Abstract
Epilepsy is a chronic neurological disorder caused by abnormal changes in the functions of neuronal circuits and manifested by seizures. It affects patients of all age, substantially worsens the quality of life for the patients as well as their families, and imposes a huge economic burden on the healthcare system. Historically, efforts for discovering and developing antiseizure therapies have been focused on modulating the functions of receptors, transporters, and enzymes expressed by neurons. These drug development efforts have paid off, as we have over 25 antiseizure drugs available in the clinic. However, these drugs mainly provide symptomatic relief from seizures and often cause serious adverse effects. Importantly, almost one-third of patients with epilepsy do not have their seizures adequately controlled by available drugs. To address this problem, researchers are investigating cellular and molecular mechanisms fundamental to the optimal function of neuronal circuits. Evidence shows that disruptions in these mechanisms cause impairment in neuroglial interactions, uncontrolled inflammation, aberrant synaptogenesis, and neurodegeneration in genetic and acquired epilepsies. Many novel therapeutic targets have been identified to target these mechanisms for developing new antiseizure drugs. In addition, the field is exploring new drug targets which may impede the development of epilepsy. We have summarized some of these novel targets in this brief review.
Commentary
Epilepsy is a devastating neurological disorder affecting nearly 65 million people worldwide. The etiology of epilepsy is heterogeneous in nature and a variety of factors—such as infection, stroke, traumatic brain injury, brain tumors, cerebral ischemia, and importantly, mutations in genes that are crucial for the development, migration, and function of neurons and glia—may cause seizures and lead to the development of epilepsy. Over 25 drugs are clinically available for the symptomatic treatment of the seizures that are the defining criteria of epilepsy. However, most of these drugs tend to optimize the balance between excitatory and inhibitory neurotransmission by modulating the functions of ion channels, receptors, enzymes, and transporters expressed by neurons (1). Importantly, around one-third of epilepsy patients are still pharmacoresistant, and many patients who respond to antiseizure drugs (ASDs) often present with significant adverse effects (2). In addition, people with epilepsy also suffer from co-morbidities such as anxiety, depression, and cognitive impairment. Many ASDs either do not address or, in some instances, aggravate these co-morbidities. Therefore, future ASD development efforts should 1) include novel mechanisms of action for ASDs, 2) evaluate potential therapeutic targets for disease modification, and 3) identify potential therapeutic interventions for the prevention of epilepsy. There are several promising targets for developing novel drugs that might prevent or impede epileptogenesis. Some of these novel targets are discussed in this brief review.
Transforming Growth Factor β (TGFβ)
Sustained damage to the blood brain barrier (BBB) is associated with increased incidence of seizures (3). Animal studies show that the activation of the TGFβ signaling pathway, mainly in astrocytes by albumin extravasated into brain parenchyma following BBB injury, disrupts regulatory functions of astrocytes and causes hyperexcitability in cortical and limbic structures (4). Direct activation of the TGFβ signaling pathway by TGFβ1 also causes epileptiform activity in rats, whereas TGFβ receptor blockers ameliorate TGFβ1- or albumin-induced epileptiform activity (5). Mice overexpressing TGFβ1 specifically in astrocytes develop seizures (6). It was recently found that losartan, which inhibits TGFβ1 signaling, decreased the number of rats developing albumin-induced chronic spontaneous seizures and inhibited epileptogenesis in this vascular injury-induced model of epilepsy (7). It is evident from these animal studies that inhibition of the TGFβ signaling pathway might be an effective strategy to inhibit seizures and epileptogenesis, especially in conditions where the blood brain barrier is compromised (8). Importantly, losartan is an FDA-approved drug widely prescribed for the treatment of cardiovascular diseases and would be a good candidate for epilepsy prevention trials in patients.
Tyrosine Receptor Kinase B (TrkB)
TrkB is a receptor for brain-derived neurotrophic factor (BDNF). Targeted deletions of either BDNF or the TrkB gene, or the pharmacological inhibition of TrkB signaling, suppresses kindling-induced epileptogenesis in rodents (9–11). In contrast, mice overexpressing BDNF or TrkB develop severe chemoconvulsant-induced seizures and intrahippocampal administration of BDNF causes spontaneous behavioral seizures in rats (12–14). Seizures also induce the expression of TrkB in the hippocampus of rodents and uncontrolled BDNF-TrkB signaling may further exacerbate seizures (15). Given that activation of TrkB signaling is associated with the development of seizures and epilepsy, inhibitors of TrkB or its downstream signaling partners may be a promising antiseizure or antiepileptogenic strategy. However, global inhibition of TrkB might be counterproductive as TrkB signaling is also required for neuronal survival and growth (16). Mice treated with a novel inhibitor of TrkB, which specifically inhibits phospholipase-γ1 (PLCγ1)-mediated effects of TrkB following kainic acid-induced status epilepticus (SE), had a significantly reduced number of chronic seizures without compromising neuroprotective functions of TrkB (17). Clinical potential of TrkB-PLCγ1 inhibitors for the prevention of epilepsy should be validated in additional animal models of epilepsy and may eventually prove useful in the clinic.
Cytokines
Development of seizures and epilepsy in animal models and in humans following a variety of brain insults is correlated with neuroinflammation and increased expression of cytokines in brain (18). Cytokines can modulate neurotransmission by affecting the levels of excitatory and inhibitory neurotransmitters and their receptors, enzymes involved in the metabolism of neurotransmitters, and cellular signaling proteins (19). Interleukin-1 (IL-1), interleukin-6 (IL-6), and tumor necrosis factor-α (TNFα) are the most widely investigated cytokines for their roles in causing inflammation, ictogenesis, and epilep-togenesis. Targeting these cytokines or their receptors and downstream signaling partners may provide a useful strategy for new antiseizure drugs.
IL-1β
Activation of the IL-1β signaling cascade following an epileptogenic brain insult causes neuronal hyperexcitability and long-term changes in the genes associated with aberrant neuronal sprouting, neurogenesis, inflammation, and glial dysfunction that reduce the threshold for chronic spontaneous seizures in animal models (20). Indeed, intrahippocampal injection of IL-1β prolonged the duration of kainic acid-induced seizures, whereas inhibition of IL-1β signaling by an IL-1R antagonist or by selective inhibition of IL-1β converting enzyme (ICE)/capsase-1 (which produces the biologically active form of IL-1β) decreased seizures in a number of animal models (21–23). Clinical studies also show increased expression of IL-1β, ICE, and IL-1β signal transduction molecules in glia and neurons in epileptic foci of patients with pharmacoresistant seizures and in neurological conditions associated with epilepsy, such as cortical malformation and Rasmussen's encephalitis (24, 25). A stage IIA clinical trial with VX-765 was completed in patients with epilepsy which found VX-765 safe and well tolerated; however, this caspase-1 inhibitor is not currently being developed for patients. Nevertheless, IL-1β remains an attractive molecular target for seizure suppression and potential disease modification.
TNFα
TNFα serves many regulatory functions in synaptic plasticity, neurotransmission and neuroglial interaction (26). Preclinical investigation suggests that there is a dichotomous role of TNFα in the pathophysiology of seizures and epilepsy. Studies using various mice strains deficient in TNFα or TNF receptors (TNFRs) show that TNFR1 and TNFR2 mediate pro- and anticonvulsant effects of TNFα, respectively, in animal models of epilepsy (27–29). Similarly, selective chronic activation of TNFR1 increased the numbers of kainic acid-induced seizures in rats (30). Anti-TNFα antibodies are widely prescribed for many peripheral inflammatory conditions, and several novel small molecules and biologics targeting central TNFα and TNFRs are under development that might be useful to treat seizures occurring as a consequence of neuroinflammation and to interfere in the development of epilepsy (31).
IL-6
Activation of IL-6 signaling may alter the expression of many receptor gated ion channel subunits through the JAK/STAT pathway (19). Increase in the levels of IL-6 can also influence synaptogenesis and tilt the balance of excitation and inhibition towards hyperexcitability (32). Animal studies investigating the role of IL-6 in seizure generation present diametric results. IL-6 decreased the latency and increased the duration of pentylenetetrazole-induced seizures in adult rats (33), whereas it increased the latency and decreased the duration of hyperthermia-induced seizures in developing rats (34). Mice overexpressing IL-6 in astrocytes have spontaneous seizures (35), while significantly fewer IL-6−/− mice infected with Theiler's virus develop acute seizures (28). In contrast, a significantly higher proportion of IL-6−/− mice exhibit kainic acid-induced seizures (36). Many variables related to animal model and experimental paradigm might have caused ambiguity in these results. Future efforts should be directed toward developing and evaluating the antiseizure effects of IL-6 modulators.
Complement proteins
The complement system is suspected to contribute to seizures and epileptogenesis in patients with mesial temporal lobe epilepsy (37), Rasmussen's encephalitis (38), and tuberous sclerosis complex (39). Activation of complement system occurs in animal models of limbic epilepsy (37). Complement 5a receptor 1 (C5aR1) was found to be upregulated in the kainic acid and pilocarpine models of epilepsy, and the C5aR1 antagonist, PMX53, reduced electrographic spontaneous seizures in the kainic acid model, and decreased the severity and mortality associated with pilocarpine-induced SE (40). PMX53 also had anticonvulsant effects in the 6 Hz and corneal kindling models of acute seizures (40). Several anticomplement therapies for a number of inflammatory conditions are currently under clinical development and might be beneficial in the treatment of seizures (41).
Cyclooxygenase-2 (COX-2) and Prostaglandins (PGs)
COX-2 is an inducible rate-limiting enzyme in the synthesis of prostanoids that include various prostaglandins (PGs) and thromboxane A2 (TXA2). The expression level of various prostanoids and their receptors contributes to the regulation of inflammation and vascular responses under pathophysiological conditions (42). COX-2 expression is rapidly increased in the brain following seizures in both animal models and humans (43). Overexpression of COX-2 in neurons increases the severity of kainic acid-induced seizures in mice (44), whereas COX-2 knockout mice have a reduced incidence of hippocampal seizures following repetitive electrical stimulation of the perforant path (45). Importantly, COX-2 inhibitors such as celecoxib and aspirin decrease pilocarpine-induced chronic spontaneous seizures and neuronal damage in rats when treated after the period of SE (46, 47). Given the regulation of vascular diameter by prostanoids, especially by PGI2 and TXA2, long-term inhibition of COX-2 may cause cardiovascular adverse effects (42). Also, animal studies have demonstrated that PGE2 signaling via EP1 and EP2 receptors are crucially involved in causing neuroinflammation and cell death following seizures. Therefore, specific inhibitors of PGE2 signaling via EP1 and EP2 could be a promising strategy for the treatment of epilepsy (48).
microRNA (miRNA)
Gene expression profiling studies have found changes in the expression levels of a number of miRNAs in animal models of epilepy and in resected brain tissues from patients with epilepsy. Importantly, mice deficient in the expression of miR-128 developed fatal epilepsy, whereas overexpression of miR-128 suppressed kainic acid-induced seizures (49). Pretreatment of mice with ant-134 (miR-134 antagomirs), which silences miR-134, reduced severity of kainic acid-induced seizures and imparted long-term neuroprotection (50). In addition, ant-134 also decreased the occurrence of chronic spontaneous seizures after kainate-induced SE, which suggests that targeting miRNAs may provide disease-modifying anti-epileptogenic effects (50). Although the mechanisms by which miRNAs may contribute to the development of hyperexcitable neural networks are largely unknown, the evidence so far suggests that miRNAs regulate the expression of glutamate receptors and the morphology of dendritic spines (51). Some miRNAs may also modulate neuroimmune interactions by inducing changes in inflammatory molecules (51). Indeed, recent work in the intra-amygdala mouse model of TLE demonstrated that short-term treatment with an miR-146a mimic that inhibits IL-1R1/TLR4 intracellular signaling could dramatically reduce the seizure frequency observed in mice several weeks after treatment. This suggests that a transient block of IL-R1 and TLR4 signaling by a miRNA mimic could be disease modifying (52), thus demonstrating the utility of miRNA targets of inflammatory signals. Given the importance of miRNAs in controlling gene expression of numerous proteins contributing to epileptogenesis, they may prove to be important therapeutic targets.
Galanin
Galanin is one of several neuropeptides that has demonstrated antiseizure activity (53). Activation of galanin receptor subtypes 1 and 2 (GalR1 and GalR2) confers antiseizure effects, whereas the role of subtype 3 (GalR3) in brain hyperexcitability and seizure activity has not been fully elucidated (53). Thus far, the potential for a novel galanin therapy has been explored by use of systemically administered, receptor subtype-preferring analogs (54). However, as systemic administration of GalR1-preferring analogs produces hyperglycemia resulting from receptor-mediated insulin inhibition in the pancreas (55, 56), GalR2 has emerged as the more likely candidate for galanin-based therapy. The lead GalR2-preferring analog, NAX 810-2, is metabolically stable and reduces seizures following sytemic and i.v. administration in the mouse 6 Hz and corneal kindling models (57, 58). Further, this lead analog is active using the mouse 6 Hz 44 mA stimulus intensity, which suggests that this novel analog may be useful in the treatment of pharmacoresistant epilepsy.
Cannabinoids
The use of marijuana (Cannabis sativa) and marijuana-derived compounds (phytocannabinoids) to treat various neurological conditions has been described previously (59). More recently, largely due to the success of cannabidiol (CBD) in preclinical and clinical settings, CBD has emerged as an attractive ASD candidate for the treatment of various forms of epilepsy. CBD protects against seizures in a variety of preclinical seizure models including electrically and chemically induced acute seizures and SE (59). Currently, CBD has orphan drug designation for Dravet Syndrome, infantile spasms, Lennox-Gastaut syndrome, and tuberous sclerosis complex. The lead candidate, Epidiolex (GW Pharmaceuticals, Carlsbad, CA), a liquid formulation of purified CBD, was tested in a Phase III clinical trial in children with Dravet Syndrome and found to result in a 22.8% reduction in median frequency of convulsive seizures per month compared to placebo treatment (60). In a similar manner, the phytocannabinoid cannabidivarin (CBDV) is also being investigated (clinical and preclinical development) by GW Pharmaceuticals as a potential therapy for pharmacoresistant epilepsy (58). As interest in these molecules increases, it is anticipated that additional studies evaluating efficacy will continue.
Metabotropic Glutamate Receptors
Activation of metabotropic glutamate receptors, and in particular subtypes 2 and 3 (mGlu2, mGlu3), can affect excitatory synaptic transmission (61) and agonists of these receptors can block or reduce seizures in a variety of seizure models (62, 63). Further, as these receptors possess both orthosteric and allosteric modulator sites, positive allosteric modulators (PAMs) of mGlu2 have emerged as a potential therapy for a variety of neurologic conditions, including epilepsy. Moreover, as mGlu2 receptors are expressed pre- and perisynaptically (64), they are uniquely positioned to respond to excess glutamate levels that may occur during seizures. mGlu2 PAMs (e.g., JNJ-42153605 and JNJ-40411813) have been evaluated both alone and in combination with the ASD levetiracetam (LEV; 65). Both of these compounds are efficacious in the mouse 6 Hz model at both the 32 mA and the more pharmacoresistant 44 mA stimulus intensities (65). Further, in contrast to LEV, which is dramatically less potent at the 44 mA stimulus intensity (at least 18-fold vs 32 mA), mGlu2 PAMs are effective in this model (44 mA) at doses only 1.5- to 2-fold greater than efficacious doses using the 32 mA stimulus intensity (65). This suggests that these compounds present a potentially novel therapy for pharmaco-resistant epilepsy. When combined with LEV, mGlu2 PAMs show a potentially synergistic effect (65), suggesting a combination therapy strategy that may be effective for pharmacoresistant seizures.
Conclusion
Currently available ASDs generally target receptor gated or voltage gated ion channels and are beneficial to a large number of epilepsy patients. Despite this advancement, significant unmet medical need exits for pharmacoresistant cases of epilepsy and epilepsy-associated comorbidites. As far as antiseizure/antiepileptic targets are concerned, it appears that the low-hanging fruits have already been plucked and future efforts should focus on exploring molecular mechanisms fundamental in regulating synatpic and network activity. As the basic mechanisms of the epilepsies are understood, especially with respect to the genetic epilepsies, novel molecular targets will be identified and provide unique opportunities to address the problems of refractory epilepsy and epileptogenesis.
Emerging evidence strongly suggests that CNS inflammation is a common occurrence in animals and patients with epilepsy, and there may be a causative link between uncontrolled inflammation and epilepsy. Testing clinical safety and efficacy of anti-inflammtory agents in epilepsy should be prioritized. VX-765, which is the only known anti-inflammatory molecule underwent early stage clinical trials in chronic epilepsy, was safe and well tolerated, and therefore, warrants larger efficacy trials. Since inflammation occurs in many CNS diseases, anti-inflammatory agents may have a wider application and that may incentivize pharmaceutical companies to develop novel anti-inflammatory therapies. Cannabinoid-based therapies have also generated well-deserved interest among epileptologists due to remarkable improvement in managing intractable epilepsy and quality of life in some patients. The results of a recent clinical trial have shown that cannabidiol is efficacious in controlling seizures in pediatric patients with Dravet's syndrome, suggesting that more trials in other epilepsies is warranted (60). Future studies should also test the combination of molecules acting on novel targets described here with the existing ASDs, as they may impart synergistic effects given their unique mechanisms of action and provide higher safety and efficacy for the treatment of epilepsy. Basic research has contributed enormously to the identification of novel targets in different types of epilepsy and we anticipate that future work will continue to provide hope to the patient with epilepsy and their families and caregivers.
Supplementary Material
References
- 1. Loscher W, Klitgaard H, Twyman RE, Schmidt D.. New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov 2013; 12: 757– 776. [DOI] [PubMed] [Google Scholar]
- 2. Loscher W, Schmidt D.. Modern antiepileptic drug development has failed to deliver: Ways out of the current dilemma. Epilepsia 2011; 52: 657– 678. [DOI] [PubMed] [Google Scholar]
- 3. Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U, Friedman A.. Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci 2004; 24: 7829– 7836. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Kim SY, Buckwalter M, Soreq H, Vezzani A, Kaufer D.. Blood-brain barrier dysfunction-induced inflammatory signaling in brain pathology and epileptogenesis. Epilepsia 2012; 53 suppl 6: 37– 44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Cacheaux LP, Ivens S, David Y, Lakhter AJ, Bar-Klein G, Shapira M, Heinemann U, Friedman A, Kaufer D.. Transcriptome profiling reveals TGF-beta signaling involvement in epileptogenesis. J Neurosci 2009; 29: 8927– 8935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Wyss-Coray T, Feng L, Masliah E, Ruppe MD, Lee HS, Toggas SM, Rockenstein EM, Mucke L.. Increased central nervous system production of extracellular matrix components and development of hydrocephalus in transgenic mice overexpressing transforming growth factor-beta 1. Am J Pathol 1995; 147: 53– 67. [PMC free article] [PubMed] [Google Scholar]
- 7. Bar-Klein G, Cacheaux LP, Kamintsky L, Prager O, Weissberg I, Schoknecht K, Cheng P, Kim SY, Wood L, Heinemann U, Kaufer D, Friedman A.. Losartan prevents acquired epilepsy via TGF-beta signaling suppression. Ann Neurol 2014; 75: 864– 875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Bar-Klein G, Lublinsky S, Kamintsky L, Noyman I, Veksler R, Dalipaj H, Senatorov VV Jr, Swissa E, Rosenbach D, Elazary N, Milikovsky DZ, Milk N, Kassirer M, Rosman Y, Serlin Y, Eisenkraft A, Chassidim Y, Parmet Y, Kaufer D, Friedman A.. Imaging blood-brain barrier dysfunction as a biomarker for epileptogenesis. Brain 2017; 140: 1692– 1705. [DOI] [PubMed] [Google Scholar]
- 9. Kokaia M, Ernfors P, Kokaia Z, Elmér E, Jaenisch R, Lindvall O.. Suppressed epileptogenesis in BDNF mutant mice. Exp Neurol 1995; 133: 215– 224. [DOI] [PubMed] [Google Scholar]
- 10. He XP, Kotloski R, Nef S, Luikart BW, Parada LF, McNamara JO.. Conditional deletion of TrkB but not BDNF prevents epileptogenesis in the kindling model. Neuron 2004; 43: 31– 42. [DOI] [PubMed] [Google Scholar]
- 11. Binder DK, Routbort MJ, Ryan TE, Yancopoulos GD, McNamara JO.. Selective inhibition of kindling development by intraventricular administration of TrkB receptor body. J Neurosci 1999; 19: 1424– 1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Scharfman HE, Goodman JH, Sollas AL, Croll SD.. Spontaneous limbic seizures after intrahippocampal infusion of brain-derived neurotrophic factor. Exp Neurol 2002; 174: 201– 214. [DOI] [PubMed] [Google Scholar]
- 13. Croll SD, Suri C, Compton DL, Simmons MV, Yancopoulos GD, Lindsay RM, Wiegand SJ, Rudge JS, Scharfman HE.. Brain-derived neurotrophic factor transgenic mice exhibit passive avoidance deficits, increased seizure severity and in vitro hyperexcitability in the hippocampus and entorhinal cortex. Neuroscience 1999; 93: 1491– 1506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Lahteinen S, Pitkänen A, Koponen E, Saarelainen T, Castrén E.. Exacerbated status epilepticus and acute cell loss, but no changes in epileptogenesis, in mice with increased brain-derived neurotrophic factor signaling. Neuroscience 2003; 122: 1081– 1092. [DOI] [PubMed] [Google Scholar]
- 15. He XP, Minichiello L, Klein R, McNamara JO.. Immunohistochemical evidence of seizure-induced activation of trkB receptors in the mossy fiber pathway of adult mouse hippocampus. J Neurosci 2002; 22: 7502– 7508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Atwal JK, Massie B, Miller FD, Kaplan DR.. The TrkB-Shc site signals neuronal survival and local axon growth via MEK and P13-kinase. Neuron 2000; 27: 265– 277. [DOI] [PubMed] [Google Scholar]
- 17. Gu B, Huang YZ, He XP, Joshi RB, Jang W, McNamara JO.. A peptide uncoupling BDNF receptor TrkB from phospholipase Cγ1 prevents epilepsy induced by status epilepticus. Neuron 2015; 88: 484– 491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Dey A, Kang X, Qiu J, Du Y, Jiang J.. Anti-inflammatory small molecules to treat seizures and epilepsy: From bench to bedside. Trends Pharmacol Sci 2016; 37: 463– 484. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Wilcox KS, Vezzani A.. Does brain inflammation mediate pathological outcomes in epilepsy? Adv Exp Med Biol 2014; 813: 169– 183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Vezzani A, Maroso M, Balosso S, Sanchez MA, Bartfai T.. IL-1 receptor/toll-like receptor signaling in infection, inflammation, stress and neurodegeneration couples hyperexcitability and seizures. Brain Behav Immun 2011; 25: 1281– 1289. [DOI] [PubMed] [Google Scholar]
- 21. Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, De Simoni MG, Sperk G, Andell-Jonsson S, Lundkvist J, Iverfeldt K, Bartfai T.. Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. Proc Natl Acad Sci U S A 2000; 97: 11534– 11539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Ravizza T, Lucas SM, Balosso S, Bernardino L, Ku G, Noé F, Malva J, Randle JC, Allan S, Vezzani A.. Inactivation of caspase-1 in rodent brain: A novel anticonvulsive strategy. Epilepsia 2006; 47: 1160– 1168. [DOI] [PubMed] [Google Scholar]
- 23. Ravizza T, Noé F, Zardoni D, Vaghi V, Sifringer M, Vezzani A.. Interleukin converting enzyme inhibition impairs kindling epileptogenesis in rats by blocking astrocytic IL-1beta production. Neurobiol Dis 2008; 31: 327– 333. [DOI] [PubMed] [Google Scholar]
- 24. Aronica E, Crino PB.. Inflammation in epilepsy: Clinical observations. Epilepsia 2011; 52 suppl 3: 26– 32. [DOI] [PubMed] [Google Scholar]
- 25. Vezzani A, French J, Bartfai T, Baram TZ. . The role of inflammation in epilepsy. Nat Rev Neurol 2011; 71: 31– 40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Probert L. TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience, 2015; 302: 2– 22. [DOI] [PubMed] [Google Scholar]
- 27. Baloss S, Ravizza T, Perego C, Peschon J, Campbell IL, De Simoni MG, Vezzani A.. Tumor necrosis factor-alpha inhibits seizures in mice via p75 receptors. Ann Neurol 2005; 57: 804– 812. [DOI] [PubMed] [Google Scholar]
- 28. Kirkman NJ, Libbey JE, Wilcox KS, White HS, Fujinami RS.. Innate but not adaptive immune responses contribute to behavioral seizures following viral infection. Epilepsia 2010; 51: 454– 464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Patel DC, Wallis G, Dahle EJ, McElroy PB, Thomson KE, Tesi RJ, Szymkowski DE, West PJ, Smeal RM, Patel M, Fujinami RS, White HS, Wilcox KS.. Hippocampal TNFa signaling contributes to seizure generation in an infection-induced mouse model of limbic epilepsy. eNeuro 2017; 4 doi: 10.1523/ENEURO.0105-17.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Weinberg MS, Blake BL, McCown TJ.. Opposing actions of hippocampus TNFalpha receptors on limbic seizure susceptibility. Exp Neurol 2013; 247: 429– 437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Fischer R, Kontermann R, Maier O.. Targeting sTNF/TNFR1 signaling as a new therapeutic strategy. Antibodies 2015; 4: 48– 70. [Google Scholar]
- 32. Wei H, Chadman KK, McCloskey DP, Sheikh AM, Malik M, Brown WT, Li X.. Brain IL-6 elevation causes neuronal circuitry imbalances and mediates autism-like behaviors. Biochim Biophys Acta 2012; 1822: 831– 842. [DOI] [PubMed] [Google Scholar]
- 33. Kalueff AV, Lehtimaki KA, Ylinen A, Honkaniemi J, Peltola J.. Intranasal administration of human IL-6 increases the severity of chemically induced seizures in rats. Neurosci Lett 2004; 365: 106– 110. [DOI] [PubMed] [Google Scholar]
- 34. Fukuda M, Morimoto T, Suzuki Y, Shinonaga C, Ishida Y.. Interleukin-6 attenuates hyperthermia-induced seizures in developing rats. Brain Dev 2007; 29: 644– 648. [DOI] [PubMed] [Google Scholar]
- 35. Campbell IL, Abraham CR, Masliah E, Kemper P, Inglis JD, Oldstone MB, Mucke L.. Neurologic disease induced in transgenic mice by cerebral overexpression of interleukin 6. Proc Natl Acad Sci U S A 1993; 90: 10061– 10065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Penkowa M, Molinero A, Carrasco J, Hidalgo J.. Interleukin-6 deficiency reduces the brain inflammatory response and increases oxidative stress and neurodegeneration after kainic acid-induced seizures. Neuroscience 2001; 102: 805– 818. [DOI] [PubMed] [Google Scholar]
- 37. Aronica E, Boer K, van Vliet EA, Redeker S, Baayen JC, Spliet WG, van Rijen PC, Troost D, da Silva FH, Wadman WJ, Gorter JA.. Complement activation in experimental and human temporal lobe epilepsy. Neurobiol Dis 2007; 263: 497– 511. [DOI] [PubMed] [Google Scholar]
- 38. Whitney KD, Andrews PI, McNamara JO.. Immunoglobulin G and complement immunoreactivity in the cerebral cortex of patients with Rasmussen's encephalitis. Neurology 1999; 53: 699– 708. [DOI] [PubMed] [Google Scholar]
- 39. Boer K, Crino PB, Gorter JA, Nellist M, Jansen FE, Spliet WG, van Rijen PC, Wittink FR, Breit TM, Troost D, Wadman WJ, Aronica E.. Gene expression analysis of tuberous sclerosis complex cortical tubers reveals increased expression of adhesion and inflammatory factors. Brain Pathol 2010; 20: 704– 719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Benson MJ, Thomas NK, Talwar S, Hodson MP, Lynch JW, Woodruff TM, Borges K.. A novel anticonvulsant mechanism via inhibition of complement receptor C5ar1 in murine epilepsy models. Neurobiol Dis 2015; 76: 87– 97. [DOI] [PubMed] [Google Scholar]
- 41. Morgan BP, Harris CL.. Complement, a target for therapy in inflammatory and degenerative diseases. Nat Rev Drug Discov 2015; 14: 857– 877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Ricciotti E, FitzGerald GA.. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 2011; 31: 986– 1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Rojas A, Jiang J, Ganesh T, Yang MS, Lelutiu N, Gueorguieva P, Dingledine R.. Cyclooxygenase-2 in epilepsy. Epilepsia 2014; 55: 17– 25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kelley KA, Ho L, Winger D, Freire-Moar J, Borelli CB, Aisen PS, Pasinetti GM.. Potentiation of excitotoxicity in transgenic mice overexpressing neuronal cyclooxygenase-2. Am J Pathol 1999; 155: 995– 1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Takemiya T, Suzuki K, Sugiura H, Yasuda S, Yamagata K, Kawakami Y, Maru E.. Inducible brain COX-2 facilitates the recurrence of hippocampal seizures in mouse rapid kindling. Prostaglandins Other Lipid Mediat 2003; 71: 205– 216. [DOI] [PubMed] [Google Scholar]
- 46. Jung KH, Chu K, Lee ST, Kim J, Sinn DI, Kim JM, Park DK, Lee JJ, Kim SU, Kim M, Lee SK, Roh JK.. Cyclooxygenase-2 inhibitor, celecoxib, inhibits the altered hippocampal neurogenesis with attenuation of spontaneous recurrent seizures following pilocarpine-induced status epilepticus. Neurobiol Dis 2006; 23: 237– 246. [DOI] [PubMed] [Google Scholar]
- 47. Ma L, Cui XL, Wang Y, Li XW, Yang F, Wei D, Jiang W.. Aspirin attenuates spontaneous recurrent seizures and inhibits hippocampal neuronal loss, mossy fiber sprouting and aberrant neurogenesis following pilocarpine-induced status epilepticus in rats. Brain Res 2012; 1469: 103– 113. [DOI] [PubMed] [Google Scholar]
- 48. Varvel NH, Jiang J, Dingledine R.. Candidate drug targets for prevention or modification of epilepsy. Annu Rev Pharmacol Toxicol 2015; 55: 229– 247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Tan CL, Plotkin JL, Venø MT, von Schimmelmann M, Feinberg P, Mann S, Handler A, Kjems J, Surmeier DJ, O'Carroll D, Greengard P, Schaefer A.. MicroRNA-128 governs neuronal excitability and motor behavior in mice. Science 2013; 342: 1254– 1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Jimenez-Mateos EM, Engel T, Merino-Serrais P, McKiernan RC, Tanaka K, Mouri G, Sano T, O'Tuathaigh C, Waddington JL, Prenter S, Delanty N, Farrell MA, O'Brien DF, Conroy RM, Stallings RL, DeFelipe J, Henshall DC.. Silencing microRNA-134 produces neuroprotective and prolonged seizure-suppressive effects. Nat Med 2012; 18: 1087– 1094. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Reschke CR, Henshall DC.. microRNA and epilepsy. Adv Exp Med Biol 2015; 888: 41– 70. [DOI] [PubMed] [Google Scholar]
- 52. Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, Cerovic M, Hill C, Ferrari M, Zucchetti M, Molteni M, Rossetti C, Brambilla R, Steve White H, D'Incalci M, Aronica E, Vezzani A.. Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis 2017; 99: 12– 23. [DOI] [PubMed] [Google Scholar]
- 53. Clynen E, Swijsen A, Raijmakers M, Hoogland G, Rigo JM.. Neuropeptides as targets for the development of anticonvulsant drugs. Mol Neurobiol 2014; 50: 626– 646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Lang R, Gundlach AL, Kofler B.. The galanin peptide family: Receptor pharmacology, pleiotropic biological actions, and implications in health and disease. Pharmacol Ther 2007; 115: 177– 207. [DOI] [PubMed] [Google Scholar]
- 55. Tang G, Wang Y, Park S, Bajpayee NS, Vi D, Nagaoka Y, Birnbaumer L, Jiang M.. Go2 G protein mediates galanin inhibitory effects on insulin release from pancreatic beta cells. Proc Natl Acad Sci U S A 2012; 109: 2636– 2641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Flynn SP, White HS.. Regulation of glucose and insulin release following acute and repeated treatment with the synthetic galanin analog NAX-5055. Neuropeptides 2015; 50: 35– 42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Metcalf CS, Klein BD, McDougle DR, Zhang L, Kaufmann D, Bulaj G, White HS.. Preclinical evaluation of intravenous NAX 810-2, a novel GalR2-preferring analog, for anticonvulsant efficacy and pharmacokinetics. Epilepsia 2017; 58: 239– 246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS.. Progress report on new antiepileptic drugs: A summary of the Twelfth Eilat Conference (EILAT XII). Epilepsy Res 2015; 111: 85– 141. [DOI] [PubMed] [Google Scholar]
- 59. Devinsky O, Cilio MR, Cross H, Fernandez-Ruiz J, French J, Hill C, Katz R, Di Marzo V, Jutras-Aswad D, Notcutt WG, Martinez-Orgado J, Robson PJ, Rohrback BG, Thiele E, Whalley B, Friedman D.. Cannabidiol: Pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia 2014; 55: 791– 802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Devinsky O, Cross JH, Laux L, Marsh E, Miller I, Nabbout R, Scheffer IE, Thiele EA, Wright S; Cannabidiol in Dravet Syndrome Study Group. . Trial of cannabidiol for drug-resistant seizures in the Dravet Syndrome. N Engl J Med 2017; 376: 2011– 2020. [DOI] [PubMed] [Google Scholar]
- 61. Anwyl R. Metabotropic glutamate receptors: Electrophysiological properties and role in plasticity. Brain Res Brain Res Rev 1999; 29: 83– 120. [DOI] [PubMed] [Google Scholar]
- 62. Moldrich RX, Chapman AG, De Sarro G, Meldrum BS.. Glutamate metabotropic receptors as targets for drug therapy in epilepsy. Eur J Pharmacol 2003; 476: 3– 16. [DOI] [PubMed] [Google Scholar]
- 63. Qian F, Tang FR.. Metbotropic glutamate receptors and interacting proteins in epileptogenesis. Curr Neuropharmacol 2016; 14: 551– 562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lujan R, Roberts JD, Shigemoto R, Ohishi H, Somogyi P.. Differential plasma membrane distribution of metabotropic glutamate receptors mGluR1 alpha, mGluR2 and mGluR5, relative to neurotransmitter release sites. J Chem Neuroanat 1997; 13: 219– 241. [DOI] [PubMed] [Google Scholar]
- 65. Metcalf CS, Klein BD, Smith MD, Pruess T, Ceusters M, Lavreysen H, Pype S, Van Osselaer N, Twyman R, White HS.. Efficacy of mGlu2-positive allosteric modulators alone and in combination with levetiracetam in the mouse 6 Hz model of psychomotor seizures. Epilepsia 2017; 58: 484– 493. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.