Summary
In this exciting era, we are coming closer and closer to bringing an anti-inflammatory therapy to the clinic for the purpose of seizure prevention, modification, and/or suppression. At present, it is unclear what this approach might entail, and what form it will take. Irrespective of the therapy that ultimately reaches the clinic, there will be some commonalities with regard to clinical trials. A number of animal models have now been used to identify inflammation as a major underlying mechanism of both chronic seizures and the epileptogenic process. These models have demonstrated that specific anti-inflammatory treatments can be effective at both suppressing chronic seizures and interfering with the process of epileptogenesis. Some of these have already been evaluated in early phase clinical trials. It can be expected that there will soon be more clinical trials of both “conventional, broad spectrum” anti-inflammatory agents and novel new approaches to utilizing specific anti-inflammatory therapies with drugs or other therapeutic interventions. A summary of some of those approaches appears below, as well as a discussion of the issues facing clinical trials in this new domain.
Keywords: Antiepileptogenesis, Ketogenic diet, Fingolimod, Endocannabinoid system, Vagus nerve stimulation
Anti-inflammatory treatments may add to the therapeutic armamentarium and help to suppress seizures in epilepsies that are refractory to conventional antiepileptic drugs (AEDs). Although trial methodology for classical AEDs is well established, evaluation of anti-inflammatory treatments for epilepsy has to respect a variety of additional factors, and trial design may become more complex for reasons that will become clear in the sections below. There is expectation and hope that these treatments may even display a true antiepileptogenic effect; a trial to prevent poststroke epilepsy will be discussed in this article. The anti-inflammatory and antiepileptogenic potential of other compounds already in use, like anakinra for rheumatoid arthritis and autoinflammatory diseases and fingolimod for multiple sclerosis, is presented here. Steroids are increasingly in use for the treatment of some epileptic syndromes where an inflammatory cause is suspected. This article presents the rationale behind the administration of these well-known large-spectrum immunosuppressant drugs and weighs their expected benefits against their imminent harms. Cannabinoids modulate the nervous and immune systems by various pathways that will be discussed together with their potential as antiseizure and antiepileptogenic drugs. Nonpharmaceutical therapies for refractory epilepsy such as the ketogenic diet or vagus nerve stimulation also influence the immune system and exert anti-inflammatory effects, which will be addressed in the following chapter.
Clinical Trials of Anti-Inflammatory Agents
(Jacqueline A. French)
Inflammation may be critical to both development and perpetuation of an epileptic focus, and the underlying mechanisms for each may differ.1 Thus an anti-inflammatory drug may be effective as an antiepileptogenic agent, an anti-ictal agent that suppresses seizures in the chronic state, or both. Moreover, the impact may simply be to suppress seizures, or may be disease-modifying. Clinical trial design will differ substantially depending on the purpose of the trial. Anti-ictal studies are those studies that are performed to determine if the drug can reduce or eliminate ongoing seizures in treatment-resistant patients. These would be similar to studies performed for anti-ictal drugs with other mechanisms, but there may be some special considerations for a drug with anti-inflammatory properties. One issue would be patient selection. To date, preclinical studies have suggested that some degree of inflammation may be present in all patients with chronic epilepsy. However, it is possible that specific etiologies, such as mesial temporal sclerosis (MTS), cortical dysplasia, and tuberous sclerosis have a greater component of inflammation driving ictogenesis.2 If this is correct, it would be prudent to find patients with these specific etiologies. In addition,, inflammation may be more active at some points in the course of epilepsy than in others. It may not be simple to identify patients who have active inflammation contributing to ictogenesis, and limiting patients to those with specific etiologies may lead to poor recruitment. In future studies, it will be critical to enrich the trial population with responders, to increase effect size. Identification of imaging and/or blood biomarkers will be critical to this effort. Alternative solutions may be “enriched studies” in which patients are initially treated in an open-label fashion, and only responders undergo a randomized clinical trial (RCT), or adaptive trials, in which nonresponders are discontinued early.
Because anti-inflammatory therapies might work in a different way than standard AEDs, the timing of onset and offset of the effect may also be different. Specifically, an anti-inflammatory therapy may take time to exert its effect, and may continue to have an effect after it is withdrawn. An example is VX-765, an anti-inflammatory drug that specifically inhibits interleukin-converting enzyme (ICE), thereby reducing interleukin (IL)-1β biosynthesis and high mobility group box 1 (HMGB1) release, two molecules implicated in seizure mechanisms in experimental models.1 VX-765 is the only known anti-inflammatory agent that has been used for controlled efficacy trials in chronic focal epilepsy, although these trials were only preliminary.3 Animal studies showed a delayed start of the antiseizure effect of the anti-inflammatory molecule.4 This was ignored when designing the first study, which was very short, possibly leading to a failed trial. Only a post hoc analysis showed a probable effect, starting 1–2 weeks after drug initiation. There was also a suggestion of a delayed return to baseline seizure frequency after the drug was withdrawn. Unfortunately, the second study of this drug was terminated after only 10 subjects/arm were enrolled, due to change of priorities on the part of the pharmaceutical company that was developing the molecule. Nonetheless, some very important lessons were learned that can and should be used to inform future trials of anti-inflammatory interventions.
No trials have been done in epilepsy that specifically contain an endpoint to demonstrate disease modification. Disease modification trials have been planned and attempted in other chronic central nervous system (CNS) diseases such as Alzheimer’s and Parkinson’s disease.5,6 Trial design options include those that capitalize on delayed start, continued benefit after treatment discontinuation, and active comparison to a non-disease-modifying drug with comparison of slope of improvement over time. These trials may not be easy to perform, and there will need to be some understanding of time to effect and other drug characteristics, to increase the odds of success. Again, biomarkers would be helpful to determine improvement along the way, particularly if there might be substantial delay to effect.
Future directions
Trials to demonstrate antiepileptogenic effects would be the most difficult, and would require careful preclinical studies demonstrating a strong effect of the compound in animal models at relevant doses, duplicated in at least two studies or through a multicenter collaboration. Because most epilepsy risk factors (such as traumatic brain injury and stroke) cause epilepsy in only a subset of affected individuals (typically 10–25%), and epilepsy does not usually develop immediately, trials would need to be long, and would need to recruit a large number of patients. The burden of the therapy should match the potential expected risk reduction, and the trial would have to be planned in such a way that the population is recruitable. Many previous antiepileptogenesis trials have failed due to poor recruitment, or other design flaws.7 Availability of biomarkers could substantially increase the likelihood of success by either enriching recruitment for patients who will develop epilepsy, or providing an early indicator of benefit.
A Proposal for an Anti-epileptogenesis Trial in Post-stroke Epilepsy
(Matthias Koepp)
Stroke is the third leading cause of death and a major cause of disability in Europe; it affects one in six adults with an estimated 3–6 million stroke cases annually.8 Individuals who have had a stroke have an increased risk of epilepsy (~10%),9 with most epilepsy cases occurring within 24 months after stroke. When unprovoked seizures occur, they further impair an already compromised quality of life, worsen the degree of cognitive disability, and are associated with an increased risk of subsequent dementia.10 There are no treatments to prevent the development of these disabling comorbidities11 and there are only few effective pharmacologic interventions that facilitate poststroke recovery.12
Although progress has been made in understanding the cellular and molecular mechanisms of ischemic tissue damage, neuroprotective and regenerative treatments that could improve outcome in patients recovering from stroke are lacking. Stroke induces a complex cascade of different inflammatory mediators and cytokines, and aging, a risk factor for stroke, further exacerbates neuroinflammatory responses. There is increasing evidence that inflammatory changes in the brain after stroke also promote the development of epilepsy, that is, epileptogenesis, among many other changes through processes triggered by the pro-inflammatory cytokine interleukin (IL)-1β and inhibited by the endogenous IL-1 receptor antagonist (IL-1ra).1,13,14 Because epileptogenesis is variable and only a minority of patients develop epilepsy following stroke,15 clinical trials of potentially disease-modifying treatments would benefit greatly from the ability to identify reliably those individuals who are most likely to develop epilepsy after stroke.
Rationale for repurposing an anti-inflammatory compound
Major advances have been made in understanding basic mechanisms of epileptogenesis and epilepsy in a variety of animal models. Among etiologies of epilepsy, which are readily detectable and could permit early interventions, stroke stands out prominently, particularly because patients at-risk and associated epileptogenic mechanisms can be identified within a reasonable time from the insult. One mechanism that has emerged as having primary importance for recovery from stroke and epileptogenesis, is brain inflammation. The available preclinical data support the hypothesis that brain inflammation can play a crucial role not only in promoting epileptogenesis after stroke, but also in sustaining recurrence of seizures once an epileptic condition has become established.16
There is evidence indicating that centrally active anti-inflammatory agents can exert neuroprotective effects in animal models of seizure-related cell damage: administration of the human recombinant IL-1ra (anakinra) for 7 days after pilocarpine- or electrically induced status epilepticus (ElectrSE) yielded therapeutic drug levels in brain, and decreased both IL-1β expression in astrocytes and cell loss in rat forebrain.17 Anakinra given in combination with VX-765, a specific inhibitor of the biosynthetic enzyme of IL-1β, afforded significant neuroprotection in the ElectrSE rat model when given as an intravenous bolus (33 mg/kg, 10 mg/200 μL) followed by sustained subcutaneous infusion with osmotic minipumps (24 mg/day; 80 mg/kg/day) for 1 week starting 3 h after the onset of status epilepticus. Anakinra treatment was based on previous evidence of neuroprotection in rat and human stroke.18 Immunohistochemical analysis of brain sections at the end of treatment showed reduction in IL-1β expression in glial cells concomitant with neuroprotection in forebrain areas. However, onset of epilepsy and frequency and duration of seizures assessed 3 months after ElectrSE were not significantly modified. In subsequent studies, however, the combination of anakinra with a cyclooxygenase-2 (COX-2) inhibitor, or with BoxA, a peptide blocking the effects of the inflammatory molecule HMGB1, administered at the time of status epilepticus induction, was able to significantly decrease spontaneous seizures in rats.
Therefore, there is a clear rationale for conducting a proof-of-concept study to test the ability of IL-1 antagonists to inhibit brain inflammation in stroke patients. Demonstration of a central anti-inflammatory action of IL-1 antagonists in the relevant human population is the prerequisite for the design and execution of a randomized controlled trial to determine whether these agents are also effective in achieving inhibition of epileptogenesis, and functional recovery after insult.
Objectives for antiepileptogenesis trial
The development of new, potentially antiepileptogenic therapies has been stagnating, partly due to lack of clinically validated biomarkers to reliably predict who is most likely to develop epilepsy in a short time frame. In fact, currently available tools (e.g., routine magnetic resonance imaging [MRI] and electroencephalography [EEG]), are not accurate predictors for the risk of future seizures, or for the need to start antiepileptic drug treatment after a first unprovoked seizure.
Thus specific objectives for an antiepileptogenesis trial are the following:
-
To evaluate the efficacy of a novel therapeutic strategy in patients following stroke who are at highest risk of developing epilepsy.
As a first step, it is important to measure the development of a hyperexcitable state following an initial epileptogenic lesion, which underpins the process leading to the occurrence of late unprovoked seizures. Because it is inconceivable to test putative antiepileptogenic drugs over long periods in populations with a very low (<10% over 2 years) risk of a seizure, the challenge is to identify an enriched population of patients with the highest risk of developing epilepsy after stroke. Stroke patients who develop one or more acute symptomatic seizures represent a potentially adequate population for such a trial, as their risk of subsequently developing unprovoked seizures, that is, epilepsy, is about 30% over 2 years.19
-
To quantitate the response to anti-inflammatory treatment using imaging, electrophysiologic, and circulating biomarkers.
The “conditio-sine-qua-non” of an antiepileptogenic treatment targeting poststroke brain inflammation is to demonstrate its capacity to reduce inflammation in the CNS. This may be possible by measuring inflammatory serum biomarkers mirroring brain inflammation, but more specifically by imaging brain inflammatory changes directly using positron emission tomography (PET) ligands for translocator protein 18 kDa (TSPO), which measure in vivo microglial activation.20 TSPO is normally only lightly expressed in the brain, but it is drastically upregulated in response to neuroinflammatory stimuli. This upregulation correlates with microglial activation or infiltration of macrophages following stroke.21
To determine the capacity of biomarkers to predict the development of poststroke epilepsy.
An alternative to evaluating the efficacy of the intervention in preventing epilepsy is to assess the effect of the intervention on appropriate surrogate biomarkers of epileptogenesis with sufficient sensitivity and specificity. Even within the high-risk group, the risk of recurrent seizures will remain below 20% at 1 year. Identification of reliable biomarkers for epileptogenesis would facilitate and shorten targeted trials of novel antiepileptogenic therapies.
Future directions
The quest for appropriate surrogate biomarkers with sufficient sensitivity and specificity is essential for evaluating novel interventions during short-term trials, as well as for identifying populations at higher risk of developing epilepsy than those who can be currently selected. In patients with stroke, the risk of recurrent seizure remains below 10% at 2 years. Thus we propose to focus on an enriched population of patient with stroke and an acute symptomatic seizure, that is, those with the highest risk (~30%) of developing epilepsy.
Fingolimod (FTY720), Is It a Potential Antiepileptic Drug?
(Yvonne Naegelin)
FTY720 has been developed after being isolated from the fungus Isaria sinclairii. The synthetic compound fingolimod (Gilenya, 2-amino-2-(2-(4-octylpheyl)ethyl)propane-1,3-diol, C19H33NO2) acts mainly over sequestration of circulating lymphocytes to the lymph nodes without major alterations of their immune functions. It was meant to prevent allograft rejection without inducing a severe immunosuppression but did not show enough immunosuppressive effect in the context of tissue transplantation.22 It did so, in the context of multiple sclerosis (MS), where it was the first oral drug being approved for relapsing remitting forms, reducing the annual relapse rate by roughly 50%.23 Acceleration of homing and blocking egress from lymph nodes (over sphingosine 1-phosphate (S1P1) receptor downregulation) is thought to be the main mode of action for its efficacy in MS.24
It is well established that inflammation can be a consequence as well as a cause of epilepsy.25 In contrast to the pathogenesis of MS, where there is a marked activation of the adaptive immune system with infiltrating B and T lymphocytes,26 brain inflammation in epilepsy is dominated by innate immunity cells including activated microglia, astrocytes, as well as granulocytes and monocytes/macrophages.27 There seems to be a link between the innate and adaptive immuneresponse,28,29 possibly explaining why there is some evidence of a scarce presence of adaptive immune cells such as T or B cells in nonautoimmune forms of epilepsy such as TLE.27,28,30 In general, the involvement of the adaptive immunity appears to be much more related to the autoimmune forms of epilepsy.31
Nevertheless, the two entities might share some common pathways and therefore some treatment approaches.
FTY720 is a potent nonselective agonist at S1P1 and S1P3–5 receptors.32 FTY720 has a half-life of 6 days and reaches steady-state blood concentrations after 1–2 months of daily dosing. It reduces lymphocytes (homing) within hours after first dose administration, an effect being fully reversible within 6–8 weeks after stopping treatment.33 Its second most relevant and temporary side effect for clinical applications (negative chronotropy) is caused by targeting S1P1 and S1P3 receptors on atrial myocytes. FTY720 crosses the blood-brain barrier (BBB)34 to bind S1P receptors located on neural cells in the CNS (S1P1, -3, -5). The S1P2 receptor is located on neural cells as well but not targeted by FTY720. The S1P4 receptor is located only on lymphocytes, but the major effect on lymphocytes is induced by targeting S1P1 receptors on those cells. FTY720 is phosphorylated by sphingosine kinases to FTY720-P.35 This probably is taking place within the CNS: after oral administration of FTY720, the brain concentrations of FTY720 are 10–27-fold higher than peripheral concentrations.34 Within the brain, FTY720 and FTY720-P reach nearly the same concentrations.34
S1P receptors are also enriched on astrocytes, and these cells are key players in the generation and perpetuation of brain inflammation both in MS and epilepsy as well as in the formation and preservation of the BBB.36 FTY720-P mediates anti-inflammatory effects on astrocytes as well as extracellular-signal regulated kinase (ERK) phosphorylation by activation of S1P1.37 The ERK cascade regulates many distinct processes such as proliferation, differentiation, survival, as well as apoptosis of cells. Some data do link ERK activation to epilepsy, but its involvement is still not fully understood. In vitro experiments show a beneficial effect by FTY720-P and FTY720 on endothelial cells, and S1P modulation seems to reduce transmigration of peripheral blood mononuclear cells through the BBB.38
In summary, there are various direct effects of FTY720 on the CNS resident cells that go far beyond the homing of peripheral lymphocytes, and most of them are thought to be receptor mediated.39 These effects may contribute to the therapeutic actions of this drug.
Apart from those receptor-mediated effects, FTY720-P directly binds to histone deacetylase (HDAC), thereby possibly having a direct effect on epigenetic gene regulation.40 In addition, there seems to be an interaction with other lipids, an inhibition of the cannabinoid receptor CB1, and of phospholipase A2 activity in mast cells.41
FTY720 has also been shown to increase brain-derived neurotrophic factor (BDNF) levels and ameliorates symptoms in methyl CpG binding protein 2 (MECP2)-null mice, a model of Rett syndrome.42 Whether this effect is S1P-mediated is not completely understood. BDNF is upregulated by seizures in animal models, and in brain specimens and blood serum from human pharmacoresistant epilepsy.43 The current understanding is that BDNF can significantly impact seizures, with either beneficial44 or detrimental effects being reported. One recent study has shown both anti-inflammatory and antiepileptogenic effects of FTY720 in a lithium-pilocarpine model of epilepsy in rats.45
Because FTY720 ameliorates symptoms of MECP2-null mice,42 we are performing a clinical study to assess safety and efficacy of oral FTY720 in children with Rett syndrome (FINGORETT). The study (phase I–II) has a focus on safety and was not designed for detecting therapeutic effects on seizures of FTY720 in those children. Nevertheless, information about seizure frequencies before, under, and after treatment are being collected, and EEG studies are recorded on a regular base within the study. The final results will be available in spring of 2017.
Future directions
FTY720 by its multiple modes of action on the immune system, the epigenetic machinery and BDNF signaling may be an interesting candidate as an anti-inflammatory and disease-modifying drug for the treatment of refractory epilepsy. The good penetration across the BBB is another advantage of this drug. Approval and large clinical experience in patients with MS should facilitate and speed up the planning of a clinical trial.
Large Spectrum Anti-inflammatory Treatments: Friend or Foe?
(Federico Vigevano)
The evidence of neuroinflammation and immune mechanism involvement in the genesis of epilepsy is growing,1,25,46 thus opening the way to immune therapies for treating epilepsy. Immunotherapies are now routinely used in severe epilepsies with potential immune-mediated pathogenesis, such as Rasmussen encephalitis, anti-NMDAR (N-methyl-D-aspartate), anti-GAD (glutamic acid decarboxylase) or anti-VGKC (voltage gated potassium channel) complex encephalitis and fever-induced refractory epileptic encephalopathy in school-aged children.47,48 Beyond these clinical entities, drugs with anti-inflammatory and immunomodulatory actions are considered also in epileptic disorders lacking specific immunologic markers.49
Adrenocorticotropic hormone (ACTH) and corticosteroids (prednisone, prednisolone, and hydrocortisone) are widely used in some forms of epilepsies. ACTH and corticosteroids are mostly administered to treat patients with epileptic encephalopathy (EE), a group of severe clinical entities with heterogeneous clinical presentations and variable causes.50 EE includes prenatal causes like brain malformations, chromosomal or genetic abnormalities and neurocutaneous diseases, perinatal causes such as hypoxic ischemic injuries, and postnatal causes such as vascular or infectious insults.51 In EE, interictal epileptiform EEG abnormalities play a significant role in generating progressive deterioration in neurologic function.52 Thus the aim of treatment is not limited to seizure control; more often, the greatest challenge is to improve the child’s psychomotor development, a goal that requires suppression or reduction of interictal EEG discharges. With few exceptions, ACTH and corticosteroids are considered first-line agents in all cases of infantile spasms.53 Therapeutic efficacy has been also reported in other epileptic EE such as Lennox-Gastaut syndrome, Landau-Kleffner syndrome, continuous spike and wave during sleep (CSWS), and other forms of epilepsy resistant to conventional AEDs.50
Little is known on the mechanism of the antiepileptic action of ACTH and corticosteroids, as well as on the pathogenesis of EE. However, various hypotheses have been put forward. At present the most accredited hypothesis is that ACTH and corticosteroids exert their actions through modulation of the hypothalamic-pituitary-adrenal axis. However, the possibility of direct effects on the immune system is also considered. In vitro, ACTH and corticosteroids stimulate the growth of neuroblasts, a property that might be relevant for the treatment of EE occurring in the first year of life. ACTH and corticosteroids may also modulate various neurotransmitter systems and voltage- or receptor-gated ion channels. In particular, ACTH downregulates serotonin 5HT2 receptors in the cerebral cortex, and modulates GABA and dopamine receptors, an effect that, in animal models, is age-dependent.54
In the last few years, increasing evidence has accumulated that certain forms of severe epilepsy could have an inflammatory and/or immunologic basis. Hence, the antiseizure effects of ACTH and corticosteroids, as well as immunoglobulins, may depend on their well-known anti-inflammatory and immunosuppressant effects.55
ACTH and corticosteroids may cause significant adverse effects. In about two thirds of patients, a transient cerebral atrophy has been described, detectable by brain MRI. This finding may sometimes be a confounding factor in the diagnostic process. Transient dyskinesia with hyperkinetic movements involving face and limbs was observed during treatment with corticosteroids or ACTH. ACTH represents the first-line agent for the treatment of infantile spasms, except for spasms associated with tuberous sclerosis.56 However, conclusive data supporting the long-term efficacy of ACTH are still lacking in the literature. There are also inadequate data on the optimal dosage and duration of therapy, although short duration and low dose are preferable. The issue of possible efficacy differences between natural and synthetic ACTH is also an object of debate.57
Future directions
Immunotherapy trials for the treatment of drug-resistant epilepsy represent a future therapeutic aim. This requires a better definition of how etiopathogenetic immune mechanisms are involved in the mechanisms of epileptogenesis.
The more precise concepts of how inflammation and the immune system may cause epilepsy and/or influence the course of disease will help to refine clinical trials involving the steroids as a class of anti-inflammatory drugs displaying broad effects on the immune system. These trials should also aim at identifying those steroid compounds that may be more effective and have a better adverse event profile.
Anti-Inflammatory Aspects of the Ketogenic Diet and PUFAs
(Stéphane Auvin, Jong M. Rho)
The ketogenic diet (KD) is a high-fat, low-carbohydrate diet characterized by ketonemia, relative hypoglycemia, and high fatty acids levels. The KD is an established treatment for pharmacoresistant epilepsy, including some inflammation-induced epileptic encephalopathies such as febrile infection-related epilepsy syndrome (FIRES). Although the antiseizure mechanism(s) remain(s) unclear, new potential clinical applications for the KD are rapidly emerging, principally on the basis of its broad neuroprotective actions in various experimental models of neurologic disease, importantly those that significantly involve aberrant inflammatory responses.58
Although the KD has been investigated in multiple seizure and epilepsy models, variations on this metabolism-based treatment have also been administered in experimental models of pain, MS, Alzheimer’s and Parkinson’s diseases, neurotrauma, Autism Spectrum Disorder (ASD), and even malignant brain cancer (Table 1). Furthermore, there is now abundant evidence that the KD possesses anti-inflammatory properties, and specifically, this diet has been shown to decrease proinflammatory cytokine levels after an immune challenge.59 Although the precise mechanisms underlying such effects in these models remain unclear, there are likely multiple, parallel, and synergistic processes and molecular targets, similar to what has been proposed for epilepsy.58 For example, polyunsaturated fatty acids (PUFAs), dietary lipids that contain more than one double bond, may play an important mechanistic role in KD action. Systemic PUFA levels rise in response to KD treatment, and have been reported to block epileptiform activity in models in vitro as well as in acutely provoked seizures in rodents. There are two groups of PUFAs: the omega-3 (n-3) and the omega-6 (n-6) PUFAs. This nomenclature refers to the position of the double bond relative to the methyl terminal of the molecule.60 N-3 PUFAs can decrease the production of inflammatory eicosanoids, cytokines, and reactive oxygen species, and the expression of adhesion molecules. In addition,, n-3 PUFAs act both directly (by replacing arachidonic acid as an eicosanoid substrate and inhibiting arachidonic acid metabolism) and indirectly (by altering the expression of inflammatory genes through effects on transcription factor activation). In addition,, n-3 PUFAs, particularly eicosapentaenoic acid (EPA; 20:5n-3) and docosahexaenoic acid (DHA, 22:6n-3), also exert anti-inflammatory actions, primarily through their hydroxylated metabolites, which include resolvins and docosanoids.61 Furthermore, PUFAs can bind to and activate peroxisome proliferator-activated receptors (PPARs), including both PPARα and PPARγ. It is notable that synthetic PPARs agonists have been shown to reduce experimentally induced inflammation. This effect is the result of the inhibition of pro-inflammatory pathways involving nuclear factor kappa B (NF-κB), signal transduction and transcription-1, and nuclear factor of activated T cells.62
Table 1.
Examples of utilization of KD as an anti-inflammatory treatment for various pathologic models
References | Type of diet | Species | Model | Anti-inflammatory effect of KD |
---|---|---|---|---|
67 | 6.6:1 KD 3 weeks before Exp. |
Rats | Subcutaneous injection of complete Freund’s adjuvant into one hind paw | Decrease both swelling and plasma extravasation |
68 | Made by the lab 2 weeks before Exp. |
C57BL/6J Mice | MPTP model | Decrease of activated microglia (Iba 1 staining) Decrease of IL1β, IL-6, TNFα (ELISA of SN) |
69 | 6.3:1 KD (Bio-Serv F3666 diet) 7 days before Exp. |
C57BL/6 mice | Experimental autoimmune encephalomyelitis S.C. myelin oligodendrocyte glycoprotein (MOG)35–55 peptide + complete Freund’s adjuvant (CFA) I.V. 20 ng of pertussis toxin |
2-2.5-fold reduction in CNS-derived CD4+ cells and CD11b+ CD45+ cells (macrophage and microglia tendency toward increased CD4+ CD25+ Foxp3+ Treg cells Lymph node & CNS reduction in cytokines (IL-1β, IL-6, TNF-α, IL-12, IL-17) and chemokines (IFN-γ, MCP-1, MIP-1a, MIP-1b) |
70 | 6.3:1 KD (Bio-Serv F3666 diet) 4 weeks before Exp. |
C57BL/6 mice | Liver and white adipose tissue (WAT) | Liver: Increase of expression of Tnfα, Il-6, Emr1, Cd68, Itgam, Nlrp3 WAT: Decrease of expression of Tnfα, Il-6, Emrl, Cd68, Itgam, Nlrp3 |
58 | 3:1 KD (Ketocal) 2 weeks before Exp. |
Wistar rats | Fever model 50 μg/kg of LPS (Escherichia coli 055:B5) |
Modulate raise of body temperature Blood: Reduce IL-1β, TNF-α Brain: Reduce IL-1β mRNA |
Another KD-related mechanism involved in neuroinflammation is ketone-induced disruption of inflammasome assembly, and ketones such as β-hydroxybutyrate may modulate inflammation through actions on mitochondrial targets.63 Clearly, with growing evidence that the KD affords anti-inflammatory activity in a variety of animal models and human epileptic conditions (such as FIRES), the concept that inflammation as both a cause of and therapeutic target for epilepsy (and other neurologic conditions) is becoming more valid and worthy of further investigation.58,64
Future directions
Preclinical studies should further characterize the mechanisms underlying the anti-inflammatory effects of the KD and its substrates/mediators, not only for epilepsy but also for other neurologic disorders. One intriguing aspect is the potential inflammatory basis of comorbid conditions such as epilepsy and ASD. There is some preliminary clinical59 and growing experimental evidence65 that the KD is effective in mitigating core symptoms of ASD, and although the mechanisms underlying such behavioral benefits remain unclear, one intriguing possibility is diet-induced alterations in the gut microbiota, resulting in an anti-microbial-like effect and comparable shifts in specific bacterial abundance seen in humans with autism.66 Clearly, the KD appears to render beneficial effects in both epilepsy and ASD. Further studies would provide a stronger scientific rationale for much-needed clinical trials for testing unique and complementary treatment approaches for affected patients.
Endocannabinoid System, CBIR/CB2Rs, and Inflammation
(Orrin Devinsky)
Δ9-THC modulates the endocannabinoid system, an endogenous signaling system with hydrophobic ligands N-arachidonoyl ethanolamide (anandamide, AEA)71 and 2-arachidonoyl-glycerol (2-AG).72 These compounds are produced from postsynaptic membrane phospholipid precursors and released in an activity-dependent, “on-demand” manner. Hydrolysis of endocannabinoids produces arachidonic acid, a precursor for COX-2-mediated production of inflammatory prostaglandins.73 AEA is degraded by the enzyme fatty acid amide hydrolase (FAAH) into arachidonic acid and ethanolamide,74,75 and 2-AG is catabolized by monoacylglycerol lipase (MAGL) and α/β-hydrolase domain containing 6 (ABHD6)76 into arachidonic acid and glycerol.75–77 Inhibition of the degradative enzymes of AEA and 2-AG shunts lipid mediators away from proinflammatory prostaglandin mediators and toward anti-inflammatory endocannabinoids,68 thereby reducing neuropathic pain,78,79 Aβ-mediated inflammation, and oligodendrocyte excitotoxicity.81 In addition, 2-AG directly inhibits COX-2 function, potentially via nuclear peroxisome proliferator–activated receptor gamma (PPARγ) receptor.82
The primary targets for the endocannabinoids are the cannabinoid receptors type-1 (CB1R) and -2 (CB2R). CB1R is the primary CNS receptor, particularly in the hippocampal mossy cell-granule cell synapses, and to a lesser extent, on microglia, astrocytes, and oligodendrocytes.74 Although implicated primarily in peripheral signaling, CB2Rs are also expressed in the brain,83 mediating neuronal excitability84 and inflammation in microglia.85 Kainic acid-induced status epilepticus86 and lipopolysaccharide (LPS)-induced inflammation87 upregulate CB1R expression, either as a direct consequence of inflammation-induced sequelae or as a potential compensatory mechanism to limit prolonged hyperexcitability. Supporting a potential anti-inflammatory role of CB1R/CB2Rs, CB2R agonism decreases microglial activation, edema, excitotoxicity, oxidative stress, and cell death associated with stroke,88 germinal matrix hemorrhage,89 and traumatic brain injury. Furthermore, activation of CB1Rs91 and CB2Rs92 via synthetic agonists reduced inflammatory nociception in several animal models.
Δ9-Tetrahydrocannabinol (Δ9-THC) and inflammation
Both cannabidiol (CBD) and Δ9-tetrahydrocannabinol (Δ9-THC) produce net anti-inflammatory effects, although through different target receptor mechanisms (Table 2). Δ9-THC acts primarily as a partial agonist at CB1Rs and CB2Rs on microglia, the primary CNS immune cells. Δ9-THC reduces LPS-induced inflammation in vitro and in vivo by limiting release of the pro-inflammatory cytokines IL-1β, IL-6, IL-17, tumor necrosis factor α (TNFα), and interferon β (IFNβ), and elevating anti-inflammatory cytokines such as IL-10.88–94 These effects may be age-specific, however, as treating adolescent mice Δ9-THC exerts opposite effects when assayed later in life, increasing pro-inflammatory and decreasing anti-inflammatory cytokine release.95 In other studies, Δ9-THC limits oxidative stress in LPS-triggered inflammation,98 reduces nitrite formation following intravitreal NMDA injection,99 and decreases glutamate neurotoxicity and oxidative stress.100 Anti-oxidative effects of Δ9-THC may be independent from, or only partially mediated by, CB1Rs/CB2Rs.100,101 Other proposed anti-inflammatory targets for Δ9-THC (or related compound Δ9-tetrahydrocannabivarin, Δ9-THCV) include transient activation and desensitization of TRP (transient receptor potential) channels TRPA1, TRPV1–4,102–104 and antagonism of TRPM8,103 which may regulate postinflammatory Ca2+ influx. Similar to endocannabinoids, Δ9-THC may reduce COX-2 activation via PPARγ,105 although a Δ9-THC-mediated, CB1R-dependent increase in COX-2 and PGE2106 may be dose dependent. Collectively, Δ9-THC primarily exerts anti-inflammatory properties in animal models of inflammation (in vivo) including cerebral artery occlusion–induced ischemia,107,108 acute palmar inflammation,109 5XFAD APP transgenic (Alzheimer’s disease model) mice,106 and the myelin oligodendrocyte glycoprotein (MOG)35-55-induced experimental autoimmune encephalomyelitis model of MS.101
Table 2.
Potential anti-inflammatory mechanism of Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD)
Anti-inflammatory mechanism | References | |
---|---|---|
Δ9-THC | (1) CB1R/CB2R partial agonist | 137 |
(2) ↓pro-inflammatorycytokines (IL-1β, IL-6, IL-17, TNFα, IFNβ) ↑ anti-inflammatorycytokine (IL-10) | 93–97 | |
(3) TRP channels agonist (TRPV1-4, TRPA1), TRPM8 antagonist | 102–104 | |
(4) ↓(or↑) COX-2 activation via PPARγ, dose-dependent | 105, 106 | |
CBD | (1) Regulation of intracellular Ca2+ via mitochondrial Na+/Ca2+ exchanger, or VDAC channel | 117, 118 |
(2) Inhibition of ENT-1 transporter, ↑ adenosine at A2A receptor | 120, 121 | |
(3) ↓pro-inflammatory cytokines (IL-Iβ, IL-3, IL-6, IL-12, IL-17, TNFα, IFNβ/γ) ↑ anti-inflammatory | 94, 101, 113, 120, 121, 125, 127 | |
cytokines (IL-4, IL-10) | 102–104 | |
(4) TRP channels agonist (TRPV1–4, TRPA1), TRPM8 antagonist | 116 | |
(5) G-protein-coupled-receptor GPR55 antagonist | 105 | |
(6) ↓ COX-2activation via PPARγ | 124, 128 | |
(7) ↓ activation of microglial VCAM-I, CCL2, CCL5 transmigration of leukocytes, (mediated in part by adenosine A2A receptors) | 94, 122–124, 126 | |
(8) ↓ oxidative stress, lipid peroxidation, caspase 3 activation, ROS (iNOS, NO), nuclear stress response (Nrf2→ ↓NFκB, ↑STAT3) |
Cannabidiol (CBD) and inflammation
CBD has very low affinity at CBRs,110,111 but acts as an agonist at TRP channels (TRPV1, TRPV2, and TRPA1),103,104,110,112 5HT1α receptors,113 and glycine receptors.114 CBD is an antagonist at TRPM8 channels,102 T-type voltage-gated calcium channels,115 and the G protein-coupled receptor GPR55.116 CBD may have unique effects on inflammation through dynamic regulation of intracellular calcium stores via multiple, activity-dependent pathways.117 CBD induces a bidirectional change in intracellular calcium levels that depends on cellular excitability: slightly increasing intracellular [Ca2+] under normal physiologic Ca2+ conditions and reducing intracellular [Ca2+] under high-excitability conditions. These changes may be mediated by the mitochondrial Na+/Ca2+ exchanger.117 CBD also produces biphasic changes in intracellular calcium levels via antagonism of the mitochondrial VDAC1 channel.118 Furthermore, CBD is an agonist at PPARγ119 and competitive antagonist at the equilibrative nucleotide transporter (ENT-1), reducing adenosine uptake at baseline120 and during LPS-induced inflammation in vitro121 and in vivo.120
CBD exerts dynamic changes in intracellular signaling to reduce oxidative stress, as assayed via genome-wide microarray studies. At baseline, CBD treatment in cell cultures targets pathways implicated in oxidative stress and glutathione depletion (e.g., GCN2, PKR, and eIF2a) and nuclear oxidative stress response (e.g. Nrf2).122,123 Upon LPS stimulation, CBD triggers Nrf2 activation, inhibits NFκB and AP-1, and activates MAPK, JAK/STAT, and cell cycle regulatory pathways, producing net anti-inflammatory effects and immunosuppression.94,124 CBD regulates oxidative stress post hypoxic-ischemia107,113,125 and oxygen-glucose deprivation, in part by reducing LDH efflux, caspase-9 activation, and iNOS expression.126 Similar to Δ9-THC, CBD reduces pro-inflammatory cytokines and IFNβ/γ and increases anti-inflammatory cytokines such as IL-4 and IL-10 via a CB1R/CB2R mechanism, potentially through adenosine or PPARγ signaling.94,101,113,120,122,126,127 This mechanism also limits Th17 differentiation and function, and reduces leukocyte transmigration, partly by downregulating VCAM-1, CCL2, and CCL5, in animal models of MS.124,128 In addition, CBD increases cell survival and reduces oxidative stress in in vitro and in vivo models of Aβ (1–42)-induced inflammation.126–129
Cannabinoids and neuroprotection from seizures
The anti-inflammatory effects of the endocannabinoid system, Δ9-THC, and CBD may help to explain the neuroprotective effects of cannabinoids in animal models of seizures. In chronic (e.g., pilocarpine-induced) seizure models, synthetic CB1R agonism130 or CBD131,132 administered during the chronic phase reduces neuronal damage and oxidative stress/autophagy. In acute (e.g., kainic acid) preclinical studies, CB1R activity133 or elevated endocannabinoid signaling134,135 provides neuroprotective effects postseizure.
Future directions
CBDs are potentially promising anti-inflammatory drugs. Yet the specific nature, extent, and specificity of their immunomodulatory and immunosuppressive effects remain to be determined. The role of cannabinoids as therapeutic agents against inflammatory and autoimmune disorders, including selected epilepsy syndromes, deserves further study.
Anti-Inflammatory Mechanisms of Vagus Nerve Stimulation
(Peder S. Olofsson)
Accumulating evidence suggests that inflammation and immune system activation may play a role in epileptogenesis and lowering seizure threshold, and that anti-inflammatory treatments improve the disease outcomes.136–139 Discoveries in bioelectronic medicine, which is the convergence of neuroscience, engineering, computing, and clinical medicine, have revealed that nerve stimulation has the capacity to regulate inflammation, cytokine release, and other immune system functions.140–143 It is becoming increasingly clear that the immune system no longer can be regarded as fully autonomous, because it is regulated by neural reflex circuits.
The best characterized neural reflex that regulates cytokine release in inflammation is the so called “inflammatory reflex,” in which the vagus nerve plays a key role. Electrical vagus nerve stimulation has been used for treatment of drug-resistant epilepsy since the 1990s144 and for refractory depression for more than a decade.145 Vagus nerve stimulators have been implanted in tens of thousands of patients, and few significant adverse effects have been reported.146–148 The neurophysiologic and molecular mechanisms that underlie the therapeutic effect of vagus nerve stimulation in epilepsy and depression are, however, not well understood.
Electrical stimulation of the cervical vagus nerve reduces inflammation in a number of experimental inflammatory diseases, ranging from sepsis, over ischemia-reperfusion injury, to experimental arthritis.141,149 The bridging of neuroscience and immunology has revealed specific molecular targets of neural reflex signals in inflammation.150 Neurophysiologic and molecular mapping of the efferent arc of the inflammatory reflex has demonstrated that electrical activation of the left cervical vagus nerve triggers signals in the efferent vagus nerve that reach the celiac ganglion where the splenic nerve arises.151,152 The splenic nerve is required for relaying the signals to the spleen, where choline acetyl-transferase (ChAT)+ T cells, “CD4 TChAT,” which appear in close apposition of splenic nerve endings, release acetylcholine in response to norepinephrine.153 Acetylcholine subsequently activates α7 nicotinic acetylcholine receptors (α7nAChRs) on innate immune cells, including splenic macrophages, which significantly reduce release of proinflammatory cytokines such as TNF-α in experimental systemic inflammation.144,154 Vagus nerve stimulation also reduces infiltration of leukocytes in local inflammation,155 inhibits macrophage activation,156 and promotes a reparative phenotype of macrophages at sites of injury.157
Intriguingly, the discovery that CD4 TChAT relay neural signals afford a mechanism for providing cholinergic signals to tissues devoid of cholinergic innervation. As an example, most blood vessels are devoid of cholinergic innervation, but vascular endothelial cells express cholinergic receptors. Activation of endothelial cholinergic receptors promotes release of nitric oxide that reduces blood pressure by relaxing vascular smooth muscle cells.158 CD4 TChAT are found in murine blood, and mice deficient in CD4 TChAT show significantly increased blood pressure and their cardiovascular physiology is consistent with an increased systemic vascular resistance.159 These observations indicate that CD4 TChAT regulates blood pressure by providing a cholinergic signal to vascular endothelial cells, and it is conceivable that neurotransmitter-releasing immune cells may interact with nerves and organs to regulate physiology in additional ways not yet discovered.
The new insights on reflex control of inflammation and, in particular, on the inflammatory reflex, have spawned clinical trials for treatment of chronic human inflammatory diseases using an implantable vagus nerve stimulator developed originally for treatment of epilepsy.142 The first reports on its effects in rheumatoid arthritis and inflammatory bowel disease are encouraging, with improved clinical scores and attenuated release of pro-inflammatory cytokines.160,161 The study population sizes were, however, limited, and results from larger clinical trials that may improve our understanding of the usefulness of vagus nerve stimulation in treatment inflammatory diseases are eagerly awaited.
In light of the accumulating evidence that inflammation plays a role in epileptogenesis and seizure threshold reduction, and the anti-inflammatory effects of vagus nerve stimulation, it is tempting to speculate that one mechanism that underlies the therapeutic effect of vagus nerve stimulation in epilepsy involves attenuation of inflammation. In a recent study, Varvel et al. observed that infiltrating monocytes exacerbate neuronal damage and increase morbidity after status epilepticus.28 Spleen can deploy a significant portion of the monocytes recruited to sites of inflammation.158,162 Vagus nerve stimulation reduces cytokine release from innate immune cells in spleen and may shift macrophage phenotype from pro-inflammatory to reparative.143 It would be interesting to investigate whether vagus nerve stimulation can reduce epileptogenesis by reducing local neuroinflammation perhaps by local direct effects in the CNS and/or by reducing the pro-inflammatory activity of infiltrating immune cells.
Future directions
Despite the convincing physiologic effects of vagus nerve stimulation in experimental models of inflammatory diseases and the comprehensive understanding of some of the molecular pathways in neural regulation of inflammation, the mechanistic basis for the therapeutic effect of vagus nerve stimulation on epilepsy is currently unclear. Further studies of the neurophysiology and molecular mechanisms that underlie the neural control of immune cell activity both in the periphery and in the CNS are warranted to improve our understanding of the pathogenesis of neuroinflammation and epilepsy.
Conclusions and Future Directions
During the past years, it has been recognized that anti-inflammatory mechanisms play an important role in the antiseizure effect of established therapies for epilepsy, like the KD and vagus nerve stimulation. In addition, a variety of drugs, like CBDs, are under scrutiny for their use as antiseizure drugs, which work through a substantial anti-inflammatory mechanism and which also may have a potential antiepileptogenic effect. When testing these two issues, future trials need to adopt more sophisticated designs with enrollment of “enriched” patients. Treatment of therapy-resistant patients with CBDs or specific anti-inflammatory drugs already approved for other indications, like fingolimod or anakinra, needs to adhere to such a protocol. Prevention of epilepsy after stroke or traumatic brain injury may be another opportune clinical situation for conducting antiepileptogenesis trials. Patterns of adverse events of anti-inflammatory drugs should be closely monitored because they are likely to be different from those observed in classic AEDs. Fostering a network of experimental and clinical research collaborations from academia, industry, and funding institutions is germane for further progress in the translation of preclinical research results into clinical studies for the sake of patients at risk of developing the disease or with difficult-to-treat epilepsy.
Key Points.
Specific anti-inflammatory treatments have been effective at suppressing experimental chronic seizures and interfering with epileptogenesis
Some of these drugs have already been evaluated in early phase clinical trials in pharmacoresistant epilepsies
Nonpharmaceutical therapies for refractory epilepsy like the ketogenic diet or vagus nerve stimulation influence the immune system
Novel clinical trial design trials should contain an end-point to demonstrate disease modification
Acknowledgments
Additional contributors to the studies described in this manuscript are the following: Evan C. Rosenberg (NYU, Department of Neuroscience and Physiology, and New York, NY, USA); Emilio Perucca (National Neurological Institute IRCCS, C. Mondino Foundation, Pavia, Italy); Philippe Ryvlin (Département des Neurosciences Cliniques, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland); Torbjorn Tomson (Department of Neurology, Karolinska University Hospital, Stockholm, Sweden); Eugen Trinka (Universitätsklinikum der Paracelsus Medizinischen Privatuniversität, Salzburg, Austria); Laura Papetti (Neurology Unit, Dept Neuroscience, Bambino Gesù Children Hospital, Rome, Italy).
Biography
Dr. Jacqueline French is professor of neurology at NYU Langone Medical Center.
Footnotes
Disclosure
The authors declare that they have no conflicts of interest to disclose. We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines.
References
- 1.Vezzani A, Aronica E, Mazarati A, Pittman QJ. Epilepsy and brain inflammation. Exp Neurol. 2013;244:11–21. doi: 10.1016/j.expneurol.2011.09.033. [DOI] [PubMed] [Google Scholar]
- 2.Iyer A, Zurolo E, Spliet WG, et al. Evaluation of the innate and adaptive immunity in type I and type II focal cortical dysplasias. Epilepsia. 2010;51:1763–1773. doi: 10.1111/j.1528-1167.2010.02547.x. [DOI] [PubMed] [Google Scholar]
- 3.http://investors.vrtx.com/releasedetail.cfm?ReleaseID=555967
- 4.Maroso M, Balosso S, Ravizza T, et al. Interleukin-1β biosynthesis inhibition reduces acute seizures and drug resistant chronic epileptic activity in mice. Neurotherapeutics. 2011;8:304–315. doi: 10.1007/s13311-011-0039-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bhattaram VA, Siddiqui O, Kapcala LP, Gobburu JV. Endpoints and analyses to discern disease-modifying drug effects in early Parkinson’s disease. AAPS J. 2009;11:456–464. doi: 10.1208/s12248-009-9123-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Zhang RY, Leon AC, Chuang-Stein C, Romano SJ. A new proposal for randomized start design to investigate disease-modifying therapies for Alzheimer disease. Clin Trials. 2011;8:5–14. doi: 10.1177/1740774510392255. [DOI] [PubMed] [Google Scholar]
- 7.Schmidt D. Is antiepileptogenesis a realistic goal in clinical trials? Concerns and new horizons. Epileptic Disord. 2012;14:105–113. doi: 10.1684/epd.2012.0512. [DOI] [PubMed] [Google Scholar]
- 8.Gustavsson A, Svensson M, Jacobi F, et al. Cost of disorders of the brain in Europe 2010. Eur Neuropsychopharmacol. 2011;21:718–779. doi: 10.1016/j.euroneuro.2011.08.008. [DOI] [PubMed] [Google Scholar]
- 9.Cordonnier C, Hénon H, Derambure P, et al. Early epileptic seizures after stroke are associated with increased risk of new-onset dementia. J Neurol Neurosurg Psychiatry. 2007;78:514–516. doi: 10.1136/jnnp.2006.105080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Trinka E, Brigo F. Antiepileptogenesis in humans: disappointing clinical evidence and ways to move forward. Curr Opin Neurol. 2014;27:227–235. doi: 10.1097/WCO.0000000000000067. [DOI] [PubMed] [Google Scholar]
- 11.Dickens AM, Vainio S, Marjamäki P, et al. Detection of microglial activation in an acute model of neuroinflammation using PET and radiotracers 11C-(R)-PK11195 and 18F-GE-180. J Nucl Med. 2014;55:466–472. doi: 10.2967/jnumed.113.125625. [DOI] [PubMed] [Google Scholar]
- 12.Chollet F, Tardy J, Albucher JF, et al. Fluoxetine for motor recovery after acute ischaemic stroke (FLAME): a randomised placebo-controlled trial. Lancet Neurol. 2011;10:123–130. doi: 10.1016/S1474-4422(10)70314-8. [DOI] [PubMed] [Google Scholar]
- 13.Devinsky O, Vezzani A, Najjar S, et al. Glia and epilepsy: excitability and inflammation. Trends Neurosci. 2013;36:174–184. doi: 10.1016/j.tins.2012.11.008. [DOI] [PubMed] [Google Scholar]
- 14.Clausen F, Hånell A, Israelsson C, et al. Neutralization of interleukin-1β reduces cerebral edema and tissue loss and improves late cognitive outcome following traumatic brain injury in mice. Eur J Neurosci. 2011;34:110–123. doi: 10.1111/j.1460-9568.2011.07723.x. [DOI] [PubMed] [Google Scholar]
- 15.Gulyas B, Toth M, Vas A, et al. Visualising neuroinflammation in post-stroke patients: a comparative PET study with the TSPO molecular imaging biomarkers [11C]PK11195 and [11C]vinpocetine. Curr Radiopharm. 2012;5:19–28. doi: 10.2174/1874471011205010019. [DOI] [PubMed] [Google Scholar]
- 16.Fleischmann RM, Tesser J, Schiff MH, et al. Safety of extended treatment with anakinra in patients with rheumatoid arthritis. Ann Rheum Dis. 2006;65:1006–1012. doi: 10.1136/ard.2005.048371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Noe FM, Polascheck N, Frigerio F, et al. Pharmacological blockade of IL-1β/IL-1 receptor type 1 axis during epileptogenesis provides neuroprotection in two rat models of temporal lobe epilepsy. Neurobiol Dis. 2013;59:183–193. doi: 10.1016/j.nbd.2013.07.015. [DOI] [PubMed] [Google Scholar]
- 18.Clark SR, McMahon CJ, Gueorguieva I, et al. Interleukin-1 receptor antagonist penetrates human brain at experimentally therapeutic concentrations. J Cereb Blood Flow Metab. 2008;28:387–394. doi: 10.1038/sj.jcbfm.9600537. [DOI] [PubMed] [Google Scholar]
- 19.Hesdorffer DC, Benn EK, Cascino GD, Hauser WA. Is a first acute symptomatic seizure epilepsy? Mortality and risk for recurrent seizure. Epilepsia. 2009;50:1102–1108. doi: 10.1111/j.1528-1167.2008.01945.x. [DOI] [PubMed] [Google Scholar]
- 20.Pradillo JM, Denes A, Greenhalgh AD, et al. Delayed administration of interleukin-1 receptor antagonist reduces ischemic brain damage and inflammation in comorbid rats. J Cereb Blood Flow Metab. 2012;32:1810–1819. doi: 10.1038/jcbfm.2012.101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Banwell V, Sena ES, Macleod MR. Systematic review and stratified meta-analysis of the efficacy of interleukin-1 receptor antagonist in animal models of stroke. J Stroke Cerebrovasc Dis. 2009;18:269–276. doi: 10.1016/j.jstrokecerebrovasdis.2008.11.009. [DOI] [PubMed] [Google Scholar]
- 22.Brinkmann V. FTY720: mechanism of action and potential benefit in organ transplantation. Yonsei Med J. 2004;45:991–997. doi: 10.3349/ymj.2004.45.6.991. [DOI] [PubMed] [Google Scholar]
- 23.Kappos L, Radue EW, O’Connor P, et al. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N Engl J Med. 2010;362:387–401. doi: 10.1056/NEJMoa0909494. [DOI] [PubMed] [Google Scholar]
- 24.Chun J, Hartung HP. Mechanism of action of oral fingolimod (FTY720) in multiple sclerosis. Clin Neuropharmacol. 2010;33:91–101. doi: 10.1097/WNF.0b013e3181cbf825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011;7:31–40. doi: 10.1038/nrneurol.2010.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Lassmann H, van Horssen J, Mahad D. Progressive multiple sclerosis: pathology and pathogenesis. Nat Rev Neurol. 2012;8:647–656. doi: 10.1038/nrneurol.2012.168. [DOI] [PubMed] [Google Scholar]
- 27.Ravizza T, Gagliardi B, Noé F, et al. Innate and adaptive immunity during epileptogenesis and spontaneous seizures: evidence from experimental models and human temporal lobe epilepsy. Neurobiol Dis. 2008;29:142–160. doi: 10.1016/j.nbd.2007.08.012. [DOI] [PubMed] [Google Scholar]
- 28.Varvel NH, et al. Infiltrating monocytes promote brain inflammation and exacerbate neuronal damage after status epilepticus. Proc Natl Acad Sci USA. 2016;113:E5665–E5674. doi: 10.1073/pnas.1604263113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nguyen MD, Julien JP, Rivest S. Innate immunity: the missing link in neuroprotection and neurodegeneration? Nat Rev Neurosci. 2002;3:216–227. doi: 10.1038/nrn752. [DOI] [PubMed] [Google Scholar]
- 30.Zattoni M, et al. Brain infiltration of leukocytes contributes to the pathophysiology of temporal lobe epilepsy. J Neurosci. 2011;31:4037–4050. doi: 10.1523/JNEUROSCI.6210-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Bauer J, Vezzani A, Bien CG. Epileptic encephalitis: the role of the innate and adaptive immune system. Brain Pathol. 2012;22:412–421. doi: 10.1111/j.1750-3639.2012.00580.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Brinkmann V. Sphingosine 1-phosphate receptors in health and disease: mechanistic insights from gene deletion studies and reverse pharmacology. Pharmacol Ther. 2007;115:84–105. doi: 10.1016/j.pharmthera.2007.04.006. [DOI] [PubMed] [Google Scholar]
- 33.Massberg S, von Andrian UH. Fingolimod and sphingosine-1-phosphate-modifiers of lymphocyte migration. N Engl J Med. 2006;355:1088–1091. doi: 10.1056/NEJMp068159. [DOI] [PubMed] [Google Scholar]
- 34.Foster CA, Howard LM, Schweitzer A, et al. Brain penetration of the oral immunomodulatory drug FTY720 and its phosphorylation in the central nervous system during experimental autoimmune encephalomyelitis: consequences for mode of action in multiple sclerosis. J Pharmacol Exp Ther. 2007;323:469–475. doi: 10.1124/jpet.107.127183. [DOI] [PubMed] [Google Scholar]
- 35.Mizugishi K, Yamashita T, Oliveira A, et al. Essential role for sphingosine kinases in neural and vascular development. Mol Cell Biol. 2005;25:11113–11121. doi: 10.1128/MCB.25.24.11113-11121.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.De Keyser J, Mostert JP, Koch MW. Dysfunctional astrocytes as key players in the pathogenesis of central nervous system disorders. J Neurol Sci. 2008;267:3–16. doi: 10.1016/j.jns.2007.08.044. [DOI] [PubMed] [Google Scholar]
- 37.Osinde M, Mullershausen F, Dev KK. Phosphorylated FTY720 stimulates ERK phosphorylation in astrocytes via S1P receptors. Neuropharmacology. 2007;52:1210–1218. doi: 10.1016/j.neuropharm.2006.11.010. [DOI] [PubMed] [Google Scholar]
- 38.Spampinato SF, Obermeier B, Cotleur A, et al. Sphingosine 1 phosphate at the blood brain barrier: can the modulation of S1P receptor 1 influence the response of endothelial cells and astrocytes to inflammatory stimuli? PLoS ONE. 2015;10:e0133392. doi: 10.1371/journal.pone.0133392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hunter SF, Bowen JD, Reder AT. The direct effects of fingolimod in the central nervous system: Implications for relapsing multiple sclerosis. CNS Drugs. 2016;30:135–147. doi: 10.1007/s40263-015-0297-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hait NC, Wise LE, Allegood JC, et al. Active, phosphorylated fingolimod inhibits histone deacetylases and facilitates fear extinction memory. Nat Neurosci. 2014;17:971–980. doi: 10.1038/nn.3728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Brunkhorst R, Vutukuri R, Pfeilschifter W. Fingolimod for the treatment of neurological diseases-state of play and future perspectives. Front Cell Neurosci. 2014;8:e283. doi: 10.3389/fncel.2014.00283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Deogracias R, Yazdani M, Dekkers MP, et al. Fingolimod, a sphingosine-1 phosphate receptor modulator, increases BDNF levels and improves symptoms of a mouse model of Rett syndrome. Proc Natl Acad Sci USA. 2012;109:14230–14235. doi: 10.1073/pnas.1206093109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chen NC, Chuang YC, Huang CW, et al. Interictal serum brain-derived neurotrophic factor level reflects white matter integrity, epilepsy severity, and cognitive dysfunction in chronic temporal lobe epilepsy. Epilepsy Behav. 2016;59:147–154. doi: 10.1016/j.yebeh.2016.02.029. [DOI] [PubMed] [Google Scholar]
- 44.Eftekhari S, Mehrabi S, Karimzadeh F, et al. Brain derived neurotrophic factor modification of epileptiform burst discharges in a temporal lobe epilepsy model. Basic Clin Neurosci. 2016;7:115–120. doi: 10.15412/J.BCN.03070205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Gao F, Liu Y, Li X, et al. Fingolimod (FTY720) inhibits neuroinflammation and attenuates spontaneous convulsions in lithium-pilocarpine induced status epilepticus in rat model. Pharmacol Biochem Behav. 2012;103:187–196. doi: 10.1016/j.pbb.2012.08.025. [DOI] [PubMed] [Google Scholar]
- 46.Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96:70–82. doi: 10.1016/j.neuropharm.2014.10.027. [DOI] [PubMed] [Google Scholar]
- 47.Toeldano M, Pittock SJ. Autoimmune epilepsy. Semin Neurol. 2015;35:245–258. doi: 10.1055/s-0035-1552625. [DOI] [PubMed] [Google Scholar]
- 48.Kramer U, Chi CS, Lin KL, et al. Febrile infection-related epilepsy syndrome (FIRES): pathogenesis, treatment, and outcome. A multicenter study on 77 children. Epilepsia. 2011;52:1956–1965. doi: 10.1111/j.1528-1167.2011.03250.x. [DOI] [PubMed] [Google Scholar]
- 49.Von Rhein B, Wagner J, Widman G, et al. Suspected antibody-negative autoimmune limbic encephalitis: outcome of immunotherapy. Acta Neurol Scand. 2017;135:134–141. doi: 10.1111/ane.12575. [DOI] [PubMed] [Google Scholar]
- 50.Vigevano F, Arzimanoglou A, Plouin P, Specchio N. Therapeutic approach to epileptic encephalopathies. Epilepsia. 2013;54(Suppl 8):S45–S50. doi: 10.1111/epi.12423. [DOI] [PubMed] [Google Scholar]
- 51.Helbig I, Tayoun AA. Understanding genotypes and phenotypes in epileptic encephalopathies. Mol Synromol. 2016;7:172–181. doi: 10.1159/000448530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Avanzini G, Depaulis A, Tassinari A, de Curtis M. Do seizures and epileptic activity worsen epilepsy and deteriorate cognitive function? Epilepsia. 2013;54(Suppl 8):S14–S21. doi: 10.1111/epi.12418. [DOI] [PubMed] [Google Scholar]
- 53.Iyer A, Appleton R. Improving outcomes in infantile spasms: role of pharmacotherapy. Paediatr Drugs. 2016;18:357–366. doi: 10.1007/s40272-016-0188-y. [DOI] [PubMed] [Google Scholar]
- 54.Brunson KL, Avishai-Eliner S, Baram TZ. ACTH treatment of infantile spasms: mechanisms of its effects in modulation of neuronal excitability. Int Rev Neurobiol. 2002;49:185–197. doi: 10.1016/s0074-7742(02)49013-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Melvin JJ, Huntley Hardison H. Immunomodulatory treatments of epilepsy. Semin Pediatr Neurol. 2014;21:232–237. doi: 10.1016/j.spen.2014.08.001. [DOI] [PubMed] [Google Scholar]
- 56.Wilmshurst JM, Gaillard WD, Vinayan KP, et al. Summary of recommendations for the management of infantile seizures: Task Force Report for the ILAE Commission of Pediatrics. Epilepsia. 2015;56:1185–1197. doi: 10.1111/epi.13057. [DOI] [PubMed] [Google Scholar]
- 57.Shumiloff NA, Lam WM, Manasco KB. Adrenocorticotropic hormone for the treatment of West Syndrome in children. Ann Pharmacother. 2013;47:744–754. doi: 10.1345/aph.1R535. [DOI] [PubMed] [Google Scholar]
- 58.Dupuis N, Auvin S. Anti-inflammatory effects of a ketogenic diet: Implications for new indications. In: Masino SA, editor. Ketogenic diet and metabolic therapies: Expanded roles in health and disease. New York: Oxford University Press; 2016. pp. 147–155. [Google Scholar]
- 59.Dupuis N, Curatolo N, Benoist J-F, Auvin S. Ketogenic diet exhibits anti-inflammatory properties. Epilepsia. 2015;56:e95–e98. doi: 10.1111/epi.13038. [DOI] [PubMed] [Google Scholar]
- 60.Taha AY, Burnham WM, Auvin S. Polyunsaturated fatty acids and epilepsy. Epilepsia. 2010;51:1348–1358. doi: 10.1111/j.1528-1167.2010.02654.x. [DOI] [PubMed] [Google Scholar]
- 61.Hong S, Gronert K, Devchand PR, et al. Novel docosatrienes and 17s-resolvins generated from docosahexaenoic acid in murine brain, human blood, and glial cells - autacoids in anti-inflammation. J Biol Chem. 2003;278:14677–14687. doi: 10.1074/jbc.M300218200. [DOI] [PubMed] [Google Scholar]
- 62.Blanquart C, Barbier O, Fruchart JC, Tschopp J. Peroxisome proliferator-activated receptors: Regulation of transcriptional activities and roles in inflammation. J Steroid Biochem Mol Biol. 2003;85:267–273. doi: 10.1016/s0960-0760(03)00214-0. [DOI] [PubMed] [Google Scholar]
- 63.Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in nlrp3 inflammasome activation. Nature. 2011;469:221–225. doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
- 64.Evangeliou A, Vlachonikolis I, Mihailidou H, et al. Application of a ketogenic diet in children with autistic behavior: Pilot study. J Child Neurol. 2003;18:113–118. doi: 10.1177/08830738030180020501. [DOI] [PubMed] [Google Scholar]
- 65.Ruskin DN, Svedova J, Cote JL, et al. Ketogenic diet improves core symptoms of autism in btbr mice. PLoS ONE. 2013;8:e65021. doi: 10.1371/journal.pone.0065021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Newell C, Bomhof MR, Reimer RA, et al. Ketogenic diet modifies the gut microbiota in a murine model of autism spectrum disorder. Mol Autism. 2016;7:37. doi: 10.1186/s13229-016-0099-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ruskin DN, Kawamura M, Masino SA. Reduced pain and inflammation in juvenile and adult rats fed a ketogenic diet. PLoS ONE. 2009;4:e8349. doi: 10.1371/journal.pone.0008349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yang XX, Cheng BH. Neuroprotective and anti-inflammatory activities of ketogenic diet on mptp-induced neurotoxicity. J Mol Neurosci. 2010;42:145–153. doi: 10.1007/s12031-010-9336-y. [DOI] [PubMed] [Google Scholar]
- 69.Kim DY, Hao JW, Liu RL, et al. Inflammation-mediated memory dysfunction and effects of a ketogenic diet in a murine model of multiple sclerosis. PLoS ONE. 2012;7:e35476. doi: 10.1371/journal.pone.0035476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Asrih M, Altirriba J, Rohner-Jeanrenaud F, Jornayvaz FR. Ketogenic diet impairs fgf21 signaling and promotes differential inflammatory responses in the liver and white adipose tissue. PLoS ONE. 2015;10:e0126364. doi: 10.1371/journal.pone.0126364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.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]
- 72.Sugiura T, Kondo S, Sukagawa A, et al. 2-Arachidonoylglycerol: a possible endogenous cannabinoid receptor ligand in brain. Biochem Biophys Res Commun. 1995;215:89–97. doi: 10.1006/bbrc.1995.2437. [DOI] [PubMed] [Google Scholar]
- 73.Nomura DK, Morrison BE, Blankman JL, et al. Endocannabinoid hydrolysis generates brain prostaglandins that promote neuroinflammation. Science. 2011;334:809–813. doi: 10.1126/science.1209200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Di Marzo V, Melck D, Bisogno T, De Petrocellis L. Endocannabinoids: endogenous cannabinoid receptor ligands with neuromodulatory action. Trends Neurosci. 1998;21:521–528. doi: 10.1016/s0166-2236(98)01283-1. Erratum in: Trends Neurosci 1999:80. [DOI] [PubMed] [Google Scholar]
- 75.Sugiura T, Kobayashi Y, Oka S, et al. Biosynthesis and degradation of anandamide and 2-arachidonoylglycerol and their possible physiological significance. Prostaglandins Leukot Essent Fatty Acids. 2002;66:173–192. doi: 10.1054/plef.2001.0356. [DOI] [PubMed] [Google Scholar]
- 76.Marrs WR, Blankman JL, Horne EA, et al. The serine hydrolase ABHD6 controls the accumulation and efficacy of 2-AG at cannabinoid receptors. Nat Neurosci. 2010;13:951–957. doi: 10.1038/nn.2601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Stella N, Schweitzer P, Piomelli D. A second endogenous cannabinoid that modulates long-term potentiation. Nature. 1997;388:773–778. doi: 10.1038/42015. [DOI] [PubMed] [Google Scholar]
- 78.Crowe MS, Leishman E, Banks ML, et al. Combined inhibition of monoacylglycerol lipase and cyclooxygenases synergistically reduces neuropathic pain in mice. Br J Pharmacol. 2015;172:1700–1712. doi: 10.1111/bph.13012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Grim TW, Ghosh S, Hsu KL, et al. Combined inhibition of FAAH and COX produces enhanced anti-allodynic effects in mouse neuropathic and inflammatory pain models. Pharmacol Biochem Behav. 2014;124:405–411. doi: 10.1016/j.pbb.2014.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Benito C, Tolón RM, Castillo AI, et al. beta-Amyloid exacerbates inflammation in astrocytes lacking fatty acid amide hydrolase through a mechanism involving PPAR-alpha, PPAR-gamma and TRPV1, but not CB(1) or CB(2) receptors. Br J Pharmacol. 2012;166:1474–1489. doi: 10.1111/j.1476-5381.2012.01889.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bernal-Chico A, Canedo M, Manterola A, et al. Blockade of monoacylglycerol lipase inhibits oligodendrocyte excitotoxicity and prevents demyelination in vivo. Glia. 2015;63:163–176. doi: 10.1002/glia.22742. [DOI] [PubMed] [Google Scholar]
- 82.Du H, Chen X, Zhang J, Chen C. Inhibition of COX-2 expression by endocannabinoid 2-arachidonoylglycerol is mediated via PPAR-gamma. Br J Pharmacol. 2011;163:1533–1549. doi: 10.1111/j.1476-5381.2011.01444.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Li Y, Kim J. Neuronal expression of CB2 cannabinoid receptor mRNAs in the mouse hippocampus. Neuroscience. 2015;311:253–267. doi: 10.1016/j.neuroscience.2015.10.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kim J, Li Y. Chronic activation of CB2 cannabinoid receptors in the hippocampus increases excitatory synaptic transmission. J Physiol. 2015;593:871–886. doi: 10.1113/jphysiol.2014.286633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Atwood BK, Mackie K. CB2: a cannabinoid receptor with an identity crisis. BrJ Pharmacol. 2010;160:467–479. doi: 10.1111/j.1476-5381.2010.00729.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Laurén HB, Lopez-Picon FR, Brandt AM, et al. Transcriptome analysis of the hippocampal CA1 pyramidal cell region after kainic acid-induced status epilepticus in juvenile rats. PLoS ONE. 2010;5:e10733. doi: 10.1371/journal.pone.0010733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hu H, Ho W, Mackie K, et al. Brain CB(1) receptor expression following lipopolysaccharide-induced inflammation. Neuroscience. 2012;227:211–222. doi: 10.1016/j.neuroscience.2012.09.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zarruk JG, Fernández-López D, García-Yébenes I, et al. Cannabinoid type 2 receptor activation downregulates stroke-induced classic and alternative brain macrophage/microglial activation concomitant to neuroprotection. Stroke. 2012;43:211–219. doi: 10.1161/STROKEAHA.111.631044. [DOI] [PubMed] [Google Scholar]
- 89.Tang J, Tao Y, Tan L, et al. Cannabinoid receptor 2 attenuates microglial accumulation and brain injury following germinal matrix hemorrhage via ERK dephosphorylation in vivo and in vitro. Neuropharmacology. 2015;95:424–433. doi: 10.1016/j.neuropharm.2015.04.028. [DOI] [PubMed] [Google Scholar]
- 90.Amenta PS, Jallo JI, Tuma RF, Elliott MB. A cannabinoid type 2 receptor agonist attenuates blood-brain barrier damage and neurodegeneration in a murine model of traumatic brain injury. J Neurosci Res. 2012;90:2293–2305. doi: 10.1002/jnr.23114. [DOI] [PubMed] [Google Scholar]
- 91.Gutierrez T, Farthing JN, Zvonok AM, et al. Activation of peripheral cannabinoid CB1 and CB2 receptors suppresses the maintenance of inflammatory nociception: a comparative analysis. Br J Pharmacol. 2007;150:153–163. doi: 10.1038/sj.bjp.0706984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kinsey SG, Mahadevan A, Zhao B, et al. The CB2 cannabinoid receptor-selective agonist O-3223 reduces pain and inflammation without apparent cannabinoid behavioral effects. Neuropharmacology. 2011;60:244–251. doi: 10.1016/j.neuropharm.2010.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Xie J, Xiao D, Xu Y, et al. Up-regulation of immunomodulatory effects of mouse bone-marrow derived mesenchymal stem cells by tetrahydrocannabinol pre-treatment involving cannabinoid receptor CB2. Oncotarget. 2016;7:6436–6447. doi: 10.18632/oncotarget.7042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kozela E, Pietr M, Juknat A, et al. Cannabinoids Delta(9)-tetrahydrocannabinol and cannabidiol differentially inhibit the lipopolysaccharide-activated NF-kappaB and interferon-beta/STAT proinflammatory pathways in BV-2 microglial cells. J Biol Chem. 2010;285:1616–1626. doi: 10.1074/jbc.M109.069294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Moretti S, Franchi S, Castelli M, et al. Exposure of adolescent mice to Delta-9-tetrahydrocannabinol induces long-lasting modulation of pro- and anti-Inflammatory cytokines in hypothalamus and hippocampus similar to that observed for peripheral macrophages. J Neuroimmune Pharmacol. 2015;10:371–379. doi: 10.1007/s11481-015-9592-2. [DOI] [PubMed] [Google Scholar]
- 96.Puffenbarger RA, Boothe AC, Cabral GA. Cannabinoids inhibit LPS-inducible cytokine mRNA expression in rat microglial cells. Glia. 2000;29:58–69. [PubMed] [Google Scholar]
- 97.Ehrhart J, Obregon D, Mori T, et al. Stimulation of cannabinoid receptor 2 (CB2) suppresses microglial activation. J Neuroinflammation. 2005;2:29. doi: 10.1186/1742-2094-2-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Coffey RG, Snella E, Johnson K, Pross S. Inhibition of macrophage nitric oxide production by tetrahydrocannabinol in vivo and in vitro. Int J Immunopharmacol. 1996;18:749–752. doi: 10.1016/s0192-0561(97)85557-9. [DOI] [PubMed] [Google Scholar]
- 99.El-Remessy AB, Khalil IE, Matragoon S, et al. Neuroprotective effect of (−)Delta9-tetrahydrocannabinol and cannabidiol in N-methyl-D-aspartate-induced retinal neurotoxicity: involvement of peroxynitrite. Am J Pathol. 2003;163:1997–2008. doi: 10.1016/s0002-9440(10)63558-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Hampson AJ, Grimaldi M, Axelrod J, Wink D. Cannabidiol and (−) Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci USA. 1998;95:8268–8273. doi: 10.1073/pnas.95.14.8268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Kozela E, Juknat A, Kaushansky N, et al. Cannabinoids decrease the th17 inflammatory autoimmune phenotype. J Neuroimmune Pharmacol. 2013;8:1265–1276. doi: 10.1007/s11481-013-9493-1. [DOI] [PubMed] [Google Scholar]
- 102.De Petrocellis L, Vellani V, Schiano-Moriello A, et al. Plant-derived cannabinoids modulate the activity of transient receptor potential channels of ankyrin type-1 and melastatin type-8. J Pharmacol Exp Ther. 2008;325:1007–1015. doi: 10.1124/jpet.107.134809. [DOI] [PubMed] [Google Scholar]
- 103.De Petrocellis L, Ligresti A, Moriello AS, et al. Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol. 2011;163:1479–1494. doi: 10.1111/j.1476-5381.2010.01166.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Qin N, Neeper MP, Liu Y, et al. TRPV2 is activated by cannabidiol and mediates CGRP release in cultured rat dorsal root ganglion neurons. J Neurosci. 2008;28:6231–6238. doi: 10.1523/JNEUROSCI.0504-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Fishbein-Kaminietsky M, Gafni M, Sarne Y. Ultralow doses of cannabinoid drugs protect the mouse brain from inflammation-induced cognitive damage. J Neurosci Res. 2014;92:1669–1677. doi: 10.1002/jnr.23452. [DOI] [PubMed] [Google Scholar]
- 106.Chen R, Zhang J, Fan N, et al. Delta9-THC-caused synaptic and memory impairments are mediated through COX-2 signaling. Cell. 2013;155:1154–1165. doi: 10.1016/j.cell.2013.10.042. Erratum in: Cell. 2014; 156:618. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Hayakawa K, Mishima K, Nozako M, et al. Delayed treatment with cannabidiol has a cerebroprotective action via a cannabinoid receptor-independent myeloperoxidase-inhibiting mechanism. J Neuro chem. 2007;102:1488–1496. doi: 10.1111/j.1471-4159.2007.04565.x. [DOI] [PubMed] [Google Scholar]
- 108.Zani A, Braida D, Capurro V, Sala M. Delta9-tetrahydrocannabinol (THC) and AM 404 protect against cerebral ischaemia in gerbils through a mechanism involving cannabinoid and opioid receptors. Br J Pharmacol. 2007;152:1301–1311. doi: 10.1038/sj.bjp.0707514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bolognini D, Costa B, Maione S, et al. The plant cannabinoid Delta9-tetrahydrocannabivarin can decrease signs of inflammation and inflammatory pain in mice. Br J Pharmacol. 2010;160:677–687. doi: 10.1111/j.1476-5381.2010.00756.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bisogno T, Hanus L, De Petrocellis L, et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol. 2001;134:845–852. doi: 10.1038/sj.bjp.0704327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Jones NA, Hill AJ, Smith I, et al. Cannabidiol displays antiepileptiform and antiseizure properties in vitro and in vivo. J Pharmacol Exp Ther. 2010;332:569–577. doi: 10.1124/jpet.109.159145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Costa B, Giagnoni G, Franke C, et al. Vanilloid TRPV1 receptor mediates the antihyperalgesic effect of the nonpsychoactive cannabinoid, cannabidiol, in a rat model of acute inflammation. Br J Pharmacol. 2004;143:247–250. doi: 10.1038/sj.bjp.0705920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Pazos MR, Mohammed N, Lafuente H, et al. Mechanisms of cannabidiol neuroprotection in hypoxic-ischemic newborn pigs: role of 5HT(1A) and CB2 receptors. Neuropharmacology. 2013;71:282–291. doi: 10.1016/j.neuropharm.2013.03.027. [DOI] [PubMed] [Google Scholar]
- 114.Ahrens J, Demir R, Leuwer M, et al. The nonpsychotropic cannabinoid cannabidiol modulates and directly activates alpha-1 and alpha-1-beta glycine receptor function. Pharmacology. 2009;83:217–222. doi: 10.1159/000201556. [DOI] [PubMed] [Google Scholar]
- 115.Ross HR, Napier I, Connor M. Inhibition of recombinant human T-type calcium channels by Delta9-tetrahydrocannabinol and cannabidiol. J Biol Chem. 2008;283:16124–16134. doi: 10.1074/jbc.M707104200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ross RA. The enigmatic pharmacology of GPR55. Trends Pharmacol Sci. 2009;30:156–163. doi: 10.1016/j.tips.2008.12.004. [DOI] [PubMed] [Google Scholar]
- 117.Ryan D, Drysdale AJ, Lafourcade C, et al. Cannabidiol targets mitochondria to regulate intracellular Ca2+ levels. J Neurosci. 2009;29:2053–2063. doi: 10.1523/JNEUROSCI.4212-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Rimmerman N, Juknat A, Kozela E, et al. The non-psychoactive plant cannabinoid, cannabidiol affects cholesterol metabolism-related genes in microglial cells. Cell Mol Neurobiol. 2011;31:921–930. doi: 10.1007/s10571-011-9692-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.O’Sullivan SE, Kendall DA. Cannabinoid activation of peroxisome proliferator-activated receptors: potential for modulation of inflammatory disease. Immunobiology. 2010;215:611–616. doi: 10.1016/j.imbio.2009.09.007. [DOI] [PubMed] [Google Scholar]
- 120.Carrier EJ, Auchampach JA, Hillard CJ. Inhibition of an equilibrative nucleoside transporter by cannabidiol: a mechanism of cannabinoid immunosuppression. Proc Natl Acad Sci USA. 2006;103:7895–7900. doi: 10.1073/pnas.0511232103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Liou GI, Auchampach JA, Hillard CJ, et al. Mediation of cannabidiol anti-inflammation in the retina by equilibrative nucleoside transporter and A2A adenosine receptor. Invest Ophthalmol Vis Sci. 2008;49:5526–5531. doi: 10.1167/iovs.08-2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Juknat A, Pietr M, Kozela E, et al. Differential transcriptional profiles mediated by exposure to the cannabinoids cannabidiol and Delta9-tetrahydrocannabinol in BV-2 microglial cells. Br J Pharmacol. 2012;165:2512–2528. doi: 10.1111/j.1476-5381.2011.01461.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Mecha M, Torrao AS, Mestre L, et al. Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death Dis. 2012;3:e331. doi: 10.1038/cddis.2012.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Kozela E, Juknat A, Gao F, et al. Pathways and gene networks mediating the regulatory effects of cannabidiol, a nonpsychoactive cannabinoid, in autoimmune T cells. J Neuroinflammation. 2016;13:136. doi: 10.1186/s12974-016-0603-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Pazos MR, Cinquina V, Gomez A, et al. Cannabidiol administration after hypoxia-ischemia to newborn rats reduces long-term brain injury and restores neurobehavioral function. Neuropharmacology. 2012;63:776–783. doi: 10.1016/j.neuropharm.2012.05.034. [DOI] [PubMed] [Google Scholar]
- 126.Castillo A, Tolon MR, Fernandez-Ruiz J, et al. The neuroprotective effect of cannabidiol in an in vitro model of newborn hypoxic-ischemic brain damage in mice is mediated by CB(2) and adenosine receptors. Neurobiol Dis. 2010;37:434–440. doi: 10.1016/j.nbd.2009.10.023. [DOI] [PubMed] [Google Scholar]
- 127.De Filippis D, Esposito G, Cirillo C, et al. Cannabidiol reduces intestinal inflammation through the control of neuroimmune axis. PLoS ONE. 2011;6:e28159. doi: 10.1371/journal.pone.0028159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Mecha M, Feliú A, Iñigo PM, et al. Cannabidiol provides long-lasting protection against the deleterious effects of inflammation in a viral model of multiple sclerosis: a role for A2A receptors. Neurobiol Dis. 2013;59:141–150. doi: 10.1016/j.nbd.2013.06.016. [DOI] [PubMed] [Google Scholar]
- 129.Janefjord E, Maag JL, Harvey BS, Smid SD. Cannabinoid effects on beta amyloid fibril and aggregate formation, neuronal and microglial-activated neurotoxicity in vitro. Cell Mol Neurobiol. 2014;34:31–42. doi: 10.1007/s10571-013-9984-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Di Maio R, Cannon JR, Greenamyre JT. Post-status epilepticus treatment with the cannabinoid agonist WIN 55,212-2 prevents chronic epileptic hippocampal damage in rats. Neurobiol Dis. 2015;73:356–365. doi: 10.1016/j.nbd.2014.10.018. [DOI] [PubMed] [Google Scholar]
- 131.Hosseinzadeh M, Nikseresht S, Khodagholi F, et al. Cannabidiol post-treatment alleviates rat epileptic-related behaviors and activates hippocampal cell autophagy pathway along with antioxidant defense in chronic phase of pilocarpine-induced seizure. J Mol Neurosci. 2016;58:432–440. doi: 10.1007/s12031-015-0703-6. [DOI] [PubMed] [Google Scholar]
- 132.Mao K, You C, Lei D, Zhang H. High dosage of cannabidiol (CBD) alleviates pentylenetetrazole-induced epilepsy in rats by exerting an anticonvulsive effect. Int J Clin Exp Med. 2015;8:8820–8827. [PMC free article] [PubMed] [Google Scholar]
- 133.Monory K, Massa F, Egertová M, et al. The endocannabinoid system controls key epileptogenic circuits in the hippocampus. Neuron. 2006;51:455–466. doi: 10.1016/j.neuron.2006.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Karanian DA, Karim SL, Wood JT, et al. Endocannabinoid enhancement protects against kainic acid-induced seizures and associated braindamage. J Pharmacol Exp Ther. 2007;322:1059–1066. doi: 10.1124/jpet.107.120147. [DOI] [PubMed] [Google Scholar]
- 135.Karanian DA, Brown QB, Makriyannis A, et al. Dual modulation of endocannabinoid transport and fatty acid amide hydrolase protects against excitotoxicity. J Neurosci. 2005;25:7813–7820. doi: 10.1523/JNEUROSCI.2347-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Alyu F, Dikmen M. Inflammatory aspects of epileptogenesis: contribution of molecular inflammatory mechanisms. Acta Neuropsychiatr. 2017;29:1–16. doi: 10.1017/neu.2016.47. [DOI] [PubMed] [Google Scholar]
- 137.Iori V, Iyer AM, Ravizza T, et al. Blockade of the IL-1R1/TLR4path-way mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis. 2016;99:12–23. doi: 10.1016/j.nbd.2016.12.007. [DOI] [PubMed] [Google Scholar]
- 138.Kenney-Jung DL, Vezzani A, Kahoud RJ, et al. Febrile infection-related epilepsy syndrome treated with anakinra. Ann Neurol. 2016;80:939–945. doi: 10.1002/ana.24806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.van Baalen A, Vezzani A, Hausler M, Kluger G. Febrile infection-related epilepsy syndrome: clinical review and hypotheses of epileptogenesis. Neuropediatrics. 2016;48:5–18. doi: 10.1055/s-0036-1597271. [DOI] [PubMed] [Google Scholar]
- 140.Hanes WM, Olofsson PS, Talbot S, et al. Neuronal circuits modulate antigen flow through lymph nodes. Bioelectron Med. doi: 10.15424/bioelectronmed.2016.00001. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Olofsson PS, Rosas-Ballina M, Levine YA, Tracey KJ. Rethinking inflammation: neural circuits in the regulation of immunity. Immunol Rev. 2012;248:188–204. doi: 10.1111/j.1600-065X.2012.01138.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Steinberg BE, Sundman E, Terrando N, et al. Neural control of inflammation: implications for perioperative and critical care. Anesthesiology. 2016;124:1174–1189. doi: 10.1097/ALN.0000000000001083. [DOI] [PubMed] [Google Scholar]
- 143.Tracey KJ. Shock medicine. Sci Am. 2015;312:28–35. [Google Scholar]
- 144.Penry JK, Dean JC. Prevention of intractable partial seizures by intermittent vagal stimulation in humans: preliminary results. Epilepsia. 1990;31(Suppl 2):S40–S43. doi: 10.1111/j.1528-1157.1990.tb05848.x. [DOI] [PubMed] [Google Scholar]
- 145.Rush AJ, Marangell LB, Sackeim HA, et al. Vagus nerve stimulation for treatment-resistant depression: a randomized, controlled acute phase trial. Biol Psychiatry. 2005;58:347–354. 13. doi: 10.1016/j.biopsych.2005.05.025. [DOI] [PubMed] [Google Scholar]
- 146.Ben-Menachem E. Vagus nerve stimulation, side effects, and longterm safety. J Clin Neurophysiol. 2001;18:415–418. doi: 10.1097/00004691-200109000-00005. [DOI] [PubMed] [Google Scholar]
- 147.Bonaz B, Sinniger V, Pellissier S. Vagus nerve stimulation: a new promising therapeutic tool in inflammatory bowel disease. J Intern Med. 2017;127:2118–2132. doi: 10.1111/joim.12611. [DOI] [PubMed] [Google Scholar]
- 148.Englot DJ, Hassnain KH, Rolston JD, et al. Quality-of-life metrics with vagus nerve stimulation for epilepsy from provider survey data. Epilepsy Behav. 2016;66:4–9. doi: 10.1016/j.yebeh.2016.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Andersson U, Tracey KJ. Reflex principles of immunological homeostasis. Annu Rev Immunol. 2012;30:313–335. doi: 10.1146/annurev-immunol-020711-075015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Inoue T, Abe C, Sung SS, et al. Vagus nerve stimulation mediates protection from kidney ischemia-reperfusion injury through alpha7-nAChR+ splenocytes. J Clin Invest. 2016;126:1939–1952. doi: 10.1172/JCI83658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Borovikova LV, Ivanova S, Zhang M, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–462. doi: 10.1038/35013070. [DOI] [PubMed] [Google Scholar]
- 152.Olofsson PS, Levine YA, Caravaca A, et al. Single-pulse and unidirectional electrical activation of the cervical vagus nerve reduces TNF in endotoxemia. Bioelectron Med. 2015;2:37–42. [Google Scholar]
- 153.Rosas-Ballina M, Olofsson PS, Ochani M, et al. Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science. 2011;334:98–101. doi: 10.1126/science.1209985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Olofsson PS, Katz DA, Rosas-Ballina M, et al. alpha7 nicotinic acetylcholine receptor (alpha7nAChR) expression in bone marrow-derived non-T cells is required for the inflammatory reflex. Mol Med. 2012;18:539–543. doi: 10.2119/molmed.2011.00405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421:384–388. doi: 10.1038/nature01339. [DOI] [PubMed] [Google Scholar]
- 156.Saeed RW, Varma S, Peng-Nemeroff T, et al. Cholinergic stimulation blocks endothelial cell activation and leukocyte recruitment during inflammation. J Exp Med. 2005;201:1113–1123. doi: 10.1084/jem.20040463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.de Jonge WJ, van der Zanden EP, The FO, et al. Stimulation of the vagus nerve attenuates macrophage activation by activating the Jak2-STAT3 signaling pathway. Nat Immunol. 2005;6:844–851. doi: 10.1038/ni1229. [DOI] [PubMed] [Google Scholar]
- 158.Fleming I, Busse R. NO: the primary EDRF. J Mol Cell Cardiol. 1999;31:5–14. doi: 10.1006/jmcc.1998.0839. [DOI] [PubMed] [Google Scholar]
- 159.Olofsson PS, Steinberg BE, Sobbi R, et al. Blood pressure regulation by CD4+ lymphocytes expressing choline acetyltransferase. Nat Biotechnol. 2016;34:1066–1071. doi: 10.1038/nbt.3663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Bonaz B, Sinniger V, Hoffmann D, et al. Chronic vagus nerve stimulation in Crohn’s disease: a 6-month follow-up pilot study. Neurogastroenterol Motil. 2016;28:455–462. doi: 10.1111/nmo.12792. [DOI] [PubMed] [Google Scholar]
- 161.Koopman FA, Chavan SS, Miljko S, et al. Vagus nerve stimulation inhibits cytokine production and attenuates disease severity in rheumatoid arthritis. Proc Natl Acad Sci USA. 2016;113:8284–8289. doi: 10.1073/pnas.1605635113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Swirski FK, Nahrendorf M, Etzrodt M, et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science. 2009;325:612–616. doi: 10.1126/science.1175202. [DOI] [PMC free article] [PubMed] [Google Scholar]