This is an open access publication, available online and distributed under the terms of a Creative Commons Attribution-Non Commercial-No Derivatives 4.0 International licence (CC BY-NC-ND 4.0), a copy of which is available at https://creativecommons.org/licenses/by-nc-nd/4.0/. Subject to this license, all rights are reserved.
NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.
Noebels JL, Avoli M, Rogawski MA, et al., editors. Jasper's Basic Mechanisms of the Epilepsies. 5th edition. New York: Oxford University Press; 2024. doi: 10.1093/med/9780197549469.003.0064
Abstract
Drug and therapy screening in models of epilepsy have made considerable advances in recent years. Rodent models of seizures are widely used in drug development and have helped to evaluate several approved antiseizure drugs (ASDs) over the last four decades. However, current ASDs are only fully effective in approximately two-thirds of patients, with the remaining populations experiencing drug-refractory seizures. Advances in understanding of additional model systems have provided new avenues for screening and evaluation of novel therapies in epilepsy. Zebrafish and Drosophila systems allow for high-throughput (vs. rodent assays) screening and drug repurposing studies and can be adapted for specific genetic populations. Similarly, mouse models based on genetic mutations corresponding to human pathology can be used and manipulated to study pathophysiology and screen novel compounds. Special populations of epilepsy, such as individuals developing seizures arising from central nervous system infection, can be modeled by viral-induced epilepsy models. The application of chemically induced seizures has been streamlined into moderate throughput assays in mice subjected to intra-amygdala or intra-hippocampal kainic acid, generating mice with repetitive seizures and other behavioral characteristics consistent with temporal lobe epilepsy. Finally, patient-derived induced pluripotent stem cells offer an opportunity to study patient-derived organoids, in the context of patient-specific genetic backgrounds, where drug screening can be used as a personalized medicine approach. These new approaches to drug development offer opportunities to complement and improve upon traditional screening approaches in rodents, in an effort to improve approaches to hard-to-treat epilepsy conditions.
Introduction
Recent developments in our understanding of the etiologies of many types of epilepsy, combined with a host of new experimental model systems, have brought significant changes in the way that new therapies are developed and assessed in preclinical studies. This is therefore an exciting time in epilepsy therapy development. It is hypothesized that with sufficient knowledge of the mechanisms underlying epilepsy, the development of appropriate biomarkers (Simonato et al., 2021), and utilization of the right model systems, new therapies will be identified that go beyond simply stopping seizures and are also disease modifying. In this chapter, we will discuss several new experimental approaches, from cell-based assays to whole organisms, that are being used to assess the potential efficacy of new therapies in stopping seizures. While therapy development has traditionally referred to the development of small molecules, a variety of alternative approaches, many of which are described in numerous chapters in this edition, can be explored, developed, and validated with the model systems described in this chapter. Biologics, antisense oligonucleotides, neuromodulation approaches (e.g., electrical stimulation, focused ultrasound, transcranial magnetic stimulation, and optogenetics), diets, viral transfection, and cell-based transplantation therapies are all amenable to these novel preclinical approaches and hold significant promise for addressing the unmet clinical needs of the person with epilepsy or at risk for developing epilepsy.
Zebrafish and Other Model Organisms
Armed with the information derived from the explosion in identification of genetic mutations that underlie epilepsy, the development of the use of zebrafish larvae models has been incredibly successful in high-throughput approaches for therapy discovery. Zebrafish models offer numerous advantages in drug development, including the ability to use CRISPR-Cas9 systems to rapidly introduce epilepsy conferring mutations efficiently, rapid phenotypic characterization, and the ability to use a combination of approaches such as behavioral assays, motion detection, imaging, and electrophysiology to validate the ability of promising compounds to stop not only convulsive activity but also electrographic seizure events and comorbid behaviors. Of note, this approach can provide a rapid path for FDA-approved drugs to enter clinical trials for repurposing in the treatment of epilepsy. This was recently accomplished using a zebrafish larvae model of Dravet syndrome (DS), a catastrophic epileptic encephalopathy that often results in therapy-resistant seizures that increase the risk of sudden unexplained death in epilepsy (SUDEP). In the majority of patients, DS results as a consequence of loss-of-function mutations in the SCN1A gene, which encodes the pore-forming α-subunit of a voltage-gated sodium channel (Nav1.1) (Harkin et al., 2007). A loss-of-function missense mutation in the zebrafish ortholog, scn1Lab, was found to induce convulsive behavior and electrographic ictal-like activity in mutant larvae as early as 3 days post fertilization (dpf) and were thus amenable to screening hundreds of compounds. Through this initial ground-breaking work, Baraban and colleagues identified several FDA-approved compounds that were efficacious in blocking both the convulsive behaviors and electrographic ictal activity, which paved the way for clinical trials for clemizole, an antihistamine that also has activity at serotonergic receptors (Baraban et al., 2013; Dinday et al., 2015). Additional hits of FDA-approved drugs also act through a number of mechanisms at the serotonergic receptor, which as a consequence largely of this work, is now viewed as a novel target for the treatment of seizures in DS (Griffin et al., 2017).
While there are significant advantages to screening for ASDs in zebrafish, like all model systems, there are also limitations in the utility of this approach. For example, as recently noted (Griffin et al., 2021), if only behavioral motor assays are used for phenotypic analysis, in the absence of electrophysiology or calcium imaging, phenotypes may not be accurately assessed. In addition, as is true for mouse models, phenotypic penetrance of seizures may be different than in humans because of gene-gene interactions, developmental regulation of gene expression, and lack of cortical development (Griffin et al., 2021). Nevertheless, the ability to use zebrafish in high-throughput assays has put them at the forefront of drug discovery in epilepsy and other disorders (Shcheglovitov et al., 2021).
Drosophila melanogaster Models of Epilepsy
As has been done in zebrafish, Drosophila have also been used to generate mutations found in the orthologs of human epilepsy-conferring genes. Drosophila breed rapidly, have numerous conserved genes, are easy to genetically manipulate, and can provide high-throughput screening opportunities. Drug administration is straightforward and can be achieved by administration in food, and the ability to induce seizures can then be evaluated in the presence and absence of ASDs. For a comprehensive review of the use of Drosophila to model developmental epileptic encephalopathies (DEEs), see Takai et al. (2020). While to date, no novel compounds or targets have been identified using Drosophila as a model organism, the ease with which basic mechanisms can be identified, coupled with high-throughput approaches and ease of genetic manipulations, make it likely that this model system, like zebrafish, can also yield promising novel approaches to therapy discovery.
Mouse Models of Genetic Epilepsy and Therapy Development
As is the case with model organisms, induced pluripotent stem cells (iPSCs), and other in vitro neuronal preparations, it is now straightforward to introduce epilepsy-conferring mutations into mice, a commonly used species for preclinical therapy development. The highly conserved nature of proteins between mice and humans, a complex cortex, and the ability to monitor seizure activity with video electroencephalogram (EEG) combine to provide the research community with tractable animal models to not only determine the mechanisms underlying seizures because of specific mutations but also to develop targeted therapies for the treatment of seizures. As next-generation sequencing has identified epilepsy-conferring mutations in more than 80 genes, the number of animal models has also risen dramatically, and several recent reviews comprehensively summarize the face validity of many of the available models (Fallah et al., 2020; Griffin et al., 2018; Maljevic et al., 2017; Wang et al., 2021). Mouse models for Fragile X syndrome, Rett syndrome, CDKL5 deficiency disorder, tuberous sclerosis, and those for numerous DS mutations have been shown to have spontaneous seizures and relevant comorbidities (Fallah et al., 2020; Wong et al., 2016). With respect to ASD preclinical pharmacology, perhaps the most common mouse genetic models used to date are those with DS mutations. Mouse models with a variety of DS-conferring mutations are available, wherein mice are observed to have hyperthermia-induced seizures, spontaneous seizures, behavioral comorbidities, and SUDEP (Griffin et al., 2018). Hyperthermia-induced seizures allow for precise timing of drug delivery (Fig. 64–2), as seizures are induced within minutes of a rise in core body temperature and have been used extensively in several different mouse models of DS to evaluate the ability of different prototype and investigational compounds to prevent seizures (Anderson et al., 2019, 2022; Cao et al., 2012; Hawkins et al., 2017a, 2017b, 2021; Kaplan et al., 2017; Kharouf et al., 2020; Silenieks et al., 2019).

Figure 64–2.
A diagram of the Dravet mouse hyperthermia assay. Seizures are induced when there is a rise in the core body temperature of a Dravet syndrome mouse. This model can be used to evaluate the ability of a compound to increase the temperature at which an (more...)
A DS mouse model is now in use in the screening paradigm of the contract site of the epilepsy therapy screening program (ETSP) as both hyperthermia-induced seizures as well as spontaneous seizures are observed in the Scn1aA1783V/WT mouse. This represents the first time in the history of the program wherein a genetic model of human epilepsy is in use. The Scn1aA1783V/WT is a conditional knock-in, loss-of-function mutation with both hyperthermia-induced and spontaneous seizures that reliably persist into adulthood. Thus, this mouse represents a useful model of DS in which to screen for novel therapies. The effect of a battery of prototype ASDs on hyperthermia-induced seizures in this mouse model was recently published (Pernici et al., 2021a). While clobazam, levetiracetam, and tiagabine were found to effectively shift the temperature at which seizures occur to significantly higher temperatures, indicating potential therapeutic value, the sodium channel blocker, carbamazepine, shifted the temperature at which seizures occur to a lower temperature, which is consistent with findings in patients and other mouse models of DS (Anderson et al., 2019; Hawkins et al., 2017a). Numerous other ASDs tested in this model had no effect on the temperature at which seizures occur, indicating that this model represents a refractory model of hyperthermia-induced seizures and may be useful in finding novel investigational compounds. Additionally, a combination dosing approach, wherein clobazam and valproic acid are administered to mice, along with an additional investigational compound, seeks to mimic the recommended clinical treatment algorithm for patients with DS (Cross et al., 2019). Under those testing conditions, stiripentol was efficacious in combination, whereas it was without effect when administered alone in this assay. This suggests that the combined use of clobazam and valproic acid with an add-on investigational compound may be a useful strategy for screening novel therapies in this refractory mouse model of DS. The complex pharmacology of such a treatment regimen would necessitate the use of pharmacokinetic evaluation, especially before proceeding to subchronic dosing regimens.
As has been noted previously (Griffin et al., 2018), it is currently unclear what relationship exists between evoked hyperthermia-induced seizures and true epilepsy, where spontaneous, unprovoked seizures occur. Therefore, pharmacology studies in animal models wherein spontaneous seizures are observed may prove useful as a secondary screen following identification of active compounds in hyperthermia-induced seizures. To that end, the contract site of the ETSP has also evaluated spontaneous seizures that occur in both male and female Scn1aA1783/WT mice (Pernici et al., 2021b). Male Scn1aA1783V/WT mice exhibit significantly increased seizure frequency (1.1 ± 0.7 seizures/day) as compared to female mice (0.7 ± 0.6 seizures/day), and the seizures often occur in clusters, with the potential to have several days between seizures. These baseline seizure frequency data have allowed us to perform a power analysis to determine the optimal length of time to treat mice with ASDs. We estimate that to detect a 50% reduction in seizures and achieve 80% power at a 95% significance level, in a combined cohort of males and females, we will need an n = 24 mice with 10 days of treatment. This accounts for both seizure clustering and low frequency of seizures. Given the capacity of the contract site of the ETSP, these labor-intensive video EEG experiments can be performed to test the most promising novel compounds for the ability to prevent spontaneous seizures in this mouse model of DS. A limitation of these subchronic experiments, however, is that there is a need for high-quality pharmacokinetic data to inform treatment regimens for 10 days of administration to mice. Numerous injections per day are not always feasible or practical, and not all compounds are orally available if delivered in food (Anderson et al., 2022). Nevertheless, subchronic administration may reveal potential adverse effects that are not always observable following single acute administration and can provide important evidence regarding the ability of a novel compound to reduce spontaneous seizures.
While there are numerous examples of compounds that are effective in mouse models of seizures and epilepsy failing to ultimately reach the clinic, rodent testing has identified numerous compounds that are now approved for clinical use as epilepsy treatments (Kehne et al., 2017). Additionally, those approved compounds were often first identified using naïve animals with acute seizures induced by electrical stimulation, such as the maximal electroshock seizure (MES) and 6 Hz tests. What we currently do not know is whether using genetic mouse models for therapy discovery will yield clinically successful drugs in discrete patient populations. However, this hypothesis will certainly be tested going forward. Although it is not currently feasible to generate mouse models of all relevant epilepsy-conferring mutations (indeed, hundreds of pathogenic mutations are associated with DS for SCN1A alone; Harkin et al., 2007), recent advances in the use of model systems and iPSCs can provide high-throughput screening approaches that can narrow the search for viable molecular targets and novel therapies for the treatment of epilepsy that arises as a consequence of genetic mutations (Griffin et al., 2021; Shcheglovitov et al., 2021). Once targets and compounds and therapeutic approaches are identified, further testing in genetic rodent models of epilepsy may provide important evidence supporting the development of these approaches and move them closer to use in the clinic.
Theiler’s Murine Encephalomyelitis Virus Mouse Model
Viral infections can produce seizures and, in some cases, promote the development of epilepsy (Eeg-Olofsson, 2003; Ficker et al., 1998; Pardo et al., 2014). Injection of Theiler’s murine encephalomyelitis virus (TMEV) intracortically in mice has been used as a model of viral-induced seizures and temporal lobe epilepsy (TLE) (Libbey et al., 2008; Stewart et al., 2010a, 2010b). Both spontaneous and handling-induced seizures occur following central infection with the Daniels strain of TMEV (Broer et al., 2016; Patel et al., 2019; Stewart et al., 2010a; Waltl et al., 2018a, 2018b). Further, this mouse model has been used as a means to screen compounds with antiseizure and anti-inflammatory mechanisms of action (Barker-Haliski et al., 2015; Cusick et al., 2013; Metcalf et al., 2021; Patel et al., 2019), and it has been incorporated into the screening program for the National Institute of Neurological Disorders and Stroke (NINDS) ETSP. Seizures resulting from central nervous system (CNS) infection may result from central inflammation and/or fever (Pardo et al., 2014). While seizures may be treated with traditional ASDs, drugs with anti-inflammatory mechanisms may also be used. Therefore, this animal model offers the opportunity to not only differentiate ASDs for their efficacy against infection-related seizures, but it may also help to identify compounds with unique mechanisms.
Using a standard screening approach wherein mice are subjected to twice-daily drug injections followed by handling sessions and quantification of behavioral seizures, several medications have been evaluated. Typically, handling-induced seizures begin 2–3 days following infection, peaking around 5 days post infection and stop by 8–10 days post infection (Fig. 64–1A). Spontaneous seizures have also been observed to occur in the weeks following initial infection (Fig. 64–1B). However, the rate of spontaneous seizures observed is relatively low and requires large numbers of mice monitored under continuous video-EEG in order to evaluate potential therapies for their effects on spontaneous seizures in this model. However, handling-induced seizures occur at a high rate (50%–70% of mice infected) (Metcalf et al., 2021) and are amenable to screening. ASDs administered during the acute infection period (day 3–7 post infection) with varying mechanisms of action were effective in reducing seizures in this model. When available, the median effective dose (ED50) in the 6 Hz seizure model was used to inform dose selection, and when efficacy was observed at well-tolerated doses, additional doses were administered in order to evaluate dose–response relationships. Sodium channel blockers were differentially effective, with lacosamide and phenytoin significantly reducing seizures, whereas carbamazepine and lamotrigine were ineffective (Metcalf et al., 2021). Similarly, compounds acting on GABA were also differentially effective, with tiagabine and phenobarbital reducing seizures, whereas clonazepam was ineffective (Metcalf et al., 2021). Gabapentin, levetiracetam, topiramate, and valproic acid were also effective in reducing handling-induced seizures (Metcalf et al., 2021). However, while several ASDs reduced handling-induced seizures in this model, none were able to completely eliminate seizures.

Figure 64–1.
Mouse model of viral-induced epilepsy. A. C57BL/6J mice are intracerebrally injected with Theiler’s murine encephalomyelitis virus (TMEV), a neurotropic virus member of the Picornaviridae family that can cause encephalitis in infected mice. Between (more...)
TMEV infection in mice results in substantial elevations in cytokines (Kirkman et al., 2010; Libbey et al., 2011), and compounds that reduce inflammation may reduce seizures in this model. Further, counteracting the innate immune response in this model that contributes to seizures represents a unique antiseizure mechanism that may not be observed in other rodent models (e.g., acute electrically or chemically induced seizures). Nonsteroidal anti-inflammatory drugs (diclofenac, ibuprofen), steroids (prednisone, dexamethasone), the cyclooxygenase-2 inhibitor celecoxib, and the microglial inhibitor minocycline were administered during the acute infection period. Doses used had previously been observed to reduce inflammation or inflammation-related behaviors (e.g., pain responses) in other studies. Of these, a mild effect was observed with celecoxib and prednisone, and moderate efficacy was observed with dexamethasone and minocycline (Barker-Haliski et al., 2016; Metcalf et al., 2021).
In summary, TMEV infection and handling-induced seizures represents a unique model for screening compounds with antiseizure and/or anti-inflammatory mechanisms of action. Efficacy conferred on handling-induced seizures may be followed with confirmatory studies using video-EEG. However, since video-EEG studies are much more labor intensive, this approach should be reserved for the most promising compounds. ASDs reducing seizures in this model are likely effective due to their primary modes of action, although some may directly or indirectly reduce inflammation and thereby reduce seizures in this model. For example, levetiracetam was effective in this model across a wide range of doses and is also known to reduce inflammation (Stienen et al., 2011; Thone et al., 2012). Additional studies would be required, however, to differentiate antiseizure and anti-inflammatory effects for compounds where both potential mechanisms are present. Anti-inflammatory compounds evaluated thus far have generally been those with broad mechanisms. However, TMEV infection produces elevated reactive oxygen species, TGF-β, IL-6, TNFα, and IL-1β (Cusick et al., 2013; Kaufer et al., 2018; Kirkman et al., 2010; Libbey et al., 2008, 2010, 2011; Patel et al., 2017), and compounds acting specifically on these processes and cytokines may prove useful in reducing behavioral seizures and long-term outcomes (Bhuyan et al., 2015; Loewen et al., 2016; Wilcox et al., 2014). Identification of compounds with pronounced efficacy in this model may provide guidelines for treatment not only of infection-induced epilepsy but also seizure conditions where inflammation is pronounced.
Intra-Amygdala Kainate and Intra-Hippocampal Kainate Mouse Models
As discussed elsewhere in this text, delivery of the chemoconvulsant kainate (KA) to rodents induces status epilepticus (SE) and the subsequent development of mesial TLE (MTLE). KA can be administered via several routes (e.g., systemic, intra-nasal, intra-hippocampal, and intra-amygdala), each possessing unique experimental advantages and disadvantages, resulting in varying homology to the neuropathological features of MTLE in humans (Rusina et al., 2021). Both systemic administration of KA to rats and intra-hippocampal microinjection of KA to mice produce models of MTLE that are actively being used by the ETSP to evaluate investigational compounds for their effects on pharmacoresistant spontaneous recurrent seizures (SRSs) and hippocampal paroxysmal discharges (HPDs), respectively (Thomson et al., 2020; Wilcox et al., 2020). However, these models are not a panacea for all of the features of MTLE, nor do they represent the only models tractable enough to be useful in the further differentiation of investigational compounds under the auspices of the ETSP. Accordingly, additional KA-mediated SE models are continuing to be evaluated for inclusion in the ETSP pharmacoresistance drug screening efforts.
The intra-amygdala kainate (IAK) model, originally developed in rats (Ben-Ari et al., 1979, 1980) and adapted for mice in the early 2000s (Araki et al., 2002), has been used by a number of laboratories to investigate neuropathology, epileptogenesis, and MTLE resulting from focal microinjection of KA (Iori et al., 2017; Jimenez-Pacheco et al., 2013; Kondratiuk et al., 2015; Li et al., 2008, 2013a; Welzel et al., 2020). Similar to the systemic KA rat model, microinjection of KA in the mouse amygdala leads to SE, a seizure-free latent period, and ultimately the development of behavioral and electrographic SRSs. However, unlike models that employ systemic injections of KA and lead to widespread damage, IAK microinjection in mice causes limited and primarily unilateral hippocampal damage similar to human MTLE (Mouri et al., 2008). Furthermore, IAK mouse models offer several advantages that make them particularly well suited for assessing the efficacy of antiseizure therapies (e.g., SRSs manifest after a short latent period, occur with a relatively stable frequency, and mice require smaller quantities of often precious experimental compounds) (Jimenez-Pacheco et al., 2016; West et al., 2022). For these reasons, a mouse variant of the IAK microinjection model of MTLE has been developed (Fig. 64–3), pharmacologically characterized using standard ASDs, and adopted by the ETSP (West et al., 2022). Using five ASDs representing three mechanistic classes, this model was shown to produce SRSs that are resistant to two sodium channel-inhibiting ASDs (phenytoin and carbamazepine) and partially sensitive to GABA receptor-modulating ASDs (diazepam and phenobarbital) or a mixed-mechanism ASD (valproate). Although a more comprehensive examination of the effects of additional ASDs, including additional dose–response relationships combined with pharmacokinetic evaluations of plasma and brain concentrations, will be necessary in order to fully appraise this model, these data alone make a compelling case for the designation of SRSs in these mice as pharmacoresistant.

Figure 64–3.
Microinjection of kainic acid (KA) into the mouse amygdala induces status epilepticus (SE) and the subsequent development of spontaneous recurrent seizures (SRS). A. Schematic showing coordinates for injection cannula and CA1 electrode implantation sites. (more...)
Similar to the IAK model, the intra-hippocampal kainate (IHK) model shares the principal advantage of creating a focal lesion with limited and primarily unilateral damage akin to human MTLE. Microinjection of KA into the hippocampus has been performed in rats (Schwarcz et al., 1978), mice (Bouilleret et al., 1999), guinea pigs (Carriero et al., 2012), and cats (Tanaka et al., 1992). Furthermore, KA can be injected into either the dorsal or ventral hippocampus (Zeidler et al., 2018). As discussed elsewhere in this text, a unique feature of the IHK mouse model is the occurrence of relatively high-frequency electrographic seizures in the forms of high-voltage sharp waves (HVSWs) and hippocampal paroxysmal discharges (HPDs) (Klee et al., 2017; Twele et al., 2017). The high frequency of these electrographic events facilitates the screening of novel investigational ASDs because these experiments do not require multiday continuous video-EEG evaluations as required when measuring drug effects on the lower-frequency behavioral (electroclinical) SRSs. That said, the pharmacological sensitivity of behavioral seizures in IHK models is still of great interest, but using these models for this purpose has been challenging. For instance, while the rat IHK model produces frequent behavioral SRSs (Klee et al., 2017; Rattka et al., 2013), the evaluation of precious investigational compounds, which may have limited supply, is expensive (if at all possible) in relatively large rats. Conversely, mice require less quantity of a compound in order to perform subchronic administration experiments, but behavioral SRSs in the IHK mouse model have historically been reported to be infrequent and highly variable (Bielefeld et al., 2017; Klee et al., 2017; Welzel et al., 2020). Promising with regards to the mouse IHK model is a recent report by Lisgaras and Scharfman that describes a new implementation of the mouse IHK model exhibiting robust and frequent (2–4 per day) behavioral SRSs (Lisgaras et al., 2022). It is expected that this version of the IHK mouse, with its higher frequency electroclinical SRSs, may be adequate for the evaluation of their SRS sensitivities to classical and novel pharmacological interventions.
Induced Pluripotent Stem Cells and the Future
Pluripotent stem cells derived from patients have emerged as a means to study neuronal excitability and pathogenesis in a patient-specific manner. Traditional screening models in rodents are generally performed using relatively homogenous animal populations (e.g., inbred mouse strains) and do not reproduce any patient-specific pathophysiology contributing to seizures. By contrast, iPSC-derived organoids may offer a patient-specific opportunity to study epilepsy and antiseizure pharmacology (Shcheglovitov et al., 2021). Patient fibroblasts obtained from skin biopsies, for example, are subjected to a mixture of regulatory factors (e.g., Yamanaka factors) that induce pluripotency and then neural tissue generation (Grainger et al., 2018). iPSCs can be differentiated into a number of different cell types, including, in the context of epilepsy, neurons and glial cells (Cunningham et al., 2014; Krencik et al., 2011; Liu et al., 2013b; Maroof et al., 2013; Yu et al., 2014). For example, in addition to neurons and interneurons (Kim et al., 2014; Liu et al., 2013b), several studies have demonstrated that stem-cell-derived astrocytes reproduce physiologic functions of this cell type, including synaptic localization, gliotransmission, and calcium propagation (Hill et al., 2016; Shaltouki et al., 2013). Further, the heterogeneity of iPSC cultures recapitulates key network phenotypes, including a balance between excitation and inhibition and drug responsiveness (Ishii et al., 2017; Pei et al., 2016; Tukker et al., 2018). Further, iPSCs incorporate the patient’s genetic background, allowing for evaluation of different mutations types (variants), such as those that occur in DS. Several laboratories have generated iPSCs for patients with DS (Isom, 2017; Liu et al., 2013c; Sun et al., 2016, 2018; Tanaka et al., 2018; Tidball et al., 2020), which may lead to bench-to-bedside screening and therapy (Tidball et al., 2020). Thus, this approach can be used to both identify physiologic changes inherent with specific mutations and variants, such as changes in ion channel responses (Livide et al., 2015; Ricciardi et al., 2012; Tidball et al., 2020), and screen-specific treatments.
Under certain growing conditions, iPSCs will self-organize into spheroid structures (brain organoids) and, given sufficient time and growth conditions, become reminiscent of brain structures such as the hippocampus (Yu et al., 2014). Brain organoids are a monolayer in culture and consist of heterogenous cell populations of cells that can be further isolated and studied under various experimental conditions. For example, iPSC-derived forebrain neurons from patients with DS have been used to study ion channel conductance and demonstrate hyperexcitable phenotypes (Jiao et al., 2013; Liu et al., 2013c). Furthermore, three-dimensional brain organoids supported by hydrogels or other matrices have been generated from human iPSCs, recapitulating distinct brain structures and functions (Hirose et al., 2020).
Finally, advances in genome editing technology have further enhanced the utility of iPSCs and brain organoids. For example, the use of CRISPR/Cas9 approaches has allowed for reprogramming of iPSC lines for various purposes and the development of isogenic cell lines (e.g., cell lines where the mutation of interest is absent) (Tidball et al., 2017). Isogenic controls can be a critical missing piece from initial studies using patient-derived iPSC approaches, as they are required for proper interpretation of outcomes (Rowe et al., 2019). Therefore, gene editing has not only filled this gap but will continue to inform iPSC organoid studies.
Summary and Conclusions
While traditional animal models of seizures (e.g., MES, 6 Hz, and kindling models) have helped to identify and develop numerous clinically available ASDs over the last four decades, there continues to be an unmet clinical need to treat seizures in the nearly 37% of patients who remain refractory to existing therapies (Chen et al., 2018). The advent of the genetic revolution has greatly increased our understanding of the etiology of many types of epilepsy, and the ability to introduce pathogenic gene variants into a number of organisms, such as zebrafish and mice, provides the research community with an array of new assays to identify novel therapeutics. In addition, the use of spontaneously seizing animals to screen for these therapies has greatly changed the nature of preclinical evaluation of drugs. While these labor-intensive studies often require large numbers of animals to sufficiently power experiments, some groups have suggested utilizing multicenter preclinical studies to increase throughput and rigor and reproducibility (Pitkanen et al., 2019). Overall, it is hypothesized that the use of more etiologically relevant in vivo and in vitro assays will aid in the discovery of new therapies to treat those people with refractory epilepsy. Time will tell if this hypothesis holds.
Acknowledgments
This work was supported by R37NS065434 (KSW).
Disclosure Statement
The authors declare no relevant conflicts.
References
- Anderson LL, Absalom NL, Abelev SV, Low IK, Doohan PT, Martin LJ, Chebib M, McGregor IS, Arnold JC. Coadministered cannabidiol and clobazam: Preclinical evidence for both pharmacodynamic and pharmacokinetic interactions. Epilepsia. 2019;60(11):2224–34. doi: 10.1111/epi.16355. PMID: 31625159. [PMC free article: PMC6900043] [PubMed: 31625159]
- Anderson LL, Doohan PT, Hawkins NA, Bahceci D, Garai S, Thakur GA, Kearney JA, Arnold JC. The endocannabinoid system impacts seizures in a mouse model of Dravet syndrome. Neuropharmacology. 2022;205:108897. doi: 10.1016/j.neuropharm.2021.108897. PMID: 34822817. [PMC free article: PMC9514665] [PubMed: 34822817]
- Araki T, Simon RP, Taki W, Lan JQ, Henshall DC. Characterization of neuronal death induced by focally evoked limbic seizures in the C57BL/6 mouse. J Neurosci Res. 2002;69(5):614–21. doi: 10.1002/jnr.10356. PMID: 12210827. [PubMed: 12210827]
- Baraban SC, Dinday MT, Hortopan GA. Drug screening in Scn1a zebrafish mutant identifies clemizole as a potential Dravet syndrome treatment. Nat Commun. 2013;4:2410. doi: 10.1038/ncomms3410. PMID: 24002024. [PMC free article: PMC3891590] [PubMed: 24002024]
- Barker-Haliski ML, Dahle EJ, Heck TD, Pruess TH, Vanegas F, Wilcox KS, White HS. Evaluating an etiologically relevant platform for therapy development for temporal lobe epilepsy: effects of carbamazepine and valproic acid on acute seizures and chronic behavioral comorbidities in the Theiler’s murine encephalomyelitis virus mouse model. J Pharmacol Exp Ther. 2015;353(2):318–29. doi: 10.1124/jpet.114.222513. PMID: 25755209. [PMC free article: PMC4407718] [PubMed: 25755209]
- Barker-Haliski ML, Heck TD, Dahle EJ, Vanegas F, Pruess TH, Wilcox KS, White HS. Acute treatment with minocycline, but not valproic acid, improves long-term behavioral outcomes in the Theiler’s virus model of temporal lobe epilepsy. Epilepsia. 2016;57(12):1958–67. doi: 10.1111/epi.13577. PMID: 27739576. [PMC free article: PMC5154893] [PubMed: 27739576]
- Ben-Ari Y, Lagowska J, Tremblay E, Le Gal La Salle G. A new model of focal status epilepticus: intra-amygdaloid application of kainic acid elicits repetitive secondarily generalized convulsive seizures. Brain Res. 1979;163(1):176–9. doi: 10.1016/0006-8993(79)90163-x. PMID: 427540. [PubMed: 427540]
- Ben-Ari Y, Tremblay E, Ottersen OP. Injections of kainic acid into the amygdaloid complex of the rat: An electrographic, clinical and histological study in relation to the pathology of epilepsy. Neuroscience. 1980;5(3):515–28. doi: 10.1016/0306-4522(80)90049-4. [PubMed: 6892841]
- Bhuyan P, Patel DC, Wilcox KS, Patel M. Oxidative stress in murine Theiler’s virus-induced temporal lobe epilepsy. Exp Neurol. 2015;271:329–34. doi: 10.1016/j.expneurol.2015.06.012. PMID: 26079647. [PMC free article: PMC4848026] [PubMed: 26079647]
- Bielefeld P, Sierra A, Encinas JM, Maletic-Savatic M, Anderson A, Fitzsimons CP. A Standardized Protocol for Stereotaxic Intrahippocampal Administration of Kainic Acid Combined with Electroencephalographic Seizure Monitoring in Mice. Front Neurosci. 2017;11:160. doi: 10.3389/fnins.2017.00160. PMID: 28405182. [PMC free article: PMC5370320] [PubMed: 28405182]
- Bouilleret V, Ridoux V, Depaulis A, Marescaux C, Nehlig A, Le Gal La Salle G. Recurrent seizures and hippocampal sclerosis following intrahippocampal kainate injection in adult mice: electroencephalography, histopathology and synaptic reorganization similar to mesial temporal lobe epilepsy. Neuroscience. 1999;89(3):717–29. doi: 10.1016/s0306-4522(98)00401-1. PMID: 10199607. [PubMed: 10199607]
- Broer S, Kaufer C, Haist V, Li L, Gerhauser I, Anjum M, Bankstahl M, Baumgartner W, Loscher W. Brain inflammation, neurodegeneration and seizure development following picornavirus infection markedly differ among virus and mouse strains and substrains. Exp Neurol. 2016;279:57–74. doi: 10.1016/j.expneurol.2016.02.011. PMID: 26892877. [PubMed: 26892877]
- Cao D, Ohtani H, Ogiwara I, Ohtani S, Takahashi Y, Yamakawa K, Inoue Y. Efficacy of stiripentol in hyperthermia-induced seizures in a mouse model of Dravet syndrome. Epilepsia. 2012;53(7):1140–5. doi: 10.1111/j.1528-1167.2012.03497.x. PMID: 22578034. [PubMed: 22578034]
- Carriero G, Arcieri S, Cattalini A, Corsi L, Gnatkovsky V, de Curtis M. A guinea pig model of mesial temporal lobe epilepsy following nonconvulsive status epilepticus induced by unilateral intrahippocampal injection of kainic acid. Epilepsia. 2012;53(11):1917–27. doi: 10.1111/j.1528-1167.2012.03669.x. PMID: 22998690. [PubMed: 22998690]
- Chen Z, Brodie MJ, Liew D, Kwan P. Treatment Outcomes in Patients With Newly Diagnosed Epilepsy Treated With Established and New Antiepileptic Drugs: A 30-Year Longitudinal Cohort Study. JAMA Neurol. 2018;75(3):279–86. doi: 10.1001/jamaneurol.2017.3949. PMID: 29279892. [PMC free article: PMC5885858] [PubMed: 29279892]
- Cross JH, Caraballo RH, Nabbout R, Vigevano F, Guerrini R, Lagae L. Dravet syndrome: Treatment options and management of prolonged seizures. Epilepsia. 2019;60 Suppl 3:S39–S48. doi: 10.1111/epi.16334. PMID: 31904119. [PubMed: 31904119]
- Cunningham M, Cho JH, Leung A, Savvidis G, Ahn S, Moon M, Lee PK, Han JJ, Azimi N, Kim KS, Bolshakov VY, Chung S. hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice. Cell Stem Cell. 2014;15(5):559–73. doi: 10.1016/j.stem.2014.10.006. PMID: 25517465. [PMC free article: PMC4270101] [PubMed: 25517465]
- Cusick MF, Libbey JE, Patel DC, Doty DJ, Fujinami RS. Infiltrating macrophages are key to the development of seizures following virus infection. Journal of virology. 2013;87(3):1849–60. doi: 10.1128/JVI.02747-12. PMID: 23236075. [PMC free article: PMC3554195] [PubMed: 23236075]
- DePaula-Silva AB, Bell LA, Wallis GJ, Wilcox KS. Inflammation Unleashed in Viral-Induced Epilepto-genesis. Epilepsy currents. 2021;21(6):433–40. doi: 10.1177/15357597211040939. PMID: 34924851. [PMC free article: PMC8652320] [PubMed: 34924851]
- Dinday MT, Baraban SC. Large-Scale Phenotype-Based Antiepileptic Drug Screening in a Zebrafish Model of Dravet Syndrome. eNeuro. 2015;2(4). doi: 10.1523/ENEURO.0068-15.2015. PMID: 26465006. [PMC free article: PMC4596025] [PubMed: 26465006]
- Eeg-Olofsson O. Virological and immunological aspects of seizure disorders. Brain Dev. 2003;25(1):9–13. doi: 10.1016/s0387-7604(02)00162-6. PMID: 12536027. [PubMed: 12536027]
- Fallah MS, Eubanks JH. Seizures in Mouse Models of Rare Neurodevelopmental Disorders. Neuroscience. 2020;445:50–68. doi: 10.1016/j.neuroscience.2020.01.041. PMID: 32059984. [PubMed: 32059984]
- Ficker DM, So EL, Shen WK, Annegers JF, O’Brien PC, Cascino GD, Belau PG. Population-based study of the incidence of sudden unexplained death in epilepsy. Neurology. 1998;51(5):1270–4. doi: 10.1212/wnl.51.5.1270. PMID: 9818844. [PubMed: 9818844]
- Grainger AI, King MC, Nagel DA, Parri HR, Coleman MD, Hill EJ. In vitro Models for Seizure-Liability Testing Using Induced Pluripotent Stem Cells. Front Neurosci. 2018;12:590. doi: 10.3389/fnins.2018.00590. PMID: 30233290. [PMC free article: PMC6127295] [PubMed: 30233290]
- Griffin A, Carpenter C, Liu J, Paterno R, Grone B, Hamling K, Moog M, Dinday MT, Figueroa F, Anvar M, Ononuju C, Qu T, Baraban SC. Phenotypic analysis of catastrophic childhood epilepsy genes. Commun Biol. 2021;4(1):680. doi: 10.1038/s42003-021-02221-y. PMID: 34083748. [PMC free article: PMC8175701] [PubMed: 34083748]
- Griffin A, Hamling KR, Hong S, Anvar M, Lee LP, Baraban SC. Preclinical Animal Models for Dravet Syndrome: Seizure Phenotypes, Comorbidities and Drug Screening. Frontiers in Pharmacology. 2018;9. doi: 10.3389/fphar.2018.00573. [PMC free article: PMC5994396] [PubMed: 29915537]
- Griffin A, Hamling KR, Knupp K, Hong S, Lee LP, Baraban SC. Clemizole and modulators of serotonin signalling suppress seizures in Dravet syndrome. Brain. 2017;140(3):669–83. doi: 10.1093/brain/aww342. PMID: 28073790. [PMC free article: PMC6075536] [PubMed: 28073790]
- Harkin LA, McMahon JM, Iona X, Dibbens L, Pelekanos JT, Zuberi SM, Sadleir LG, Andermann E, Gill D, Farrell K, Connolly M, Stanley T, Harbord M, Andermann F, Wang J, Batish SD, Jones JG, Seltzer WK, Gardner A, Infantile Epileptic Encephalopathy Referral C, Sutherland G, Berkovic SF, Mulley JC, Scheffer IE. The spectrum of SCN1A-related infantile epileptic encephalopathies. Brain. 2007;130(Pt 3):843–52. doi: 10.1093/brain/awm002. PMID: 17347258. [PubMed: 17347258]
- Hawkins NA, Anderson LL, Gertler TS, Laux L, George AL, Jr., Kearney JA. Screening of conventional anticonvulsants in a genetic mouse model of epilepsy. Ann Clin Transl Neurol. 2017a;4(5):326–39. doi: 10.1002/acn3.413. PMID: 28491900. [PMC free article: PMC5420810] [PubMed: 28491900]
- Hawkins NA, Jurado M, Thaxton TT, Duarte SE, Barse L, Tatsukawa T, Yamakawa K, Nishi T, Kondo S, Miyamoto M, Abrahams BS, During MJ, Kearney JA. Soticlestat, a novel cholesterol 24-hydroxylase inhibitor, reduces seizures and premature death in Dravet syndrome mice. Epilepsia. 2021;62(11):2845–57. doi: 10.1111/epi.17062. PMID: 34510432. [PMC free article: PMC9291096] [PubMed: 34510432]
- Hawkins NA, Lewis M, Hammond RS, Doherty JJ, Kearney JA. The synthetic neuroactive steroid SGE-516 reduces seizure burden and improves survival in a Dravet syndrome mouse model. Sci Rep. 2017b;7(1):15327. doi: 10.1038/s41598-017-15609-w. PMID: 29127345. [PMC free article: PMC5681541] [PubMed: 29127345]
- Hill E, Nagel D, Parri R, Coleman M. Stem cell-derived astrocytes: are they physiologically credible? J Physiol. 2016;594(22):6595–606. doi: 10.1113/JP270658. PMID: 26634807. [PMC free article: PMC5108894] [PubMed: 26634807]
- Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci. 2020;108:103535. doi: 10.1016/j.mcn.2020.103535. PMID: 32758699. [PubMed: 32758699]
- Iori V, Iyer AM, Ravizza T, Beltrame L, Paracchini L, Marchini S, Cerovic M, Hill C, Ferrari M, Zucchetti M, Molteni M, Rossetti C, Brambilla R, Steve White H, D’Incalci M, Aronica E, Vezzani A. Blockade of the IL-1R1/TLR4 pathway mediates disease-modification therapeutic effects in a model of acquired epilepsy. Neurobiol Dis. 2017;99(1095-953X (Electronic)):12–23. doi: 10.1016/j.nbd.2016.12.007. PMID: 27939857. [PubMed: 27939857]
- Ishii MN, Yamamoto K, Shoji M, Asami A, Kawamata Y. Human induced pluripotent stem cell (hiPSC)-derived neurons respond to convulsant drugs when co-cultured with hiPSC-derived astrocytes. Toxicology. 2017;389:130–8. doi: 10.1016/j.tox.2017.06.010. PMID: 28666936. [PubMed: 28666936]
- Isom LL. Opposing Phenotypes in Dravet Syndrome Patient-Derived Induced Pluripotent Stem Cell Neurons: Can Everyone Be Right? Epilepsy Curr. 2017;17(4):244–7. doi: 10.5698/1535-7597.17.4.244. PMID: 29225534. [PMC free article: PMC5716123] [PubMed: 29225534]
- Jiao J, Yang Y, Shi Y, Chen J, Gao R, Fan Y, Yao H, Liao W, Sun XF, Gao S. Modeling Dravet syndrome using induced pluripotent stem cells (iPSCs) and directly converted neurons. Hum Mol Genet. 2013;22(21):4241–52. doi: 10.1093/hmg/ddt275. PMID: 23773995. [PubMed: 23773995]
- Jimenez-Pacheco A, Diaz-Hernandez M, Arribas-Blazquez M, Sanz-Rodriguez A, Olivos-Ore LA, Artalejo AR, Alves M, Letavic M, Miras-Portugal MT, Conroy RM, Delanty N, Farrell MA, O’Brien DF, Bhattacharya A, Engel T, Henshall DC. Transient P2X7 Receptor Antagonism Produces Lasting Reductions in Spontaneous Seizures and Gliosis in Experimental Temporal Lobe Epilepsy. J Neurosci. 2016;36(22):5920–32. doi: 10.1523/JNEUROSCI.4009-15.2016. PMID: 27251615. [PMC free article: PMC6601816] [PubMed: 27251615]
- Jimenez-Pacheco A, Mesuret G, Sanz-Rodriguez A, Tanaka K, Mooney C, Conroy R, Miras-Portugal MT, Diaz-Hernandez M, Henshall DC, Engel T. Increased neocortical expression of the P2X7 receptor after status epilepticus and anticonvulsant effect of P2X7 receptor antagonist A-438079. Epilepsia. 2013;54(9):1551–61. doi: 10.1111/epi.12257. [PubMed: 23808395]
- Kaplan JS, Stella N, Catterall WA, Westenbroek RE. Cannabidiol attenuates seizures and social deficits in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A. 2017;114(42):11229–34. doi: 10.1073/pnas.1711351114. PMID: 28973916. [PMC free article: PMC5651774] [PubMed: 28973916]
- Kaufer C, Chhatbar C, Broer S, Waltl I, Ghita L, Gerhauser I, Kalinke U, Loscher W. Chemokine receptors CCR2 and CX3CR1 regulate viral encephalitis-induced hippocampal damage but not seizures. Proc Natl Acad Sci U S A. 2018;115(38):E8929–E38. doi: 10.1073/pnas.1806754115. PMID: 30181265. [PMC free article: PMC6156634] [PubMed: 30181265]
- Kehne JH, Klein BD, Raeissi S, Sharma S. The National Institute of Neurological Disorders and Stroke (NINDS) Epilepsy Therapy Screening Program (ETSP). Neurochem Res. 2017;42(7):1894–903. doi: 10.1007/s11064-017-2275-z. PMID: 28462454. [PMC free article: PMC5504134] [PubMed: 28462454]
- Kharouf Q, Pinares-Garcia P, Romanelli MN, Reid CA. Testing broad-spectrum and isoform-preferring HCN channel blockers for anticonvulsant properties in mice. Epilepsy Res. 2020;168:106484. doi: 10.1016/j.eplepsyres.2020.106484. PMID: 33099130. [PubMed: 33099130]
- Kim TG, Yao R, Monnell T, Cho JH, Vasudevan A, Koh A, Peeyush KT, Moon M, Datta D, Bolshakov VY, Kim KS, Chung S. Efficient specification of interneurons from human pluripotent stem cells by dorsoventral and rostrocaudal modulation. Stem Cells. 2014;32(7):1789–804. doi: 10.1002/stem.1704. PMID: 24648391. [PMC free article: PMC4147720] [PubMed: 24648391]
- Kirkman NJ, Libbey JE, Wilcox KS, White HS, Fujinami RS. Innate but not adaptive immune responses contribute to behavioral seizures following viral infection. Epilepsia. 2010;51(3):454–64. doi: 10.1111/j.1528-1167.2009.02390.x. PMID: 19845729. [PMC free article: PMC3046460] [PubMed: 19845729]
- Klee R, Brandt C, Tollner K, Loscher W. Various modifications of the intrahippocampal kainate model of mesial temporal lobe epilepsy in rats fail to resolve the marked rat-to-mouse differences in type and frequency of spontaneous seizures in this model. Epilepsy Behav. 2017;68:129–40. doi: 10.1016/j.yebeh.2016.11.035. PMID: 28167446. [PubMed: 28167446]
- Kondratiuk I, Plucinska G, Miszczuk D, Wozniak G, Szydlowska K, Kaczmarek L, Filipkowski RK, Lukasiuk K. Epileptogenesis following Kainic Acid-Induced Status Epilepticus in Cyclin D2 Knock-Out Mice with Diminished Adult Neurogenesis. PloS one. 2015;10(5):e0128285. doi: 10.1371/journal.pone.0128285. PMID: 26020770. [PMC free article: PMC4447381] [PubMed: 26020770]
- Krencik R, Zhang SC. Directed differentiation of functional astroglial subtypes from human pluripotent stem cells. Nat Protoc. 2011;6(11):1710–7. doi: 10.1038/nprot.2011.405. PMID: 22011653. [PMC free article: PMC3198813] [PubMed: 22011653]
- Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, Boe AF, Boguski MS, Brockway KS, Byrnes EJ, Chen L, Chen L, Chen TM, Chin MC, Chong J, Crook BE, Czaplinska A, Dang CN, Datta S, Dee NR, Desaki AL, Desta T, Diep E, Dolbeare TA, Donelan MJ, Dong HW, Dougherty JG, Duncan BJ, Ebbert AJ, Eichele G, Estin LK, Faber C, Facer BA, Fields R, Fischer SR, Fliss TP, Frensley C, Gates SN, Glattfelder KJ, Halverson KR, Hart MR, Hohmann JG, Howell MP, Jeung DP, Johnson RA, Karr PT, Kawal R, Kidney JM, Knapik RH, Kuan CL, Lake JH, Laramee AR, Larsen KD, Lau C, Lemon TA, Liang AJ, Liu Y, Luong LT, Michaels J, Morgan JJ, Morgan RJ, Mortrud MT, Mosqueda NF, Ng LL, Ng R, Orta GJ, Overly CC, Pak TH, Parry SE, Pathak SD, Pearson OC, Puchalski RB, Riley ZL, Rockett HR, Rowland SA, Royall JJ, Ruiz MJ, Sarno NR, Schaffnit K, Shapovalova NV, Sivisay T, Slaughterbeck CR, Smith SC, Smith KA, Smith BI, Sodt AJ, Stewart NN, Stumpf KR, Sunkin SM, Sutram M, Tam A, Teemer CD, Thaller C, Thompson CL, Varnam LR, Visel A, Whitlock RM, Wohnoutka PE, Wolkey CK, Wong VY, Wood M, Yaylaoglu MB, Young RC, Youngstrom BL, Yuan XF, Zhang B, Zwingman TA, Jones AR. Genome-wide atlas of gene expression in the adult mouse brain. Nature. 2007;445(7124):168–76. doi: 10.1038/nature05453. PMID: 17151600. [PubMed: 17151600]
- Li T, Ren G, Lusardi T, Wilz A, Lan JQ, Iwasato T, Itohara S, Simon RP, Boison D. Adenosine kinase is a target for the prediction and prevention of epileptogenesis in mice. The Journal of Clinical Investigation. 2008;118(2):571–82. doi: 10.1172/JCI33737. [PMC free article: PMC2157568] [PubMed: 18172552]
- Libbey JE, Kennett NJ, Wilcox KS, White HS, Fujinami RS. Lack of correlation of central nervous system inflammation and neuropathology with the development of seizures following acute virus infection. Journal of virology. 2011;85(16):8149–57. doi: 10.1128/JVI.00730-11. PMID: 21680509. [PMC free article: PMC3147986] [PubMed: 21680509]
- Libbey JE, Kirkman NJ, Smith MC, Tanaka T, Wilcox KS, White HS, Fujinami RS. Seizures following picornavirus infection. Epilepsia. 2008;49(6):1066–74. doi: 10.1111/j.1528-1167.2008.01535.x. PMID: 18325012. [PubMed: 18325012]
- Libbey JE, Kirkman NJ, Wilcox KS, White HS, Fujinami RS. Role for complement in the development of seizures following acute viral infection. Journal of virology. 2010;84(13):6452–60. doi: 10.1128/JVI.00422-10. PMID: 20427530. [PMC free article: PMC2903264] [PubMed: 20427530]
- Lisgaras CP, Scharfman HE. Robust chronic convulsive seizures, high frequency oscillations, and human seizure onset patterns in an intrahippocampal kainic acid model in mice. Neurobiology of Disease. 2022:105637. doi: 10.1016/j.nbd.2022.105637. [PMC free article: PMC9034729] [PubMed: 35091040]
- Liu G, Gu B, He XP, Joshi RB, Wackerle HD, Rodriguiz RM, Wetsel WC, McNamara JO. Transient inhibition of TrkB kinase after status epilepticus prevents development of temporal lobe epilepsy. Neuron. 2013a;79(1):31–8. doi: 10.1016/j.neuron.2013.04.027. PMID: 23790754. [PMC free article: PMC3744583] [PubMed: 23790754]
- Liu Y, Liu H, Sauvey C, Yao L, Zarnowska ED, Zhang SC. Directed differentiation of forebrain GABA interneurons from human pluripotent stem cells. Nat Protoc. 2013b;8(9):1670–9. doi: 10.1038/nprot.2013.106. PMID: 23928500. [PMC free article: PMC4121169] [PubMed: 23928500]
- Liu Y, Lopez-Santiago LF, Yuan Y, Jones JM, Zhang H, O’Malley HA, Patino GA, O’Brien JE, Rusconi R, Gupta A, Thompson RC, Natowicz MR, Meisler MH, Isom LL, Parent JM. Dravet syndrome patient-derived neurons suggest a novel epilepsy mechanism. Ann Neurol. 2013c;74(1):128–39. doi: 10.1002/ana.23897. PMID: 23821540. [PMC free article: PMC3775921] [PubMed: 23821540]
- Livide G, Patriarchi T, Amenduni M, Amabile S, Yasui D, Calcagno E, Lo Rizzo C, De Falco G, Ulivieri C, Ariani F, Mari F, Mencarelli MA, Hell JW, Renieri A, Meloni I. GluD1 is a common altered player in neuronal differentiation from both MECP2-mutated and CDKL5-mutated iPS cells. Eur J Hum Genet. 2015;23(2):195–201. doi: 10.1038/ejhg.2014.81. PMID: 24916645. [PMC free article: PMC4172451] [PubMed: 24916645]
- Loewen JL, Barker-Haliski ML, Dahle EJ, White HS, Wilcox KS. Neuronal Injury, Gliosis, and Glial Proliferation in Two Models of Temporal Lobe Epilepsy. J Neuropathol Exp Neurol. 2016;75(4):366–78. doi: 10.1093/jnen/nlw008. PMID: 26945036. [PMC free article: PMC5009480] [PubMed: 26945036]
- Maljevic S, Reid CA, Petrou S. Models for discovery of targeted therapy in genetic epileptic encephalopathies. J Neurochem. 2017;143(1):30–48. doi: 10.1111/jnc.14134. PMID: 28742937. [PubMed: 28742937]
- Maroof AM, Keros S, Tyson JA, Ying SW, Ganat YM, Merkle FT, Liu B, Goulburn A, Stanley EG, Elefanty AG, Widmer HR, Eggan K, Goldstein PA, Anderson SA, Studer L. Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells. Cell Stem Cell. 2013;12(5):559–72. doi: 10.1016/j.stem.2013.04.008. PMID: 23642365. [PMC free article: PMC3681523] [PubMed: 23642365]
- Metcalf CS, Vanegas F, Underwood T, Johnson K, West PJ, Smith MD, Wilcox KS. Screening of prototype antiseizure and anti-inflammatory compounds in the Theiler’s murine encephalomyelitis virus model of epilepsy. Epilepsia open. 2021. doi: 10.1002/epi4.12550. PMID: 34668659. [PMC free article: PMC8886069] [PubMed: 34668659]
- Mouri G, Jimenez-Mateos E, Engel T, Dunleavy M, Hatazaki S, Paucard A, Matsushima S, Taki W, Henshall DC. Unilateral hippocampal CA3-predominant damage and short latency epileptogenesis after intra-amygdala microinjection of kainic acid in mice. Brain Res. 2008;1213(0006-8993 (Print)):140–51. doi: 10.1016/j.brainres.2008.03.061. PMID: 18455706. [PubMed: 18455706]
- Pardo CA, Nabbout R, Galanopoulou AS. Mechanisms of epileptogenesis in pediatric epileptic syndromes: Rasmussen encephalitis, infantile spasms, and febrile infection-related epilepsy syndrome (FIRES). Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. 2014;11(2):297–310. doi: 10.1007/s13311-014-0265-2. PMID: 24639375. [PMC free article: PMC3996116] [PubMed: 24639375]
- Patel DC, Wallis G, Dahle EJ, McElroy PB, Thomson KE, Tesi RJ, Szymkowski DE, West PJ, Smeal RM, Patel M, Fujinami RS, White HS, Wilcox KS. Hippocampal TNFalpha Signaling Contributes to Seizure Generation in an Infection-Induced Mouse Model of Limbic Epilepsy. eNeuro. 2017;4(2). doi: 10.1523/ENEURO.0105-17.2017. PMID: 28497109. [PMC free article: PMC5422919] [PubMed: 28497109]
- Patel DC, Wallis G, Fujinami RS, Wilcox KS, Smith MD. Cannabidiol reduces seizures following CNS infection with Theiler’s murine encephalomyelitis virus. Epilepsia open. 2019;4(3):431–42. doi: 10.1002/epi4.12351. PMID: 31440724. [PMC free article: PMC6698680] [PubMed: 31440724]
- Pei Y, Peng J, Behl M, Sipes NS, Shockley KR, Rao MS, Tice RR, Zeng X. Comparative neurotoxicity screening in human iPSC-derived neural stem cells, neurons and astrocytes. Brain Res. 2016;1638(Pt A):57–73. doi: 10.1016/j.brainres.2015.07.048. PMID: 26254731. [PMC free article: PMC5032144] [PubMed: 26254731]
- Pernici CD, Mensah JA, Dahle EJ, Johnson KJ, Handy L, Buxton L, Smith MD, West PJ, Metcalf CS, Wilcox KS. Development of an antiseizure drug screening platform for Dravet syndrome at the NINDS contract site for the Epilepsy Therapy Screening Program. Epilepsia. 2021a;62(7):1665–76. doi: 10.1111/epi.16925. PMID: 34002394. [PMC free article: PMC8360068] [PubMed: 34002394]
- Pernici CD, Spink A, Dahle EJ, Johnson KJ, Metcalf CS, West PJ, Wilcox KS. Evaluation of spontaneous seizure activity, sex-dependent differences, behavioral comorbidities, and alterations in CA1 neuron firing properties in a mouse model of Dravet Syndrome. bioRxiv. 2021b:2021.06.16.448684. doi: 10.1101/2021.06.16.448684.
- Pitkanen A, O’Brien TJ, Staba R. Preface—Practical and theoretical considerations for performing a multi-center preclinical biomarker discovery study of post-traumatic epileptogenesis: lessons learned from the EpiBioS4Rx consortium. Epilepsy Res. 2019;156:106080. doi: 10.1016/j.eplepsyres.2019.01.007. PMID: 30685321. [PMC free article: PMC6799991] [PubMed: 30685321]
- Rattka M, Brandt C, Loscher W. The intrahippocampal kainate model of temporal lobe epilepsy revisited: epileptogenesis, behavioral and cognitive alterations, pharmacological response, and hippoccampal damage in epileptic rats. Epilepsy Res. 2013;103(2-3):135–52. doi: 10.1016/j.eplepsyres.2012.09.015. PMID: 23196211. [PubMed: 23196211]
- Ricciardi S, Ungaro F, Hambrock M, Rademacher N, Stefanelli G, Brambilla D, Sessa A, Magagnotti C, Bachi A, Giarda E, Verpelli C, Kilstrup-Nielsen C, Sala C, Kalscheuer VM, Broccoli V. CDKL5 ensures excitatory synapse stability by reinforcing NGL-1-PSD95 interaction in the postsynaptic compartment and is impaired in patient iPSC-derived neurons. Nat Cell Biol. 2012;14(9):911–23. doi: 10.1038/ncb2566. PMID: 22922712. [PMC free article: PMC6485419] [PubMed: 22922712]
- Rowe RG, Daley GQ. Induced pluripotent stem cells in disease modelling and drug discovery. Nat Rev Genet. 2019;20(7):377–88. doi: 10.1038/s41576-019-0100-z. PMID: 30737492. [PMC free article: PMC6584039] [PubMed: 30737492]
- Rusina E, Bernard C, Williamson A. The Kainic Acid Models of Temporal Lobe Epilepsy. eNeuro. 2021;8(2). doi: 10.1523/ENEURO.0337-20.2021. PMID: 33658312. [PMC free article: PMC8174050] [PubMed: 33658312]
- Schwarcz R, Zaczek R, Coyle JT. Microinjection of kainic acid into the rat hippocampus. Eur J Pharmacol. 1978;50(3):209–20. doi: 10.1016/0014-2999(78)90353-9. PMID: 689077. [PubMed: 689077]
- Shaltouki A, Peng J, Liu Q, Rao MS, Zeng X. Efficient generation of astrocytes from human pluripotent stem cells in defined conditions. Stem Cells. 2013;31(5):941–52. doi: 10.1002/stem.1334. PMID: 23341249. [PubMed: 23341249]
- Shcheglovitov A, Peterson RT. Screening Platforms for Genetic Epilepsies-Zebrafish, iPSC-Derived Neurons, and Organoids. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics. 2021;18(3):1478–89. doi: 10.1007/s13311-021-01115-5. PMID: 34595731. [PMC free article: PMC8608971] [PubMed: 34595731]
- Silenieks LB, Carroll NK, Van Niekerk A, Van Niekerk E, Taylor C, Upton N, Higgins GA. Evaluation of Selective 5-HT(2C) Agonists in Acute Seizure Models. ACS Chem Neurosci. 2019;10(7):3284–95. doi: 10.1021/acschemneuro.8b00739. PMID: 31082204. [PubMed: 31082204]
- Simonato M, Agoston DV, Brooks-Kayal A, Dulla C, Fureman B, Henshall DC, Pitkanen A, Theodore WH, Twyman RE, Kobeissy FH, Wang KK, Whittemore V, Wilcox KS. Identification of clinically relevant biomarkers of epileptogenesis—a strategic roadmap. Nat Rev Neurol. 2021;17(4):231–42. doi: 10.1038/s41582-021-00461-4. PMID: 33594276. [PubMed: 33594276]
- Stewart KA, Wilcox KS, Fujinami RS, White HS. Development of postinfection epilepsy after Theiler’s virus infection of C57BL/6 mice. J Neuropathol Exp Neurol. 2010a;69(12):1210–9. doi: 10.1097/NEN.0b013e3181ffc420. PMID: 21107134. [PMC free article: PMC3077028] [PubMed: 21107134]
- Stewart KA, Wilcox KS, Fujinami RS, White HS. Theiler’s virus infection chronically alters seizure susceptibility. Epilepsia. 2010b;51(8):1418–28. doi: 10.1111/j.1528-1167.2009.02405.x. PMID: 20002148. [PubMed: 20002148]
- Stienen MN, Haghikia A, Dambach H, Thone J, Wiemann M, Gold R, Chan A, Dermietzel R, Faustmann PM, Hinkerohe D, Prochnow N. Anti-inflammatory effects of the anticonvulsant drug levetiracetam on electrophysiological properties of astroglia are mediated via TGFbeta1 regulation. Br J Pharmacol. 2011;162(2):491–507. doi: 10.1111/j.1476-5381.2010.01038.x. PMID: 20955362. [PMC free article: PMC3031068] [PubMed: 20955362]
- Sun Y, Dolmetsch RE. Investigating the Therapeutic Mechanism of Cannabidiol in a Human Induced Pluripotent Stem Cell (iPSC)-Based Model of Dravet Syndrome. Cold Spring Harb Symp Quant Biol. 2018;83:185–91. doi: 10.1101/sqb.2018.83.038174. PMID: 31186344. [PubMed: 31186344]
- Sun Y, Pasca SP, Portmann T, Goold C, Worringer KA, Guan W, Chan KC, Gai H, Vogt D, Chen YJ, Mao R, Chan K, Rubenstein JL, Madison DV, Hallmayer J, Froehlich-Santino WM, Bernstein JA, Dolmetsch RE. A deleterious Nav1.1 mutation selectively impairs telencephalic inhibitory neurons derived from Dravet Syndrome patients. Elife. 2016;5. doi: 10.7554/eLife.13073. PMID: 27458797. [PMC free article: PMC4961470] [PubMed: 27458797]
- Takai A, Yamaguchi M, Yoshida H, Chiyonobu T. Investigating Developmental and Epileptic Encephalopathy Using Drosophila melanogaster. Int J Mol Sci. 2020;21(17). doi: 10.3390/ijms21176442. PMID: 32899411. [PMC free article: PMC7503973] [PubMed: 32899411]
- Tanaka T, Tanaka S, Fujita T, Takano K, Fukuda H, Sako K, Yonemasu Y. Experimental complex partial seizures induced by a microinjection of kainic acid into limbic structures. Progress in neurobiology. 1992;38(3):317–34. doi: 10.1016/0301-0082(92)90023-8. PMID: 1312245. [PubMed: 1312245]
- Tanaka Y, Sone T, Higurashi N, Sakuma T, Suzuki S, Ishikawa M, Yamamoto T, Mitsui J, Tsuji H, Okano H, Hirose S. Generation of D1-1 TALEN isogenic control cell line from Dravet syndrome patient iPSCs using TALEN-mediated editing of the SCN1A gene. Stem Cell Res. 2018;28:100–4. doi: 10.1016/j.scr.2018.01.036. PMID: 29453127. [PubMed: 29453127]
- Thomson KE, Metcalf CS, Newell TG, Huff J, Edwards SF, West PJ, Wilcox KS. Evaluation of subchronic administration of antiseizure drugs in spontaneously seizing rats. Epilepsia. 2020;61(6):1301–11. doi: 10.1111/epi.16531. PMID: 32420627. [PMC free article: PMC7383749] [PubMed: 32420627]
- Thone J, Ellrichmann G, Faustmann PM, Gold R, Haghikia A. Anti-inflammatory effects of levetiracetam in experimental autoimmune encephalomyelitis. Int Immunopharmacol. 2012;14(1):9–12. doi: 10.1016/j.intimp.2012.05.021. PMID: 22691576. [PubMed: 22691576]
- Tidball AM, Dang LT, Glenn TW, Kilbane EG, Klarr DJ, Margolis JL, Uhler MD, Parent JM. Rapid Generation of Human Genetic Loss-of-Function iPSC Lines by Simultaneous Reprogramming and Gene Editing. Stem Cell Reports. 2017;9(3):725–31. doi: 10.1016/j.stemcr.2017.07.003. PMID: 28781079. [PMC free article: PMC5599229] [PubMed: 28781079]
- Tidball AM, Lopez-Santiago LF, Yuan Y, Glenn TW, Margolis JL, Clayton Walker J, Kilbane EG, Miller CA, Martina Bebin E, Scott Perry M, Isom LL, Parent JM. Variant-specific changes in persistent or resurgent sodium current in SCN8A-related epilepsy patient-derived neurons. Brain : a journal of neurology. 2020;143(10):3025–40. doi: 10.1093/brain/awaa247. PMID: 32968789. [PMC free article: PMC7780473] [PubMed: 32968789]
- Tukker AM, Wijnolts FMJ, de Groot A, Westerink RHS. Human iPSC-derived neuronal models for in vitro neurotoxicity assessment. Neurotoxicology. 2018;67:215–25. doi: 10.1016/j.neuro.2018.06.007. PMID: 29909083. [PubMed: 29909083]
- Twele F, Schidlitzki A, Tollner K, Loscher W. The intrahippocampal kainate mouse model of mesial temporal lobe epilepsy: Lack of electrographic seizure-like events in sham controls. Epilepsia open. 2017;2(2):180–7. doi: 10.1002/epi4.12044. PMID: 29588947. [PMC free article: PMC5719860] [PubMed: 29588947]
- Waltl I, Kaufer C, Broer S, Chhatbar C, Ghita L, Gerhauser I, Anjum M, Kalinke U, Loscher W. Macrophage depletion by liposome-encapsulated clodronate suppresses seizures but not hippocampal damage after acute viral encephalitis. Neurobiol Dis. 2018a;110:192–205. doi: 10.1016/j.nbd.2017.12.001. PMID: 29208406. [PubMed: 29208406]
- Waltl I, Kaufer C, Gerhauser I, Chhatbar C, Ghita L, Kalinke U, Loscher W. Microglia have a protective role in viral encephalitis-induced seizure development and hippocampal damage. Brain Behav Immun. 2018b;74:186–204. doi: 10.1016/j.bbi.2018.09.006. PMID: 30217535. [PMC free article: PMC7111316] [PubMed: 30217535]
- Wang W, Frankel WN. Overlaps, gaps, and complexities of mouse models of Developmental and Epileptic Encephalopathy. Neurobiol Dis. 2021;148:105220. doi: 10.1016/j.nbd.2020.105220. PMID: 33301879. [PMC free article: PMC8547712] [PubMed: 33301879]
- Welzel L, Schidlitzki A, Twele F, Anjum M, Löscher W. A face-to-face comparison of the intra-amygdala and intrahippocampal kainate mouse models of mesial temporal lobe epilepsy and their utility for testing novel therapies. Epilepsia. 2020;61(1):157–70. doi: 10.1111/epi.16406. [PubMed: 31828786]
- West PJ, Thomson K, Billingsley P, Pruess T, Rueda C, Saunders GW, Smith MD, Metcalf CS, Wilcox KS. Spontaneous recurrent seizures in an intra-amygdala kainate microinjection model of temporal lobe epilepsy are differentially sensitive to antiseizure drugs. Exp Neurol. 2022;349:113954. doi: 10.1016/j.expneurol.2021.113954. PMID: 34922908. [PMC free article: PMC8815304] [PubMed: 34922908]
- Wilcox KS, Vezzani A. Does brain inflammation mediate pathological outcomes in epilepsy? Adv Exp Med Biol. 2014;813:169–83. doi: 10.1007/978-94-017-8914-1_14. PMID: 25012376. [PMC free article: PMC4867105] [PubMed: 25012376]
- Wilcox KS, West PJ, Metcalf CS. The current approach of the Epilepsy Therapy Screening Program contract site for identifying improved therapies for the treatment of pharmacoresistant seizures in epilepsy. Neuropharmacology. 2020;166:107811. doi: 10.1016/j.neuropharm.2019.107811. PMID: 31790717. [PMC free article: PMC7054975] [PubMed: 31790717]
- Williams PA, White AM, Clark S, Ferraro DJ, Swiercz W, Staley KJ, Dudek FE. Development of spontaneous recurrent seizures after kainate-induced status epilepticus. J Neurosci. 2009;29(7):2103–12. doi: 10.1523/JNEUROSCI.0980-08.2009. PMID: 19228963. [PMC free article: PMC2897752] [PubMed: 19228963]
- Wong M, Roper SN. Genetic animal models of malformations of cortical development and epilepsy. J Neurosci Methods. 2016;260:73–82. doi: 10.1016/j.jneumeth.2015.04.007. PMID: 25911067. [PMC free article: PMC4618289] [PubMed: 25911067]
- Yu DX, Di Giorgio FP, Yao J, Marchetto MC, Brennand K, Wright R, Mei A, McHenry L, Lisuk D, Grasmick JM, Silberman P, Silberman G, Jappelli R, Gage FH. Modeling hippocampal neurogenesis using human pluripotent stem cells. Stem Cell Reports. 2014;2(3):295–310. doi: 10.1016/j.stemcr.2014.01.009. PMID: 24672753. [PMC free article: PMC3964286] [PubMed: 24672753]
- Zeidler Z, Brandt-Fontaine M, Leintz C, Krook-Magnuson C, Netoff T, Krook-Magnuson E. Targeting the Mouse Ventral Hippocampus in the Intrahippocampal Kainic Acid Model of Temporal Lobe Epilepsy. eNeuro. 2018;5(4). doi: 10.1523/ENEURO.0158-18.2018. PMID: 30131968. [PMC free article: PMC6102375] [PubMed: 30131968]
- Abstract
- Introduction
- Zebrafish and Other Model Organisms
- Drosophila melanogaster Models of Epilepsy
- Mouse Models of Genetic Epilepsy and Therapy Development
- Theiler’s Murine Encephalomyelitis Virus Mouse Model
- Intra-Amygdala Kainate and Intra-Hippocampal Kainate Mouse Models
- Induced Pluripotent Stem Cells and the Future
- Summary and Conclusions
- Acknowledgments
- Disclosure Statement
- References
- A face-to-face comparison of the intra-amygdala and intrahippocampal kainate mouse models of mesial temporal lobe epilepsy and their utility for testing novel therapies.[Epilepsia. 2020]A face-to-face comparison of the intra-amygdala and intrahippocampal kainate mouse models of mesial temporal lobe epilepsy and their utility for testing novel therapies.Welzel L, Schidlitzki A, Twele F, Anjum M, Löscher W. Epilepsia. 2020 Jan; 61(1):157-170. Epub 2019 Dec 11.
- Spontaneous recurrent seizures in an intra-amygdala kainate microinjection model of temporal lobe epilepsy are differentially sensitive to antiseizure drugs.[Exp Neurol. 2022]Spontaneous recurrent seizures in an intra-amygdala kainate microinjection model of temporal lobe epilepsy are differentially sensitive to antiseizure drugs.West PJ, Thomson K, Billingsley P, Pruess T, Rueda C, Saunders GW, Smith MD, Metcalf CS, Wilcox KS. Exp Neurol. 2022 Mar; 349:113954. Epub 2021 Dec 17.
- Development of an antiseizure drug screening platform for Dravet syndrome at the NINDS contract site for the Epilepsy Therapy Screening Program.[Epilepsia. 2021]Development of an antiseizure drug screening platform for Dravet syndrome at the NINDS contract site for the Epilepsy Therapy Screening Program.Pernici CD, Mensah JA, Dahle EJ, Johnson KJ, Handy L, Buxton L, Smith MD, West PJ, Metcalf CS, Wilcox KS. Epilepsia. 2021 Jul; 62(7):1665-1676. Epub 2021 May 17.
- Review Animal Models of Pharmacoresistant Epilepsy.[Jasper's Basic Mechanisms of t...]Review Animal Models of Pharmacoresistant Epilepsy.Guignet M, Steve White H. Jasper's Basic Mechanisms of the Epilepsies. 2024
- Review Animal Models of Seizures and Epilepsy: Past, Present, and Future Role for the Discovery of Antiseizure Drugs.[Neurochem Res. 2017]Review Animal Models of Seizures and Epilepsy: Past, Present, and Future Role for the Discovery of Antiseizure Drugs.Löscher W. Neurochem Res. 2017 Jul; 42(7):1873-1888. Epub 2017 Mar 13.
- New Models for Assessment of Antiseizure Activity - Jasper's Basic Mechanisms of...New Models for Assessment of Antiseizure Activity - Jasper's Basic Mechanisms of the Epilepsies
Your browsing activity is empty.
Activity recording is turned off.
See more...