Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2019

Open Access 01.12.2019 | Review

Cyclooxygenase-2 (COX-2) inhibitors: future therapeutic strategies for epilepsy management

verfasst von: Chitra Rawat, Samiksha Kukal, Ujjwal Ranjan Dahiya, Ritushree Kukreti

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2019

Abstract

Epilepsy, a common multifactorial neurological disease, affects about 69 million people worldwide constituting nearly 1% of the world population. Despite decades of extensive research on understanding its underlying mechanism and developing the pharmacological treatment, very little is known about the biological alterations leading to epileptogenesis. Due to this gap, the currently available antiepileptic drug therapy is symptomatic in nature and is ineffective in 30% of the cases. Mounting evidences revealed the pathophysiological role of neuroinflammation in epilepsy which has shifted the focus of epilepsy researchers towards the development of neuroinflammation-targeted therapeutics for epilepsy management. Markedly increased expression of key inflammatory mediators in the brain and blood-brain barrier may affect neuronal function and excitability and thus may increase seizure susceptibility in preclinical and clinical settings. Cyclooxygenase-2 (COX-2), an enzyme synthesizing the proinflammatory mediators, prostaglandins, has widely been reported to be induced during seizures and is considered to be a potential neurotherapeutic target for epilepsy management. However, the efficacy of such therapy involving COX-2 inhibition depends on various factors viz., therapeutic dose, time of administration, treatment duration, and selectivity of COX-2 inhibitors. This article reviews the preclinical and clinical evidences supporting the role of COX-2 in seizure-associated neuroinflammation in epilepsy and the potential clinical use of COX-2 inhibitors as a future strategy for epilepsy treatment.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AA
Arachidonic acid
ABC
ATP-binding cassette
AED
Antiepileptic drug
BBB
Blood-brain barrier
CNS
Central nervous system
COX
Cyclooxygenase
CSF
Cerebrospinal fluid
DRTLE
Drug-resistant temporal lobe epilepsy
ECF
Extracellular fluid
GABA
γ-Aminobutyric acid
GPCR
G protein-coupled receptor
HS
Hippocampal sclerosis
KA
Kainic acid
LPS
Lipopolysaccharide
NMDA
N-Methyl-d-aspartate
NO
Nitric oxide
NSAID
Non-steroidal anti-inflammatory drug
PG
Prostaglandin
P-gp
P-glycoprotein
PTZ
Pentylenetetrazol
SE
Status epilepticus
TLE
Temporal lobe epilepsy

Background

Epilepsy, a common neurological disorder, is characterized by (1) at least two unprovoked seizures occurring > 24 h apart, (2) one unprovoked seizure and a probability of further seizures similar to the general recurrence risk (at least 60%) after two unprovoked seizures, occurring over the next 10 years, or (3) diagnosis of an epilepsy syndrome [1]. The underlying pathophysiology behind the disease progression is still unknown, and therefore, the only available remedy is to control the frequency of seizures using the symptomatic treatment, antiepileptic drugs (AEDs). However, nearly 30% of the patients do not respond to the available AEDs [2], emphasizing the need to develop better effective therapies which can target epileptogenesis.
Investigations to elucidate the mechanisms involved in epileptogenesis provided strong evidences on the crucial role of inflammation as the cause as well as consequence of seizure and epilepsy development [3]. In this regard, inflammatory molecules such as cytokines, chemokines, and prostaglandins (PGs) are often observed to be released by the brain and brain capillary endothelial cells affecting neuronal function and excitability in preclinical and clinical set-ups [4]. Blocking the undesired inflammatory signaling using anti-inflammatory molecules may provide novel strategies to treat epilepsy [5]. The inflammatory molecules might, therefore, serve as therapeutic targets to develop better effective medications for epilepsy management.
Over the past two decades, cyclooxygenase-2 (COX-2), being the central link to various inflammatory processes, has received much attention due to its involvement in seizure generation and epilepsy development. COX-2 has been reported to be upregulated in different cells within the brain following seizure induction leading to increased production of proinflammatory mediators, PGs, which further aggravates seizure severity [6]. Besides, in vivo evidences suggest that COX-2 induction following seizure may upregulate the multidrug efflux transporter P-glycoprotein (P-gp) at the blood-brain barrier (BBB) causing reduced delivery of administered AEDs to the brain target site, thus leading to poor efficacy [7]. Such studies proposed that inhibition of COX-2, genetic or pharmacological, might reduce seizure severity and pharmacoresistance to AEDs and thus could be exploited as a future strategy for epilepsy treatment.
In this article, we review the preclinical and clinical evidences supporting the role of COX-2 in seizure-associated neuroinflammation in epilepsy and its regulatory effect governing patient’s response to AEDs. This review also addresses the potential therapeutic use of COX-2 inhibitors as (1) anticonvulsants for epilepsy management or (2) adjunctives to AED therapy to overcome pharmacoresistance.

Inflammation in epilepsy

Neuroinflammation comprises activation of microglia, astrocytes, brain capillary endothelial cells, and circulating peripheral immune cells along with the production of inflammatory mediators, initiated in response to a variety of stimulus such as traumatic brain injury, brain infection, and autoimmunity. Evidences revealing the altered expression of different cytokines, chemokines, and other immune-related molecules in epilepsy indicated inflammation as a crucial factor contributing in its pathogenesis. Increased glial cellular expression of a cytokine IL-1α was observed in patients with drug-resistant temporal lobe epilepsy (DRTLE) with complex partial seizures [8]. An equally potent inflammatory cytokine IL1B was also found to be associated with epilepsy when a polymorphism in that gene responsible for increasing the production of IL-1β was detected in patients with temporal lobe epilepsy (TLE) [9]. Besides, cerebrospinal fluid (CSF) as well as serum samples from patients experiencing seizures exhibited increased levels of different cytokines such as IL-1β, IL-6, IL-1Ra, and IFNγ, substantiating the role of such cytokines in seizure sustenance [1013]. A recent follow-up study compared central and peripheral levels of IL-1β and IL-6 in sera of drug-resistant patients before surgical treatment and 1 year after surgery when most patients were either seizure-free or had reduced seizures [14]. The respective levels were found to decrease in the absence of seizures after surgical treatment indicating seizure-induced inflammation. Gene expression profiling of surgically removed hippocampal tissue from patients with TLE revealed upregulation of several chemokines, CCL2, CCL3, and CCL4 along with the chemokine receptor, CXCR4 [15]. The chemokine ligand CX3CL1 was also observed to be upregulated in the hippocampus and the adjacent cortex of epileptic rats as well as in temporal neocortex of patients with TLE [16]. CX3CL1 was further reported to be elevated in the CSF and serum of the same patients compared to the non-epileptic group. Expression of another C-X-C chemokine motif ligand CXCL13 and its receptor CXCR5 were also altered in brain tissues of patients with DRTLE [17]. These alterations were associated with changes in the molecules regulating the cytokines. Severe neuronal loss and persistent overexpression of NFκB-p65, a key regulator of acute inflammatory reactions, was noticed in reactive astrocytes in human medial TLE with hippocampal sclerosis (HS) [18]. The findings were strengthened by Das et al. who revealed an upregulation of NF-κB-p65 along with COX-2 enzyme and TGF-β in the hippocampal region as the key molecular events associated with histopathological changes observed in DRTLE [19]. Such clinical evidences and their similarities with the findings of rodent studies promoted the use of in vivo animal models to determine the putative mechanism underlying the shared link between inflammation and seizures.
Accumulating evidences in in vivo experimental models suggested the role of inflammation as either the cause or the consequence of epilepsy contributing to its pathophysiology [3]. Findings from the studies performed in rodent epilepsy models showed activation of hippocampal astrocytes and glial cells along with the elevation of inflammatory mediators in the hippocampus. A transient time-dependent increase in expression of important inflammatory cytokines IL-1β, IL-6, and TNFα was observed in the hippocampus of electrically induced limbic status epilepticus (SE) rat model [20]. SE induction in mouse altered the expression of chemokine receptors, CCR3 and CCR2A, and their ligands in the brain and thereby may weaken the neuroprotective mechanisms [21]. Furthermore, microarray analysis of different brain regions of rat model of TLE during epileptogenesis indicated alterations in inflammatory molecules such as interleukins in the acute, latent, and chronic phase of epilepsy [22]. Besides the cytokines, induction of the proinflammatory enzyme COX-2 was observed in hippocampal and neocortical neurons upon hippocampal kindling in rats, suggesting COX-2 induction to be a key signaling event in epileptogenesis [23]. A remarkable rise in the production of COX enzyme products, i.e., PGs, was observed along with the increased COX-2 expression, following seizure induction in rodents [6, 24]. Such studies illustrate inflammation as a consequence of seizure induction and epilepsy.
Systemic administration of lipopolysaccharide (LPS), an inducer of inflammation, prior to SE induction increased hippocampal vulnerability to seizure-induced neuronal injury in immature rats, suggesting the involvement of inflammation in seizure etiology [25]. LPS administration in immature rats also increased kindling progression indicating LPS-induced inflammation to enhance epileptogenesis [26]. Administration of high doses of IL-1β, a pyrogenic proinflammatory cytokine, induced febrile seizures in only IL-1β receptor-expressing mice whereas IL-1β receptor-deficient mice were resistant to seizure generation [27]. Brain transcriptome profiling in Wistar rats after epileptogenic treatment revealed TGF-β signaling as a novel inflammatory cascade involved in influencing the generation of epileptiform activity [28]. Endogenous nitric oxide (NO), another proinflammatory mediator, increased seizure activity in mice brain slices and reported as a key promoting factor for initiation of seizure-like events [29]. Similarly, intrahippocampal injection of HMGB1, a cytokine-like proinflammatory molecule, increased seizure frequency in epileptic rats [30]. However, the effect abrogated in case of TLR-4 mutant mice, recognizing the involvement of HMGB1-TLR4 inflammatory axis in generating seizures. Furthermore, expression of different inflammatory mediators TLR4, ATF-3, and IL-8 in the epileptic brain tissue of patients with mesial TLE correlated with their seizure frequency, supporting the etiologic role of inflammation in seizure generation [31]. Therefore, inflammation plays a reciprocal role in epilepsy, i.e., an outcome of seizure activity as well as a contributing factor in the disease development.

Inflammation: a potential therapeutic target

Current anticonvulsive therapy primarily consists of AEDs which only control the frequency of seizures and hence considered symptomatic. AEDs function by exerting a number of concurrent mechanisms involving both the excitatory and inhibitory synapses. Common modes of action include targeting ion channels and neuroreceptors embedded in the cell membrane to regulate neuronal excitability. Most of the AEDs such as phenytoin, carbamazepine, valproate, and topiramate act as sodium channel blockers by stabilizing the inactivated state of these channels, thus preventing the neuronal depolarization and excitability [32]. On the contrary, activation of potassium channels by retigabine causes a generalized reduction in neuronal excitability by driving the membrane potential to a hyperpolarized state [33]. Gabapentin and pregabalin act on pre-synaptic calcium channels and blocks calcium-mediated release of neurotransmitter across the synapse, preventing the synaptic conduction [34]. Simultaneously, inhibition of glutamate receptor by topiramate prevents the effect of the excitatory neurotransmitter, glutamate, while agonists of the inhibitory neurotransmitter, γ-aminobutyric acid (GABA), such as benzodiazepines activate GABA receptor preventing generation of neuronal action potential. Valproate and vigabatrin increase GABA turnover by blocking its degradation [32, 33]. Levetiracetam works by binding to the synaptic vesicle protein, SV2A, causing a reduction in the vesicular release of neurotransmitter, thus differing in its mechanism from other AEDs [34].
Despite the availability of a wide spectrum of AEDs with diverse pharmacological targets, 30% of patients experience multidrug resistance in epilepsy [35]. Various hypotheses have been proposed to address the molecular mechanism behind this drug resistance. Drug target hypothesis suggested that resistance to AEDs is caused by genetic or acquired alterations at their target sites (ion channels or neuroreceptors associated with neuronal excitability) affecting the pharmacodynamics of the drug [36]. Genetic polymorphisms in the target genes such as SCN1A and GABRA1 have been observed to be associated with AED dose requirements or AED resistance [3740]. Transporter hypothesis proposed that increased expression of ATP-binding cassette (ABC) efflux transporters at the BBB interferes with the pharmacokinetics of the AEDs leading to their decreased concentration at the target site [41]. Brain endothelial cells and other brain cells of patients with refractory epilepsy have been reported to have increased expression of ABC transporters compared to healthy individuals [4244]. A third recent hypothesis known as intrinsic severity hypothesis correlated AED resistance with the severity of the disease [45]. The higher the seizure frequency, the more difficult it is to treat with the available AEDs [46, 47]. Another recent hypothesis called the methylation hypothesis proposed that seizures can mediate epigenetic modifications that result in persistent genomic methylation, histone density, and posttranslational modifications, as well as noncoding RNA-based changes leading to pharmacoresistance in epilepsy [48].
In view of the above hypotheses, till date, nothing conclusive has been gained to address pharmacoresistance in epilepsy, suggesting a gap in the current pharmacological research which often overlooks inflammation as a primary therapeutic target in managing epilepsy. Findings of studies examining whether the currently available AEDs possess anti-inflammatory action are quite debatable. Levetiracetam displayed anti-inflammatory property by reducing immunoreactivity of astrocytic and glial IL-1β system in epileptic rat hippocampus; on the other hand, valproate was unable to show such effects [49]. However, whether the drug has primary anti-inflammatory properties or secondary to reduced seizure activity is still a question. Pre-administration of vinpocetine and carbamazepine before LPS also reduced the brain mRNA levels of IL-1β and TNF-α in rats while valproate again showed neutral behavior [50]. The same study also showed complete prevention of seizure activity along with decreased IL-1β and TNF-α mRNA levels in the groups pre-administered with the two AEDs before the pro-convulsive drug, 4-aminopyridine, suggesting that the anti-inflammatory action of these drugs might be achieved primary to reduced epileptic activity. Further investigation on such evidences is required to better understand the mechanism of anticonvulsive agents. Contrary to these findings, Verotti et al. observed that both carbamazepine and valproate induced inflammation by increasing interleukin and chemokine levels in children with epilepsy on 1 year of monotherapy [51]. The conflicting findings on the effect of these AEDs on inflammation hint towards the need of a better efficacious anti-inflammatory treatment to manage epilepsy.
Given the indispensable role of inflammation in epilepsy pathogenesis, inflammatory molecules may be considered as important therapeutic targets for epilepsy management; however, evidence on the efficacy of such therapy is limited. Vezzani et al. demonstrated powerful anticonvulsant action of IL-1Ra, an endogenous IL-1 receptor antagonist, by blocking the proinflammatory action of IL-1β in mice [52]. Another study observed a delay in onset of seizure and decrease in its duration when the administered caspase-1 inhibitor reduced IL-1β levels in SE rats [53]. The similar finding was observed in a more recent study, where epigenetic impairment of IL-1β/TLR4 pathway reduced seizure frequency by approximately 50% while the widely prescribed AED, carbamazepine, was ineffective, again representing the anti-inflammatory therapy as a pivotal strategy to manage drug-resistant epilepsy [54]. Selective PG EP2 receptor antagonist also reduced SE-induced neuronal injury by preventing COX-2 upregulation in rats [55]. Supporting this, administration of rofecoxib, a COX-2 inhibitor, potentiates the anticonvulsant activity of subeffective dose of tiagabine against pentylenetetrazol (PTZ)-induced convulsions in mice [56]. Therefore, inhibition of such inflammatory molecules may serve as an effective treatment strategy for drug-resistant epilepsy.
Inflammation is a complex biological process involving several proinflammatory as well as anti-inflammatory mechanisms. While application of a polypharmaceutical approach may have unintended consequences through drug-drug interactions [57], targeting an inflammatory cascade which involves both pro- and anti-inflammatory molecules may be exactly what would produce the desired effects by reducing inflammation, provided the anti-inflammatory mechanisms remain intact. Therefore, identification of such cascade involving different inflammatory processes is essential to produce broad-spectrum efficacious treatment. Notably, most of the published findings on crosstalk or signaling pathways of inflammation converge at the common proinflammatory gene, COX-2 (Fig. 1). COX-2 is rapidly induced after a pro-inflammatory event with a subsequent release of PGs, potent mediators of inflammatory responses. IL-1β released by activated microglia during inflammation of the central nervous system (CNS) was shown to induce COX-2 and biosynthesis of its proinflammatory product, PGE2, in mice astrocytes [58] and in human neuroblastoma cells [59]. Increased levels of HMGB1 found within inflamed synovium of rheumatoid arthritis patients potentiate IL-1β to stimulate COX-2 and prostanoid synthesis [60]. Similarly, the proinflammatory cytokine, TNFα, also induces COX-2 expression and PGE2 release resulting in enhanced vascular permeability and cytoskeletal changes in brain capillary endothelial cells [61]. The signaling pathways connecting such cytokines to COX-2 expression may involve regulatory kinases such as SPK, TK, PKC, NF-κB, ERK, and MAPK [58, 6264]. The chemokine, CXCL1, induced COX-2 via ERK, thus mediating astroglial-neuronal interaction to enhance sensitization towards neuropathic pain [65]. More recently, activation of COX-2 and PG production by TGF-β1 via ALK5/SMAD and MEK/ERK pathway in dental pulp cells have been demonstrated to be an early event in tissue inflammation and regeneration [66]. Besides the cytokines, TLR4 was also found to regulate COX-2 in TLR4-positive human intestinal epithelial cells which expressed higher COX-2 levels upon LPS exposure compared to TLR4-negative cells [67]. Simultaneously, in vivo induction of colitis showed increased COX-2 expression in wild-type mice compared to TLR4-deficient mice. Besides, the anti-inflammatory cascades involving IL-10, IL-4, and IL-1Ra have also been shown to regulate COX-2 in different cells [6870]. Moreover, administration of COX-2 inhibitors, ibuprofen and celecoxib, following traumatic brain injury in rats showed no significant difference in brain IL-10 indicating anti-inflammatory mechanisms to remain intact in presence of COX-2 inhibitors [71]. COX-2, therefore, serves as a downstream molecule to several inflammatory processes. In addition, change in the COX-2 activity itself has also been reported to alter the pro-inflammatory pathways. Treatment with a selective COX-2 inhibitor, SC-58125, reduced synovial inflammation by reducing local and systemic IL-6 levels, thus reversing paw edema in adjuvant-induced arthritis rat model showing regulation of IL-6 by COX-2-derived PGs [72]. Consistent with this, increased PGE2 production and IL-6 levels in IL-1β-stimulated human fibroblast-like synoviocytes from patients with disk displacement were reversed on in vitro treatment with COX-2 inhibitors, celecoxib, and indomethacin [73]. Indomethacin also reduced IL-1β and TNF-α expression in hippocampus of pilocarpine-induced SE rat model [74] demonstrating a regulatory effect of COX-2 on these inflammatory molecules. COX-2, therefore, acts as a central signaling molecule for various inflammatory processes and could be explored as a potential therapeutic target for the management of numerous diseases including epilepsy.

Cyclooxygenases (COX)

Prostaglandin-endoperoxide synthases (PTGS), also known as COX, are enzymes which synthesize prostanoids involving PGs and thromboxane from their substrate arachidonic acid (AA). Mammalian COX enzyme group consists of two widely studied and well-understood oxygenases, COX-1 and COX-2, responsible for the synthesis of the proinflammatory mediators, PGs. With the help of mutagenesis, X-ray crystallography, and kinetic studies, their structure and mechanism of action are well established [75]. The two isoforms COX-1 and COX-2 catalyzing the synthesis of inflammatory molecules are almost similar in weight with 60–65% sequence similarity; however, they differ in their localization and expression. While COX-1 is constitutively expressed in nearly all tissues, COX-2 is an inducible enzyme primarily localized to immune cells such as macrophages and leucocytes and upregulated in pathological conditions [76]. Further, COX-1 is believed to be responsible for homeostatic PG production whereas COX-2 produces PGs which are generally pathophysiological in nature [77]. Exposure to endotoxins, cytokines, and mitogens induces COX-2 in different cell types such as chondrocytes and macrophages, suggesting the involvement of COX-2 in inflammatory responses. In this regard, administration of COX-2 inhibitory drugs is considered as the treatment to suppress inflammation in various acute and chronic conditions. The last two decades have seen tremendous research on the role of COX-2 in various neuroinflammatory diseases including epilepsy and its applicability as a neurotherapeutic target.

COX-2-mediated neuroinflammation in epilepsy

COX-2-mediated neuroinflammation is a subject of broad and current interest in basic epilepsy research. Yamagata et al., for the first time, demonstrated immediate induction of COX-2 mRNA and protein in rat hippocampus and cerebral cortex following a maximal electroconvulsive seizure [78]. They also demonstrated this seizure-mediated COX-2 induction to be regulated by N-methyl-d-aspartate (NMDA) receptor-dependent synaptic activity. They further proposed that COX-2 induction during seizure leads to the activation of PG signaling pathway, consequently triggering secondary damage to the brain and amplifying disease severity. Thereafter, several studies replicated the findings in other rat brain tissues, striatum, brain stem and cerebellum, besides hippocampus and cerebral cortex in kainic acid (KA) as well as electroconvulsive shock-induced seizures, suggesting delayed neuronal damage of an interconnected neuronal network during COX-2 activation [7981]. COX-2 induction, in turn, facilitated recurrence of hippocampal seizures in a mouse model of kindling by synthesizing more PGE2 [6]. Production of PGE2, the major product of COX-2, was observed to be increased along with COX-2 induction following seizures, stimulating neuronal loss in rodent models of epilepsy [82, 83]. Furthermore, combining PGE2 with subconvulsant dose of PTZ caused seizures whereas administration of PGE2 antibodies attenuated PTZ-induced seizures in rats, supporting a key role of PGE2 production in triggering seizure and maintaining its threshold [84]. PGE2 binds to G protein-coupled receptors (GPCRs), namely EP1, EP2, EP3, and EP4, with highest affinity for the EP1 receptor. Activation of EP receptors results in increased calcium ion influx which in turn enhances glutamate release presynaptically [85, 86]. Genetic ablation or pharmacological inhibition of EP1 receptor in mice did not affect the seizure threshold but prevented the likelihood of SE and aggravation of seizure severity, indicating the involvement of EP1 receptor in seizure exacerbation [87, 88]. Unlike EP1, studies on the involvement of EP2 receptor in neuroprotection from seizures had shown quite debatable findings. EP2 antagonism using 3-aryl-acrylamide derivatives attenuated SE-induced neuroinflammation and neuronal injury in rodents [55, 8991]. Conflictingly, few other studies found systemic administration of EP2 agonists to have significant anticonvulsant effect to PTZ- and pilocarpine-induced seizures [92, 93]. This dual effect of EP2 modulation on neuronal activity depends on the latency to seizure onset and thus shows neuroprotection within a tightly regulated therapeutic window [94, 95]. Such studies question if EP receptors can be considered as potential therapeutic targets in neurological diseases.
Following seizure insults, the developing rat brain showed either no or minor change in COX-2 expression; in contrast, a pronounced increase in COX-2 expression in the adult rat brain was observed [96, 97]. This suggests that COX-2 induction following seizure is age-dependent and the mechanisms regulating its expression and functions are immature in the developing brain [97]. Interestingly, electrically induced SE showed biphasic upregulation of COX-2 in rat hippocampus, an immediate induction at 1 day after SE and induction during spontaneous recurrent seizures (SRS) at the chronic phase, 4–5 months after SE [98]. Moreover, systemic KA administration demonstrated a similar biphasic increase in PG production in the rat hippocampus consisting of an initial burst in the first 30 min and a sustained late-phase production due to COX-2 induction even with a limited AA supply [24]. This suggests that amidst the acute phase and the chronic phase, some recovery mechanisms are activated, which can prevent the exacerbation of seizure-induced neuronal damage, but failed to prevent epileptogenesis.
In respect of human subjects, so far, only four studies have investigated COX-2 expression profile in the hippocampal tissue from patients with DRTLE [19, 98100] (Table 1). These studies revealed strong immunohistochemical expression of COX-2 in neurons, astrocytes, and microglial cells of hippocampal sclerotic tissue. While Desjardins et al. suggested the implication of COX-2 induction in the pathogenesis of HS in TLE [101], Weidner et al. found no difference in COX-2 expression between TLE with HS and TLE without HS groups implying COX-2 induction to be independent of presence/absence of HS [100]. More recently, epilepsy researchers began working on the relevance of genetic aspects of COX-2 in people experiencing seizures by investigating associated genetic variants [101].
Table 1
Clinical studies reporting COX-2 involvement in epilepsy
Reference
Study type
Tissue
Cells
Study subjects
Reference
Desjardins et al. [99]
Expression
Hippocampus
Astrocytes and neurons
5 sclerotic and 2 non-sclerotic DRTLE
Induction of astrocytic COX-2 in patients with HS suggesting its implication in the pathogenesis of HS in epilepsy
Holtman et al. [98]
Expression
Hippocampus
Astrocytes and neurons
6 sclerotic and 4 non-sclerotic DRTLE and 5 controls
Higher astrocytic and neuronal COX-2 in patients with HS compared to non-HS and controls
Das et al. [19]
Expression
Hippocampus
Astrocytes and neurons
6 sclerotic DRTLE and 3 sudden-death controls
Increased COX-2 in patients suggesting its crucial role in TLE pathogenesis
Hung et al. [101]
Genetic
Whole blood
White blood cells
35 children with febrile seizures and 31 controls
A single SNP, rs689466, localized at 5′-1192 of the PTGS2 gene was significantly association with febrile seizures
Weidner et al. [100]
Expression
Hippocampus
Microglia, astrocytes and neurons
16 sclerotic and 17 non-sclerotic DRTLE
Higher microglial and neuronal COX-2 expression than astrocytic COX-2
No difference in COX-2 levels among sclerotic and non-sclerotic samples

COX-2-mediated blood-brain barrier disruption

Regulating the trafficking of circulating molecules between the blood and the brain, there exists a selectively permeable monolayer of brain capillary endothelial cells called BBB. Disruption of this barrier is associated with neurological ailments where it can be a cause or appear as a consequence of the disease [102]. Activation of NMDA receptor by the excitatory neurotransmitter, glutamate, disrupted BBB function and permeability in human brain capillary endothelial cells [103] whereas its inhibition by the antagonist, MK-801, prevented BBB breakdown in isolated rat brain capillary endothelial cells [104], suggesting neuronal over-excitation to cause BBB disruption. Several studies demonstrated BBB disruption to be related to seizure occurrence in humans and rodents [105109]. It was proposed that the resultant disruption leads to BBB leakage, further causing efflux of administered AEDs [110]. Since the brain as well as brain endothelial cells express several well-characterized ABC efflux transporters such as P-glycoprotein (P-gp), BCRP, MRP1, MRP4, and MRP5 [111], these are often associated with AED resistance attributed to BBB leakage in epilepsy [7]. Of these transporters, P-gp has been widely studied for causing multidrug resistance in epilepsy due to its upregulation during seizure. Several investigations revealed upregulation of P-gp in the brain and BBB due to seizure activity in rodent models [112117] as well as in human patients with refractory epilepsy [117121]. Rats with drug-resistant seizures were also shown to exhibit enhanced brain P-gp expression compared to those with drug-responsive seizures [122]. Consequently, the prescribed AEDs gets effluxed out into the circulation by the upregulated P-gp, even before reaching the drug target, thereby causing pharmacoresistance.
The underlying molecular mechanism behind the regulation of seizure-induced P-gp is currently being investigated in in vivo and in vitro model systems to achieve the goal of decreasing pharmacoresistance and establishing highly efficacious treatment for epilepsy. P-gp had been observed to be upregulated by glutamate-mediated NMDA receptor activation in brain capillary endothelial cells [123125]. This glutamate-mediated P-gp upregulation was prevented by exposure to selective COX-2 inhibitors such as celecoxib and NS-398 and non-selective inhibitor, indomethacin heptyl ester [124126], suggesting the involvement of COX-2 in regulating P-gp expression. Brain capillary endothelial cells from pilocarpine-induced SE rat model also showed high P-gp expression and activity which was blocked by treatment with COX-2 inhibitors, indomethacin and celecoxib [124, 126], revealing the role of COX-2 in seizure-induced P-gp upregulation. Similarly, administration of COX-2 inhibitors, SC-58236 and NS-398, both counteracted the SE-associated increase in P-gp expression in the parahippocampal cortex and the ventral hippocampus in rats [127]. Besides, blocking the EP1 receptor by the antagonist, SC-51089, in pilocarpine-induced SE rat model prevented seizure-induced P-gp upregulation [128]. These investigations indicated that activation of COX-2/EP receptor signaling during seizure is somehow causing upregulation of P-gp, thereby leading to decreased drug delivery to the brain and enhanced resistance to drugs. Direct inhibition of P-gp may improve seizure control; however, its pan inhibition may lead to deleterious effects [129]. Moreover, when a known P-gp inhibitor, verapamil, was administered to pediatric patients, children with drug-resistant epilepsy receiving AED polytherapy showed no significant difference in seizure control compared to those receiving placebo along with AED polytherapy [130]. Likewise, inhibition of multiple EP receptors simultaneously may produce adverse effects. This signifies the importance of targeting the upstream regulatory molecule, COX-2, for a potential future strategy for epilepsy treatment.

COX-2 inhibitors: therapeutic strategies

Non-steroidal anti-inflammatory drugs (NSAIDs), the currently available COX inhibitory drugs, prevent the formation of PGs by competitively inhibiting the activity of COX enzymes. NSAIDs are of two types: selective, which inhibit only COX-2 (e.g., celecoxib and rofecoxib), and non-selective, which inhibit both COX-1 and COX-2 (e.g., aspirin, ibuprofen, and indomethacin). These inhibitors work by varying degrees of reversible (example, ibuprofen and indomethacin) or irreversible (example, celecoxib, rofecoxib, and aspirin) competitive inhibition. NSAIDs help in mitigating different manifestations of allergic reactions and provide antipyretic, analgesic, and anti-platelet effects in acute and chronic conditions; however, they also have several undesirable effects. Due to the protective role of COX-1 in the maintenance of the stomach lining by preventing it from stomach acid, its inhibition sometimes causes gastric problems. In response to this challenge, new generational NSAIDs, specific to COX-2, were developed. However, the selective COX-2 inhibitors, coxibs, can increase the risk of adverse renal and cardiovascular events and thus require improvisation for better efficacy without any complications.

COX-2 inhibitors as anticonvulsants

To ascertain the anticonvulsive therapeutic potential of COX-2 inhibition, several studies investigated the effect of COX-2 inhibitors on seizure activity and development in animal models of epilepsy (Table 2). The findings differ with different seizure and treatment conditions. Dhir et al. 2006a examined the effect of selective COX-2 inhibitors, nimesulide and rofecoxib, administered 45 min prior to an epileptic challenge in different mice models [138]. They found that the inhibitors prolonged the mean onset time of convulsions and decreased the seizure duration and the percentage mortality rate against bicuculline- and picrotoxin-induced seizures; however, they did not affect maximal electroshock-induced seizures, suggesting varied efficacy of COX-2 inhibitors in different types of convulsive challenge. Another study by the same group revealed the two selective COX-2 inhibitors to be more efficacious than non-selective COX-2 inhibitors, aspirin and naproxen [140], in antagonizing the effect of PTZ-induced seizures. The authors also found that nimesulide provides a neuroprotective effect by controlling the biochemical alterations caused by PTZ-induced chemical kindling in mice [141]. Administration of 2 mg/kg and 4 mg/kg of rofecoxib increased the seizure threshold; however, a lower dose of 1 mg/kg of rofecoxib failed to do the same, indicating a dose-dependent effect [145]. Pretreatment of selective COX-2 inhibitor, celecoxib, 60 min prior to seizure induction also demonstrated anticonvulsant effects in the PTZ-induced rat model [84]. Conversely, several studies suggest that pretreatment of selective or non-selective COX-2 inhibitors may have an inverse effect by acting as proconvulsants [132134, 139, 151], and a postictal COX-2 inhibition would rather have a neuroprotective effect [132, 142, 144, 149, 150]. After PTZ challenge to male Wistar rats, the selective COX-2 inhibitor, etoricoxib, showed an anticonvulsant effect at a dose of 1 mg/kg which got reduced or reversed at 10 mg/kg of dose displaying the neuroprotective effect within a narrow therapeutic dose window [137].
Table 2
Preclinical evidences supporting or opposing clinical application of COX-2 inhibitors for epilepsy treatment
Selectivity
Drug
Type of convulsive challenge
Supporting evidences
Opposing evidences
Selective
Celecoxib
Electrical stimulation
[131]
[87]
Flurothyl
[96]
Kainic acid
[132]
[133, 134]
Pentylenetetrazol
[84]
Pilocarpine
[126, 135]
Etoricoxib
Genetic model
[136]
Pentylenetetrazol
[137]
Nimesulide
Bicuculline
[138]
Electrical stimulation
[6, 23]
Kainic acid
[134, 139]
Pentylenetetrazol
[140, 141]
Picrotoxin
[138]
NS-398
Kainic acid
[83]
[133]
Pilocarpine
[127, 142]
Parecoxib
Pilocarpine
[143]
Rofecoxib
Kainic acid
[144]
Pentylenetetrazol
[56, 140, 145]
[146]
SC-58125
Kainic acid
[82]
SC-58236
Electrical stimulation
[98, 147]
Pilocarpine
[127]
Non-selective
Aspirin
Electrical stimulation
[148]
Kainic acid
[133]
Pilocarpine
[149, 150]
[151]
Ibuprofen
Electrical stimulation
[148]
Indomethacin
Electrical stimulation
[148]
Kainic acid
[133, 151]
Others (metamizole, paracetamol, piroxicam, ketoprofen)
Electrical stimulation
[148]
Kainic acid
[134]
Celecoxib administration 1 day after pilocarpine-induced SE reduced the likelihood of developing SRS and prevented hippocampal neuronal damage in rats [135]. Early long-term treatment of another coxib drug, etoricoxib, also displayed antiepileptogenic effect by reducing the development of absence seizures in the genetic WAG/Rij rat model of absence epilepsy [136]. However, several reports contradict these findings. Administration of rofecoxib 5 days prior to epileptic challenge showed no effect on PTZ-kindling development [146]. A 3-day treatment with the selective COX-2 inhibitor, SC-58326, starting 1 day before electrically induced SE increased rat mortality in models of TLE [147] while a 7-day treatment starting 4 h after SE induction effectively reduced PGE2 production but did not prevent seizure development or neuronal damage [98]. Similarly, an 18-day administration of the selective COX-2 inhibitor, parecoxib, following pilocarpine-induced SE prevented the subsequent increase in PGE2 and reduced seizure severity in the rat hippocampus and piriform cortex; however, it could not prevent the development, frequency, and duration of seizures [143]. These studies demonstrate an anticonvulsive but not antiepileptogenic effect of COX-2 inhibitors. It is proposed that COX-2 inhibitors display this anticonvulsive activity by reducing the production of PGE2 causing decreased activation of EP receptors which, in turn, lowers calcium ion influx and release of the excitatory neurotransmitter, glutamate, thus blocking the seizures [152] (Fig. 2a). Simultaneously, COX-2 inhibitors suppress the production of pro-inflammatory cytokines reducing inflammation [7274].
In vivo studies reporting the effect of COX-2 inhibitors on seizure activity revealed that various factors determine the anticonvulsant action of these drugs viz., dosage, time of administration, treatment duration, type of convulsive challenge, and selectivity of COX-2 inhibitors. Based on these factors, COX-2 inhibitors, alike EP receptors, can produce dichotomous effects, neuroprotective or neurotoxic [153], suggesting the need to optimize their therapeutic dose, time, treatment duration, and other pharmacokinetic and pharmacodynamic properties. However, instead of targeting multiple EP receptors, targeting the upstream COX-2 enzyme would produce a broad-spectrum effect and therefore remains the key therapeutic target for epilepsy treatment.

COX-2 inhibitors as adjunctive to AED therapy

As discussed earlier, COX-2 may regulate the expression of the multidrug transporter, P-gp, which is found to be overexpressed in drug-resistant epilepsy. COX-2 upregulation following seizure produces higher levels of PGE2, which when bound to the EP1 receptor, activates a signaling cascade involving the transcription factor, NF-κB, leading to increased expression of P-gp [7]. Overexpression of P-gp at the BBB may result in enhanced the efflux of the prescribed AED/s even before reaching the target site in the brain, causing pharmacoresistance. Blocking seizure-mediated P-gp overexpression in the brain capillary endothelial cells, therefore, may facilitate better drug delivery to the brain and improve drug efficacy provided the prescribed AED is a substrate of P-gp. Several studies have investigated different AEDs as potential substrates of P-gp. Phenytoin, phenobarbital, lamotrigine, and levetiracetam have widely been observed to be weak but potential substrates of P-gp while carbamazepine, felbamate, and ethosuximide had no substrate interaction with the transporter [154]. Other drugs have shown either contradictory findings or remain insufficiently investigated in this regard.
Overexpression of P-gp at the BBB decreased brain uptake of phenytoin at specific limbic brain regions of chronic epileptic rats [155]. Its direct inhibition using three different P-gp inhibitors resulted in increased phenytoin concentration in brain extracellular fluid (ECF) in rats [156]. Treatment with selective P-gp inhibitor, tariquidar, improved seizure control in chronic epileptic rats due to increased phenytoin delivery to the brain [155, 157]. Despite these beneficial findings of direct P-gp inhibition, its pan inhibition may lead to deleterious effect [129], thus an alternative indirect approach is required for its suppression. Since COX-2 serves as a transcriptional regulator of P-gp, inhibiting COX-2 may assist in achieving enhanced efficacy of prescribed AEDs (Fig. 2b). In vitro data revealed prevention of glutamate-induced P-gp upregulation by selective and non-selective COX-2 inhibitors in rodent and human brain capillary endothelial cells [124126]. COX-2 inhibitors also blocked SE-induced P-gp upregulation in the brain capillaries of epileptic rats, revealing the role of COX-2 inhibitors in preventing seizure-induced P-gp upregulation [124, 126]. Furthermore, brain uptake of phenytoin was significantly enhanced by sub-chronic COX-2 inhibition via suppressing P-gp expression in chronic epileptic rats, suggesting that COX-2 inhibitors may help in increasing drug delivery to the target sites in the brain [127]. To relate this effect of COX-2 inhibitors with the drug efficacy, Schlichtiger et al. investigated the outcome of celecoxib treatment in phenobarbital-treated responder and non-responder epileptic rats [131]. A 6-day treatment with celecoxib significantly reduced P-gp expression as well as frequency of SRS in both the responders and non-responders suggesting the potential role of COX-2 inhibition in increasing AED efficacy via downregulating P-gp. Rofecoxib also potentiated the anticonvulsant activity of the AED, tiagabine, against PTZ-induced seizures in mice [56]. Non-selective NSAIDs such aspirin, ibuprofen, indomethacin, metamizole, paracetamol, and piroxicam also enhanced the anticonvulsive activity of valproate while the anticonvulsive activity of phenytoin was increased only by ibuprofen and piroxicam against maximal electroshock-induced seizures in mice [148]. Therefore, besides their anticonvulsive effect, COX-2 inhibitors also show a great promise towards being an adjunctive therapy for improving the efficacy of administered AEDs.

Human clinical studies

Despite decades of extensive research on the beneficial role of COX-2 inhibition in controlling seizure and drug-resistance in epilepsy, the selective COX-2 inhibitors have, so far, not been clinically tested in patients with epilepsy due to their severe adverse effects. However, a recent report by Lim et al. [158] investigated the effect of celecoxib on the neuronal excitability and electrophysiological properties of the brain of healthy volunteers. Though the authors found no effect of celecoxib on the neuronal excitability of the healthy volunteers, the study is limited due to the absence of inflammation in the healthy subjects to observe the effect of the anti-inflammatory drug, celecoxib. In regard to the non-selective COX-2 inhibitors, aspirin, having relatively fewer side effects, has been investigated independently or in adjunction to AED therapy for controlling seizures in epilepsy and related syndromes (Table 3) to substantiate the findings of preclinical studies reporting efficient seizure reduction upon administration of COX-2 inhibitors (Table 2). Most of these studies were performed on patients with the rare neurological disorder, Sturge-Weber syndrome, limiting the current knowledge on the applicability of these drugs for other seizure disorders or epilepsy. Long-term continuous use of aspirin in patients with Sturge-Weber syndrome resulted in seizure freedom for at least 1 year in five of six patients [160]. An internet-based survey involving patients with Sturge-Weber syndrome receiving aspirin reported seizure reduction in 21 of 34 patients [161], suggesting the use of low-dose aspirin to be safe and beneficial. The findings were further validated by Lance et al. who observed seizure control in 91% of the patients with the syndrome receiving low-dose aspirin with minimal side effects [162]. Patients with focal epilepsy receiving aspirin also showed significantly fewer seizures compared to age-, sex- and disease-matched controls not receiving aspirin [163]. They found an inverse correlation between aspirin doses and seizure frequency. However, a randomized, double-blind, placebo-controlled trial failed to demonstrate a preventive effect of another non-selective NSAID, ibuprofen, on the number of febrile seizure recurrences in 230 children at increased risk [159] (Table 3). Therefore, studies revealing the anticonvulsive effect of aspirin require replication in a large-sample, randomized, controlled trial to substantiate the effect and applicability of COX-2 inhibitors as a therapeutic approach in epilepsy management.
Table 3
Clinical evidences using non-selective NSAIDs in patients experiencing seizures
Reference
NSAID
Use
Study subjects
Effects
van Stuijvenberg et al. [159]
Ibuprofen
Independent
Randomized, double-blind, placebo-controlled study in 230 children with febrile seizures (111 on ibuprofen and 119 on placebo)
Failed to reduce number of seizure recurrences in children at increased risk
Udani et al. [160]
Aspirin
Not reported
9 children with Sturge-Weber syndrome (6 with long-term continuous aspirin therapy and 3 with intermittent use of aspirin)
Seizure freedom for at least one year in 8 of 9 children
Bay et al. [161]
Aspirin
In adjunction to AED therapy
Internet-based survey in 34 subjects with Sturge-Weber syndrome receiving aspirin
Seizure reduction in 21 of 34 patients
Lance et al. [162]
Aspirin
In adjunction to AED therapy
58 subjects with Sturge-Weber syndrome receiving aspirin
Seizure control in 91% of the patients
Godfred et al. [163]
Aspirin
Independent
46 subjects with focal epilepsy (23 receiving aspirin and 23 not receiving aspirin)
Fewer seizures in patients on aspirin therapy than patients not receiving it

Conclusion

Increasing evidences on the role of inflammation in epilepsy pathogenesis are encouraging researchers to gather information on the clinical use of neuroinflammation-targeted therapeutics and to develop their better, improved, efficacious analogues. COX-2, a proinflammatory enzyme interconnecting various inflammatory processes, is widely being investigated as a therapeutic target in epilepsy. Several preclinical and clinical studies demonstrated COX-2 induction during a seizure event and in epilepsy. Pharmacological inhibition of COX-2 enzyme using selective and non-selective COX-2 inhibitors not only resulted in reduced seizure recurrence and disease severity but also increased the efficacy of administered AEDs, suggesting their two probable modes of action, (1) anticonvulsive and (2) adjunctive to AED therapy. However, the efficacy of COX-2 inhibitors depends on various factors such as viz., therapeutic dose, time of administration, treatment duration, and their selectivity and is often accompanied by mild or severe adverse effects, thus prompting further investigations on improvising the efficacy and optimizing their use without any complications. In addition, future studies should also focus on investigating the anticonvulsive effect of COX-2 inhibitors in large-sample, randomized, controlled trials to substantiate the clinical application of COX-2 inhibitors as a future therapeutic strategy for epilepsy management.

Acknowledgements

We thank Dr. Anurag Agrawal, Director, Institute of Genomics and Integrative Biology (IGIB), for his motivation and unconditional support. CR and URD acknowledge University Grants Commission (UGC), and SK acknowledges Department of Biotechnology (DBT) for providing fellowships.
Not applicable.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55:475–82.PubMedCrossRef Fisher RS, Acevedo C, Arzimanoglou A, Bogacz A, Cross JH, Elger CE, et al. ILAE official report: a practical clinical definition of epilepsy. Epilepsia. 2014;55:475–82.PubMedCrossRef
2.
Zurück zum Zitat Laxer KD, Trinka E, Hirsch LJ, Cendes F, Langfitt J, Delanty N, et al. The consequences of refractory epilepsy and its treatment. Epilepsy Behav. 2014;37:59–70.PubMedCrossRef Laxer KD, Trinka E, Hirsch LJ, Cendes F, Langfitt J, Delanty N, et al. The consequences of refractory epilepsy and its treatment. Epilepsy Behav. 2014;37:59–70.PubMedCrossRef
3.
Zurück zum Zitat Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011;7:31–40.PubMedCrossRef Vezzani A, French J, Bartfai T, Baram TZ. The role of inflammation in epilepsy. Nat Rev Neurol. 2011;7:31–40.PubMedCrossRef
4.
Zurück zum Zitat Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96:70–82.PubMedCrossRef Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96:70–82.PubMedCrossRef
5.
Zurück zum Zitat 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;37:463–84.PubMedPubMedCentralCrossRef 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;37:463–84.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Takemiya T, Suzuki K, Sugiura H, Yasuda S, Yamagata K, Kawakami Y, et al. Inducible brain COX-2 facilitates the recurrence of hippocampal seizures in mouse rapid kindling. Prostaglandins Other Lipid Mediat. 2003;71:205–16.PubMedCrossRef Takemiya T, Suzuki K, Sugiura H, Yasuda S, Yamagata K, Kawakami Y, et al. Inducible brain COX-2 facilitates the recurrence of hippocampal seizures in mouse rapid kindling. Prostaglandins Other Lipid Mediat. 2003;71:205–16.PubMedCrossRef
7.
Zurück zum Zitat Potschka H. Modulating P-glycoprotein regulation: future perspectives for pharmacoresistant epilepsies? Epilepsia. 2010;51:1333–47.PubMedCrossRef Potschka H. Modulating P-glycoprotein regulation: future perspectives for pharmacoresistant epilepsies? Epilepsia. 2010;51:1333–47.PubMedCrossRef
8.
Zurück zum Zitat Sheng JG, Boop FA, Mrak RE, Griffin WS. Increased neuronal β-amyloid precursor protein expression in human temporal lobe epilepsy: association with interleukin-1α immunoreactivity. J Neurochem. 1994;63:1872–9.PubMedPubMedCentralCrossRef Sheng JG, Boop FA, Mrak RE, Griffin WS. Increased neuronal β-amyloid precursor protein expression in human temporal lobe epilepsy: association with interleukin-1α immunoreactivity. J Neurochem. 1994;63:1872–9.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Kanemoto K, Kawasaki J, Miyamoto T, Obayashi H, Nishimura M. Interleukin (IL)1beta, IL-1alpha, and IL-1 receptor antagonist gene polymorphisms in patients with temporal lobe epilepsy. Ann Neurol. 2000;47:571–4.PubMedCrossRef Kanemoto K, Kawasaki J, Miyamoto T, Obayashi H, Nishimura M. Interleukin (IL)1beta, IL-1alpha, and IL-1 receptor antagonist gene polymorphisms in patients with temporal lobe epilepsy. Ann Neurol. 2000;47:571–4.PubMedCrossRef
10.
Zurück zum Zitat Ichiyama T, Nishikawa M, Yoshitomi T, Hayashi T, Furukawa S. Tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 in cerebrospinal fluid from children with prolonged febrile seizures. Comparison with acute encephalitis/encephalopathy. Neurology. 1998;50:407–11.PubMedCrossRef Ichiyama T, Nishikawa M, Yoshitomi T, Hayashi T, Furukawa S. Tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 in cerebrospinal fluid from children with prolonged febrile seizures. Comparison with acute encephalitis/encephalopathy. Neurology. 1998;50:407–11.PubMedCrossRef
11.
Zurück zum Zitat Shi LM, Chen RJ, Zhang H, Jiang CM, Gong J. Cerebrospinal fluid neuron specific enolase, interleukin-1β and erythropoietin concentrations in children after seizures. Childs Nerv Syst. 2017;33:805–11.PubMedCrossRef Shi LM, Chen RJ, Zhang H, Jiang CM, Gong J. Cerebrospinal fluid neuron specific enolase, interleukin-1β and erythropoietin concentrations in children after seizures. Childs Nerv Syst. 2017;33:805–11.PubMedCrossRef
12.
Zurück zum Zitat Uludag IF, Duksal T, Tiftikcioglu BI, Zorlu Y, Ozkaya F, Kirkali G. IL-1β, IL-6 and IL1Ra levels in temporal lobe epilepsy. Seizure. 2015;26:22–5.PubMedCrossRef Uludag IF, Duksal T, Tiftikcioglu BI, Zorlu Y, Ozkaya F, Kirkali G. IL-1β, IL-6 and IL1Ra levels in temporal lobe epilepsy. Seizure. 2015;26:22–5.PubMedCrossRef
13.
Zurück zum Zitat Wang Y, Wang D, Guo D. Interictal cytokine levels were correlated to seizure severity of epileptic patients: a retrospective study on 1218 epileptic patients. J Transl Med. 2015;13:378.PubMedPubMedCentralCrossRef Wang Y, Wang D, Guo D. Interictal cytokine levels were correlated to seizure severity of epileptic patients: a retrospective study on 1218 epileptic patients. J Transl Med. 2015;13:378.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Lorigados Pedre L, Morales Chacón LM, Pavón Fuentes N, Robinson Agramonte MLA, Serrano Sánchez T, Cruz-Xenes RM, et al. Follow-up of peripheral IL-1β and IL-6 and relation with apoptotic death in drug-resistant temporal lobe epilepsy patients submitted to surgery. Behav Sci. 2018;8:21. Lorigados Pedre L, Morales Chacón LM, Pavón Fuentes N, Robinson Agramonte MLA, Serrano Sánchez T, Cruz-Xenes RM, et al. Follow-up of peripheral IL-1β and IL-6 and relation with apoptotic death in drug-resistant temporal lobe epilepsy patients submitted to surgery. Behav Sci. 2018;8:21.
15.
Zurück zum Zitat Lee TS, Mane S, Eid T, Zhao H, Lin A, Guan Z, et al. Gene expression in temporal lobe epilepsy is consistent with increased release of glutamate by astrocytes. Mol Med. 2007;13:1–13.PubMedPubMedCentralCrossRef Lee TS, Mane S, Eid T, Zhao H, Lin A, Guan Z, et al. Gene expression in temporal lobe epilepsy is consistent with increased release of glutamate by astrocytes. Mol Med. 2007;13:1–13.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat Xu Y, Zeng K, Han Y, Wang L, Chen D, Xi Z, et al. Altered expression of CX3CL1 in patients with epilepsy and in a rat model. Am J Pathol. 2012;180:1950–62.PubMedCrossRef Xu Y, Zeng K, Han Y, Wang L, Chen D, Xi Z, et al. Altered expression of CX3CL1 in patients with epilepsy and in a rat model. Am J Pathol. 2012;180:1950–62.PubMedCrossRef
17.
Zurück zum Zitat Li R, Ma L, Huang H, Ou S, Yuan J, Xu T, et al. Altered expression of CXCL13 and CXCR5 in intractable temporal lobe epilepsy patients and pilocarpine-induced epileptic rats. Neurochem Res. 2017;42:526–40.PubMedCrossRef Li R, Ma L, Huang H, Ou S, Yuan J, Xu T, et al. Altered expression of CXCL13 and CXCR5 in intractable temporal lobe epilepsy patients and pilocarpine-induced epileptic rats. Neurochem Res. 2017;42:526–40.PubMedCrossRef
18.
Zurück zum Zitat Crespel A, Coubes P, Rousset MC, Brana C, Rougier A, Rondouin G, et al. Inflammatory reactions in human medial temporal lobe epilepsy with hippocampal sclerosis. Brain Res. 2002;952:159–69.PubMedCrossRef Crespel A, Coubes P, Rousset MC, Brana C, Rougier A, Rondouin G, et al. Inflammatory reactions in human medial temporal lobe epilepsy with hippocampal sclerosis. Brain Res. 2002;952:159–69.PubMedCrossRef
19.
Zurück zum Zitat Das A, Wallace GC 4th, Holmes C, McDowell ML, Smith JA, Marshall JD, et al. Hippocampal tissue of patients with refractory temporal lobe epilepsy is associated with astrocyte activation, inflammation, and altered expression of channels and receptors. Neuroscience. 2012;220:237–46.PubMedPubMedCentralCrossRef Das A, Wallace GC 4th, Holmes C, McDowell ML, Smith JA, Marshall JD, et al. Hippocampal tissue of patients with refractory temporal lobe epilepsy is associated with astrocyte activation, inflammation, and altered expression of channels and receptors. Neuroscience. 2012;220:237–46.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat De Simoni MG, Perego C, Ravizza T, Moneta D, Conti M, Marchesi F, et al. Inflammatory cytokines and related genes are induced in the rat hippocampus by limbic status epilepticus. Eur J Neurosci. 2000;12:2623–33.PubMedCrossRef De Simoni MG, Perego C, Ravizza T, Moneta D, Conti M, Marchesi F, et al. Inflammatory cytokines and related genes are induced in the rat hippocampus by limbic status epilepticus. Eur J Neurosci. 2000;12:2623–33.PubMedCrossRef
21.
Zurück zum Zitat Xu JH, Long L, Tang YC, Zhang JT, Hut HT, Tang FR. CCR3, CCR2A and macrophage inflammatory protein (MIP)-1a, monocyte chemotactic protein-1 (MCP-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (PISE). Neuropathol Appl Neurobiol. 2009;35:496–514.PubMedCrossRef Xu JH, Long L, Tang YC, Zhang JT, Hut HT, Tang FR. CCR3, CCR2A and macrophage inflammatory protein (MIP)-1a, monocyte chemotactic protein-1 (MCP-1) in the mouse hippocampus during and after pilocarpine-induced status epilepticus (PISE). Neuropathol Appl Neurobiol. 2009;35:496–514.PubMedCrossRef
22.
Zurück zum Zitat Gorter JA, van Vliet EA, Aronica E, Breit T, Rauwerda H, Lopes da Silva FH, et al. Potential new antiepileptogenic targets indicated by microarray analysis in a rat model for temporal lobe epilepsy. J Neurosci 2006;26(43):11083–11110. Gorter JA, van Vliet EA, Aronica E, Breit T, Rauwerda H, Lopes da Silva FH, et al. Potential new antiepileptogenic targets indicated by microarray analysis in a rat model for temporal lobe epilepsy. J Neurosci 2006;26(43):11083–11110.
23.
Zurück zum Zitat Tu B, Bazan NG. Hippocampal kindling epileptogenesis upregulates neuronal cyclooxygenase-2 expression in neocortex. Exp Neurol. 2003;179:167–75.PubMedCrossRef Tu B, Bazan NG. Hippocampal kindling epileptogenesis upregulates neuronal cyclooxygenase-2 expression in neocortex. Exp Neurol. 2003;179:167–75.PubMedCrossRef
24.
Zurück zum Zitat Yoshikawa K, Kita Y, Kishimoto K, Shimizu T. Profiling of eicosanoid production in the rat hippocampus during kainic acid-induced seizure: dual phase regulation and differential involvement of COX-1 and COX-2. J Biol Chem. 2006;281:14663–9.PubMedCrossRef Yoshikawa K, Kita Y, Kishimoto K, Shimizu T. Profiling of eicosanoid production in the rat hippocampus during kainic acid-induced seizure: dual phase regulation and differential involvement of COX-1 and COX-2. J Biol Chem. 2006;281:14663–9.PubMedCrossRef
25.
Zurück zum Zitat Auvin S, Shin D, Mazarati A, Nakagawa J, Miyamoto J, Sankar R. Inflammation exacerbates seizure-induced injury in the immature brain. Epilepsia. 2007;48(Suppl 5):27–34.PubMedCrossRef Auvin S, Shin D, Mazarati A, Nakagawa J, Miyamoto J, Sankar R. Inflammation exacerbates seizure-induced injury in the immature brain. Epilepsia. 2007;48(Suppl 5):27–34.PubMedCrossRef
26.
Zurück zum Zitat Auvin S, Shin D, Mazarati A, Sankar R. Inflammation induced by LPS enhances epileptogenesis in immature rat and may be partially reversed by IL1RA. Epilepsia. 2010;51(Suppl 3):34–8.PubMedPubMedCentralCrossRef Auvin S, Shin D, Mazarati A, Sankar R. Inflammation induced by LPS enhances epileptogenesis in immature rat and may be partially reversed by IL1RA. Epilepsia. 2010;51(Suppl 3):34–8.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Dube C, Vezzani A, Behrens M, Bartfai T, Baram TZ. Interleukin-1beta contributes to the generation of experimental febrile seizures. Ann Neurol. 2005;57:152–5.PubMedPubMedCentralCrossRef Dube C, Vezzani A, Behrens M, Bartfai T, Baram TZ. Interleukin-1beta contributes to the generation of experimental febrile seizures. Ann Neurol. 2005;57:152–5.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Cacheaux LP, Ivens S, David Y, Lakhter AJ, Bar-Klein G, Shapira M, et al. Transcriptome profiling reveals TGF-beta signaling involvement in epileptogenesis. J Neurosci. 2009;29:8927–35.PubMedPubMedCentralCrossRef Cacheaux LP, Ivens S, David Y, Lakhter AJ, Bar-Klein G, Shapira M, et al. Transcriptome profiling reveals TGF-beta signaling involvement in epileptogenesis. J Neurosci. 2009;29:8927–35.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Kovacs R, Rabanus A, Otahal J, Patzak A, Kardos J, Albus K, et al. Endogenous nitric oxide is a key promoting factor for initiation of seizure-like events in hippocampal and entorhinal cortex slices. J Neurosci. 2009;29:8565–77.PubMedPubMedCentralCrossRef Kovacs R, Rabanus A, Otahal J, Patzak A, Kardos J, Albus K, et al. Endogenous nitric oxide is a key promoting factor for initiation of seizure-like events in hippocampal and entorhinal cortex slices. J Neurosci. 2009;29:8565–77.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, Iyer AM, et al. Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med. 2010;16:413–9.PubMedCrossRef Maroso M, Balosso S, Ravizza T, Liu J, Aronica E, Iyer AM, et al. Toll-like receptor 4 and high-mobility group box-1 are involved in ictogenesis and can be targeted to reduce seizures. Nat Med. 2010;16:413–9.PubMedCrossRef
31.
Zurück zum Zitat Pernhorst K, Herms S, Hoffmann P, Cichon S, Schulz H, Sander T, et al. TLR4, ATF-3 and IL8 inflammation mediator expression correlates with seizure frequency in human epileptic brain tissue. Seizure. 2013;22:675–8.PubMedCrossRef Pernhorst K, Herms S, Hoffmann P, Cichon S, Schulz H, Sander T, et al. TLR4, ATF-3 and IL8 inflammation mediator expression correlates with seizure frequency in human epileptic brain tissue. Seizure. 2013;22:675–8.PubMedCrossRef
32.
Zurück zum Zitat Brodie MJ, Sills GJ. Combining antiepileptic drugs--rational polytherapy? Seizure. 2011;20:369–75.PubMedCrossRef Brodie MJ, Sills GJ. Combining antiepileptic drugs--rational polytherapy? Seizure. 2011;20:369–75.PubMedCrossRef
34.
Zurück zum Zitat Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A, et al. The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci U S A. 2004;101:9861–6.PubMedPubMedCentralCrossRef Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A, et al. The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci U S A. 2004;101:9861–6.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Loscher W, Klitgaard H, Twyman RE, Schmidt D. New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov. 2013;12:757–76.PubMedCrossRef Loscher W, Klitgaard H, Twyman RE, Schmidt D. New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov. 2013;12:757–76.PubMedCrossRef
36.
Zurück zum Zitat Schmidt D, Loscher W. New developments in antiepileptic drug resistance: an integrative view. Epilepsy Curr. 2009;9:47–52.PubMedCrossRef Schmidt D, Loscher W. New developments in antiepileptic drug resistance: an integrative view. Epilepsy Curr. 2009;9:47–52.PubMedCrossRef
37.
Zurück zum Zitat Tate SK, Depondt C, Sisodiya SM, Cavalleri GL, Schorge S, Soranzo N, et al. Genetic predictors of the maximum doses patients receive during clinical use of the anti-epileptic drugs carbamazepine and phenytoin. Proc Natl Acad Sci U S A. 2005;102:5507–12.PubMedPubMedCentralCrossRef Tate SK, Depondt C, Sisodiya SM, Cavalleri GL, Schorge S, Soranzo N, et al. Genetic predictors of the maximum doses patients receive during clinical use of the anti-epileptic drugs carbamazepine and phenytoin. Proc Natl Acad Sci U S A. 2005;102:5507–12.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Abe T, Seo T, Ishitsu T, Nakagawa T, Hori M, Nakagawa K. Association between SCN1A polymorphism and carbamazepine-resistant epilepsy. Br J Clin Pharmacol. 2008;66:304–7.PubMedPubMedCentralCrossRef Abe T, Seo T, Ishitsu T, Nakagawa T, Hori M, Nakagawa K. Association between SCN1A polymorphism and carbamazepine-resistant epilepsy. Br J Clin Pharmacol. 2008;66:304–7.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Kumari R, Lakhan R, Kalita J, Misra UK, Mittal B. Association of alpha subunit of GABAA receptor subtype gene polymorphisms with epilepsy susceptibility and drug resistance in north Indian population. Seizure. 2010;19:237–41.PubMedCrossRef Kumari R, Lakhan R, Kalita J, Misra UK, Mittal B. Association of alpha subunit of GABAA receptor subtype gene polymorphisms with epilepsy susceptibility and drug resistance in north Indian population. Seizure. 2010;19:237–41.PubMedCrossRef
40.
Zurück zum Zitat Kwan P, Poon WS, Ng HK, Kang DE, Wong V, Ng PW, et al. Multidrug resistance in epilepsy and polymorphisms in the voltage-gated sodium channel genes SCN1A, SCN2A, and SCN3A: correlation among phenotype, genotype, and mRNA expression. Pharmacogenet Genomics. 2008;18:989–98.PubMedCrossRef Kwan P, Poon WS, Ng HK, Kang DE, Wong V, Ng PW, et al. Multidrug resistance in epilepsy and polymorphisms in the voltage-gated sodium channel genes SCN1A, SCN2A, and SCN3A: correlation among phenotype, genotype, and mRNA expression. Pharmacogenet Genomics. 2008;18:989–98.PubMedCrossRef
41.
Zurück zum Zitat Kwan P, Brodie MJ. Potential role of drug transporters in the pathogenesis of medically intractable epilepsy. Epilepsia. 2005;46:224–35.PubMedCrossRef Kwan P, Brodie MJ. Potential role of drug transporters in the pathogenesis of medically intractable epilepsy. Epilepsia. 2005;46:224–35.PubMedCrossRef
42.
Zurück zum Zitat Dombrowski SM, Desai SY, Marroni M, Cucullo L, Goodrich K, Bingaman W, et al. Overexpression of multiple drug resistance genes in endothelial cells from patients with refractory epilepsy. Epilepsia. 2001;42:1501–6.PubMedCrossRef Dombrowski SM, Desai SY, Marroni M, Cucullo L, Goodrich K, Bingaman W, et al. Overexpression of multiple drug resistance genes in endothelial cells from patients with refractory epilepsy. Epilepsia. 2001;42:1501–6.PubMedCrossRef
43.
Zurück zum Zitat Banerjee Dixit A, Sharma D, Srivastava A, Banerjee J, Tripathi M, Prakash D, et al. Upregulation of breast cancer resistance protein and major vault protein in drug resistant epilepsy. Seizure. 2017;47:9–12.PubMedCrossRef Banerjee Dixit A, Sharma D, Srivastava A, Banerjee J, Tripathi M, Prakash D, et al. Upregulation of breast cancer resistance protein and major vault protein in drug resistant epilepsy. Seizure. 2017;47:9–12.PubMedCrossRef
44.
Zurück zum Zitat Kubota H, Ishihara H, Langmann T, Schmitz G, Stieger B, Wieser HG, et al. Distribution and functional activity of P-glycoprotein and multidrug resistance-associated proteins in human brain microvascular endothelial cells in hippocampal sclerosis. Epilepsy Res. 2006;68:213–28.PubMedCrossRef Kubota H, Ishihara H, Langmann T, Schmitz G, Stieger B, Wieser HG, et al. Distribution and functional activity of P-glycoprotein and multidrug resistance-associated proteins in human brain microvascular endothelial cells in hippocampal sclerosis. Epilepsy Res. 2006;68:213–28.PubMedCrossRef
45.
Zurück zum Zitat Rogawski MA. The intrinsic severity hypothesis of pharmacoresistance to antiepileptic drugs. Epilepsia. 2013;54(Suppl 2):33–40.PubMedCrossRef Rogawski MA. The intrinsic severity hypothesis of pharmacoresistance to antiepileptic drugs. Epilepsia. 2013;54(Suppl 2):33–40.PubMedCrossRef
46.
Zurück zum Zitat Kwan P, Brodie MJ. Early identification of refractory epilepsy. N Engl J Med. 2000;342:314–9.PubMedCrossRef Kwan P, Brodie MJ. Early identification of refractory epilepsy. N Engl J Med. 2000;342:314–9.PubMedCrossRef
47.
Zurück zum Zitat Rawat C, Guin D, Talwar P, Grover S, Baghel R, Kushwaha S, et al. Clinical predictors of treatment outcome in North Indian patients on antiepileptic drug therapy: a prospective observational study. Neurol India. 2018;66:1052–9.PubMedCrossRef Rawat C, Guin D, Talwar P, Grover S, Baghel R, Kushwaha S, et al. Clinical predictors of treatment outcome in North Indian patients on antiepileptic drug therapy: a prospective observational study. Neurol India. 2018;66:1052–9.PubMedCrossRef
48.
Zurück zum Zitat Kobow K, El-Osta A, Blumcke I. The methylation hypothesis of pharmacoresistance in epilepsy. Epilepsia. 2013;54(Suppl 2):41–7.PubMedCrossRef Kobow K, El-Osta A, Blumcke I. The methylation hypothesis of pharmacoresistance in epilepsy. Epilepsia. 2013;54(Suppl 2):41–7.PubMedCrossRef
49.
Zurück zum Zitat Kim JE, Choi HC, Song HK, Jo SM, Kim DS, Choi SY, et al. Levetiracetam inhibits interleukin-1 beta inflammatory responses in the hippocampus and piriform cortex of epileptic rats. Neurosci Lett. 2010;471:94–9.PubMedCrossRef Kim JE, Choi HC, Song HK, Jo SM, Kim DS, Choi SY, et al. Levetiracetam inhibits interleukin-1 beta inflammatory responses in the hippocampus and piriform cortex of epileptic rats. Neurosci Lett. 2010;471:94–9.PubMedCrossRef
50.
Zurück zum Zitat Gomez CD, Buijs RM, Sitges M. The anti-seizure drugs vinpocetine and carbamazepine, but not valproic acid, reduce inflammatory IL-1β and TNF-α expression in rat hippocampus. J Neurochem. 2014;130:770–9.PubMedCrossRef Gomez CD, Buijs RM, Sitges M. The anti-seizure drugs vinpocetine and carbamazepine, but not valproic acid, reduce inflammatory IL-1β and TNF-α expression in rat hippocampus. J Neurochem. 2014;130:770–9.PubMedCrossRef
51.
Zurück zum Zitat Verrotti A, Basciani F, Trotta D, Greco R, Morgese G, Chiarelli F. Effect of anticonvulsant drugs on interleukins-1, -2 and -6 and monocyte chemoattractant protein-1. Clin Exp Med. 2001;1:133–6.PubMedCrossRef Verrotti A, Basciani F, Trotta D, Greco R, Morgese G, Chiarelli F. Effect of anticonvulsant drugs on interleukins-1, -2 and -6 and monocyte chemoattractant protein-1. Clin Exp Med. 2001;1:133–6.PubMedCrossRef
52.
Zurück zum Zitat Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, et al. Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. Proc Natl Acad Sci U S A. 2000;97:11534–9.PubMedPubMedCentralCrossRef Vezzani A, Moneta D, Conti M, Richichi C, Ravizza T, De Luigi A, et al. Powerful anticonvulsant action of IL-1 receptor antagonist on intracerebral injection and astrocytic overexpression in mice. Proc Natl Acad Sci U S A. 2000;97:11534–9.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Ravizza T, Lucas SM, Balosso S, Bernardino L, Ku G, Noé F, et al. Inactivation of caspase-1 in rodent brain: a novel anticonvulsive strategy. Epilepsia. 2006;47:1160–8.PubMedCrossRef Ravizza T, Lucas SM, Balosso S, Bernardino L, Ku G, Noé F, et al. Inactivation of caspase-1 in rodent brain: a novel anticonvulsive strategy. Epilepsia. 2006;47:1160–8.PubMedCrossRef
54.
Zurück zum Zitat Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, et al. Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis. 2017;99:12–23.PubMedCrossRef Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, et al. Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis. 2017;99:12–23.PubMedCrossRef
55.
Zurück zum Zitat Jiang J, Ganesh T, Du Y, Quan Y, Serrano G, Qui M, et al. Small molecule antagonist reveals seizure-induced mediation of neuronal injury by prostaglandin E2 receptor subtype EP2. Proc Natl Acad Sci U S A. 2012;109:3149–54.PubMedPubMedCentralCrossRef Jiang J, Ganesh T, Du Y, Quan Y, Serrano G, Qui M, et al. Small molecule antagonist reveals seizure-induced mediation of neuronal injury by prostaglandin E2 receptor subtype EP2. Proc Natl Acad Sci U S A. 2012;109:3149–54.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Dhir A, Kulkarni SK. Rofecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor potentiates the anticonvulsant activity of tiagabine against pentylenetetrazol-induced convulsions in mice. Inflammopharmacology. 2006;14:222–5.PubMedCrossRef Dhir A, Kulkarni SK. Rofecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor potentiates the anticonvulsant activity of tiagabine against pentylenetetrazol-induced convulsions in mice. Inflammopharmacology. 2006;14:222–5.PubMedCrossRef
57.
Zurück zum Zitat Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13(1):57–65.PubMedCrossRef Maher RL, Hanlon J, Hajjar ER. Clinical consequences of polypharmacy in elderly. Expert Opin Drug Saf. 2014;13(1):57–65.PubMedCrossRef
58.
Zurück zum Zitat Molina-Holgado E, Ortiz S, Molina-Holgado F, Guaza C. Induction of COX-2 and PGE(2) biosynthesis by IL-1beta is mediated by PKC and mitogen-activated protein kinases in murine astrocytes. Br J Pharmacol. 2000;131:152–9.PubMedPubMedCentralCrossRef Molina-Holgado E, Ortiz S, Molina-Holgado F, Guaza C. Induction of COX-2 and PGE(2) biosynthesis by IL-1beta is mediated by PKC and mitogen-activated protein kinases in murine astrocytes. Br J Pharmacol. 2000;131:152–9.PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat Hoozemans JJ, Veerhuis R, Janssen I, Rozemuller AJ, Eikelenboom P. Interleukin-1beta induced cyclooxygenase 2 expression and prostaglandin E2 secretion by human neuroblastoma cells: implications for Alzheimer’s disease. Exp Gerontol. 2001;36:559–70.PubMedCrossRef Hoozemans JJ, Veerhuis R, Janssen I, Rozemuller AJ, Eikelenboom P. Interleukin-1beta induced cyclooxygenase 2 expression and prostaglandin E2 secretion by human neuroblastoma cells: implications for Alzheimer’s disease. Exp Gerontol. 2001;36:559–70.PubMedCrossRef
60.
Zurück zum Zitat Leclerc P, Wahamaa H, Idborg H, Jakobsson PJ, Harris HE, Korotkova M. IL-1β/HMGB1 complexes promote the PGE2 biosynthesis pathway in synovial fibroblasts. Exp Gerontol Scand J Immunol. 2013;77:350–60.CrossRef Leclerc P, Wahamaa H, Idborg H, Jakobsson PJ, Harris HE, Korotkova M. IL-1β/HMGB1 complexes promote the PGE2 biosynthesis pathway in synovial fibroblasts. Exp Gerontol Scand J Immunol. 2013;77:350–60.CrossRef
61.
Zurück zum Zitat Mark KS, Trickler WJ, Miller DW. Tumor necrosis factor-alpha induces cyclooxygenase-2 expression and prostaglandin release in brain microvessel endothelial cells. J Pharmacol Exp Ther. 2001;297:1051–8.PubMed Mark KS, Trickler WJ, Miller DW. Tumor necrosis factor-alpha induces cyclooxygenase-2 expression and prostaglandin release in brain microvessel endothelial cells. J Pharmacol Exp Ther. 2001;297:1051–8.PubMed
62.
Zurück zum Zitat Pettus BJ, Bielawski J, Porcelli AM, Reames DL, Johnson KR, Morrow J, et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. 2003;17:1411–21.PubMedCrossRef Pettus BJ, Bielawski J, Porcelli AM, Reames DL, Johnson KR, Morrow J, et al. The sphingosine kinase 1/sphingosine-1-phosphate pathway mediates COX-2 induction and PGE2 production in response to TNF-alpha. FASEB J. 2003;17:1411–21.PubMedCrossRef
63.
Zurück zum Zitat Lin CC, Hsiao LD, Chien CS, Lee CW, Hsieh JT, Yang CM. Tumor necrosis factor-alpha-induced cyclooxygenase-2 expression in human tracheal smooth muscle cells: involvement of p42/p44 and p38 mitogen-activated protein kinases and nuclear factor-kappaB. Cell Signal. 2004;16:597–607.PubMedCrossRef Lin CC, Hsiao LD, Chien CS, Lee CW, Hsieh JT, Yang CM. Tumor necrosis factor-alpha-induced cyclooxygenase-2 expression in human tracheal smooth muscle cells: involvement of p42/p44 and p38 mitogen-activated protein kinases and nuclear factor-kappaB. Cell Signal. 2004;16:597–607.PubMedCrossRef
64.
Zurück zum Zitat Ogata S, Kubota Y, Yamashiro T, Takeuchi H, Ninomiya T, Suyama Y, et al. Signaling pathways regulating IL-1alpha-induced COX-2 expression. J Dent Res. 2007;86:186–91.PubMedCrossRef Ogata S, Kubota Y, Yamashiro T, Takeuchi H, Ninomiya T, Suyama Y, et al. Signaling pathways regulating IL-1alpha-induced COX-2 expression. J Dent Res. 2007;86:186–91.PubMedCrossRef
65.
Zurück zum Zitat Cao DL, Zhang ZJ, Xie RG, Jiang BC, Ji RR, Gao YJ. Chemokine CXCL1 enhances inflammatory pain and increases NMDA receptor activity and COX-2 expression in spinal cord neurons via activation of CXCR2. Exp Neurol. 2014;261:328–36.PubMedCrossRef Cao DL, Zhang ZJ, Xie RG, Jiang BC, Ji RR, Gao YJ. Chemokine CXCL1 enhances inflammatory pain and increases NMDA receptor activity and COX-2 expression in spinal cord neurons via activation of CXCR2. Exp Neurol. 2014;261:328–36.PubMedCrossRef
66.
Zurück zum Zitat Lin PS, Cheng RH, Chang MC, Lee JJ, Chang HH, Huang WL, et al. TGF-β1 stimulates cyclooxygenase-2 expression and PGE2 production of human dental pulp cells: role of ALK5/Smad2 and MEK/ERK signal transduction pathways. J Formos Med Assoc. 2017;116:748–54.PubMedCrossRef Lin PS, Cheng RH, Chang MC, Lee JJ, Chang HH, Huang WL, et al. TGF-β1 stimulates cyclooxygenase-2 expression and PGE2 production of human dental pulp cells: role of ALK5/Smad2 and MEK/ERK signal transduction pathways. J Formos Med Assoc. 2017;116:748–54.PubMedCrossRef
67.
Zurück zum Zitat Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, et al. Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: role in proliferation and apoptosis in the intestine. Gastroenterology. 2006;131:862–77.PubMedPubMedCentralCrossRef Fukata M, Chen A, Klepper A, Krishnareddy S, Vamadevan AS, Thomas LS, et al. Cox-2 is regulated by Toll-like receptor-4 (TLR4) signaling: role in proliferation and apoptosis in the intestine. Gastroenterology. 2006;131:862–77.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Porreca E, Reale M, Di Febbo C, Di Gioacchino M, Barbacane RC, Castellani ML, et al. Down-regulation of cyclooxygenase-2 (COX-2) by interleukin-1 receptor antagonist in human monocytes. Immunology. 1996;89:424–9.PubMedPubMedCentralCrossRef Porreca E, Reale M, Di Febbo C, Di Gioacchino M, Barbacane RC, Castellani ML, et al. Down-regulation of cyclooxygenase-2 (COX-2) by interleukin-1 receptor antagonist in human monocytes. Immunology. 1996;89:424–9.PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat Berg DJ, Zhang J, Lauricella DM, Moore SA. Il-10 is a central regulator of cyclooxygenase-2 expression and prostaglandin production. J Immunol. 2001;166:2674–80.PubMedCrossRef Berg DJ, Zhang J, Lauricella DM, Moore SA. Il-10 is a central regulator of cyclooxygenase-2 expression and prostaglandin production. J Immunol. 2001;166:2674–80.PubMedCrossRef
70.
Zurück zum Zitat Teloni R, Giannoni F, Rossi P, Nisini R, Gagliardi MC. Interleukin-4 inhibits cyclo-oxygenase-2 expression and prostaglandin E production by human mature dendritic cells. Immunology. 2007;120:83–9.PubMedPubMedCentralCrossRef Teloni R, Giannoni F, Rossi P, Nisini R, Gagliardi MC. Interleukin-4 inhibits cyclo-oxygenase-2 expression and prostaglandin E production by human mature dendritic cells. Immunology. 2007;120:83–9.PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Keshavarzi Z, Khaksari M, Razmi Z, Soltani Hekmat A, Naderi V, Rostami S. The effects of cyclooxygenase inhibitors on the brain inflammatory response following traumatic brain injury in rats. Iran J Basic Med Sci. 2012;15:1102–5.PubMedPubMedCentral Keshavarzi Z, Khaksari M, Razmi Z, Soltani Hekmat A, Naderi V, Rostami S. The effects of cyclooxygenase inhibitors on the brain inflammatory response following traumatic brain injury in rats. Iran J Basic Med Sci. 2012;15:1102–5.PubMedPubMedCentral
72.
Zurück zum Zitat Anderson GD, Hauser SD, McGarity KL, Bremer ME, Isakson PC, Gregory SA. Selective inhibition of cyclooxygenase (COX)-2 reverses inflammation and expression of COX-2 and interleukin 6 in rat adjuvant arthritis. J Clin Invest. 1996;97:2672–9.PubMedPubMedCentralCrossRef Anderson GD, Hauser SD, McGarity KL, Bremer ME, Isakson PC, Gregory SA. Selective inhibition of cyclooxygenase (COX)-2 reverses inflammation and expression of COX-2 and interleukin 6 in rat adjuvant arthritis. J Clin Invest. 1996;97:2672–9.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Kawashima M, Ogura N, Akutsu M, Ito K, Kondoh T. The anti-inflammatory effect of cyclooxygenase inhibitors in fibroblast-like synoviocytes from the human temporomandibular joint results from the suppression of PGE2 production. J Oral Pathol Med. 2013;42:499–506.PubMedPubMedCentralCrossRef Kawashima M, Ogura N, Akutsu M, Ito K, Kondoh T. The anti-inflammatory effect of cyclooxygenase inhibitors in fibroblast-like synoviocytes from the human temporomandibular joint results from the suppression of PGE2 production. J Oral Pathol Med. 2013;42:499–506.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Vieira MJ, Perosa SR, Argaaaraz GA, Silva JA Jr, Cavalheiro EA, Graca Naffah-Mazzacoratti M. Indomethacin can downregulate the levels of inflammatory mediators in the hippocampus of rats submitted to pilocarpine-induced status epilepticus. Clinics. 2014;69:621–6.PubMedPubMedCentralCrossRef Vieira MJ, Perosa SR, Argaaaraz GA, Silva JA Jr, Cavalheiro EA, Graca Naffah-Mazzacoratti M. Indomethacin can downregulate the levels of inflammatory mediators in the hippocampus of rats submitted to pilocarpine-induced status epilepticus. Clinics. 2014;69:621–6.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–82.CrossRef Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural, cellular, and molecular biology. Annu Rev Biochem. 2000;69:145–82.CrossRef
76.
Zurück zum Zitat Zidar N, Odar K, Glavac D, Jerse M, Zupanc T, Stajer D. Cyclooxygenase in normal human tissues--is COX-1 really a constitutive isoform, and COX-2 an inducible isoform? J Cell Mol Med. 2009;13:3753–63.PubMedCrossRef Zidar N, Odar K, Glavac D, Jerse M, Zupanc T, Stajer D. Cyclooxygenase in normal human tissues--is COX-1 really a constitutive isoform, and COX-2 an inducible isoform? J Cell Mol Med. 2009;13:3753–63.PubMedCrossRef
78.
Zurück zum Zitat Yamagata K, Andreasson KI, Kaufmann WE, Barnes CA, Worley PF. Expression of a mitogen-inducible cyclooxygenase in brain neurons: regulation by synaptic activity and glucocorticoids. Neuron. 1993;11:371–86.PubMedCrossRef Yamagata K, Andreasson KI, Kaufmann WE, Barnes CA, Worley PF. Expression of a mitogen-inducible cyclooxygenase in brain neurons: regulation by synaptic activity and glucocorticoids. Neuron. 1993;11:371–86.PubMedCrossRef
79.
Zurück zum Zitat Chen J, Marsh T, Zhang JS, Graham SH. Expression of cyclo-oxygenase 2 in rat brain following kainate treatment. Neuroreport. 1995;6:245–8.PubMed Chen J, Marsh T, Zhang JS, Graham SH. Expression of cyclo-oxygenase 2 in rat brain following kainate treatment. Neuroreport. 1995;6:245–8.PubMed
80.
Zurück zum Zitat Adams J, Collaço-Moraes Y, de Belleroche J. Cyclooxygenase-2 induction in cerebral cortex: an intracellular response to synaptic excitation. J Neurochem. 1996;66:6–13.PubMedCrossRef Adams J, Collaço-Moraes Y, de Belleroche J. Cyclooxygenase-2 induction in cerebral cortex: an intracellular response to synaptic excitation. J Neurochem. 1996;66:6–13.PubMedCrossRef
81.
Zurück zum Zitat Marcheselli VL, Bazan NG. Sustained induction of prostaglandin endoperoxide synthase-2 by seizures in hippocampus. Inhibition by a platelet-activating factor antagonist. J Biol Chem. 1996;271:24794–9.PubMedCrossRef Marcheselli VL, Bazan NG. Sustained induction of prostaglandin endoperoxide synthase-2 by seizures in hippocampus. Inhibition by a platelet-activating factor antagonist. J Biol Chem. 1996;271:24794–9.PubMedCrossRef
82.
Zurück zum Zitat Kawaguchi K, Hickey RW, Rose ME, Zhu L, Chen J, Graham SH. Cyclooxygenase-2 expression is induced in rat brain after kainate-induced seizures and promotes neuronal death in CA3 hippocampus. Brain Res. 2005;1050:130–7.PubMedCrossRef Kawaguchi K, Hickey RW, Rose ME, Zhu L, Chen J, Graham SH. Cyclooxygenase-2 expression is induced in rat brain after kainate-induced seizures and promotes neuronal death in CA3 hippocampus. Brain Res. 2005;1050:130–7.PubMedCrossRef
83.
Zurück zum Zitat Takemiya T, Maehara M, Matsumura K, Yasuda S, Sugiura H, Yamagata K. Prostaglandin E2 produced by late induced COX-2 stimulates hippocampal neuron loss after seizure in the CA3 region. Neurosci Res. 2006;56:103–10.PubMedCrossRef Takemiya T, Maehara M, Matsumura K, Yasuda S, Sugiura H, Yamagata K. Prostaglandin E2 produced by late induced COX-2 stimulates hippocampal neuron loss after seizure in the CA3 region. Neurosci Res. 2006;56:103–10.PubMedCrossRef
84.
Zurück zum Zitat Oliveira MS, Furian AF, Royes LF, Fighera MR, Fiorenza NG, Castelli M, et al. Cyclooxygenase-2/PGE2 pathway facilitates pentylenetetrazol-induced seizures. Epilepsy Res. 2008;79:14–21.PubMedCrossRef Oliveira MS, Furian AF, Royes LF, Fighera MR, Fiorenza NG, Castelli M, et al. Cyclooxygenase-2/PGE2 pathway facilitates pentylenetetrazol-induced seizures. Epilepsy Res. 2008;79:14–21.PubMedCrossRef
85.
Zurück zum Zitat Nishihara I, Minami T, Watanabe Y, Ito S, Hayaishi O. Prostaglandin E2 stimulates glutamate release from synaptosomes of rat spinal cord. Neurosci Lett. 1995;196:57–60.PubMedCrossRef Nishihara I, Minami T, Watanabe Y, Ito S, Hayaishi O. Prostaglandin E2 stimulates glutamate release from synaptosomes of rat spinal cord. Neurosci Lett. 1995;196:57–60.PubMedCrossRef
86.
Zurück zum Zitat Sang N, Zhang J, Marcheselli V, Bazan NG, Chen C. Postsynaptically synthesized prostaglandin E2 (PGE2) modulates hippocampal synaptic transmission via a presynaptic PGE2 EP2 receptor. J Neurosci. 2005;25:9858–70.PubMedPubMedCentralCrossRef Sang N, Zhang J, Marcheselli V, Bazan NG, Chen C. Postsynaptically synthesized prostaglandin E2 (PGE2) modulates hippocampal synaptic transmission via a presynaptic PGE2 EP2 receptor. J Neurosci. 2005;25:9858–70.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Fischborn SV, Soerensen J, Potschka H. Targeting the prostaglandin E2 EP1 receptor and cyclooxygenase-2 in the amygdala kindling model in mice. Epilepsy Res. 2010;91:57–65.PubMedCrossRef Fischborn SV, Soerensen J, Potschka H. Targeting the prostaglandin E2 EP1 receptor and cyclooxygenase-2 in the amygdala kindling model in mice. Epilepsy Res. 2010;91:57–65.PubMedCrossRef
88.
Zurück zum Zitat Rojas A, Gueorguieva P, Lelutiu N, Quan Y, Shaw R, Dingledine R. The prostaglandin EP1 receptor potentiates kainate receptor activation via a protein kinase C pathway and exacerbates status epilepticus. Neurobiol Dis. 2014;70:74–89.PubMedPubMedCentralCrossRef Rojas A, Gueorguieva P, Lelutiu N, Quan Y, Shaw R, Dingledine R. The prostaglandin EP1 receptor potentiates kainate receptor activation via a protein kinase C pathway and exacerbates status epilepticus. Neurobiol Dis. 2014;70:74–89.PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Jiang J, Quan Y, Ganesh T, Pouliot WA, Dudek FE, Dingledine R. Inhibition of the prostaglandin receptor EP2 following status epilepticus reduces delayed mortality and brain inflammation. Proc Natl Acad Sci U S A. 2013;110:3591–6.PubMedPubMedCentralCrossRef Jiang J, Quan Y, Ganesh T, Pouliot WA, Dudek FE, Dingledine R. Inhibition of the prostaglandin receptor EP2 following status epilepticus reduces delayed mortality and brain inflammation. Proc Natl Acad Sci U S A. 2013;110:3591–6.PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat Rojas A, Ganesh T, Lelutiu N, Gueorguieva P, Dingledine R. Inhibition of the prostaglandin EP2 receptor is neuroprotective and accelerates functional recovery in a rat model of organophosphorus induced status epilepticus. Neuropharmacology. 2015;93:15–27.PubMedPubMedCentralCrossRef Rojas A, Ganesh T, Lelutiu N, Gueorguieva P, Dingledine R. Inhibition of the prostaglandin EP2 receptor is neuroprotective and accelerates functional recovery in a rat model of organophosphorus induced status epilepticus. Neuropharmacology. 2015;93:15–27.PubMedPubMedCentralCrossRef
91.
Zurück zum Zitat Rojas A, Ganesh T, Manji Z, O'neill T, Dingledine R. Inhibition of the prostaglandin E2 receptor EP2 prevents status epilepticus-induced deficits in the novel object recognition task in rats. Neuropharmacology. 2016;110:419–30.PubMedPubMedCentralCrossRef Rojas A, Ganesh T, Manji Z, O'neill T, Dingledine R. Inhibition of the prostaglandin E2 receptor EP2 prevents status epilepticus-induced deficits in the novel object recognition task in rats. Neuropharmacology. 2016;110:419–30.PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat Oliveira MS, Furian AF, Rambo LM, Ribeiro LR, Royes LF, Ferreira J, et al. Modulation of pentylenetetrazol-induced seizures by prostaglandin E2 receptors. Neuroscience. 2008;152:1110–8.PubMedCrossRef Oliveira MS, Furian AF, Rambo LM, Ribeiro LR, Royes LF, Ferreira J, et al. Modulation of pentylenetetrazol-induced seizures by prostaglandin E2 receptors. Neuroscience. 2008;152:1110–8.PubMedCrossRef
93.
Zurück zum Zitat Santos AC, Temp FR, Marafiga JR, Pillat MM, Hessel AT, Ribeiro LR, et al. EP2 receptor agonist ONO-AE1-259-01 attenuates pentylenetetrazole- and pilocarpine-induced seizures but causes hippocampal neurotoxicity. Epilepsy Behav. 2017;73:180–8.PubMedCrossRef Santos AC, Temp FR, Marafiga JR, Pillat MM, Hessel AT, Ribeiro LR, et al. EP2 receptor agonist ONO-AE1-259-01 attenuates pentylenetetrazole- and pilocarpine-induced seizures but causes hippocampal neurotoxicity. Epilepsy Behav. 2017;73:180–8.PubMedCrossRef
94.
95.
Zurück zum Zitat Jiang J, Yang MS, Quan Y, Gueorguieva P, Ganesh T, Dingledine R. Therapeutic window for cyclooxygenase-2 related anti-inflammatory therapy after status epilepticus. Neurobiol Dis. 2015;76:126–36.PubMedPubMedCentralCrossRef Jiang J, Yang MS, Quan Y, Gueorguieva P, Ganesh T, Dingledine R. Therapeutic window for cyclooxygenase-2 related anti-inflammatory therapy after status epilepticus. Neurobiol Dis. 2015;76:126–36.PubMedPubMedCentralCrossRef
96.
97.
Zurück zum Zitat Järvelä JT, Lopez-Picon FR, Holopainen IE. Age-dependent cyclooxygenase-2 induction and neuronal damage after status epilepticus in the postnatal rat hippocampus. Epilepsia. 2008;49:832–41.PubMedCrossRef Järvelä JT, Lopez-Picon FR, Holopainen IE. Age-dependent cyclooxygenase-2 induction and neuronal damage after status epilepticus in the postnatal rat hippocampus. Epilepsia. 2008;49:832–41.PubMedCrossRef
98.
Zurück zum Zitat Holtman L, van Vliet EA, van Schaik R, Queiroz CM, Aronica E, Gorter JA. Effects of SC58236, a selective COX-2 inhibitor, on epileptogenesis and spontaneous seizures in a rat model for temporal lobe epilepsy. Epilepsy Res. 2009;84:56–66.PubMedCrossRef Holtman L, van Vliet EA, van Schaik R, Queiroz CM, Aronica E, Gorter JA. Effects of SC58236, a selective COX-2 inhibitor, on epileptogenesis and spontaneous seizures in a rat model for temporal lobe epilepsy. Epilepsy Res. 2009;84:56–66.PubMedCrossRef
99.
Zurück zum Zitat Desjardins P, Sauvageau A, Bouthillier A, Navarro D, Hazell AS, Rose C, et al. Induction of astrocytic cyclooxygenase-2 in epileptic patients with hippocampal sclerosis. Neurochem Int. 2003;42:299–303.PubMedCrossRef Desjardins P, Sauvageau A, Bouthillier A, Navarro D, Hazell AS, Rose C, et al. Induction of astrocytic cyclooxygenase-2 in epileptic patients with hippocampal sclerosis. Neurochem Int. 2003;42:299–303.PubMedCrossRef
100.
Zurück zum Zitat Weidner LD, Kannan P, Mitsios N, Kang SJ, Hall MD, Theodore WH, et al. The expression of inflammatory markers and their potential influence on efflux transporters in drug-resistant mesial temporal lobe epilepsy tissue. Epilepsia. 2018;59:1507–17.PubMedPubMedCentralCrossRef Weidner LD, Kannan P, Mitsios N, Kang SJ, Hall MD, Theodore WH, et al. The expression of inflammatory markers and their potential influence on efflux transporters in drug-resistant mesial temporal lobe epilepsy tissue. Epilepsia. 2018;59:1507–17.PubMedPubMedCentralCrossRef
101.
Zurück zum Zitat Hung KL, Liang JS, Wang JS, Chen HJ, Lin LJ, Lu JF. Association of a novel GABRG2 splicing variation and a PTGS2/COX-2 single nucleotide polymorphism with Taiwanese febrile seizures. Epilepsy Res. 2017;129:1–7.PubMedCrossRef Hung KL, Liang JS, Wang JS, Chen HJ, Lin LJ, Lu JF. Association of a novel GABRG2 splicing variation and a PTGS2/COX-2 single nucleotide polymorphism with Taiwanese febrile seizures. Epilepsy Res. 2017;129:1–7.PubMedCrossRef
102.
Zurück zum Zitat Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57:178–201.PubMedCrossRef Zlokovic BV. The blood-brain barrier in health and chronic neurodegenerative disorders. Neuron. 2008;57:178–201.PubMedCrossRef
103.
Zurück zum Zitat Collard CD, Park KA, Montalto MC, Alapati S, Buras JA, Stahl GL, et al. Neutrophil-derived glutamate regulates vascular endothelial barrier function. J Biol Chem. 2002;277:14801–11.PubMedCrossRef Collard CD, Park KA, Montalto MC, Alapati S, Buras JA, Stahl GL, et al. Neutrophil-derived glutamate regulates vascular endothelial barrier function. J Biol Chem. 2002;277:14801–11.PubMedCrossRef
104.
Zurück zum Zitat Koenig H, Trout JJ, Goldstone AD, Lu CY. Capillary NMDA receptors regulate blood-brain barrier function and breakdown. Brain Res. 1992;588:297–303.PubMedCrossRef Koenig H, Trout JJ, Goldstone AD, Lu CY. Capillary NMDA receptors regulate blood-brain barrier function and breakdown. Brain Res. 1992;588:297–303.PubMedCrossRef
105.
Zurück zum Zitat Roch C, Leroy C, Nehlig A, Namer IJ. Magnetic resonance imaging in the study of the lithium-pilocarpine model of temporal lobe epilepsy in adult rats. Epilepsia. 2002;43:325–35.PubMedCrossRef Roch C, Leroy C, Nehlig A, Namer IJ. Magnetic resonance imaging in the study of the lithium-pilocarpine model of temporal lobe epilepsy in adult rats. Epilepsia. 2002;43:325–35.PubMedCrossRef
106.
Zurück zum Zitat Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U, et al. Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci. 2004;24:7829–36.PubMedPubMedCentralCrossRef Seiffert E, Dreier JP, Ivens S, Bechmann I, Tomkins O, Heinemann U, et al. Lasting blood-brain barrier disruption induces epileptic focus in the rat somatosensory cortex. J Neurosci. 2004;24:7829–36.PubMedPubMedCentralCrossRef
107.
Zurück zum Zitat van Vliet EA, da Costa AS, Redeker S, van Schaik R, Aronica E, Gorter JA. Blood-brain barrier leakage may lead to progression of temporal lobe epilepsy. Brain. 2007;130:521–34.PubMedCrossRef van Vliet EA, da Costa AS, Redeker S, van Schaik R, Aronica E, Gorter JA. Blood-brain barrier leakage may lead to progression of temporal lobe epilepsy. Brain. 2007;130:521–34.PubMedCrossRef
108.
Zurück zum Zitat Marchi N, Angelov L, Masaryk T, Fazio V, Granata T, Hernandez N, et al. Seizure-promoting effect of blood-brain barrier disruption. Epilepsia. 2007;48:732–42.PubMedPubMedCentralCrossRef Marchi N, Angelov L, Masaryk T, Fazio V, Granata T, Hernandez N, et al. Seizure-promoting effect of blood-brain barrier disruption. Epilepsia. 2007;48:732–42.PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Tomkins O, Shelef I, Kaizerman I, Eliushin A, Afawi Z, Misk A, et al. Blood-brain barrier disruption in post-traumatic epilepsy. J Neurol Neurosurg Psychiatry. 2008;79:774–7.PubMedCrossRef Tomkins O, Shelef I, Kaizerman I, Eliushin A, Afawi Z, Misk A, et al. Blood-brain barrier disruption in post-traumatic epilepsy. J Neurol Neurosurg Psychiatry. 2008;79:774–7.PubMedCrossRef
110.
Zurück zum Zitat Marchi N, Granata T, Ghosh C, Janigro D. Blood-brain barrier dysfunction and epilepsy: pathophysiologic role and therapeutic approaches. Epilepsia. 2012;53:1877–86.PubMedPubMedCentralCrossRef Marchi N, Granata T, Ghosh C, Janigro D. Blood-brain barrier dysfunction and epilepsy: pathophysiologic role and therapeutic approaches. Epilepsia. 2012;53:1877–86.PubMedPubMedCentralCrossRef
111.
Zurück zum Zitat Warren MS, Zerangue N, Woodford K, Roberts LM, Tate EH, Feng B, et al. Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human. Pharmacol Res. 2009;59:404–13.PubMedCrossRef Warren MS, Zerangue N, Woodford K, Roberts LM, Tate EH, Feng B, et al. Comparative gene expression profiles of ABC transporters in brain microvessel endothelial cells and brain in five species including human. Pharmacol Res. 2009;59:404–13.PubMedCrossRef
112.
Zurück zum Zitat Rizzi M, Caccia S, Guiso G, Richichi C, Gorter JA, Aronica E, et al. Limbic seizures induce P-glycoprotein in rodent brain: functional implications for pharmacoresistance. J Neurosci. 2002;22:5833–9.PubMedPubMedCentralCrossRef Rizzi M, Caccia S, Guiso G, Richichi C, Gorter JA, Aronica E, et al. Limbic seizures induce P-glycoprotein in rodent brain: functional implications for pharmacoresistance. J Neurosci. 2002;22:5833–9.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Seegers U, Potschka H, Löscher W. Expression of the multidrug transporter P-glycoprotein in brain capillary endothelial cells and brain parenchyma of amygdala-kindled rats. Epilepsia. 2002;43:675–84.PubMedCrossRef Seegers U, Potschka H, Löscher W. Expression of the multidrug transporter P-glycoprotein in brain capillary endothelial cells and brain parenchyma of amygdala-kindled rats. Epilepsia. 2002;43:675–84.PubMedCrossRef
114.
Zurück zum Zitat van Vliet E, Aronica E, Redeker S, Marchi N, Rizzi M, Vezzani A, et al. Selective and persistent upregulation of mdr1b mRNA and P-glycoprotein in the parahippocampal cortex of chronic epileptic rats. Epilepsy Res. 2004;60:203–13.PubMedCrossRef van Vliet E, Aronica E, Redeker S, Marchi N, Rizzi M, Vezzani A, et al. Selective and persistent upregulation of mdr1b mRNA and P-glycoprotein in the parahippocampal cortex of chronic epileptic rats. Epilepsy Res. 2004;60:203–13.PubMedCrossRef
115.
Zurück zum Zitat Volk HA, Burkhardt K, Potschka H, Chen J, Becker A, Löscher W. Neuronal expression of the drug efflux transporter P-glycoprotein in the rat hippocampus after limbic seizures. Neuroscience. 2004;123:751–9.PubMedCrossRef Volk HA, Burkhardt K, Potschka H, Chen J, Becker A, Löscher W. Neuronal expression of the drug efflux transporter P-glycoprotein in the rat hippocampus after limbic seizures. Neuroscience. 2004;123:751–9.PubMedCrossRef
116.
Zurück zum Zitat Liu X, Yang Z, Yang J, Yang H. Increased P-glycoprotein expression and decreased phenobarbital distribution in the brain of pentylenetetrazole-kindled rats. Neuropharmacology. 2007;53:657–63.PubMedCrossRef Liu X, Yang Z, Yang J, Yang H. Increased P-glycoprotein expression and decreased phenobarbital distribution in the brain of pentylenetetrazole-kindled rats. Neuropharmacology. 2007;53:657–63.PubMedCrossRef
117.
Zurück zum Zitat Hartz AM, Pekcec A, Soldner EL, Zhong Y, Schlichtiger J, Bauer B. P-gp protein expression and transport activity in rodent seizure models and human epilepsy. Mol Pharm. 2017;14:999–1011.PubMedPubMedCentralCrossRef Hartz AM, Pekcec A, Soldner EL, Zhong Y, Schlichtiger J, Bauer B. P-gp protein expression and transport activity in rodent seizure models and human epilepsy. Mol Pharm. 2017;14:999–1011.PubMedPubMedCentralCrossRef
118.
Zurück zum Zitat Tishler DM, Weinberg KI, Hinton DR, Barbaro N, Annett GM, Raffel C. MDR1 gene expression in brain of patients with medically intractable epilepsy. Epilepsia. 1995;36:1–6.PubMedCrossRef Tishler DM, Weinberg KI, Hinton DR, Barbaro N, Annett GM, Raffel C. MDR1 gene expression in brain of patients with medically intractable epilepsy. Epilepsia. 1995;36:1–6.PubMedCrossRef
119.
Zurück zum Zitat Sisodiya SM, Lin WR, Harding BN, Squier MV, Thom M. Drug resistance in epilepsy: expression of drug resistance proteins in common causes of refractory epilepsy. Brain. 2002;125:22–31.PubMedCrossRef Sisodiya SM, Lin WR, Harding BN, Squier MV, Thom M. Drug resistance in epilepsy: expression of drug resistance proteins in common causes of refractory epilepsy. Brain. 2002;125:22–31.PubMedCrossRef
120.
Zurück zum Zitat Liu JY, Thom M, Catarino CB, Martinian L, Figarella-Branger D, Bartolomei F, et al. Neuropathology of the blood-brain barrier and pharmaco-resistance in human epilepsy. Brain. 2012;135:3115–33.CrossRef Liu JY, Thom M, Catarino CB, Martinian L, Figarella-Branger D, Bartolomei F, et al. Neuropathology of the blood-brain barrier and pharmaco-resistance in human epilepsy. Brain. 2012;135:3115–33.CrossRef
121.
Zurück zum Zitat Feldmann M, Asselin MC, Liu J, Wang S, McMahon A, Anton-Rodriguez J, et al. P-glycoprotein expression and function in patients with temporal lobe epilepsy: a case-control study. Lancet Neurol. 2013;12:777–85.PubMedCrossRef Feldmann M, Asselin MC, Liu J, Wang S, McMahon A, Anton-Rodriguez J, et al. P-glycoprotein expression and function in patients with temporal lobe epilepsy: a case-control study. Lancet Neurol. 2013;12:777–85.PubMedCrossRef
122.
Zurück zum Zitat Volk HA, Löscher W. Multidrug resistance in epilepsy: rats with drug-resistant seizures exhibit enhanced brain expression of P-glycoprotein compared with rats with drug-responsive seizures. Brain. 2005;128:1358–68.PubMedCrossRef Volk HA, Löscher W. Multidrug resistance in epilepsy: rats with drug-resistant seizures exhibit enhanced brain expression of P-glycoprotein compared with rats with drug-responsive seizures. Brain. 2005;128:1358–68.PubMedCrossRef
123.
Zurück zum Zitat Zhu HJ, Liu GQ. Glutamate up-regulates P-glycoprotein expression in rat brain microvessel endothelial cells by an NMDA receptor-mediated mechanism. Life Sci. 2004;75:1313–22.PubMedCrossRef Zhu HJ, Liu GQ. Glutamate up-regulates P-glycoprotein expression in rat brain microvessel endothelial cells by an NMDA receptor-mediated mechanism. Life Sci. 2004;75:1313–22.PubMedCrossRef
124.
Zurück zum Zitat Bauer B, Hartz AM, Pekcec A, Toellner K, Miller DS, Potschka H. Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cyclooxygenase-2 signaling. Mol Pharmacol. 2008;73:1444–53.PubMedCrossRef Bauer B, Hartz AM, Pekcec A, Toellner K, Miller DS, Potschka H. Seizure-induced up-regulation of P-glycoprotein at the blood-brain barrier through glutamate and cyclooxygenase-2 signaling. Mol Pharmacol. 2008;73:1444–53.PubMedCrossRef
125.
Zurück zum Zitat Avemary J, Salvamoser JD, Peraud A, Rémi J, Noachtar S, Fricker G, et al. Dynamic regulation of P-glycoprotein in human brain capillaries. Mol Pharm. 2013;10:3333–41.PubMedCrossRef Avemary J, Salvamoser JD, Peraud A, Rémi J, Noachtar S, Fricker G, et al. Dynamic regulation of P-glycoprotein in human brain capillaries. Mol Pharm. 2013;10:3333–41.PubMedCrossRef
126.
Zurück zum Zitat Zibell G, Unkrüer B, Pekcec A, Hartz AM, Bauer B, Miller DS, et al. Prevention of seizure-induced up-regulation of endothelial P-glycoprotein by COX-2 inhibition. Neuropharmacology. 2009;56:849–55.PubMedCrossRef Zibell G, Unkrüer B, Pekcec A, Hartz AM, Bauer B, Miller DS, et al. Prevention of seizure-induced up-regulation of endothelial P-glycoprotein by COX-2 inhibition. Neuropharmacology. 2009;56:849–55.PubMedCrossRef
127.
Zurück zum Zitat van Vliet EA, Zibell G, Pekcec A, Schlichtiger J, Edelbroek PM, Holtman L, et al. COX-2 inhibition controls P-glycoprotein expression and promotes brain delivery of phenytoin in chronic epileptic rats. Neuropharmacology. 2010;58:404–12.PubMedCrossRef van Vliet EA, Zibell G, Pekcec A, Schlichtiger J, Edelbroek PM, Holtman L, et al. COX-2 inhibition controls P-glycoprotein expression and promotes brain delivery of phenytoin in chronic epileptic rats. Neuropharmacology. 2010;58:404–12.PubMedCrossRef
128.
Zurück zum Zitat Pekcec A, Unkrüer B, Schlichtiger J, Soerensen J, Hartz AM, Bauer B, et al. Targeting prostaglandin E2 EP1 receptors prevents seizure-associated P-glycoprotein up-regulation. J Pharmacol Exp Ther. 2009;330:939–47.PubMedCrossRef Pekcec A, Unkrüer B, Schlichtiger J, Soerensen J, Hartz AM, Bauer B, et al. Targeting prostaglandin E2 EP1 receptors prevents seizure-associated P-glycoprotein up-regulation. J Pharmacol Exp Ther. 2009;330:939–47.PubMedCrossRef
129.
Zurück zum Zitat Rojas A, Jiang J, Ganesh T, Yang MS, Lelutiu N, Gueorguieva P, et al. Cyclooxygenase-2 in epilepsy. Epilepsia. 2014;55:17–25.PubMedCrossRef Rojas A, Jiang J, Ganesh T, Yang MS, Lelutiu N, Gueorguieva P, et al. Cyclooxygenase-2 in epilepsy. Epilepsia. 2014;55:17–25.PubMedCrossRef
130.
Zurück zum Zitat Elkhayat HA, Aly RH, Elagouza IA, El-Kabarity RH, Galal YI. Role of P-glycoprotein inhibitors in children with drug-resistant epilepsy. Acta Neurol Scand. 2017;136:639–44.PubMedCrossRef Elkhayat HA, Aly RH, Elagouza IA, El-Kabarity RH, Galal YI. Role of P-glycoprotein inhibitors in children with drug-resistant epilepsy. Acta Neurol Scand. 2017;136:639–44.PubMedCrossRef
131.
Zurück zum Zitat Schlichtiger J, Pekcec A, Bartmann H, Winter P, Fuest C, Soerensen J, et al. Celecoxib treatment restores pharmacosensitivity in a rat model of pharmacoresistant epilepsy. Br J Pharmacol. 2010;160:1062–71.PubMedPubMedCentralCrossRef Schlichtiger J, Pekcec A, Bartmann H, Winter P, Fuest C, Soerensen J, et al. Celecoxib treatment restores pharmacosensitivity in a rat model of pharmacoresistant epilepsy. Br J Pharmacol. 2010;160:1062–71.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Gobbo OL, O’Mara SM. Post-treatment, but not pre-treatment, with the selective cyclooxygenase-2 inhibitor celecoxib markedly enhances functional recovery from kainic acid-induced neurodegeneration. Neuroscience. 2004;125:317–27.PubMedCrossRef Gobbo OL, O’Mara SM. Post-treatment, but not pre-treatment, with the selective cyclooxygenase-2 inhibitor celecoxib markedly enhances functional recovery from kainic acid-induced neurodegeneration. Neuroscience. 2004;125:317–27.PubMedCrossRef
133.
Zurück zum Zitat Baik EJ, Kim EJ, Lee SH, Moon C. Cyclooxygenase-2 selective inhibitors aggravate kainic acid induced seizure and neuronal cell death in the hippocampus. Brain Res. 1999;843:118–29.PubMedCrossRef Baik EJ, Kim EJ, Lee SH, Moon C. Cyclooxygenase-2 selective inhibitors aggravate kainic acid induced seizure and neuronal cell death in the hippocampus. Brain Res. 1999;843:118–29.PubMedCrossRef
134.
Zurück zum Zitat Kim HJ, Chung JI, Lee SH, Jung YS, Moon CH, Baik EJ. Involvement of endogenous prostaglandin F2alpha on kainic acid-induced seizure activity through FP receptor: the mechanism of proconvulsant effects of COX-2 inhibitors. Brain Res. 2008;1193:153–61.PubMedCrossRef Kim HJ, Chung JI, Lee SH, Jung YS, Moon CH, Baik EJ. Involvement of endogenous prostaglandin F2alpha on kainic acid-induced seizure activity through FP receptor: the mechanism of proconvulsant effects of COX-2 inhibitors. Brain Res. 2008;1193:153–61.PubMedCrossRef
135.
Zurück zum Zitat Jung KH, Chu K, Lee ST, Kim J, Sinn DI, Kim JM, et al. Cyclooxygenase-2 inhibitor, celecoxib, inhibits the altered hippocampal neurogenesis with attenuation of spontaneous recurrent seizures following pilocarpine-induced status epilepticus. Neurobiol Dis. 2006;23:237–46.PubMedCrossRef Jung KH, Chu K, Lee ST, Kim J, Sinn DI, Kim JM, et al. Cyclooxygenase-2 inhibitor, celecoxib, inhibits the altered hippocampal neurogenesis with attenuation of spontaneous recurrent seizures following pilocarpine-induced status epilepticus. Neurobiol Dis. 2006;23:237–46.PubMedCrossRef
136.
Zurück zum Zitat Citraro R, Leo A, Marra R, De Sarro G, Russo E. Antiepileptogenic effects of the selective COX-2 inhibitor etoricoxib, on the development of spontaneous absence seizures in WAG/Rij rats. Brain Res Bull. 2015;113:1–7.PubMedCrossRef Citraro R, Leo A, Marra R, De Sarro G, Russo E. Antiepileptogenic effects of the selective COX-2 inhibitor etoricoxib, on the development of spontaneous absence seizures in WAG/Rij rats. Brain Res Bull. 2015;113:1–7.PubMedCrossRef
137.
Zurück zum Zitat Katyal J, Kumar H, Gupta YK. Anticonvulsant activity of the cyclooxygenase-2 (COX-2) inhibitor etoricoxib in pentylenetetrazole-kindled rats is associated with memory impairment. Epilepsy Behav. 2015;44:98–103.PubMedCrossRef Katyal J, Kumar H, Gupta YK. Anticonvulsant activity of the cyclooxygenase-2 (COX-2) inhibitor etoricoxib in pentylenetetrazole-kindled rats is associated with memory impairment. Epilepsy Behav. 2015;44:98–103.PubMedCrossRef
138.
Zurück zum Zitat Dhir A, Naidu PS, Kulkarni SK. Effect of cyclooxygenase-2 (COX-2) inhibitors in various animal models (bicuculline, picrotoxin, maximal electroshock-induced convulsions) of epilepsy with possible mechanism of action. Indian J Exp Biol. 2006;44:286–91.PubMed Dhir A, Naidu PS, Kulkarni SK. Effect of cyclooxygenase-2 (COX-2) inhibitors in various animal models (bicuculline, picrotoxin, maximal electroshock-induced convulsions) of epilepsy with possible mechanism of action. Indian J Exp Biol. 2006;44:286–91.PubMed
139.
Zurück zum Zitat Kunz T, Oliw EH. Nimesulide aggravates kainic acid-induced seizures in the rat. Pharmacol Toxicol. 2001;88:271–6.PubMedCrossRef Kunz T, Oliw EH. Nimesulide aggravates kainic acid-induced seizures in the rat. Pharmacol Toxicol. 2001;88:271–6.PubMedCrossRef
140.
Zurück zum Zitat Dhir A, Naidu PS, Kulkarni SK. Effect of cyclooxygenase inhibitors on pentylenetetrazol (PTZ)-induced convulsions: possible mechanism of action. Prog Neuro-Psychopharmacol Biol Psychiatry. 2006;30:1478–85.CrossRef Dhir A, Naidu PS, Kulkarni SK. Effect of cyclooxygenase inhibitors on pentylenetetrazol (PTZ)-induced convulsions: possible mechanism of action. Prog Neuro-Psychopharmacol Biol Psychiatry. 2006;30:1478–85.CrossRef
141.
Zurück zum Zitat Dhir A, Naidu PS, Kulkarni SK. Neuroprotective effect of nimesulide, a preferential COX-2 inhibitor, against pentylenetetrazol (PTZ)-induced chemical kindling and associated biochemical parameters in mice. Seizure. 2007;16:691–7.PubMedCrossRef Dhir A, Naidu PS, Kulkarni SK. Neuroprotective effect of nimesulide, a preferential COX-2 inhibitor, against pentylenetetrazol (PTZ)-induced chemical kindling and associated biochemical parameters in mice. Seizure. 2007;16:691–7.PubMedCrossRef
142.
Zurück zum Zitat Trandafir CC, Pouliot WA, Dudek FE, Ekstrand JJ. Co-administration of subtherapeutic diazepam enhances neuroprotective effect of COX-2 inhibitor, NS-398, after lithium pilocarpine-induced status epilepticus. Neuroscience. 2015;284:601–10.PubMedCrossRef Trandafir CC, Pouliot WA, Dudek FE, Ekstrand JJ. Co-administration of subtherapeutic diazepam enhances neuroprotective effect of COX-2 inhibitor, NS-398, after lithium pilocarpine-induced status epilepticus. Neuroscience. 2015;284:601–10.PubMedCrossRef
143.
Zurück zum Zitat Polascheck N, Bankstahl M, Löscher W. The COX-2 inhibitor parecoxib is neuroprotective but not antiepileptogenic in the pilocarpine model of temporal lobe epilepsy. Exp Neurol. 2010;224:219–33.PubMedCrossRef Polascheck N, Bankstahl M, Löscher W. The COX-2 inhibitor parecoxib is neuroprotective but not antiepileptogenic in the pilocarpine model of temporal lobe epilepsy. Exp Neurol. 2010;224:219–33.PubMedCrossRef
144.
Zurück zum Zitat Kunz T, Oliw EH. The selective cyclooxygenase-2 inhibitor rofecoxib reduces kainate-induced cell death in the rat hippocampus. Eur J Neurosci. 2001;13:569–75.PubMedCrossRef Kunz T, Oliw EH. The selective cyclooxygenase-2 inhibitor rofecoxib reduces kainate-induced cell death in the rat hippocampus. Eur J Neurosci. 2001;13:569–75.PubMedCrossRef
145.
Zurück zum Zitat Akula KK, Dhir A, Kulkarni SK. Rofecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor increases pentylenetetrazol seizure threshold in mice: possible involvement of adenosinergic mechanism. Epilepsy Res. 2008;78:60–70.PubMedCrossRef Akula KK, Dhir A, Kulkarni SK. Rofecoxib, a selective cyclooxygenase-2 (COX-2) inhibitor increases pentylenetetrazol seizure threshold in mice: possible involvement of adenosinergic mechanism. Epilepsy Res. 2008;78:60–70.PubMedCrossRef
146.
Zurück zum Zitat Claycomb RJ, Hewett SJ, Hewett JA. Prophylactic, prandial rofecoxib treatment lacks efficacy against acute PTZ-induced seizure generation and kindling acquisition. Epilepsia. 2011;52:273–83.PubMedPubMedCentral Claycomb RJ, Hewett SJ, Hewett JA. Prophylactic, prandial rofecoxib treatment lacks efficacy against acute PTZ-induced seizure generation and kindling acquisition. Epilepsia. 2011;52:273–83.PubMedPubMedCentral
147.
Zurück zum Zitat Holtman L, van Vliet EA, Edelbroek PM, Aronica E, Gorter JA. Cox-2 inhibition can lead to adverse effects in a rat model for temporal lobe epilepsy. Epilepsy Res. 2010;91:49–56.PubMedCrossRef Holtman L, van Vliet EA, Edelbroek PM, Aronica E, Gorter JA. Cox-2 inhibition can lead to adverse effects in a rat model for temporal lobe epilepsy. Epilepsy Res. 2010;91:49–56.PubMedCrossRef
148.
Zurück zum Zitat Kaminski R, Kozicka M, Parada-turska J, Dziki M, Kleinrok Z, Turski WA, et al. Effect of non-steroidal anti-inflammatory drugs on the anticonvulsive activity of valproate and diphenylhydantoin against maximal electroshock-induced seizures in mice. Pharmacol Res. 1998;37:375–81.PubMedCrossRef Kaminski R, Kozicka M, Parada-turska J, Dziki M, Kleinrok Z, Turski WA, et al. Effect of non-steroidal anti-inflammatory drugs on the anticonvulsive activity of valproate and diphenylhydantoin against maximal electroshock-induced seizures in mice. Pharmacol Res. 1998;37:375–81.PubMedCrossRef
149.
Zurück zum Zitat Ma L, Cui XL, Wang Y, Li XW, Yang F, Wei D, et al. 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;1469:103–13.PubMedCrossRef Ma L, Cui XL, Wang Y, Li XW, Yang F, Wei D, et al. 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;1469:103–13.PubMedCrossRef
150.
Zurück zum Zitat Zhu K, Hu M, Yuan B, Liu JX, Liu Y. Aspirin attenuates spontaneous recurrent seizures in the chronically epileptic mice. Neurol Res. 2017;39:744–57.PubMedCrossRef Zhu K, Hu M, Yuan B, Liu JX, Liu Y. Aspirin attenuates spontaneous recurrent seizures in the chronically epileptic mice. Neurol Res. 2017;39:744–57.PubMedCrossRef
151.
Zurück zum Zitat Jeong KH, Kim JY, Choi YS, Lee MY, Kim SY. Influence of aspirin on pilocarpine-induced epilepsy in mice. Korean J Physiol Pharmacol. 2013;17:15–21.PubMedPubMedCentralCrossRef Jeong KH, Kim JY, Choi YS, Lee MY, Kim SY. Influence of aspirin on pilocarpine-induced epilepsy in mice. Korean J Physiol Pharmacol. 2013;17:15–21.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Lin TY, Lu CW, Wang CC, Huang SK, Wang SJ. Cyclooxygenase 2 inhibitor celecoxib inhibits glutamate release by attenuating the PGE2/EP2 pathway in rat cerebral cortex endings. J Pharmacol Exp Ther. 2014;351:134–45.PubMedCrossRef Lin TY, Lu CW, Wang CC, Huang SK, Wang SJ. Cyclooxygenase 2 inhibitor celecoxib inhibits glutamate release by attenuating the PGE2/EP2 pathway in rat cerebral cortex endings. J Pharmacol Exp Ther. 2014;351:134–45.PubMedCrossRef
153.
Zurück zum Zitat An Y, Belevych N, Wang Y, Zhang H, Herschman H, Chen Q, et al. Neuronal and nonneuronal COX-2 expression confers neurotoxic and neuroprotective phenotypes in response to excitotoxin challenge. J Neurosci Res. 2014;92:486–95.PubMedCrossRef An Y, Belevych N, Wang Y, Zhang H, Herschman H, Chen Q, et al. Neuronal and nonneuronal COX-2 expression confers neurotoxic and neuroprotective phenotypes in response to excitotoxin challenge. J Neurosci Res. 2014;92:486–95.PubMedCrossRef
154.
Zurück zum Zitat Zhang C, Kwan P, Zuo Z, Baum L. The transport of antiepileptic drugs by P-glycoprotein. Adv Drug Deliv Rev. 2012;64:930–42.PubMedCrossRef Zhang C, Kwan P, Zuo Z, Baum L. The transport of antiepileptic drugs by P-glycoprotein. Adv Drug Deliv Rev. 2012;64:930–42.PubMedCrossRef
155.
Zurück zum Zitat van Vliet EA, van Schaik R, Edelbroek PM, Voskuyl RA, Redeker S, Aronica E, et al. Region-specific overexpression of P-glycoprotein at the blood-brain barrier affects brain uptake of phenytoin in epileptic rats. J Pharmacol Exp Ther. 2007;322:141–7.PubMedCrossRef van Vliet EA, van Schaik R, Edelbroek PM, Voskuyl RA, Redeker S, Aronica E, et al. Region-specific overexpression of P-glycoprotein at the blood-brain barrier affects brain uptake of phenytoin in epileptic rats. J Pharmacol Exp Ther. 2007;322:141–7.PubMedCrossRef
156.
Zurück zum Zitat Potschka H, Löscher W. In vivo evidence for P-glycoprotein-mediated transport of phenytoin at the blood-brain barrier of rats. Epilepsia. 2001;42:1231–40.PubMedCrossRef Potschka H, Löscher W. In vivo evidence for P-glycoprotein-mediated transport of phenytoin at the blood-brain barrier of rats. Epilepsia. 2001;42:1231–40.PubMedCrossRef
157.
Zurück zum Zitat van Vliet EA, van Schaik R, Edelbroek PM, Redeker S, Aronica E, Wadman WJ, et al. Inhibition of the multidrug transporter P-glycoprotein improves seizure control in phenytoin-treated chronic epileptic rats. Epilepsia. 2006;47:672–80.PubMedCrossRef van Vliet EA, van Schaik R, Edelbroek PM, Redeker S, Aronica E, Wadman WJ, et al. Inhibition of the multidrug transporter P-glycoprotein improves seizure control in phenytoin-treated chronic epileptic rats. Epilepsia. 2006;47:672–80.PubMedCrossRef
158.
Zurück zum Zitat Lim JA, Jung KY, Park B, Kim TJ, Jun JS, Kim KT, et al. Impact of a selective cyclooxygenase-2 inhibitor, celecoxib, on cortical excitability and electrophysiological properties of the brain in healthy volunteers: a randomized, double-blind, placebo-controlled study. PLoS One. 2019;14:e0212689.PubMedPubMedCentralCrossRef Lim JA, Jung KY, Park B, Kim TJ, Jun JS, Kim KT, et al. Impact of a selective cyclooxygenase-2 inhibitor, celecoxib, on cortical excitability and electrophysiological properties of the brain in healthy volunteers: a randomized, double-blind, placebo-controlled study. PLoS One. 2019;14:e0212689.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat van Stuijvenberg M, Derksen-Lubsen G, Steyerberg EW, Habbema JD, Moll HA. Randomized, controlled trial of ibuprofen syrup administered during febrile illnesses to prevent febrile seizure recurrences. Pediatrics. 1998;102:E51.PubMedCrossRef van Stuijvenberg M, Derksen-Lubsen G, Steyerberg EW, Habbema JD, Moll HA. Randomized, controlled trial of ibuprofen syrup administered during febrile illnesses to prevent febrile seizure recurrences. Pediatrics. 1998;102:E51.PubMedCrossRef
160.
Zurück zum Zitat Udani V, Pujar S, Munot P, Maheshwari S, Mehta N. Natural history and magnetic resonance imaging follow-up in 9 Sturge-Weber Syndrome patients and clinical correlation. J Child Neurol. 2007;22:479–83.PubMedCrossRef Udani V, Pujar S, Munot P, Maheshwari S, Mehta N. Natural history and magnetic resonance imaging follow-up in 9 Sturge-Weber Syndrome patients and clinical correlation. J Child Neurol. 2007;22:479–83.PubMedCrossRef
161.
Zurück zum Zitat Bay MJ, Kossoff EH, Lehmann CU, Zabel TA, Comi AM. Survey of aspirin use in Sturge-Weber syndrome. J Child Neurol. 2011;26:692–702.PubMedCrossRef Bay MJ, Kossoff EH, Lehmann CU, Zabel TA, Comi AM. Survey of aspirin use in Sturge-Weber syndrome. J Child Neurol. 2011;26:692–702.PubMedCrossRef
162.
Zurück zum Zitat Lance EI, Sreenivasan AK, Zabel TA, Kossoff EH, Comi AM. Aspirin use in Sturge-Weber syndrome: side effects and clinical outcomes. J Child Neurol. 2013;28:213–8.PubMedCrossRef Lance EI, Sreenivasan AK, Zabel TA, Kossoff EH, Comi AM. Aspirin use in Sturge-Weber syndrome: side effects and clinical outcomes. J Child Neurol. 2013;28:213–8.PubMedCrossRef
163.
Zurück zum Zitat Godfred RM, Parikh MS, Haltiner AM, Caylor LM, Sepkuty JP, Doherty MJ. Does aspirin use make it harder to collect seizures during elective video-EEG telemetry? Epilepsy Behav. 2013;27:115–7.PubMedCrossRef Godfred RM, Parikh MS, Haltiner AM, Caylor LM, Sepkuty JP, Doherty MJ. Does aspirin use make it harder to collect seizures during elective video-EEG telemetry? Epilepsy Behav. 2013;27:115–7.PubMedCrossRef
Metadaten
Titel
Cyclooxygenase-2 (COX-2) inhibitors: future therapeutic strategies for epilepsy management
verfasst von
Chitra Rawat
Samiksha Kukal
Ujjwal Ranjan Dahiya
Ritushree Kukreti
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2019
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-019-1592-3

Weitere Artikel der Ausgabe 1/2019

Journal of Neuroinflammation 1/2019 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Demenzkranke durch Antipsychotika vielfach gefährdet

Demenz Nachrichten

Der Einsatz von Antipsychotika gegen psychische und Verhaltenssymptome in Zusammenhang mit Demenzerkrankungen erfordert eine sorgfältige Nutzen-Risiken-Abwägung. Neuen Erkenntnissen zufolge sind auf der Risikoseite weitere schwerwiegende Ereignisse zu berücksichtigen.

Schlaganfall: frühzeitige Blutdrucksenkung im Krankenwagen ohne Nutzen

31.05.2024 Apoplex Nachrichten

Der optimale Ansatz für die Blutdruckkontrolle bei Patientinnen und Patienten mit akutem Schlaganfall ist noch nicht gefunden. Ob sich eine frühzeitige Therapie der Hypertonie noch während des Transports in die Klinik lohnt, hat jetzt eine Studie aus China untersucht.

Nicht Creutzfeldt Jakob, sondern Abführtee-Vergiftung

29.05.2024 Hyponatriämie Nachrichten

Eine ältere Frau trinkt regelmäßig Sennesblättertee gegen ihre Verstopfung. Der scheint plötzlich gut zu wirken. Auf Durchfall und Erbrechen folgt allerdings eine Hyponatriämie. Nach deren Korrektur kommt es plötzlich zu progredienten Kognitions- und Verhaltensstörungen.