypertension [80]. Also, in hepatic encephalopathy, believed to be partly derived from a systematic inflammatory response, a positive modulation of the disease was observed with exogenous 2-AG treatment [81]. Furthermore, AEA and 2-AG were increased in a murine model of hepatic ischemia [82]. These findings suggest an important role for ECs in various liver diseases.
In a study from Hegde et al., the effects of AEA were investigated in a concanavalin A (ConA)-induced hepatitis model in C57BL/6 mice [83]. It was found that when the mice were treated with CB1/CB2 mixed agonists such as CP55.939 and WIN55212, ConA-induced hepatitis could be suppressed, and that this suppression was lost upon the blocking of either cannabinoid receptor [83]. Treatment with AEA, which has a stronger binding affinity at CB1 receptors than CB2, was able to cause a dose dependent reduction in ConA-induced hepatitis as well as a decrease in TNF-α levels [83]. To further show that AEA could affect the severity of T-cell mediated hepatitis, ConA-induced hepatitis was studied in FAAH knockout mice. It was found that FAAH knockout mice showed resistance to the development of hepatitis [83]. AEA was also shown to cause hepatic stellate cell apoptosis which was found to be associated with a resolution of liver fibrosis [84,85]. Siegmund et al. also found 2-AG to cause apoptosis in the same hepatic stellate cells a year later [86]. However, when the CB2 receptor was preferentially activated and CB1 was blocked pharmacologically with SR141716A, liver fibrosis was reduced [87]. This suggests that activation of the CB1 receptor is beneficial in the modulation of acute rather than chronic liver disease as it has not yet been found to help directly in fibrotic conditions.
Rheumatoid arthritis
Rheumatoid arthritis (RA) is a common form of systemic inflammatory autoimmune disease [88, 89]. RA presents seemingly benign symptom such as pain, swelling, or stiffness of the joints; however, in a matter of months, the synovium is attacked causing hypertrophy [90]. The pathology of the synovium includes inflammation due to infiltration of T and B cells among other lymphocytes [89]. It is believed that T cells do not directly cause synovial deformation rather they work to initiate the systemic inflammation affecting the cytokine levels in the synovium [91]. TNF-α and IL-1 are the largest contributors to the cytokine profile in RA and have been proven as necessary factors in synovial destruction inasmuch as treatment with antibodies for TNF-α and IL-1 mitigated RA symptoms [92,93]. Classically, RA has been treated with nonsteroidal anti-inflammatory drugs in combination with disease modifying antirheumatic drugs, including methotrexate and cyclosporine [89]. More recently, treatments using biological response modifiers such as anti-TNF agents have been gaining favor, but all current treatments have the potential to cause side effects or toxicity with long-term use [89].
Research showing the impact of endogenous cannabinoids in RA is sparse. Recently, Naidu et al. looked into how FAAH inhibition affected inflammatory pain [94]. It was found that when WIN 55212–2, a CB1/CB2 mixed agonist, was given along with LPS, edema and pain sensitivity were reduced [95]. Along this same line, when FAAH was pharmacologically and irreversibly inhibited, by URB597 in a murine LPS paw edema model, a reduction was found in the TNF-α as well as in paw edema [94]. AEA may be important in the anti-inflammatory response observed upon FAAH inhibition; suggesting that as blockade of FAAH increased the anti-inflammatory response and analgesic effects, it could be useful in RA treatment [94]. Another study from Eros et al. examined how dietary phosphatidylcholine (PC) can affect a collagen-induced arthritis (CIA) murine model of RA [96]. Eros et al. found that when PC, a precursor of AEA, was given prior to onset of clinical signs of inflammation, CIA-induced pain were abrogated, and that damage to the synovium, cartilage, and bone was decreased when compared to disease control and treatment after the onset of symptoms [96]. Upon further examination, these data suggested that AEA may cause reductions in pain and inflammation in RA models either through exogenous additions or via inhibition of endogenous degradation.
With AEA being known to preferentially work through the CB1 receptor, these data provide indirect evidence of the importance of peripheral CB1. However, a more direct connection for the CB1 receptor in reducing cartilage destruction in RA has recently been found [97]. It was shown that adhesion of synovial fibroblasts was necessary for migration to and invasion of cartilage [97]. Lowin et al. has found that AEA, either through FAAH inhibition or exogenous treatment, causes adhesion of synovial fibroblasts to fibronectin, in a CB1-dependent manner, decreasing the migratory potential of these cells [97]. Together, these data strengthen the claim that the peripheral CB1 receptor is not only involved but important in the immunosuppression of autoimmune disease.
Furthermore, a study was done to directly assess the EC system in the synovial fluid and tissue of RA patients [98]. Richardson et al. found that AEA and 2-AG, while found to be below detectable levels in healthy controls, were both present in RA synovial fluid, and that PEA and virodamine were highly expressed in healthy samples and decreased in RA samples [98]. As no healthy control synovial tissue was collected for comparison, it can only be said that the levels of all four ECs were expressed more in the tissue (pico mole/gram) than in the fluid (pico mole/milliliter), which showed that levels in synovial fluid are not reflective of EC synthesis in the synovium [98]. Taken together, these data suggested that the EC system does play a part in RA with the loss of PEA possibly contributing to disease progression [98].
CONCLUSION
At a cellular level, ECs impact both the innate and adaptive immune systems. The findings that exogenous addition of ECs cause significant decreases in T cell proliferation and pro-inflammatory cytokine levels, suggests that as a whole, ECs, while elevated during autoimmune disease progression, appear to be enacting an anti-inflammatory tone. In particular AEA seems to be protective as inhibition of the degradative enzyme FAAH reduces disease severity in many T cell-mediated autoimmune disorders. The role of AEA in disease modulation is tied to the peripheral expression of the CB1 receptor. Inasmuch as CB2 expression on immune cells and its role has been widely reported, the importance of CB1 in immunological disorders has often been overlooked especially because it is not highly expressed on naive immune cells. However, due to the significant increase in CB1 expression on activated T cells, it should be considered an integral component of the EC system’s response to autoimmunity-induced inflammation. T cells play a critical role in the pathogenesis of autoimmune disorders by either directly causing tissue damage through production of proinflammatory cytokines, or through activation of other cells such as macrophages and B cells thereby driving the progression of the disease. This review looked at the CB1 receptor in various autoimmune diseases, primarily where T cells are considered disease mediators, finding that activation of CB1 correlates with reduced symptomology. This suggests that FAAH inhibitors or AEA could be used as therapeutics in certain autoimmune disease models. While a plethora of research has been conducted to better understand the EC system as a whole, the direct impact of this system in autoimmune disease models is lacking. A continuation of research into varied and diverse disease models will lead to a more thorough understanding of the effects and potential applications of ECs to prevent and treat inflammatory and autoimmune disease. Also, while it is commonly believed that the peripheral immune cells express CB2 receptors, the role of CB1 expression on immune cells is not well understood. Studies on CB1-mediated regulation of the immune response may help to better understand the role of psychotropic drugs, which act through CB1 receptors, on immune regulation.
Acknowledgments
This work was supported in part by the National Institute of Health Grants R01 MH094755, P01 AT003961 and P20 GM103641 to PN, R01 ES019313 to PN and MN, and R01 AT006888 and the Veterans Administration Merit Award BX001357 to MN.
Abbreviations
- 2-AG
2-Arachidonoyl glycerol
- AEA
Anandamide
- AIH
Autoimmune hepatitis
- CB1
Cannabinoid receptor 1
- CB2
Cannabinoid receptor 2
- CIA
Collagen-induced arthritis
- CNS
Central nervous system
- ConA
Concanavalin A
- CSF
Cerebral spinal fluid
- EAE
Experimental autoimmune encephalomyelitis
- EC
Endocannabinoid
- FAAH
Fatty acid amide hydrolysis
- GPR
G-protein coupled receptor
- MDSC
Myeloid derived suppressor cells(s)
- MOG
Myelin oligodendrocyte glycoprotein
- MS
Multiple sclerosis
- NAPE
N-arachidonoyl phosphatidylethanolamine
- PEA
Palmitoylethanolamide
- PC
Phosphatidylcholine
- RA
Rheumatoid arthritis
- RRMS
Relapse-remitting multiple sclerosis
- SPMS
Secondary-progressive multiple sclerosis
- THC
Δ9-Tetrahydrocannabinol
- TMEV-IDD
Theiler’s murine encephalomyelitis virus-induced demyelinating disease
Footnotes
Declaration of Interest
The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the article.
References
- 1.Chaplin DD. Overview of the immune response. J Allergy Clin Immunol. 2010;125(2 Suppl 2):S3–23. doi: 10.1016/j.jaci.2009.12.980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Tanasescu R, Constantinescu CS. Cannabinoids and the immune system: an overview. Immunobiology. 2010;215(8):588–597. doi: 10.1016/j.imbio.2009.12.005. [DOI] [PubMed] [Google Scholar]
- 3.Gaoni Y, Mechoulam R. Isolation, Structure, and Partial Synthesis of an Active Constituent of Hashish. J Am Chem Soc. 1964;86(8):1646–1647. [Google Scholar]
- 4.Matsuda LA, Lolait SJ, Brownstein MJ, et al. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346(6284):561–564. doi: 10.1038/346561a0. [DOI] [PubMed] [Google Scholar]
- 5.Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1992;365:61–65. doi: 10.1038/365061a0. [DOI] [PubMed] [Google Scholar]
- 6.Rossi S, Bernardi G, Centonze D. The cannabinoid system in the inflammatory and neurodegenerative processes of multiple sclerosis and amyotrophic lateral sclerosis. Exp Neurol. 2010;224:92–102. doi: 10.1016/j.expneurol.2010.03.030. [DOI] [PubMed] [Google Scholar]
- 7.Devane WA, Hanus L, Breuer A, et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science. 1992;258:1946–1949. doi: 10.1126/science.1470919. [DOI] [PubMed] [Google Scholar]
- 8.Mechoulam R, Ben-Shabat S, Hanus L, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol. 1995;50(1):83–90. doi: 10.1016/0006-2952(95)00109-d. [DOI] [PubMed] [Google Scholar]
- 9.Sugiura T, Kondo S, Sukagawa A, et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995;215(1):89–97. doi: 10.1006/bbrc.1995.2437. [DOI] [PubMed] [Google Scholar]
- 10.Pandey R, Mousawy K, Nagarkatti M, Nagarkatti P. Endocannabinoids and immune regulation. Pharmacol Res. 2009;60:85–92. doi: 10.1016/j.phrs.2009.03.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Di Marzo V, De Petrocellis L, Bisogno T. The biosynthesis, fate and pharmacological properties of endocannabinoids. Handbook Exp Pharmacol. 2005;168:147–185. doi: 10.1007/3-540-26573-2_5. [DOI] [PubMed] [Google Scholar]
- 12.Liu J, Wang L, Harvey-White J, et al. A biosynthetic pathway for anandamide. Proc Natl Acad Sci U S A. 2006;103(36):13345–13350. doi: 10.1073/pnas.0601832103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bisogno T. Endogenous cannabinoids: structure and metabolism. J Neuroendocrinol. 2008;20(Suppl 1):1–9. doi: 10.1111/j.1365-2826.2008.01676.x. [DOI] [PubMed] [Google Scholar]
- 14.Fowler CJ. The pharmacology of the cannabinoid system–a question of efficacy and selectivity. Mol Neurobiol. 2007;36(1):15–25. doi: 10.1007/s12035-007-0001-6. [DOI] [PubMed] [Google Scholar]
- 15.Cravatt BF, Lichtman AH. The endogenous cannabinoid system and its role in nociceptive behavior. J Neurobiol. 2004;61(1):149–160. doi: 10.1002/neu.20080. [DOI] [PubMed] [Google Scholar]
- 16.Ueda N. Endocannabinoid hydrolases. Prostaglandins Other Lipid Mediat. 2002;68–69:521–534. doi: 10.1016/s0090-6980(02)00053-9. [DOI] [PubMed] [Google Scholar]
- 17.Dinh TP, Freund TF, Piomelli D. A role for monoglyceride lipase in 2-arachidonoylglycerol inactivation. Chem Phys Lipids. 2002;121(1–2):149–158. doi: 10.1016/s0009-3084(02)00150-0. [DOI] [PubMed] [Google Scholar]
- 18.Fowler CJ, Holt S, Nilsson O, et al. The endocannabinoid signaling system: pharmacological and therapeutic aspects. Pharmacol Biochem Behav. 2005;81(2):248–262. doi: 10.1016/j.pbb.2005.01.023. [DOI] [PubMed] [Google Scholar]
- 19.Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel endocannabinoid, virodhamine, with antagonist activity at the CB1 receptor. J Pharmacol Exp Ther. 2002;301(3):1020–1024. doi: 10.1124/jpet.301.3.1020. [DOI] [PubMed] [Google Scholar]
- 20.Toth A, Blumberg PM, Boczan J. Anandamide and the vanilloid receptor (TRPV1) Vitam Horm. 2009;81:389–419. doi: 10.1016/S0083-6729(09)81015-7. [DOI] [PubMed] [Google Scholar]
- 21.Ryberg E, Larsson N, Sjogren S, et al. The orphan receptor GPR55 is a novel cannabinoid receptor. Br J Pharmacol. 2007;152(7):1092–1101. doi: 10.1038/sj.bjp.0707460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Cabral GA, Raborn ES, Griffin L, et al. CB2 receptors in the brain: role in central immune function. Br J Pharmacol. 2008;153(2):240–251. doi: 10.1038/sj.bjp.0707584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Calignano A, La Rana G, Giuffrida A, Piomelli D. Control of pain initiation by endogenous cannabinoids. Nature. 1998;394(6690):277–281. doi: 10.1038/28393. [DOI] [PubMed] [Google Scholar]
- 24.Facci L, Dal Toso R, Romanello S, et al. Mast cells express a peripheral cannabinoid receptor with differential sensitivity to anandamide and palmitoylethanolamide. Proc Natl Acad Sci U S A. 1995;92(8):3376–3380. doi: 10.1073/pnas.92.8.3376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zygmunt PM, Petersson J, Andersson DA, et al. Vanilloid receptors on sensory nerves mediate the vasodilator action of anandamide. Nature. 1999;400(6743):452–457. doi: 10.1038/22761. [DOI] [PubMed] [Google Scholar]
- 26.Mezey E, Toth ZE, Cortright DN, et al. Distribution of mRNA for vanilloid receptor subtype 1 (VR1), and VR1-like immunoreactivity, in the central nervous system of the rat and human. Proc Natl Acad Sci U S A. 2000;97(7):3655–3660. doi: 10.1073/pnas.060496197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Saunders CI, Fassett RG, Geraghty DP. Up-regulation of TRPV1 in mononuclear cells of end-stage kidney disease patients increases susceptibility to N-arachidonoyl-dopamine (NADA)-induced cell death. Biochim Biophys Acta. 2009;1792(10):1019–1026. doi: 10.1016/j.bbadis.2009.07.008. [DOI] [PubMed] [Google Scholar]
- 28.Sharir H, Abood ME. Pharmacological characterization of GPR55, a putative cannabinoid receptor. Pharmacol Ther. 2010;126(3):301–313. doi: 10.1016/j.pharmthera.2010.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Oka S, Nakajima K, Yamashita A, et al. Identification of GPR55 as a lysophosphatidylinositol receptor. Biochem Biophys Res Commun. 2007;362(4):928–934. doi: 10.1016/j.bbrc.2007.08.078. [DOI] [PubMed] [Google Scholar]
- 30.Moldrich G, Wenger T. Localization of the CB1 cannabinoid receptor in the rat brain. An immuno-histochemical study. Peptides. 2000;21(11):1735–1742. doi: 10.1016/s0196-9781(00)00324-7. [DOI] [PubMed] [Google Scholar]
- 31.Parolaro D. Presence and functional regulation of cannabinoid receptors in immune cells. Life Sci. 1999;65:637–644. doi: 10.1016/s0024-3205(99)00286-6. [DOI] [PubMed] [Google Scholar]
- 32.Onaivi ES, Ishiguro H, Gong JP, et al. Discovery of the presence and functional expression of cannabinoid CB2 receptors in brain. Ann N Y Acad Sci. 2006;1074:514–536. doi: 10.1196/annals.1369.052. [DOI] [PubMed] [Google Scholar]
- 33.Barnett-Norris J, Hurst DP, Lynch DL, et al. Conformational memories and the endocannabinoid binding site at the cannabinoid CB1 receptor. J Med Chem. 2002;45(17):3649–3659. doi: 10.1021/jm0200761. [DOI] [PubMed] [Google Scholar]
- 34.Song ZH, Bonner TI. A lysine residue of the cannabinoid receptor is critical for receptor recognition by several agonists but not WIN55212–2. Mol Pharmacol. 1996;49(5):891–896. [PubMed] [Google Scholar]
- 35.Bouaboula M, Dussossoy D, Casellas P. Regulation of peripheral cannabinoid receptor CB2 phosphorylation by the inverse agonist SR 144528. Implications for receptor biological responses. J Biol Chem. 1999;274(29):20397–20405. doi: 10.1074/jbc.274.29.20397. [DOI] [PubMed] [Google Scholar]
- 36.Song ZH, Slowey CA, Hurst DP, Reggio PH. The difference between the CB(1) and CB(2) cannabinoid receptors at position 5.46 is crucial for the selectivity of WIN55212–2 for CB(2) Mol Pharmacol. 1999;56(4):834–840. [PubMed] [Google Scholar]
- 37.Feng W, Song ZH. Effects of D3.49A, R3.50A, and A6.34E mutations on ligand binding and activation of the cannabinoid-2 (CB2) receptor. Biochem Pharmacol. 2003;65(7):1077–1085. doi: 10.1016/s0006-2952(03)00005-4. [DOI] [PubMed] [Google Scholar]
- 38.McAllister SD, Rizvi G, Anavi-Goffer S, et al. An aromatic microdomain at the cannabinoid CB(1) receptor constitutes an agonist/inverse agonist binding region. J Med Chem. 2003;46(24):5139–5152. doi: 10.1021/jm0302647. [DOI] [PubMed] [Google Scholar]
- 39.Croxford JL, Yamamura T. Cannabinoids and the immune system: potential for the treatment of inflammatory diseases? J Neuroimmunol. 2005;166(1–2):3–18. doi: 10.1016/j.jneuroim.2005.04.023. [DOI] [PubMed] [Google Scholar]
- 40.Daigle TL, Kwok ML, Mackie K. Regulation of CB1 cannabinoid receptor internalization by a promiscuous phosphorylation-dependent mechanism. J Neurochem. 2008;106(1):70–82. doi: 10.1111/j.1471-4159.2008.05336.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Ribeiro A, Ferraz-de-Paula V, Pinheiro ML, et al. Anandamide prior to sensitization increases cell-mediated immunity in mice. Int Immunopharmacol. 2010;10(4):431–439. doi: 10.1016/j.intimp.2009.12.017. [DOI] [PubMed] [Google Scholar]
- 42.Borner C, Hollt V, Kraus J. Activation of human T cells induces upregulation of cannabinoid receptor type 1 transcription. Neuroimmunomodulation. 2007;14(6):281–286. doi: 10.1159/000117809. [DOI] [PubMed] [Google Scholar]
- 43.Lee M, Yang KH, Kaminski NE. Effects of putative cannabinoid receptor ligands, anandamide and 2-arachidonyl-glycerol, on immune function in B6C3F1 mouse splenocytes. J Pharmacol Exp Ther. 1995;275(2):529–536. [PubMed] [Google Scholar]
- 44.Rockwell CE, Raman P, Kaplan BL, Kaminski NE. A COX-2 metabolite of the endogenous cannabinoid, 2-arachidonyl glycerol, mediates suppression of IL-2 secretion in activated Jurkat T cells. Biochem Pharmacol. 2008;76(3):353–361. doi: 10.1016/j.bcp.2008.05.005. [DOI] [PubMed] [Google Scholar]
- 45.Ouyang Y, Hwang SG, Han SH, Kaminski NE. Suppression of interleukin-2 by the putative endogenous cannabinoid 2-arachidonyl-glycerol is mediated through down-regulation of the nuclear factor of activated T cells. Mol Pharmacol. 1998;53(4):676–683. doi: 10.1124/mol.53.4.676. [DOI] [PubMed] [Google Scholar]
- 46.Schwarz H, Blanco FJ, Lotz M. Anadamide, an endogenous cannabinoid receptor agonist inhibits lymphocyte proliferation and induces apoptosis. J Neuroimmunol. 1994;55(1):107–115. doi: 10.1016/0165-5728(94)90152-x. [DOI] [PubMed] [Google Scholar]
- 47.Howlett AC, Mukhopadhyay S. Cellular signal transduction by anandamide and 2-arachidonoylglycerol. Chem Phys Lipids. 2000;108(1–2):53–70. doi: 10.1016/s0009-3084(00)00187-0. [DOI] [PubMed] [Google Scholar]
- 48.Galve-Roperh I, Sanchez C, Cortes ML, et al. Anti-tumoral action of cannabinoids: involvement of sustained ceramide accumulation and extracellular signal-regulated kinase activation. Nat Med. 2000;6(3):313–319. doi: 10.1038/73171. [DOI] [PubMed] [Google Scholar]
- 49.Jackson AR, Hegde VL, Nagarkatti PS, Nagarkatti M. Characterization of endocannabinoid-mediated induction of myeloid-derived suppressor cells involving mast cells and MCP-1. J Leukoc Biol. 2014;95(4):609–619. doi: 10.1189/jlb.0613350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Di Marzo V, Bifulco M, De Petrocellis L. The endocannabinoid system and its therapeutic exploitation. Nat Rev Drug Discov. 2004;3(9):771–784. doi: 10.1038/nrd1495. [DOI] [PubMed] [Google Scholar]
- 51.Jean-Gilles L, Feng S, Tench CR, et al. Plasma endocannabinoid levels in multiple sclerosis. J Neurol Sci. 2009;287(1–2):212–215. doi: 10.1016/j.jns.2009.07.021. [DOI] [PubMed] [Google Scholar]
- 52.Loria F, Petrosino S, Mestre L, et al. Study of the regulation of the endocannabinoid system in a virus model of multiple sclerosis reveals a therapeutic effect of palmitoylethanolamide. Eur J Neurosci. 2008;28(4):633–641. doi: 10.1111/j.1460-9568.2008.06377.x. [DOI] [PubMed] [Google Scholar]
- 53.Ortega-Gutierrez S, Molina-Holgado E, Arevalo-Martin A, et al. Activation of the endocannabinoid system as therapeutic approach in a murine model of multiple sclerosis. FASEB J: official publication of the Federation of American Societies for Experimental Biology. 2005;19(10):1338–1340. doi: 10.1096/fj.04-2464fje. [DOI] [PubMed] [Google Scholar]
- 54.Zajicek JP, Apostu VI. Role of cannabinoids in multiple sclerosis. CNS Drugs. 2011;25(3):187–201. doi: 10.2165/11539000-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 55.Compston A, Coles A. Multiple sclerosis. Lancet. 2002;359(9313):1221–1231. doi: 10.1016/S0140-6736(02)08220-X. [DOI] [PubMed] [Google Scholar]
- 56.Rossi S, Bernardi G, Centonze D. The endocannabinoid system in the inflammatory and neurodegenerative processes of multiple sclerosis and of amyotrophic lateral sclerosis. Exp Neurol. 2010;224(1):92–102. doi: 10.1016/j.expneurol.2010.03.030. [DOI] [PubMed] [Google Scholar]
- 57.Klegeris A, Bissonnette CJ, McGeer PL. Reduction of human monocytic cell neurotoxicity and cytokine secretion by ligands of the cannabinoid-type CB2 receptor. Br J Pharmacol. 2003;139(4):775–786. doi: 10.1038/sj.bjp.0705304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Molina-Holgado F, Lledo A, Guaza C. Anandamide suppresses nitric oxide and TNF-alpha responses to Theiler’s virus or endotoxin in astrocytes. Neuroreport. 1997;8(8):1929–1933. doi: 10.1097/00001756-199705260-00027. [DOI] [PubMed] [Google Scholar]
- 59.Borner C, Hollt V, Sebald W, Kraus J. Transcriptional regulation of the cannabinoid receptor type 1 gene in T cells by cannabinoids. J Leukocyte Biol. 2007;81(1):336–343. doi: 10.1189/jlb.0306224. [DOI] [PubMed] [Google Scholar]
- 60.Marsicano G, Goodenough S, Monory K, et al. CB1 cannabinoid receptors and on-demand defense against excitotoxicity. Science. 2003;302(5642):84–88. doi: 10.1126/science.1088208. [DOI] [PubMed] [Google Scholar]
- 61.Felder CC, Joyce KE, Briley EM, et al. Comparison of the pharmacology and signal transduction of the human cannabinoid CB1 and CB2 receptors. Mol Pharmacol. 1995;48(3):443–450. [PubMed] [Google Scholar]
- 62.Ryan D, Drysdale AJ, Lafourcade C, et al. Cannabidiol targets mitochondria to regulate intracellular Ca2+ levels. J Neurosci. 2009;29(7):2053–2063. doi: 10.1523/JNEUROSCI.4212-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Eljaschewitsch E, Witting A, Mawrin C, et al. The endocannabinoid anandamide protects neurons during CNS inflammation by induction of MKP-1 in microglial cells. Neuron. 2006;49(1):67–79. doi: 10.1016/j.neuron.2005.11.027. [DOI] [PubMed] [Google Scholar]
- 64.Centonze D, Bari M, Rossi S, et al. The endocannabinoid system is dysregulated in multiple sclerosis and in experimental autoimmune encephalomyelitis. Brain: A Journal of Neurology. 2007;130(Pt 10):2543–2553. doi: 10.1093/brain/awm160. [DOI] [PubMed] [Google Scholar]
- 65.Witting A, Chen L, Cudaback E, et al. Experimental autoimmune encephalomyelitis disrupts endocannabinoid-mediated neuroprotection. Proc Natl Acad Sci U S A. 2006;103(16):6362–6367. doi: 10.1073/pnas.0510418103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Witting A, Walter L, Wacker J, et al. P2 × 7 receptors control 2-arachidonoylglycerol production by microglial cells. Proc Natl Acad Sci U S A. 2004;101(9):3214–3219. doi: 10.1073/pnas.0306707101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lourbopoulos A, Grigoriadis N, Lagoudaki R, et al. Administration of 2-arachidonoylglycerol ameliorates both acute and chronic experimental autoimmune encephalomyelitis. Brain Res. 2011;1390:126–141. doi: 10.1016/j.brainres.2011.03.020. [DOI] [PubMed] [Google Scholar]
- 68.Correa F, Hernangomez-Herrero M, Mestre L, et al. The endocannabinoid anandamide downregulates IL-23 and IL-12 subunits in a viral model of multiple sclerosis: evidence for a cross-talk between IL-12p70/IL-23 axis and IL-10 in microglial cells. Brain Behav Immun. 2011;25(4):736–749. doi: 10.1016/j.bbi.2011.01.020. [DOI] [PubMed] [Google Scholar]
- 69.Loria F, Petrosino S, Hernangomez M, et al. An endocannabinoid tone limits excitotoxicity in vitro and in a model of multiple sclerosis. Neurobiol Dis. 2010;37(1):166–176. doi: 10.1016/j.nbd.2009.09.020. [DOI] [PubMed] [Google Scholar]
- 70.Rossi S, Furlan R, De Chiara V, et al. Cannabinoid CB1 receptors regulate neuronal TNF-alpha effects in experimental autoimmune encephalomyelitis. Brain Behav Immun. 2011;25(6):1242–1248. doi: 10.1016/j.bbi.2011.03.017. [DOI] [PubMed] [Google Scholar]
- 71.Maccarrone M, Valensise H, Bari M, et al. Progesterone up-regulates anandamide hydrolase in human lymphocytes: role of cytokines and implications for fertility. J Immunol. 2001;166(12):7183–7189. doi: 10.4049/jimmunol.166.12.7183. [DOI] [PubMed] [Google Scholar]
- 72.Di Filippo M, Pini LA, Pelliccioli GP, et al. Abnormalities in the cerebrospinal fluid levels of endo-cannabinoids in multiple sclerosis. J Neurol, Neurosurg Psychiatry. 2008;79(11):1224–1229. doi: 10.1136/jnnp.2007.139071. [DOI] [PubMed] [Google Scholar]
- 73.Manns MP, Griffin KJ, Sullivan KF, Johnson EF. LKM-1 autoantibodies recognize a short linear sequence in P450IID6, a cytochrome P-450 monooxygenase. J Clin Invest. 1991;88(4):1370–1378. doi: 10.1172/JCI115443. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Volkmann M, Martin L, Baurle A, et al. Soluble liver antigen: isolation of a 35-kd recombinant protein (SLA-p35) specifically recognizing sera from patients with autoimmune hepatitis. Hepatology. 2001;33(3):591–596. doi: 10.1053/jhep.2001.22218. [DOI] [PubMed] [Google Scholar]
- 75.Kita H, Mackay IR, Van De Water J, Gershwin ME. The lymphoid liver: considerations on pathways to autoimmune injury. Gastroenterology. 2001;120(6):1485–1501. doi: 10.1053/gast.2001.22441. [DOI] [PubMed] [Google Scholar]
- 76.Strassburg CP. Autoimmune hepatitis. Best Pract Res Clin Gastroenterol. 2010;24(5):667–682. doi: 10.1016/j.bpg.2010.07.011. [DOI] [PubMed] [Google Scholar]
- 77.Kirk AP, Jain S, Pocock S, et al. Late results of the Royal Free Hospital prospective controlled trial of prednisolone therapy in hepatitis B surface antigen negative chronic active hepatitis. Gut. 1980;21(1):78–83. doi: 10.1136/gut.21.1.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Xu X, Liu Y, Huang S, et al. Overexpression of cannabinoid receptors CB1 and CB2 correlates with improved prognosis of patients with hepatocellular carcinoma. Cancer Genet Cytogenet. 2006;171(1):31–38. doi: 10.1016/j.cancergencyto.2006.06.014. [DOI] [PubMed] [Google Scholar]
- 79.Batkai S, Jarai Z, Wagner JA, et al. Endocannabinoids acting at vascular CB1 receptors mediate the vasodilated state in advanced liver cirrhosis. Nat Med. 2001;7(7):827–832. doi: 10.1038/89953. [DOI] [PubMed] [Google Scholar]
- 80.Ros J, Claria J, To-Figueras J, et al. Endogenous cannabinoids: a new system involved in the homeostasis of arterial pressure in experimental cirrhosis in the rat. Gastroenterology. 2002;122(1):85–93. doi: 10.1053/gast.2002.30305. [DOI] [PubMed] [Google Scholar]
- 81.Magen I, Avraham Y, Berry E, Mechoulam R. Endocannabinoids in liver disease and hepatic encephalopathy. Curr Pharm Des. 2008;14(23):2362–2369. doi: 10.2174/138161208785740063. [DOI] [PubMed] [Google Scholar]
- 82.Batkai S, Osei-Hyiaman D, Pan H, et al. Cannabinoid-2 receptor mediates protection against hepatic ischemia/reperfusion injury. FASEB J. 2007;21(8):1788–1800. doi: 10.1096/fj.06-7451com. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hegde VL, Hegde S, Cravatt BF, et al. Attenuation of experimental autoimmune hepatitis by exogenous and endogenous cannabinoids: involvement of regulatory T cells. Mol Pharmacol. 2008;74(1):20–33. doi: 10.1124/mol.108.047035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Siegmund SV, Seki E, Osawa Y, et al. Fatty acid amide hydrolase determines anandamide-induced cell death in the liver. J Biol Chem. 2006;281(15):10431–10438. doi: 10.1074/jbc.M509706200. [DOI] [PubMed] [Google Scholar]
- 85.Iredale JP, Benyon RC, Pickering J, et al. Mechanisms of spontaneous resolution of rat liver fibrosis. Hepatic stellate cell apoptosis and reduced hepatic expression of metalloproteinase inhibitors. J Clin Invest. 1998;102(3):538–549. doi: 10.1172/JCI1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Siegmund SV, Qian T, de Minicis S, et al. The endocannabinoid 2-arachidonoyl glycerol induces death of hepatic stellate cells via mitochondrial reactive oxygen species. FASEB J: official publication of the Federation of American Societies for Experimental Biology. 2007;21(11):2798–2806. doi: 10.1096/fj.06-7717com. [DOI] [PubMed] [Google Scholar]
- 87.Teixeira-Clerc F, Julien B, Grenard P, et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med. 2006;12(6):671–676. doi: 10.1038/nm1421. [DOI] [PubMed] [Google Scholar]
- 88.Akhmetshina A, Dees C, Busch N, et al. The cannabinoid receptor CB2 exerts antifibrotic effects in experimental dermal fibrosis. Arthritis Rheum. 2009;60(4):1129–1136. doi: 10.1002/art.24395. [DOI] [PubMed] [Google Scholar]
- 89.Lee DM, Weinblatt ME. Rheumatoid arthritis. Lancet. 2001;358(9285):903–911. doi: 10.1016/S0140-6736(01)06075-5. [DOI] [PubMed] [Google Scholar]
- 90.McQueen FM, Stewart N, Crabbe J, et al. Magnetic resonance imaging of the wrist in early rheumatoid arthritis reveals a high prevalence of erosions at four months after symptom onset. Ann Rheum Dis. 1998;57(6):350–356. doi: 10.1136/ard.57.6.350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Feldmann M, Brennan FM, Maini RN. Role of cytokines in rheumatoid arthritis. Ann Rev Immunol. 1996;14:397–440. doi: 10.1146/annurev.immunol.14.1.397. [DOI] [PubMed] [Google Scholar]
- 92.Williams RO, Feldmann M, Maini RN. Anti-tumor necrosis factor ameliorates joint disease in murine collagen-induced arthritis. Proc Natl Acad Sci U S A. 1992;89(20):9784–9788. doi: 10.1073/pnas.89.20.9784. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Joosten LA, Helsen MM, van de Loo FA, van den Berg WB. Anticytokine treatment of established type II collagen-induced arthritis in DBA/1 mice. A comparative study using anti-TNF alpha, anti-IL-1 alpha/beta, and IL-1Ra. Arthritis Rheum. 1996;39(5):797–809. doi: 10.1002/art.1780390513. [DOI] [PubMed] [Google Scholar]
- 94.Naidu PS, Kinsey SG, Guo TL, et al. Regulation of inflammatory pain by inhibition of fatty acid amide hydrolase. J Pharmacol Exp THer. 2010;334(1):182–190. doi: 10.1124/jpet.109.164806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Compton DR, Gold LH, Ward SJ, et al. Aminoalkylindole analogs: cannabimimetic activity of a class of compounds structurally distinct from delta 9-tetrahydrocannabinol. J Pharmacol Exp Ther. 1992;263(3):1118–1126. [PubMed] [Google Scholar]
- 96.Eros G, Ibrahim S, Siebert N, et al. Oral phosphatidylcholine pretreatment alleviates the signs of experimental rheumatoid arthritis. Arthritis Res Ther. 2009;11(2):R43. doi: 10.1186/ar2651. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Lowin T, Zhu W, Dettmer-Wilde K, Straub RH. Cortisol-mediated adhesion of synovial fibroblasts is dependent on the degradation of anandamide and activation of the endocannabinoid system. Arthritis Rheum. 2012;64(12):3867–3876. doi: 10.1002/art.37684. [DOI] [PubMed] [Google Scholar]
- 98.Richardson D, Pearson RG, Kurian N, et al. Characterisation of the cannabinoid receptor system in synovial tissue and fluid in patients with osteoarthritis and rheumatoid arthritis. Arthritis Res Ther. 2008;10(2):R43. doi: 10.1186/ar2401. [DOI] [PMC free article] [PubMed] [Google Scholar]