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/9780197549469.003.0042
Abstract
Advances in human induced pluripotent stem cell (iPSC) approaches have greatly expanded the use of patient-derived cellular models, including cortical-like neurons and brain organoids, to study genetic epilepsies. New protocols to differentiate iPSCs into various neural cell types, and to generate brain region-specific organoids, have accelerated progress. In addition, the application of gene editing techniques adds rigor to these studies and offers the opportunity to model rare genetic epilepsies by enabling correction or insertion of mutations to generate isogenic controls or virtual patients, respectively. Studies using patient-derived cells have provided insight into the mechanisms underlying seizures and associated comorbidities for an increasing number of epilepsy gene variants. In this chapter, methods for generating iPSCs, the culture and gene editing of human pluripotent stem cells (hPSCs; both iPSCs and human embryonic stem cells), and differentiation strategies to derive cortical-like neurons are described. Next, physiological assays that can be applied to study hPSC-derived neuronal and network activity are discussed. Finally, the chapter reviews studies interrogating the functional effects of specific epilepsy gene variants using hPSC-derived 2D neural and 3D brain organoid cultures, as well as the use of iPSC-derived cardiac myocytes to study the most devastating epilepsy complication, sudden unexpected death in epilepsy (SUDEP).The chapter ends with a description of current challenges and future directions for hPSC modeling of genetic epilepsies.
Introduction
Recent years have seen a surge in the discovery of epilepsy related genes and, with it, a blooming of mechanistic studies. Because of the inaccessibility of human epileptic tissue, the lack of adequate control tissue and the fact that only late-stage specimens are acquired when it is available, model systems are necessary for the study of epilepsy mechanisms and the development of novel therapies. Moreover, as epilepsies typically are associated with comorbidities involving diverse and widespread brain networks, a useful strategy is to incorporate multifaceted approaches that encompass structural, functional, and developmental features to model epilepsies and their associated comorbidities.
The mainstay of genetic epilepsy models over the past five decades has been spontaneous or genetically engineered rodent models harboring genetic abnormalities similar to those identified in human genetic epilepsies. Such models have contributed a wealth of knowledge to the epilepsy field, specifically surrounding the function of neural networks in epilepsy and the underlying in vivo pathophysiological processes. Zebrafish represent a more recent addition to the armamentarium of genetic epilepsy models and are particularly useful for high-throughput drug screening (Baraban, 2021). However, several limitations arise in the use of animal models of epilepsy. Fish and even mice possess important genetic, structural, and neurodevelopmental differences from humans. And while some rodent models exhibit a seizure phenotype that closely resembles those seen in humans, most do not completely recapitulate the complex pathophysiology of human epilepsies. Moreover, animal models for many genetic epilepsies do not exhibit spontaneous seizures. Despite extensive research using animal models, understanding of the cellular and molecular mechanisms surrounding monogenic epilepsies remains elusive.
To try to overcome some of these obstacles and expand the tools available to model epilepsies, human pluripotent stem cells (hPSCs), a term that encompasses both human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs), have become increasingly used. In 1998, the Thomson laboratory discovered that they could derive hESCs from blastocysts and that these cells could then be used to generate many different cell types in vitro (Thomson et al., 1998). The breadth of stem cell research was further enhanced in 2006 when Yamanaka and colleagues (Takahashi and Yamanaka, 2006) demonstrated that mouse somatic cells could be reprogrammed to generate iPSCs. This discovery was quickly followed by the generation of human iPSCs in 2007 (Meissner et al., 2007; Takahashi et al., 2007), and the use of human iPSCs in translational research, drug discovery, and therapeutic development has since become increasingly widespread. Skin or blood cells taken from a patient can be reprogrammed to iPSCs and used to model a genetic disease by generating the affected cell type. Germline genetic variants found in a given patient are carried on in their somatic cell-derived iPSCs, and in any subsequent progeny that are generated from these iPSCs. Since samples of the affected tissue cannot easily be taken from patients suffering from central nervous system (CNS) diseases, brain cells generated from patient iPSCs provide a useful tool to study and model the development of brain diseases, particularly neurodevelopmental disorders. In this chapter, we review the current techniques and applications of hPSC models for studying genetic epilepsies using both 2D and 3D neural culture approaches. Additionally, we highlight potential limitations and avenues for future research using hPSC model systems.
Generating iPSCs
The ability to generate patient-specific iPSC lines provides the opportunity to model a wide range of genetic epilepsies in an in vitro system. The forced expression of specific transcription factors reverts somatic cells to a pluripotent epigenetic state and silences lineage-specific genes. This process, called “reprogramming,” is typically carried out by introducing four transcription factors, Sox2, Oct3/4, Klf4, and c-Myc, into somatic cells (Takahashi et al., 2007; Takahashi and Yamanaka, 2006). These four factors are commonly referred to as the “Yamanaka factors.” Typically, reprogramming involves the use of dermal fibroblasts obtained by a skin biopsy. More recently, peripheral blood mononuclear cells derived from patient blood samples are increasingly used as a less invasive source of somatic cells for reprogramming, and kidney epithelial cells extracted from urine are another option (Okita et al., 2013; Zhou et al., 2011). These advances have broadened the source of iPSCs and have contributed to the ease and accessibility of iPSC research.
Gene Editing of iPSCs
Genome editing approaches have substantially advanced the use of hPSCs for modeling of brain disorders, including genetic epilepsies. Because the CRISPR/Cas9 endonuclease system facilitates the ability to make precise changes in the genome (Cong et al., 2013; Ran et al., 2013), this method offers several key applications for using hPSCs to model genetic epilepsies. First, the inherent variability between iPSC lines from different subjects due to genomic background affects may be abrogated by using CRISPR-based homology directed repair to correct epilepsy gene variants in patient iPSC lines and thereby generate isogenic control lines. Second, in cases where patient material is not available to generate iPSC lines, epilepsy-specific gene variants may be created in a control line to generate paired “virtual patient” and isogenic control lines. Third, CRISPR-based homology-directed repair may be used to introduce exogenous genomic sequences into a specific locus to tag genes of interest with fluorescent reporters for cell culture-based studies of intact or mutant protein localization or function (Roberts et al., 2017). Finally, CRISPR-mediated gene knockout (via the generation of out-of-frame insertions or deletions by non-homologous end joining) in a control line may be used to study genetic epilepsies caused by autosomal recessive variants or haploinsufficiency. Indeed, methods to make this latter approach more efficient by combining somatic cell reprogramming and CRISPR gene knockout have been developed and applied to generate iPSC models of genetic epilepsies (Tidball et al., 2017).
Methods of 2D Neuronal Differentiation
For studying epilepsies, the derivation of disease-relevant neural cell types from hPSCs offers unique opportunities to explore disease mechanisms and identify novel therapies. Most relevant to epilepsy are excitatory and inhibitory cortical neurons. Neuronal differentiation protocols for these cell types broadly fall into two main categories: small molecule directed differentiation and transcription factor induction. Below, we discuss these two methods applied to both excitatory and inhibitory cortical-like neurons. Similar protocols that generate hPSC-derived astrocytes and microglia have also been developed (Canals et al., 2018; Douvaras et al., 2017; Krencik and Zhang, 2011) and are not described further in the text (but see Fig. 42–1).

Figure 42–1.
Overview of schema for generating 2D in vitro cellular models using human iPSCs. Patient or healthy control somatic cells, such as fibroblasts or peripheral blood mononuclear cells (PBMCs), are reprogrammed into iPSCs using a cocktail containing the (more...)
Small molecule differentiation protocols used to produce cortical-like neurons involve the inhibition of the bone morphogenetic protein (BMP) and transforming growth factor-beta (TGF-β) signaling pathways. This method is known as dual SMAD inhibition (Chambers et al., 2009; Shi et al., 2012), as SMAD proteins are the signal transducing effectors for TGF-β/BMP signaling. Common protocols consist of monolayer or embryoid body formation with or without the use of small molecules. While monolayer protocols are less labor-intensive, embryoid body protocols provide a purer population of cortical neurons that are more mature and have longer neurites, although they do not exhibit any difference in electrophysiological properties compared to monolayer cultures (Chandrasekaran et al., 2017; Muratore et al., 2014). The cortical-like neurons exhibit electrophysiological activity after about 4 to 6 weeks from the start of differentiation. Modulation of the Sonic Hedgehog (SHH) pathway after development of a primitive neuroepithelium can bias cultures toward the generation of excitatory (with SHH pathway inhibitors) or inhibitory (using SHH activators) neurons. To derive interneurons, exposure to SHH pathway activators yields a homogeneous population of progenitors that express the medial ganglionic eminence (a key source of cortical interneuron progenitors) marker NKX2.1 by about 6 weeks (Liu et al., 2013a; Maroof et al., 2013; Nicholas et al., 2013). However, populations that are achieved through small-molecule patterning protocols for either excitatory or inhibitory cortical-like neurons typically exhibit heterogeneous stages of differentiation throughout the culture, and differentiation efficiency can often vary between cell lines. Additionally, prolonged culture is required for cells to reach maturity.
The forced expression of transcription factors in hPSCs has recently been used to overcome some of these challenges. Transcription factor induction rapidly generates a more homogeneous population of either cortical-like excitatory or inhibitory neurons with high efficiency and reproducibility (Lam et al., 2017; Pak et al., 2015; Sun et al., 2016a; Yang et al., 2017; Zhang et al., 2013). However, this rapid induction bypasses normal signaling pathways that occur during neural development and therefore may not be suitable for investigating certain neurodevelopmental disorders (Schafer et al., 2019). Cortical-like excitatory neurons are often generated by the forced expression of the transcription factor Neurogenin-2 (NGN2) alone or combined with NGN1 using a doxycycline-inducible expression system (Lam et al., 2017; Pak et al., 2015). These induced neurons show transcriptional expression profiles closer to that of neurons in superficial cortical layers, and they display short-term plasticity, synaptic function and modulation, and the ability to functionally integrate into the host after grafting. This is in contrast to the deeper layer cortical neurons that are typically generated using small-molecule patterning unless prolonged (3-month) culture durations are used (Shi et al., 2012).
Functional GABAergic interneurons with a high degree of synaptic maturity are generated by the transient expression of transcription factors ASCL1 and DLX2 with or without LHX6 and specific microRNAs (Sun et al., 2016a; Yang et al., 2017). The cells express subtype-specific GABAergic interneuron markers and functionally integrate into host circuits after in vivo transplantation. Excitatory and inhibitory neurons can be co-cultured together and provide a useful platform for studying diseases that affect neural circuitry. An ongoing focus in the field is the challenge of generating parvalbumin (PV)-expressing fast-spiking interneurons, which are thought to be critically involved in the pathophysiology of many epilepsies and other neuropsychiatric disorders (Hu et al., 2017; Jiang et al., 2016). PV interneurons are difficult to achieve in culture due to the prolonged maturity that is required to generate these cells. More recently, brain organoid models (see below) have emerged as a powerful alternative to 2D modeling as they allow for a wider variety of cell types to be generated and cultured for extended periods of time.
Physiological Assays for hPSC-Derived Neurons
As epilepsy is a disease characterized by functional changes in neuronal and neural circuit activity, the ability to assess the physiological impact of ictogenic or epileptogenic changes in hPSC-derived cultures is critically important. A number of approaches spanning the gamut from traditional electrophysiological recordings to imaging based assays have been employed to achieve this end (Fig. 42–2).

Figure 42–2.
Approaches to measure physiological activity in 2D neuronal and brain organoid cultures. A–C. Available techniques and their advantages and disadvantages. These approaches range from more traditional electrophysiology such as single electrode-based (more...)
Classical electrophysiological techniques such as patch-clamp recordings have been successfully used to identify cell intrinsic changes in membrane currents and synaptic activity that may contribute to an epilepsy phenotype (Liu et al., 2013b; Tidball et al., 2020). This approach has the advantage of providing granular and cell-type specific data, but it is also highly labor intensive and usually limited to recording from one or, at best, a few neurons simultaneously. Extracellular recordings permit simultaneous data gathering from larger populations of neurons, including both single-unit activities (i.e., extracellular action potentials) of individual neurons as well as lower frequency local field potentials (LFPs) generated by networks of interacting cells. The LFP can include contributions from neurons and non-neuronal cells such as astrocytes (Buzsaki et al., 2012; Obien et al., 2014). In the case of 2D hPSC-derived cultures, this is typically achieved by utilizing tissue culture plates embedded with multielectrode arrays (MEAs), as monolayer cultures lack sufficient depth to be compatible with more traditional recording tools such as glass electrodes. The MEA format also allows for additional advantages such as the potential for repeated measurements over time as the recording apparatus typically couples with the array at an external interface, thus preserving cellular sterility, the ability to test drugs in a multi-well format, and the ability to both stimulate and record. To achieve this, hPSC-derived neurons are typically plated directly on the MEA. Using this approach Odawara et al. were able to track the electrophysiological dynamics of hPSC-derived neurons for over 1 year, and demonstrated hypersynchronous bursts in response to pro-convulsant drugs and rescue of this with antiseizure medications (Odawara et al., 2016). Using a variation of this approach, another group generated hPSC-derived neuronal aggregates, and over the course of 3 weeks in culture, used a nine-electrode MEA apparatus to describe patterns of network connectivity and development, such as the onset of increased synchrony (Izsak et al., 2019).
Optical microscopic approaches offer another powerful set of methods to study physiological and network activity in hPSC-derived cultures. The most well-established of these is through fluorescence-based calcium indicators of neuronal electrical activity. These indicators are predicated on the principle that neuronal activity, both action potential generation and synaptic transmission that activates neurotransmitter release, results in increases in intracellular calcium stores (Grienberger and Konnerth, 2012). This change in calcium concentration can be detected by indicators that change conformation in response to calcium binding, thus resulting in a change in emitted fluorescence following excitation. Historically, the first reliable indicators were chemical calcium indicators (Grienberger and Konnerth, 2012). More recently, improvements in genetically encoded calcium indicators (GECIs) based on chimeric fluorescent proteins have become more widely used. GECIs can be specifically targeted to select neuronal populations, are amenable to chronic recordings, and have excellent signal to noise ratios (Dana et al., 2019). The development and improvement of laser scanning microscopy, including confocal microscopy and two-photon imaging techniques, has been an equally important set of technical innovations that has resulted in widespread use of these indicators primarily for in vitro imaging of rodent neuronal cultures and in vivo rodent brain imaging. These techniques have been adapted for characterizing the calcium dynamics of epilepsy associated hPSC-derived 2D neuronal cultures (Avazzadeh et al., 2019; Krey et al., 2013; Marchetto et al., 2010).
The aforementioned technical developments have popularized the use of calcium indicators; however, major limitations of this approach include that the calcium signal is only a proxy for the electrical activity of interest, and calcium indicators provide limited or no information on hyperpolarizing input or subthreshold inputs (Knopfel and Song, 2019). While improved deconvolution algorithms, coupled with newer GECIs, have been shown to estimate activity with a relatively high degree of fidelity to simultaneously recorded spontaneous or induced neuronal spiking, the other concerns with calcium indicator imaging are inherent limitations (Grewe et al., 2010; Pnevmatikakis et al., 2016; Vogelstein et al., 2010). This has motivated the development of optical voltage indicators, which directly measure neuronal electrical activity. Optical voltage imaging is challenging for a number of reasons, including, but not limited to, the need to target a small (outer) region of the plasma membrane and yet generate sufficient photons to discern activity changes, the requirement for millisecond fast imaging speeds, and the ability to integrate reporters into the plasma membrane without causing cellular stress or damage (Bando et al., 2019). Although organic voltage-sensitive dyes have been in existence for several decades, the more recent development of genetically encoded voltage indicators based on similar protein engineering principles as GECIs has resulted in improvements in targeting specificity, signal-to-noise ratio, and cell toxicity that has allowed for improved neuronal and neural circuit imaging (Piatkevich et al., 2019; Storace et al., 2015). While voltage indicators have yet to be applied to interrogate activity in epilepsy-associated, hPSC-derived neuronal cultures, this emerging technology has the potential to provide novel insights into circuit pathology in epilepsy by combining the temporal fidelity of traditional electrophysiologic recording techniques with the spatial information conferred by calcium indicator imaging.
In order to gain more detailed information about the contribution of specific cell types to circuit activity, virtually all of the optical and electrophysiological assays described above can be combined with optogenetic or chemogenetic tools to specifically manipulate neuronal populations of interest. Optogenetics utilizes bacterial opsins that respond to stimulation by specific wavelengths of light by opening ion channels, thus converting photons directly into current (Deisseroth, 2015). The delivery of these engineered proteins to specific neuronal populations allows for temporally precise and cell specific excitation and inhibition of neuronal activity. Chemogenetics in this context refers to a parallel approach using either engineered G protein–coupled receptors or ion channels that respond only to a specific and otherwise inert compound to gain chemical, instead of light-based, control of cellular activity (Atasoy and Sternson, 2018; Magnus et al., 2019). Optogenetic and chemogenetic tools have been used to manipulate and interrogate circuit activity in ex vivo and in vivo rodent models of epilepsy (Shiri et al., 2017; Yekhlef et al., 2017; Zhou et al., 2019), including their use to activate or suppress grafted hPSC-derived interneurons (Cunningham et al., 2014; Upadhya et al., 2019). These tools also are well suited to a broad range of in vitro cellular applications, including hPSC-derived cultures.
Genetic Epilepsy Modeling Using 2D hPSC Cultures
Brain cells derived from patient or gene-edited iPSCs have provided significant insight into potential mechanisms underlying many of the genetic epilepsies to date. iPSC models of genetic epilepsies have been used to explore disorders in which epilepsy is a defining feature, such as Dravet syndrome (DS) and Syntaxin Binding Protein-1 (STXBP1)-related DEE, as well as those in which epilepsy is a variable feature associated with other prominent neurodevelopmental abnormalities, including tuberous sclerosis complex (TSC) and Rett syndrome (RTT). While replicating epileptic phenotypes in vitro has proven challenging, current iPSC models produce epileptic-like activity, which includes increased spontaneous action potential firing and synchronous network bursting activity. Alterations in cellular morphology such as changes in soma size, neurite outgrowth, synapse formation, and dendritic spine length have also been observed. The breadth of epilepsies modeled using hPSCs has been reviewed in detail recently (Hirose et al., 2020). Below we discuss select genetic epilepsies and related neurodevelopmental disorders with brain specific phenotypes that have been modeled using 2D and 3D hPSC–derived cultures.
Dravet Syndrome
DS is a severe DEE caused by de novo loss-of-function variants in the SCN1A gene that encodes the Nav1.1 voltage-gated sodium channel (VGSC) (Fujiwara, 2006; Marini et al., 2011). The onset of DS is in the first year of life, usually between 4 and 10 months, and clinical features include pharmaco-resistant generalized and focal seizures, intellectual disability that is typically severe, ataxia, and increased mortality (Dravet et al., 2005; Fujiwara, 2006; Guerrini and Aicardi, 2003). DS mouse models, most commonly Scn1a heterozygous knockout mice, have provided important advances in understanding potential DS mechanisms. The mice exhibit seizures and behavioral deficits linked to decreased sodium current and hypoexcitability of interneurons (Cheah et al., 2012; Yu et al., 2006), predominantly of the PV-expressing, fast-spiking subtype (Ogiwara et al., 2007; Tai et al., 2014). However, some groups have found that certain PV interneuron deficits are transient (Favero et al., 2018), or that abnormalities exist in excitatory neurons or other interneuron subtypes in DS mouse models (Goff and Goldberg, 2019; Mistry et al., 2014; Tai et al., 2014).
Because important features of brain development differ between mouse and human (Hodge et al., 2019; Seto and Eiraku, 2019), an increasing number of investigators have used patient iPSC-derived cortical-like neurons to model DS in a human cellular context that preserves the unique genetic background of patients. Experimental results have varied considerably, with some groups finding reduced excitability (Higurashi et al., 2013; Kim et al., 2018; Sun et al., 2016b) or shifts in the voltage-dependence of sodium current (INa) activation of DS patient iPSC-derived GABAergic neurons (Schuster et al., 2019), and others reporting reduced INa density in both excitatory and inhibitory DS neurons with reduced action potential firing of interneurons (Xie et al., 2020). In contrast, two groups have described hyperexcitability of excitatory neurons or both excitatory and inhibitory neurons (Liu et al., 2013b; Jiao et al., 2013), with increased INa density in both types of neurons in one report (Liu et al., 2013b). As expected, findings in the haploinsufficiency-associated DS tend to differ from those in studies using hPSCs with putative gain-of-function disease-causing variants in two other VGSC subunit genes, SCN2A (Que et al., 2021) and SCN8A (Tidball et al., 2020).
The most likely explanations for disparate findings in DS hPSC studies include the use of varied differentiation protocols yielding different subtypes of human neurons, and the fact that recordings were obtained from neurons at differing levels of maturation. In addition, variability in the presence of modifier genes in DS patients with different genetic backgrounds and variability in the types of DS-causing genetic variants may also play a role. Future studies that use more precise differentiation protocols to generate specific cortical neuron subtypes, adequate in vitro neuronal maturation, and the increased use of CRISPR gene correction to generate isogenic controls should help to clarify DS cellular and network mechanisms. To this end, the ability to generate a pure population of late-generated, PV-expressing, fast-spiking human cortical interneurons in vitro, a technical challenge that has not yet been overcome, will be critical. Despite the caveats surrounding the use of human iPSC-derived neurons, the current findings combined with those using DS mouse models suggest that DS mechanisms involve developmental stage-specific changes in multiple neuronal subpopulations, along with subsequent compensatory changes, both of which likely influence network activity and seizure generation.
STXBP1-Related DEE
STXBP1-related DEE, a subtype of which is also known as Ohtahara syndrome (Saitsu et al., 2008), is typically diagnosed in early infancy and is accompanied by severe epilepsy and intellectual disability (Lanoue et al., 2019; Stamberger et al., 2016). STXBP1-related DEE results from haploinsufficiency of the STXBP1 gene, although a toxic gain-of-function mechanism may also be present with some variants (Saitsu et al., 2008). The STXBP1 protein (also known as Munc-18-1) plays a critical role in presynaptic vesicle release (Lanoue et al., 2019; Stamberger et al., 2016). A study using hESCs with conditional heterozygous STXBP1 mutations found that the levels of Munc-18-1 and its binding partner, Syntaxin-1, were reduced by approximately 30% (Patzke et al., 2015). Additionally, STXBP1 mutant hESC-derived neurons showed a significant reduction in spontaneous and evoked neurotransmitter release (Patzke et al., 2015). Another group developed an iPSC model using patient-derived cells and found reduced neurite outgrowth as well as mislocalization of Syntaxin-1 to the cell cytoplasm rather than the plasma membrane (Yamashita et al., 2016). iPSC-derived GABAergic neurons exhibited altered maturation with reduced numbers of spontaneous spikes and bursts (Ichise et al., 2021). Together these studies offer mechanistic insight into the development of STXBP1-related DEE and suggest that heterozygous STXBP1-related DEE variants lead to decreased neurotransmission of both excitatory and inhibitory cortical neurons.
Tuberous Sclerosis Complex
TSC is an autosomal dominant genetic disorder that affects many organ systems, including the brain, lungs, heart, kidneys and skin (Hasbani and Crino, 2018). TSC is characterized by systemic tumors called hamartomas. In the brain, these tumors result in tubers that are associated with various neurological disorders, including intellectual disability, TSC associated neuropsychiatric disorders (TAND), and epilepsy (de Vries et al., 2018; Hasbani and Crino, 2018). Behavioral disturbances and autism spectrum disorder are often present as well. TSC is caused by an inherited or de novo heterozygous loss-of-function mutation in either hamartin (TSC1) or tuberin (TSC2), which together form the TSC1/TSC2 complex (Au et al., 2007). Under normal conditions, this complex reduces mechanistic target of rapamycin complex 1 (mTORC1) activation. However, loss-of-function germline mutations of TSC1 or 2, in some instances combined with “second hit” somatic mutations, lead to overactivation of the mTOR pathway and the pathological features of TSC (Hasbani and Crino, 2018).
In addition to mouse models and histological studies of human TSC tissue, several groups have used TSC-deficient, hPSC-derived neural cells to investigate the disorder. Human hPSC-derived neurons, both from TSC patients or with gene-edited TSC1 or 2 deletion, display delayed neuronal differentiation of neural progenitor cells, impaired mitochondrial metabolism, excess mTORC1 signaling, increased proliferation, enlarged soma size, perturbed neurite outgrowth, and abnormal connectivity (Blair and Bateup, 2020; Costa et al., 2016; Ebrahimi-Fakhari et al., 2016; Li et al., 2017; Nadadhur et al., 2019; Winden et al., 2019; Zucco et al., 2018). Functionally, hPSC-derived excitatory cortical neurons containing bi-allelic mutations in TSC2 display hyperexcitable network activity and transcriptional dysregulation consistent with that of cortical tubers (Nadadhur et al., 2019; Winden et al., 2019), although another study found decreased excitability of TSC2 null hESC-derived cortical-like neurons (Costa et al., 2016). Elegant work using hPSC-derived human brain organoid models of TSC has replicated the phenotypes found in 2D models and also provides evidence to support the “second hit” theory which suggests that a second somatic mutation superimposed upon a TSC germline mutation is necessary for cortical and subcortical tuber formation (Blair et al., 2018). The morphological and functional phenotypes observed in both 2D and 3D TSC models were reversed by the early administration of the mTOR inhibitor, rapamycin (Blair et al., 2018; Costa et al., 2016), although in one instance this reversal was only partial when rapamycin was applied at a later developmental stage (Blair et al., 2018).
Other hPSC modeling studies provide evidence to support the role of glial cells in TSC pathophysiology. The neural differentiation of TSC-deficient hPSCs gives rise to increased numbers and proliferation of astrocytes in both 2D (Li et al., 2017) and 3D (Blair et al., 2018) TSC models. Recent work has also explored the significance of neuron-glial interaction using co-cultures of cortical neurons and oligodendrocytes derived from TSC iPSCs (Nadadhur et al., 2019). Interestingly, iPSC-derived cortical-like neurons cultured alone show increased network activity, while neurons co-cultured with oligodendrocytes displayed cellular hypertrophy and increased axonal density. Increased proliferation and decreased maturation of TCS iPSC-derived oligodendrocytes were also observed. Taken together, these TSC studies highlight the complex and delicate relationship between cell type interactions, mTOR pathway activity, and a wide variety of developmental processes that may contribute to epileptogenesis.
Rett Syndrome
RTT is a genetic neurodevelopmental disorder characterized by rapid regression in language and motor skills after infancy, severe intellectual disability, and behavioral disturbances, with epilepsy comorbidly presenting in 50%–90% of patients (Dolce et al., 2013). RTT is caused primarily by loss-of-function variants in the X-linked MECP2 gene and therefore occurs almost exclusively in girls (Ip et al., 2018). Several groups have developed iPSC models of RTT using patient-derived neurons and astrocytes. Patient iPSC-derived neurons display a decrease in spontaneous activity coupled with deficits in neurite outgrowth and synapse formation, as well as reduced soma size and premature senescence compared to isogenic controls (Cheung et al., 2011; Marchetto et al., 2010; Ohashi et al., 2018, Kim et al., 2019). Some of these phenotypes were rescued via treatment with insulin-like growth factor-1 (Marchetto et al., 2010) or the P53 inhibitor Pifithrin-α (Ohashi et al., 2018). Astrocytes have also been derived from RTT patient iPSCs and display perturbed glial differentiation (Kim et al., 2019). When co-cultured with healthy neurons, MECP2 mutant RTT astrocytes induce adverse effects on neuronal function and morphology, resulting in smaller somas, shorter neurite length, and reduced synaptic terminals (Williams et al., 2014). These findings highlight the importance of glia in RTT pathogenesis and the potential contribution of astrocytes to epileptogenesis.
To investigate the role of MECP2 in early brain development, Mellios and colleagues used RTT patient-derived iPSCs to create cerebral organoids and monolayer cultures (Mellios et al., 2018). They found deficits in neurogenesis, neuronal differentiation, and migration, and identified a novel signaling pathway mediated by MECP2-targeted miRNA that may play a role in neurogenesis and early brain development. A more recent RTT brain organoid study (Samarasinghe et al., 2021) that delved more into RTT-induced network disturbances is described in the brain organoid section below. These findings from 2D and 3D hPSC RTT models suggest that disruptions in early brain development could have deleterious effects on postnatal brain maturation and plasticity, and therefore be an underlying contributor to neurodevelopmental disorder pathogenesis.
Studying SUDEP Using hPSCs
The most catastrophic epilepsy complication is SUDEP (Sudden Unexpected Death in Epilepsy), which is defined as a witnessed or unwitnessed, nontraumatic, and nondrowning death in a person with epilepsy, without an identifiable structural cause and in the absence of status epilepticus (Nashef et al., 2012). Despite the devastating nature of SUDEP, no reliable clinical biomarkers exist to identify at risk patients. Potential factors contributing to SUDEP in any given case include seizure-induced central or obstructive apnea, pulmonary edema, brainstem or autonomic dysfunction, and cardiac arrhythmias (Bagnall et al., 2017; Massey et al., 2014; Schuele et al., 2007a, 2007b; Shorvon and Tomson, 2011; Surges et al., 2009, 2010). SUDEP is responsible for up to 7.5%–20% of all epilepsy deaths (Cooper et al., 2016; Schuele et al., 2007b; Shorvon and Tomson, 2011; Skluzacek et al., 2011) and genetic epilepsies, particularly severe DEEs linked to ion channel variants, have among the highest risk. For example, DS patients have a SUDEP risk as high as 20% (Cooper et al., 2016).
SUDEP has been proposed to be caused by “arrhythmias” in brain and heart, especially in ion channel genetic epilepsies, given that many epilepsy-associated ion channel variants are expressed in both brain and heart (Goldman et al., 2009). In addition, seizure activity can secondarily alter the expression of ion channels in the heart, leading to arrhythmia (reviewed in (Li et al., 2019). Many genetic DEE mouse models have been used to explore SUDEP mechanisms and display evidence for cardiac arrhythmias, including those caused by variants in Kcnq1, Scn1a, Scn8a, and Scn1b (Goldman et al., 2009; Auerbach et al., 2013; Frasier et al., 2016; Lin et al., 2015; Lopez-Santiago et al., 2007).
While genetically modified mouse models have advanced knowledge of potential SUDEP mechanisms, mouse and human cardiac physiology differs in important aspects (Nerbonne, 2014). This fact has led some investigators to use patient iPSC-derived cardiac myocytes to explore the contribution of altered cardiac excitability to SUDEP mechanisms in genetic epilepsies. In one study, iPSC cardiac myocytes were generated from SCN1A-linked DS subjects and nonepileptic controls (Frasier et al., 2018). DS patient-derived cardiac myocytes displayed increased contraction rates and transient INa density, and comparable results were found in SCN1A haploinsufficient cardiac myocytes generated by CRISPR genome editing of iPSCs. These findings are also consistent with a previous DS mouse study (Auerbach et al., 2013). The DS patient whose iPSC-derived cardiac myocytes displayed the greatest increase in INa density also showed arrhythmogenic substrates of their cardiac myocyte action potentials, and subsequent patient examination demonstrated cardiac autonomic abnormalities. Together with DS mouse studies, these results suggest that compensatory overexpression of another VGSC gene, perhaps SCN5A, in response to SCN1A haploinsufficiency leads to altered cardiac excitability in DS. Moreover, these findings indicate that hPSC-derived cardiac myocytes are a valuable model to investigate SUDEP mechanisms and to identify biomarkers in the genetic epilepsies. In addition to cardiac dysfunction, remodeling in the autonomic nervous system can contribute to sudden cardiac death (Huang et al., 2017; Tan and Verrier, 2013), and seizures may affect brainstem respiratory centers resulting in apnea (Devinsky et al., 2016). Given that both of these systems have been linked to SUDEP, the ability to generate brainstem and autonomic neurons from epilepsy patient-derived iPSCs is a key future direction for understanding SUDEP mechanisms in genetic epilepsies.
Brain Organoid Models
While human stem cell–based 2D cultures have shown value in epilepsy modeling, these systems have significant limitations, including physiological, functional, and molecular differences from in vivo human tissue (Bershteyn and Kriegstein, 2013). In particular, it is challenging to achieve complex neural circuit formation in 2D cultures for a number of reasons, including limited neuronal (in particular interneuron) diversity, maximum in vitro culture times that are usually less than 3 months, and the lack of a layered 3D structure. Given the importance of neural circuit dysfunction in the pathogenesis of epilepsy, this is a significant shortcoming of 2D models. Brain organoid cultures consist of 3D structures containing diverse populations of neurons and glia. Organoids can also recapitulate some aspects of the layered cytoarchitecture of brain and exhibit complex neural circuit activities (Lancaster et al., 2013; Samarasinghe et al., 2021; Sun et al., 2019; Trujillo et al., 2019; Watanabe et al., 2017). Moreover, although organoids cannot achieve the level of cellular or structural complexity of in vivo brain, they nonetheless present with some unique advantages compared to commonly used rodent models. For example, hPSC-derived organoids contain an enlarged subventricular zone (SVZ), with an outer subventricular zone (oSVZ) not found in rodent brain. Importantly, human brain organoids also contain a sizable population of the primarily oSVZ-associated progenitor population known as outer radial glial cells (oRGs; also known as basal radial glial cells). The oRGs are thought to be responsible for the massive expansion of human cortex, and they are relatively abundant in gyrencephalic species such as humans and nonhuman primates but are largely absent in lisencephalic species such as mice (Hansen et al., 2010; Martinez-Cerdeno et al., 2012). Given the known impact of genetic variants on epilepsy, organoid models provide a unique platform to determine the effects of these variants on cortical development and expansion.
In order to generate brain organoids, hPSCs are aggregated in suspension using a number of different approaches such that they differentiate and self-organize into 3D brain-like structures. While the available protocols for organoid generation continue to expand in this highly dynamic field (and are reviewed in detail elsewhere; Di Lullo and Kriegstein, 2017), the general approaches can be divided into two broad categories. In one set of so-called intrinsic protocols, stem cells are allowed to differentiate in the absence of specific patterning factors. The resulting organoids typically contain a wide assortment of cell types, including forebrain cortex, hindbrain, and retinal cells (Lancaster et al., 2013). In the second category of approaches are protocols that utilize the timed application of patterning factors to direct differentiation into specific brain-like regions. For example, SMAD inhibition is used in multiple protocols to limit mesoderm and endoderm formation in favor of ectoderm (Kadoshima et al., 2013; Pasca et al., 2015; Qian et al., 2016). Often, SMAD inhibition is combined with other morphogens or small molecules to generate dorsal forebrain-like brain organoids that consist primarily of glutamatergic (excitatory) neurons (Bagley et al., 2017; Kadoshima et al., 2013; Pasca et al., 2015). These dorsal “cortical” organoids notably lack inhibitory interneurons. Similarly, SHH pathway agonists both in the presence and absence of WNT inhibition have been used to generate ventralized organoids with a large population of inhibitory interneurons (Bagley et al., 2017; Watanabe et al., 2017). In addition to dorsal and ventral forebrain organoids, this approach of directed differentiation has been used to generate a growing list of additional regional specificities, including hypothalamus, cerebellum, hippocampus, and others (Muguruma et al., 2015; Qian et al., 2016; Sakaguchi et al., 2015; reviewed in Di Lullo and Kriegstein, 2017).
Modeling Genetic Epilepsies and Their Effects on Cortical Network Function With Brain Organoids
A number of studies, primarily using directed differentiation of human forebrain cortex-like organoids, have utilized 3D culture approaches to determine the effects of epilepsy-associated genetic variants on various anatomical measures, including organoid cytoarchitecture and organization, cell expression patterns, and neuronal migration. This work has been reviewed elsewhere (Baldassari et al., 2020; Nieto-Estevez and Hsieh, 2020). Recent studies have additionally examined neural circuit activities generated by organoids. One of the first to do so utilized microelectrode arrays to document the generation of neural oscillations that increased in complexity with age (Trujillo et al., 2019). Measurements were performed in cortical organoids that were retained in culture from 2 to 10 months. In a more direct study of epilepsy, Sun and colleagues generated a cortical organoid model of Angelman syndrome resulting from a mutation in the ubiquitin ligase UBE3A gene (Sun et al., 2019). Angelman syndrome is associated with intellectual disability and lifelong seizures. Cortical organoids examined after 120–150 days in vitro demonstrated evidence of neuronal hyperexcitability by patch-clamp recording, and calcium indicator imaging revealed hypersynchronous discharges in the mutant organoids that were not observed in isogenic controls.
Both of these studies involved directed differentiation of forebrain cortex-like organoids, which would not be expected to contain a significant number of inhibitory interneurons. Interestingly, transcriptomic analyses by Trujillo and colleagues did reveal the spontaneous expression of an inhibitory interneuron population by 10 months in culture (Trujillo et al., 2019), which may partly explain the increased complexity of oscillatory activity observed by these authors at later time points. In contrast, and as expected, Sun et al. observed sparse inhibitory interneurons in their culture conditions (Sun et al., 2019). Given the importance of interneurons and excitatory-inhibitory connectivity in neural circuit activity in general and the pathophysiology of epilepsy in particular, recently developed approaches to integrate these populations within a single organoid are of particular relevance to organoid modeling of epilepsy. Birey and colleagues pioneered an innovative and elegant approach to achieve this by generating “assembloids” (also referred to as organoid “fusions”) in which glutamatergic neuron-rich dorsal and interneuron-rich ventral organoids are separately generated and then combined by placing the two structures adjacent to each other such that they physically fuse together (Birey et al., 2017). This protocol appears to recapitulate in an organoid the in vivo process of tangential migration of interneurons, whereby ventrally born interneurons and interneuron progenitors migrate and integrate into dorsal cortex. The assembloid approach has since been used to model circuit activity in an organoid model of RTT resulting from mutations in the MECP2 gene (Samarasinghe et al., 2021). In this study, MECP2 mutations were shown to result in hypersynchrony by calcium indicator imaging as well as both a loss of gamma oscillations and increased high-frequency activity (previously known as high-frequency oscillations [HFOs]) by extracellular recording of local field potentials. These authors also leveraged the assembloid approach to specifically identify ventral organoid derived interneurons, as opposed to dorsal excitatory neurons, as the primary drivers of the hyperexcitable phenotype.
Challenges and Future Directions
The use of hPSC-based epilepsy models presents a number of challenges. One issue is line-to-line variability of iPSC lines both within and between subjects. This variability requires the use of several lines each from multiple patients to show the consistency of any given finding. The use of isogenic controls generated by CRISPR gene editing also helps to address potential concerns that findings are due to genomic variability rather than the gene variant being studied. Another issue is cellular maturation. Because iPSC-derived models can only be grown in 2D culture conditions for up to a few months, they often represent a more immature state of the target cell, an issue common to many different tissues derived from hPSCs. This limitation is at least partially alleviated by brain organoid models, which can be grown in culture for extended periods. Both 2D and 3D hPSC cultures also are not yet able to generate the complete repertoire of human brain region-specific cell types or the entire repertoire of neuronal subtypes (discussed more below), which may cause difficulties in modeling certain human brain disorders. iPSC-derived neural cultures also do not recapitulate the complex anatomical connectivity that is present in vivo (See Figure 42–3).

Figure 42–3.
Advantages and disadvantages of hPSC-derived 2D and 3D models of brain disorders. Patient-derived, genome-edited, and isogenic control hPSCs may be differentiated into 2D monolayer cultures or 3D brain organoids (Lam et al., 2017). In hPSC-derived 2D (more...)
Brain organoid models share some of these challenges and have some important limitations that are unique to this approach, some of which can be addressed with new protocols or considered experimental planning. These challenges can be divided into three major categories: (1) limitations related to cellular diversity, (2) limitations in cellular maturity, and (3) potentially high levels of variability in organoid generation. Limitations in cellular diversity include a lack of specific non-neuronal cell types such as immune cells or vascular endothelial cells/components in most organoid preparations, as well concerns about limited expression of certain neuronal subtypes, including specific classes of interneurons such as fast spiking PV-expressing cells. In response to the lack of non-ectoderm-derived cell types, various approaches are being developed, including independent generation and application of particular cells types such as microglia (Bodnar et al., 2021; Xu et al., 2021), as well as entirely novel methods like “organoid on a chip” models in which microfluidic devices are used to both mimic vascular activity and the paracrine/autocrine influence of endothelial cells (Cho et al., 2021). The potential lack of neuronal diversity may be addressed by other innovations such as the dorsal-ventral assembloid approach, which appears to enhance interneuron diversity (Birey et al., 2017; Samarasinghe et al., 2021).
The fidelity of brain organoids to many aspects of human brain development, including their temporal fidelity to in vivo development, can pose a challenge when attempting to model diseases that typically manifest in advanced age. As such, organoid models are clearly well suited to disease processes that manifest early in development. This does not preclude successfully modeling neurological diseases that occur later in life since preclinical manifestations may be present in cellular composition or circuit activity well in advance of phenotypic expression. Approaches to induce “premature ageing” such as seeding an organoid with pathologic tau or α-synuclein fibrils or expressing genes associated with age-related neurodegeneration are additional approaches that can make organoids a more tractable approach for modeling neurological diseases that manifest later in life (Bowles et al., 2021).
A final concern with organoid-based models is variability in organoid preparations both within and between laboratories. Part of this variability stems from the diversity of protocols for organoid generation. As the field matures, this issue is being addressed by specific multi-investigator cross-institutional efforts to compare multiple organoid approaches and select for consistency and reliability, by increasing awareness of the key factors in stem cell and organoid maintenance and culture that are critical to consistency, and more organically by the emergence of dominant protocols that are becoming commercialized and increasingly used by many laboratories. Within laboratory batch-to-batch variability in organoid preparation is also a concern. This can be addressed by careful batch testing and bulk purchase of critical media components such as knockout serum and N-2 supplement. Careful quality control by immunohistochemistry or PCR for appropriate differentiation of organoids as well complementary accounting of batch-to-batch variability in physiological measures, as recently published (Samarasinghe et al., 2021), will provide further assurance of the reliability of organoid modeling. Even with these caveats, patient hPSC-derived 2D and 3D cultures have provided extensive insight into cellular mechanisms of genetic epilepsies, and their unique properties make them extremely useful for disease modeling, regenerative therapies, and drug/toxicity screening.
Challenges notwithstanding, the rapidly advancing field of stem cell biology suggests that future developments will expand the use of hPSC models in epilepsy research. Although at present hPSC models are predominantly used for genetic epilepsies, it is conceivable that this approach will enable the human cellular modeling of acquired epilepsies. For example, the ability to direct hPSC-derived 2D or brain organoid cultures into hippocampus-like tissue (Sakaguchi et al., 2015; Sarkar et al., 2018) should offer a paradigm for studying temporal lobe epilepsy. Moreover, hPSC-derived neurons are starting to be used as an in vitro model of traumatic brain injury (Shi et al., 2021), and this approach may prove useful in the future to study posttraumatic epilepsy mechanisms or for therapy development.
Another use of brain organoid methods currently under development is as a human tissue model for screening teratogenic effects of current and potential future antiseizure medications. Brain organoids, along with future protocols to generate spinal cord organoids, should aid in our understanding of the mechanisms underlying neural tube formation and, when used as a screening assay, decrease the incidence of neural tube defects from antiseizure medications and other drugs. Another future prospect for 3D cultures that should impact epilepsy research is the development of improved region-specific organoid patterning combined with assembloids. In addition to the existing excitatory/inhibitory cortical organoid fusions described above, it is likely that thalamic-cortical (Xiang et al., 2019) and hippocampal-subpallial fusion organoids will soon be applied to study epilepsy. Lastly, progress in the incorporation into brain organoids of cell types normally found in the brain but not present using current brain organoids protocols, including microglia, endothelial cells, and meningeal cells, should improve the fidelity of modeling epilepsy with human cells. The rapid pace of progress in hPSC-based disease modeling suggests that this field will contribute to both mechanistic understanding of and precision therapy for the epilepsies.
Conclusions
hPSC models provide a renewable source of human cells that are being increasingly applied to the study of genetic epilepsies. Many hPSC-based models have already provided important advances in the mechanistic understanding of a number of DEEs. In addition to insight into ictogenic substrates, these stem cell approaches may be applied to study neurodevelopmental comorbidities and complications of epilepsy, including SUDEP. Moreover, both 2D and 3D hPSC models of genetic epilepsy have great potential for guiding precision epilepsy therapy. All of these applications, especially when used in combination with genetically modified animal models, offer the prospect of improvements in the care of persons with epilepsy.
References
- Atasoy D and Sternson SM (2018) Chemogenetic Tools for Causal Cellular and Neuronal Biology. Physiol Rev 98:391–418. [PMC free article: PMC5866359] [PubMed: 29351511]
- Au KS, Williams AT, Roach ES, Batchelor L, Sparagana SP, Delgado MR, Wheless JW, Baumgartner JE, Roa BB, Wilson CM, Smith-Knuppel TK, Cheung MY, Whittemore VH, King TM and Northrup H (2007) Genotype/phenotype correlation in 325 individuals referred for a diagnosis of tuberous sclerosis complex in the United States. Genet Med 9:88–100. [PubMed: 17304050]
- Auerbach DS, Jones J, Clawson BC, Offord J, Lenk GM, Ogiwara I, Yamakawa K, Meisler MH, Parent JM and Isom LL (2013) Altered Cardiac Electrophysiology and SUDEP in a Model of Dravet Syndrome. PLoS One 8:e77843. [PMC free article: PMC3796479] [PubMed: 24155976]
- Avazzadeh S, McDonagh K, Reilly J, Wang Y, Boomkamp SD, McInerney V, Krawczyk J, Fitzgerald J, Feerick N, O’Sullivan M, Jalali A, Forman EB, Lynch SA, Ennis S, Cosemans N, Peeters H, Dockery P, O’Brien T, Quinlan LR, Gallagher L and Shen S (2019) Increased Ca(2+) signaling in NRXN1alpha (+/-) neurons derived from ASD induced pluripotent stem cells. Mol Autism 10:52. [PMC free article: PMC6937972] [PubMed: 31893021]
- Bagley JA, Reumann D, Bian S, Levi-Strauss J and Knoblich JA (2017) Fused cerebral organoids model interactions between brain regions. Nat Methods 14:743–751. [PMC free article: PMC5540177] [PubMed: 28504681]
- Bagnall RD, Crompton DE and Semsarian C (2017) Genetic Basis of Sudden Unexpected Death in Epilepsy. Frontiers in neurology 8:348. [PMC free article: PMC5517398] [PubMed: 28775708]
- Baldassari S, Musante I, Iacomino M, Zara F, Salpietro V and Scudieri P (2020) Brain Organoids as Model Systems for Genetic Neurodevelopmental Disorders. Front Cell Dev Biol 8:590119. [PMC free article: PMC7586734] [PubMed: 33154971]
- Bando Y, Grimm C, Cornejo VH and Yuste R (2019) Genetic voltage indicators. BMC Biol 17:71. [PMC free article: PMC6739974] [PubMed: 31514747]
- Baraban SC (2021) A zebrafish-centric approach to antiepileptic drug development. Dis Model Mech 14(7):dmm049080. [PMC free article: PMC8277967] [PubMed: 34231838]
- Bershteyn M and Kriegstein AR (2013) Cerebral organoids in a dish: progress and prospects. Cell 155:19–20. [PMC free article: PMC5127703] [PubMed: 24074857]
- Birey F, Andersen J, Makinson CD, Islam S, Wei W, Huber N, Fan HC, Metzler KRC, Panagiotakos G, Thom N, O’Rourke NA, Steinmetz LM, Bernstein JA, Hallmayer J, Huguenard JR and Pasca SP (2017) Assembly of functionally integrated human forebrain spheroids. Nature 545:54–59. [PMC free article: PMC5805137] [PubMed: 28445465]
- Blair JD and Bateup HS (2020) New frontiers in modeling tuberous sclerosis with human stem cell-derived neurons and brain organoids. Dev Dyn 249:46–55. [PMC free article: PMC6995669] [PubMed: 31070828]
- Blair JD, Hockemeyer D and Bateup HS (2018) Genetically engineered human cortical spheroid models of tuberous sclerosis. Nat Med 24:1568–1578. [PMC free article: PMC6261470] [PubMed: 30127391]
- Bodnar B, Zhang Y, Liu J, Lin Y, Wang P, Wei Z, Saribas S, Zhu Y, Li F, Wang X, Yang W, Li Q, Ho WZ and Hu W (2021) Novel Scalable and Simplified System to Generate Microglia-Containing Cerebral Organoids From Human Induced Pluripotent Stem Cells. Front Cell Neurosci 15:682272. [PMC free article: PMC8288463] [PubMed: 34290591]
- Bowles KR, Silva MC, Whitney K, Bertucci T, Berlind JE, Lai JD, Garza JC, Boles NC, Mahali S, Strang KH, Marsh JA, Chen C, Pugh DA, Liu Y, Gordon RE, Goderie SK, Chowdhury R, Lotz S, Lane K, Crary JF, Haggarty SJ, Karch CM, Ichida JK, Goate AM and Temple S (2021) ELAVL4, splicing, and glutamatergic dysfunction precede neuron loss in MAPT mutation cerebral organoids. Cell 184:4547-4563 e4517. [PMC free article: PMC8635409] [PubMed: 34314701]
- Buzsaki G, Anastassiou CA and Koch C (2012) The origin of extracellular fields and currents--EEG, ECoG, LFP and spikes. Nat Rev Neurosci 13:407–420. [PMC free article: PMC4907333] [PubMed: 22595786]
- Canals I, Ginisty A, Quist E, Timmerman R, Fritze J, Miskinyte G, Monni E, Hansen MG, Hidalgo I, Bryder D, Bengzon J and Ahlenius H (2018) Rapid and efficient induction of functional astrocytes from human pluripotent stem cells. Nat Methods 15:693–696. [PubMed: 30127505]
- Chambers SM, Fasano CA, Papapetrou EP, Tomishima M, Sadelain M and Studer L (2009) Highly efficient neural conversion of human ES and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol 27:275–280. [PMC free article: PMC2756723] [PubMed: 19252484]
- Chandrasekaran A, Avci HX, Ochalek A, Rosingh LN, Molnar K, Laszlo L, Bellak T, Teglasi A, Pesti K, Mike A, Phanthong P, Biro O, Hall V, Kitiyanant N, Krause KH, Kobolak J and Dinnyes A (2017) Comparison of 2D and 3D neural induction methods for the generation of neural progenitor cells from human induced pluripotent stem cells. Stem Cell Res 25:139–151. [PubMed: 29128818]
- Cheah CS, Yu FH, Westenbroek RE, Kalume FK, Oakley JC, Potter GB, Rubenstein JL and Catterall WA (2012) Specific deletion of NaV1.1 sodium channels in inhibitory interneurons causes seizures and premature death in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A 109:14646–14651. [PMC free article: PMC3437823] [PubMed: 22908258]
- Cheung AY, Horvath LM, Grafodatskaya D, Pasceri P, Weksberg R, Hotta A, Carrel L and Ellis J (2011) Isolation of MECP2-null Rett Syndrome patient hiPS cells and isogenic controls through X-chromosome inactivation. Hum Mol Genet 20:2103–2115. [PMC free article: PMC3090191] [PubMed: 21372149]
- Cho AN, Jin Y, An Y, Kim J, Choi YS, Lee JS, Kim J, Choi WY, Koo DJ, Yu W, Chang GE, Kim DY, Jo SH, Kim J, Kim SY, Kim YG, Kim JY, Choi N, Cheong E, Kim YJ, Je HS, Kang HC and Cho SW (2021) Microfluidic device with brain extracellular matrix promotes structural and functional maturation of human brain organoids. Nat Commun 12:4730. [PMC free article: PMC8342542] [PubMed: 34354063]
- Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA and Zhang F (2013) Multiplex genome engineering using CRISPR/Cas systems. Science 339:819–823. [PMC free article: PMC3795411] [PubMed: 23287718]
- Cooper MS, McIntosh A, Crompton DE, McMahon JM, Schneider A, Farrell K, Ganesan V, Gill D, Kivity S, Lerman-Sagie T, McLellan A, Pelekanos J, Ramesh V, Sadleir L, Wirrell E and Scheffer IE (2016) Mortality in Dravet syndrome. Epilepsy Res 128:43–47. [PubMed: 27810515]
- Costa V, Aigner S, Vukcevic M, Sauter E, Behr K, Ebeling M, Dunkley T, Friedlein A, Zoffmann S, Meyer CA, Knoflach F, Lugert S, Patsch C, Fjeldskaar F, Chicha-Gaudimier L, Kiialainen A, Piraino P, Bedoucha M, Graf M, Jessberger S, Ghosh A, Bischofberger J and Jagasia R (2016) mTORC1 Inhibition Corrects Neurodevelopmental and Synaptic Alterations in a Human Stem Cell Model of Tuberous Sclerosis. Cell Rep 15:86–95. [PubMed: 27052171]
- Cunningham M, Cho JH, Leung A, Savvidis G, Ahn S, Moon M, Lee PK, Han JJ, Azimi N, Kim KS, Bolshakov VY and Chung S (2014) hPSC-derived maturing GABAergic interneurons ameliorate seizures and abnormal behavior in epileptic mice. Cell Stem Cell 15:559–573. [PMC free article: PMC4270101] [PubMed: 25517465]
- Dana H, Sun Y, Mohar B, Hulse BK, Kerlin AM, Hasseman JP, Tsegaye G, Tsang A, Wong A, Patel R, Macklin JJ, Chen Y, Konnerth A, Jayaraman V, Looger LL, Schreiter ER, Svoboda K and Kim DS (2019) High-performance calcium sensors for imaging activity in neuronal populations and microcompartments. Nat Methods 16:649–657. [PubMed: 31209382]
- de Vries PJ, Wilde L, de Vries MC, Moavero R, Pearson DA and Curatolo P (2018) A clinical update on tuberous sclerosis complex-associated neuropsychiatric disorders (TAND). Am J Med Genet C Semin Med Genet 178:309–320. [PMC free article: PMC6209538] [PubMed: 30117265]
- Deisseroth K (2015) Optogenetics: 10 years of microbial opsins in neuroscience. Nat Neurosci 18:1213–1225. [PMC free article: PMC4790845] [PubMed: 26308982]
- Devinsky O, Hesdorffer DC, Thurman DJ, Lhatoo S and Richerson G (2016) Sudden unexpected death in epilepsy: epidemiology, mechanisms, and prevention. Lancet neurology 15:1075–1088. [PubMed: 27571159]
- Di Lullo E and Kriegstein AR (2017) The use of brain organoids to investigate neural development and disease. Nat Rev Neurosci 18:573–584. [PMC free article: PMC5667942] [PubMed: 28878372]
- Dolce A, Ben-Zeev B, Naidu S and Kossoff EH (2013) Rett syndrome and epilepsy: an update for child neurologists. Pediatr Neurol 48:337–345. [PubMed: 23583050]
- Douvaras P, Sun B, Wang M, Kruglikov I, Lallos G, Zimmer M, Terrenoire C, Zhang B, Gandy S, Schadt E, Freytes DO, Noggle S and Fossati V (2017) Directed Differentiation of Human Pluripotent Stem Cells to Microglia. Stem Cell Reports 8:1516–1524. [PMC free article: PMC5470097] [PubMed: 28528700]
- Dravet C, Bureau M, Oguni H, Fukuyama Y and Cokar O (2005) Severe myoclonic epilepsy in infancy (Dravet syndrome). [PubMed: 15508915]
- Ebrahimi-Fakhari D, Saffari A, Wahlster L, Di Nardo A, Turner D, Lewis TL, Jr., Conrad C, Rothberg JM, Lipton JO, Kolker S, Hoffmann GF, Han MJ, Polleux F and Sahin M (2016) Impaired Mitochondrial Dynamics and Mitophagy in Neuronal Models of Tuberous Sclerosis Complex. Cell Rep 17:1053–1070. [PMC free article: PMC5078873] [PubMed: 27760312]
- El Bissati K, Redel H, Ting LM, Lykins JD, McPhillie MJ, Upadhya R, Woster PM, Yarlett N, Kim K and Weiss LM (2019) Novel Synthetic Polyamines Have Potent Antimalarial Activities in vitro and in vivo by Decreasing Intracellular Spermidine and Spermine Concentrations. Front Cell Infect Microbiol 9:9. [PMC free article: PMC6382690] [PubMed: 30838177]
- Favero M, Sotuyo NP, Lopez E, Kearney JA and Goldberg EM (2018) A Transient Developmental Window of Fast-Spiking Interneuron Dysfunction in a Mouse Model of Dravet Syndrome. J Neurosci 38:7912–7927. [PMC free article: PMC6125809] [PubMed: 30104343]
- Frasier CR, Wagnon JL, Bao YO, McVeigh LG, Lopez-Santiago LF, Meisler MH and Isom LL (2016) Cardiac arrhythmia in a mouse model of sodium channel SCN8A epileptic encephalopathy. Proc Natl Acad Sci U S A 113(45):12838–12843. [PMC free article: PMC5111690] [PubMed: 27791149]
- Frasier CR, Zhang H, Offord J, Dang LT, Auerbach DS, Shi H, Chen C, Goldman AM, Eckhardt LL, Bezzerides VJ, Parent JM and Isom LL (2018) Channelopathy as a SUDEP biomarker in Dravet Syndrome patient-derived cardiac myocytes. Stem cell reports 11(3):626–634. [PMC free article: PMC6135724] [PubMed: 30146492]
- Fujiwara T (2006) Clinical spectrum of mutations in SCN1A gene: severe myoclonic epilepsy in infancy and related epilepsies. Epilepsy Res 70 Suppl 1:S223-230. [PubMed: 16806826]
- Goff KM and Goldberg EM (2019) Vasoactive intestinal peptide-expressing interneurons are impaired in a mouse model of Dravet syndrome. Elife 8:e46846. [PMC free article: PMC6629374] [PubMed: 31282864]
- Goldman AM, Glasscock E, Yoo J, Chen TT, Klassen TL and Noebels JL (2009) Arrhythmia in heart and brain: KCNQ1 mutations link epilepsy and sudden unexplained death. Sci Transl Med 1:2ra6. [PMC free article: PMC2951754] [PubMed: 20368164]
- Grewe BF, Langer D, Kasper H, Kampa BM and Helmchen F (2010) High-speed in vivo calcium imaging reveals neuronal network activity with near-millisecond precision. Nat Methods 7:399–405. [PubMed: 20400966]
- Grienberger C and Konnerth A (2012) Imaging calcium in neurons. Neuron 73:862–885. [PubMed: 22405199]
- Guerrini R and Aicardi J (2003) Epileptic encephalopathies with myoclonic seizures in infants and children (severe myoclonic epilepsy and myoclonic-astatic epilepsy). J Clin Neurophysiol 20:449–461. [PubMed: 14734934]
- Hansen DV, Lui JH, Parker PR and Kriegstein AR (2010) Neurogenic radial glia in the outer subventricular zone of human neocortex. Nature 464:554–561. [PubMed: 20154730]
- Hasbani DM and Crino PB (2018) Tuberous sclerosis complex. Handb Clin Neurol 148:813–822. [PubMed: 29478616]
- Higurashi N, Uchida T, Lossin C, Misumi Y, Okada Y, Akamatsu W, Imaizumi Y, Zhang B, Nabeshima K, Mori MX, Katsurabayashi S, Shirasaka Y, Okano H and Hirose S (2013) A human Dravet syndrome model from patient induced pluripotent stem cells. Mol Brain 6:19. [PMC free article: PMC3655893] [PubMed: 23639079]
- Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T and Ishii A (2020) Application of induced pluripotent stem cells in epilepsy. Mol Cell Neurosci 108:103535. [PubMed: 32758699]
- Hodge RD, Bakken TE, Miller JA, Smith KA, Barkan ER, Graybuck LT, Close JL, Long B, Johansen N, Penn O, Yao Z, Eggermont J, Hollt T, Levi BP, Shehata SI, Aevermann B, Beller A, Bertagnolli D, Brouner K, Casper T, Cobbs C, Dalley R, Dee N, Ding SL, Ellenbogen RG, Fong O, Garren E, Goldy J, Gwinn RP, Hirschstein D, Keene CD, Keshk M, Ko AL, Lathia K, Mahfouz A, Maltzer Z, McGraw M, Nguyen TN, Nyhus J, Ojemann JG, Oldre A, Parry S, Reynolds S, Rimorin C, Shapovalova NV, Somasundaram S, Szafer A, Thomsen ER, Tieu M, Quon G, Scheuermann RH, Yuste R, Sunkin SM, Lelieveldt B, Feng D, Ng L, Bernard A, Hawrylycz M, Phillips JW, Tasic B, Zeng H, Jones AR, Koch C and Lein ES (2019) Conserved cell types with divergent features in human versus mouse cortex. Nature 573:61–68. [PMC free article: PMC6919571] [PubMed: 31435019]
- Hu JS, Vogt D, Sandberg M and Rubenstein JL (2017) Cortical interneuron development: a tale of time and space. Development 144:3867–3878. [PMC free article: PMC5702067] [PubMed: 29089360]
- Huang WA, Boyle NG and Vaseghi M (2017) Cardiac Innervation and the Autonomic Nervous System in Sudden Cardiac Death. Card Electrophysiol Clin 9:665–679. [PMC free article: PMC5777242] [PubMed: 29173409]
- Ichise E, Chiyonobu T, Ishikawa M, Tanaka Y, Shibata M, Tozawa T, Taura Y, Yamashita S, Yoshida M, Morimoto M, Higurashi N, Yamamoto T, Okano H and Hirose S (2021) Impaired neuronal activity and differential gene expression in STXBP1 encephalopathy patient iPSC-derived GABAergic neurons. Hum Mol Genet 30:1337–1348. [PubMed: 33961044]
- Ip JPK, Mellios N and Sur M (2018) Rett syndrome: insights into genetic, molecular and circuit mechanisms. Nat Rev Neurosci 19:368–382. [PMC free article: PMC6402579] [PubMed: 29740174]
- Izsak J, Seth H, Andersson M, Vizlin-Hodzic D, Theiss S, Hanse E, Agren H, Funa K and Illes S (2019) Robust Generation of Person-Specific, Synchronously Active Neuronal Networks Using Purely Isogenic Human iPSC-3D Neural Aggregate Cultures. Front Neurosci 13:351. [PMC free article: PMC6491690] [PubMed: 31068774]
- Jiang X, Lachance M and Rossignol E (2016) Involvement of cortical fast-spiking parvalbumin-positive basket cells in epilepsy. Prog Brain Res 226:81–126. [PMC free article: PMC5218834] [PubMed: 27323940]
- Jiao J, Yang Y, Shi Y, Chen J, Gao R, Fan Y, Yao H, Liao W, Sun XF and Gao S (2013) Modeling Dravet syndrome using induced pluripotent stem cells (iPSCs) and directly converted neurons. Hum Mol Genet 22:4241–4252. [PubMed: 23773995]
- Kadoshima T, Sakaguchi H, Nakano T, Soen M, Ando S, Eiraku M and Sasai Y (2013) Self-organization of axial polarity, inside-out layer pattern, and species-specific progenitor dynamics in human ES cell-derived neocortex. Proc Natl Acad Sci U S A 110:20284–20289. [PMC free article: PMC3864329] [PubMed: 24277810]
- Kim HW, Quan Z, Kim YB, Cheong E, Kim HD, Cho M, Jang J, Yoo YR, Lee JS, Kim JH, Kim YI, Kim DS and Kang HC (2018) Differential effects on sodium current impairments by distinct SCN1A mutations in GABAergic neurons derived from Dravet syndrome patients. Brain Dev 40:287–298. [PubMed: 29295803]
- Kim JJ, Savas JN, Miller MT, Hu X, Carromeu C, Lavallee-Adam M, Freitas BCG, Muotri AR, Yates JR, 3rd and Ghosh A (2019) Proteomic analyses reveal misregulation of LIN28 expression and delayed timing of glial differentiation in human iPS cells with MECP2 loss-of-function. PLoS One 14:e0212553. [PMC free article: PMC6383942] [PubMed: 30789962]
- Knopfel T and Song C (2019) Optical voltage imaging in neurons: moving from technology development to practical tool. Nat Rev Neurosci 20:719–727. [PubMed: 31705060]
- Krencik R and Zhang SC (2011) Directed differentiation of functional astroglial subtypes from human pluripotent stem cells. Nat Protoc 6:1710–1717. [PMC free article: PMC3198813] [PubMed: 22011653]
- Krey JF, Pasca SP, Shcheglovitov A, Yazawa M, Schwemberger R, Rasmusson R and Dolmetsch RE (2013) Timothy syndrome is associated with activity-dependent dendritic retraction in rodent and human neurons. Nat Neurosci 16:201–209. [PMC free article: PMC3568452] [PubMed: 23313911]
- Lam RS, Topfer FM, Wood PG, Busskamp V and Bamberg E (2017) Functional Maturation of Human Stem Cell-Derived Neurons in Long-Term Cultures. PLoS One 12:e0169506. [PMC free article: PMC5215418] [PubMed: 28052116]
- Lancaster MA, Renner M, Martin CA, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP and Knoblich JA (2013) Cerebral organoids model human brain development and microcephaly. Nature 501:373–379. [PMC free article: PMC3817409] [PubMed: 23995685]
- Lanoue V, Chai YJ, Brouillet JZ, Weckhuysen S, Palmer EE, Collins BM and Meunier FA (2019) STXBP1 encephalopathy: Connecting neurodevelopmental disorders with alpha-synucleinopathies? Neurology 93:114–123. [PubMed: 31221716]
- Li MCH, O’Brien TJ, Todaro M and Powell KL (2019) Acquired cardiac channelopathies in epilepsy: Evidence, mechanisms, and clinical significance. Epilepsia 60:1753–1767. [PubMed: 31353444]
- Li Y, Cao J, Chen M, Li J, Sun Y, Zhang Y, Zhu Y, Wang L and Zhang C (2017) Abnormal Neural Progenitor Cells Differentiated from Induced Pluripotent Stem Cells Partially Mimicked Development of TSC2 Neurological Abnormalities. Stem Cell Reports 8:883–893. [PMC free article: PMC5390135] [PubMed: 28344003]
- Lin X, O’Malley H, Chen C, Auerbach D, Foster M, Shekhar A, Zhang M, Coetzee W, Jalife J, Fishman GI, Isom L and Delmar M (2015) Scn1b deletion leads to increased tetrodotoxin-sensitive sodium current, altered intracellular calcium homeostasis and arrhythmias in murine hearts. J Physiol 593:1389–1407. [PMC free article: PMC4376420] [PubMed: 25772295]
- Liu Y, Liu H, Sauvey C, Yao L, Zarnowska ED and Zhang SC (2013a) Directed differentiation of forebrain GABA interneurons from human pluripotent stem cells. Nat Protoc 8:1670–1679. [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 and Parent JM (2013b) Dravet syndrome patient-derived neurons suggest a novel epilepsy mechanism. Ann Neurol 74:128–139. [PMC free article: PMC3775921] [PubMed: 23821540]
- Lopez-Santiago LF, Meadows LS, Ernst SJ, Chen C, Malhotra JD, McEwen DP, Speelman A, Noebels JL, Maier SK, Lopatin AN and Isom LL (2007) Sodium channel Scn1b null mice exhibit prolonged QT and RR intervals. J Mol Cell Cardiol 43:636–647. [PMC free article: PMC2099572] [PubMed: 17884088]
- Magnus CJ, Lee PH, Bonaventura J, Zemla R, Gomez JL, Ramirez MH, Hu X, Galvan A, Basu J, Michaelides M and Sternson SM (2019) Ultrapotent chemogenetics for research and potential clinical applications. Science 364(6436):eaav5282. [PMC free article: PMC7252514] [PubMed: 30872534]
- Marchetto MC, Carromeu C, Acab A, Yu D, Yeo GW, Mu Y, Chen G, Gage FH and Muotri AR (2010) A model for neural development and treatment of Rett syndrome using human induced pluripotent stem cells. Cell 143:527–539. [PMC free article: PMC3003590] [PubMed: 21074045]
- Marini C, Scheffer IE, Nabbout R, Suls A, De Jonghe P, Zara F and Guerrini R (2011) The genetics of Dravet syndrome. Epilepsia 52 Suppl 2:24–29. [PubMed: 21463275]
- 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 and Studer L (2013) Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells. Cell Stem Cell 12:559–572. [PMC free article: PMC3681523] [PubMed: 23642365]
- Martinez-Cerdeno V, Cunningham CL, Camacho J, Antczak JL, Prakash AN, Cziep ME, Walker AI and Noctor SC (2012) Comparative analysis of the subventricular zone in rat, ferret and macaque: evidence for an outer subventricular zone in rodents. PLoS One 7:e30178. [PMC free article: PMC3260244] [PubMed: 22272298]
- Massey CA, Sowers LP, Dlouhy BJ and Richerson GB (2014) Mechanisms of sudden unexpected death in epilepsy: the pathway to prevention. Nat Rev Neurol 10(5):271–282. [PMC free article: PMC4565133] [PubMed: 24752120]
- Meissner A, Wernig M and Jaenisch R (2007) Direct reprogramming of genetically unmodified fibroblasts into pluripotent stem cells. Nat Biotechnol 25:1177–1181. [PubMed: 17724450]
- Mellios N, Feldman DA, Sheridan SD, Ip JPK, Kwok S, Amoah SK, Rosen B, Rodriguez BA, Crawford B, Swaminathan R, Chou S, Li Y, Ziats M, Ernst C, Jaenisch R, Haggarty SJ and Sur M (2018) MeCP2-regulated miRNAs control early human neurogenesis through differential effects on ERK and AKT signaling. Mol Psychiatry 23:1051–1065. [PMC free article: PMC5815944] [PubMed: 28439102]
- Mistry AM, Thompson CH, Miller AR, Vanoye CG, George AL, Jr. and Kearney JA (2014) Strain- and age-dependent hippocampal neuron sodium currents correlate with epilepsy severity in Dravet syndrome mice. Neurobiol Dis 65:1–11. [PMC free article: PMC3968814] [PubMed: 24434335]
- Muguruma K, Nishiyama A, Kawakami H, Hashimoto K and Sasai Y (2015) Self-organization of polarized cerebellar tissue in 3D culture of human pluripotent stem cells. Cell reports 10:537–550. [PubMed: 25640179]
- Muratore CR, Srikanth P, Callahan DG and Young-Pearse TL (2014) Comparison and optimization of hiPSC forebrain cortical differentiation protocols. PLoS One 9:e105807. [PMC free article: PMC4148335] [PubMed: 25165848]
- Nadadhur AG, Alsaqati M, Gasparotto L, Cornelissen-Steijger P, van Hugte E, Dooves S, Harwood AJ and Heine VM (2019) Neuron-Glia Interactions Increase Neuronal Phenotypes in Tuberous Sclerosis Complex Patient iPSC-Derived Models. Stem Cell Reports 12:42–56. [PMC free article: PMC6335594] [PubMed: 30581017]
- Nashef L, So EL, Ryvlin P and Tomson T (2012) Unifying the definitions of sudden unexpected death in epilepsy. Epilepsia 53:227–233. [PubMed: 22191982]
- Nerbonne JM (2014) Mouse models of arrhythmogenic cardiovascular disease: challenges and opportunities. Current opinion in pharmacology 15:107–114. [PMC free article: PMC3984610] [PubMed: 24632325]
- Nicholas CR, Chen J, Tang Y, Southwell DG, Chalmers N, Vogt D, Arnold CM, Chen YJ, Stanley EG, Elefanty AG, Sasai Y, Alvarez-Buylla A, Rubenstein JL and Kriegstein AR (2013) Functional maturation of hPSC-derived forebrain interneurons requires an extended timeline and mimics human neural development. Cell Stem Cell 12:573–586. [PMC free article: PMC3699205] [PubMed: 23642366]
- Nieto-Estevez V and Hsieh J (2020) Human Brain Organoid Models of Developmental Epilepsies. Epilepsy Curr 20:282–290. [PMC free article: PMC7653656] [PubMed: 33017223]
- Obien ME, Deligkaris K, Bullmann T, Bakkum DJ and Frey U (2014) Revealing neuronal function through microelectrode array recordings. Front Neurosci 8:423. [PMC free article: PMC4285113] [PubMed: 25610364]
- Odawara A, Katoh H, Matsuda N and Suzuki I (2016) Physiological maturation and drug responses of human induced pluripotent stem cell-derived cortical neuronal networks in long-term culture. Sci Rep 6:26181. [PMC free article: PMC4870631] [PubMed: 27188845]
- Ogiwara I, Miyamoto H, Morita N, Atapour N, Mazaki E, Inoue I, Takeuchi T, Itohara S, Yanagawa Y, Obata K, Furuichi T, Hensch TK and Yamakawa K (2007) Nav1.1 localizes to axons of parvalbumin-positive inhibitory interneurons: a circuit basis for epileptic seizures in mice carrying an Scn1a gene mutation. J Neurosci 27:5903–5914. [PMC free article: PMC6672241] [PubMed: 17537961]
- Ohashi M, Korsakova E, Allen D, Lee P, Fu K, Vargas BS, Cinkornpumin J, Salas C, Park JC, Germanguz I, Langerman J, Chronis C, Kuoy E, Tran S, Xiao X, Pellegrini M, Plath K and Lowry WE (2018) Loss of MECP2 Leads to Activation of P53 and Neuronal Senescence. Stem Cell Reports 10:1453–1463. [PMC free article: PMC5995366] [PubMed: 29742391]
- Okita K, Yamakawa T, Matsumura Y, Sato Y, Amano N, Watanabe A, Goshima N and Yamanaka S (2013) An efficient nonviral method to generate integration-free human-induced pluripotent stem cells from cord blood and peripheral blood cells. Stem Cells 31:458–466. [PubMed: 23193063]
- Pak C, Danko T, Zhang Y, Aoto J, Anderson G, Maxeiner S, Yi F, Wernig M and Sudhof TC (2015) Human Neuropsychiatric Disease Modeling using Conditional Deletion Reveals Synaptic Transmission Defects Caused by Heterozygous Mutations in NRXN1. Cell Stem Cell 17:316–328. [PMC free article: PMC4560990] [PubMed: 26279266]
- Pasca AM, Sloan SA, Clarke LE, Tian Y, Makinson CD, Huber N, Kim CH, Park JY, O’Rourke NA, Nguyen KD, Smith SJ, Huguenard JR, Geschwind DH, Barres BA and Pasca SP (2015) Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat Methods 12:671–678. [PMC free article: PMC4489980] [PubMed: 26005811]
- Patzke C, Han Y, Covy J, Yi F, Maxeiner S, Wernig M and Sudhof TC (2015) Analysis of conditional heterozygous STXBP1 mutations in human neurons. J Clin Invest 125:3560–3571. [PMC free article: PMC4588304] [PubMed: 26280581]
- Piatkevich KD, Bensussen S, Tseng HA, Shroff SN, Lopez-Huerta VG, Park D, Jung EE, Shemesh OA, Straub C, Gritton HJ, Romano MF, Costa E, Sabatini BL, Fu Z, Boyden ES and Han X (2019) Population imaging of neural activity in awake behaving mice. Nature 574:413–417. [PMC free article: PMC6858559] [PubMed: 31597963]
- Pnevmatikakis EA, Soudry D, Gao Y, Machado TA, Merel J, Pfau D, Reardon T, Mu Y, Lacefield C, Yang W, Ahrens M, Bruno R, Jessell TM, Peterka DS, Yuste R and Paninski L (2016) Simultaneous Denoising, Deconvolution, and Demixing of Calcium Imaging Data. Neuron 89:285–299. [PMC free article: PMC4881387] [PubMed: 26774160]
- Qian X, Nguyen HN, Song MM, Hadiono C, Ogden SC, Hammack C, Yao B, Hamersky GR, Jacob F, Zhong C, Yoon KJ, Jeang W, Lin L, Li Y, Thakor J, Berg DA, Zhang C, Kang E, Chickering M, Nauen D, Ho CY, Wen Z, Christian KM, Shi PY, Maher BJ, Wu H, Jin P, Tang H, Song H and Ming GL (2016) Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell 165:1238–1254. [PMC free article: PMC4900885] [PubMed: 27118425]
- Que Z, Olivero-Acosta MI, Zhang J, Eaton M, Tukker AM, Chen X, Wu J, Xie J, Xiao T, Wettschurack K, Yunis L, Shafer JM, Schaber JA, Rochet JC, Bowman AB, Yuan C, Huang Z, Hu CD, Trader DJ, Skarnes WC and Yang Y (2021) Hyperexcitability and pharmacological responsiveness of cortical neurons derived from human iPSCs carrying epilepsy-associated sodium channel Nav1.2-L1342P genetic variant. J Neurosci 41(49):10194–10208. [PMC free article: PMC8660047] [PubMed: 34716231]
- Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA and Zhang F (2013) Genome engineering using the CRISPR-Cas9 system. Nat Protoc 8:2281–2308. [PMC free article: PMC3969860] [PubMed: 24157548]
- Roberts B, Haupt A, Tucker A, Grancharova T, Arakaki J, Fuqua MA, Nelson A, Hookway C, Ludmann SA, Mueller IA, Yang R, Horwitz R, Rafelski SM and Gunawardane RN (2017) Systematic gene tagging using CRISPR/Cas9 in human stem cells to illuminate cell organization. Molecular biology of the cell 28:2854–2874. [PMC free article: PMC5638588] [PubMed: 28814507]
- Saitsu H, Kato M, Mizuguchi T, Hamada K, Osaka H, Tohyama J, Uruno K, Kumada S, Nishiyama K, Nishimura A, Okada I, Yoshimura Y, Hirai S, Kumada T, Hayasaka K, Fukuda A, Ogata K and Matsumoto N (2008) De novo mutations in the gene encoding STXBP1 (MUNC18-1) cause early infantile epileptic encephalopathy. Nat Genet 40:782–788. [PubMed: 18469812]
- Sakaguchi H, Kadoshima T, Soen M, Narii N, Ishida Y, Ohgushi M, Takahashi J, Eiraku M and Sasai Y (2015) Generation of functional hippocampal neurons from self-organizing human embryonic stem cell-derived dorsomedial telencephalic tissue. Nat Commun 6:8896. [PMC free article: PMC4660208] [PubMed: 26573335]
- Samarasinghe RA, Miranda OA, Buth JE, Mitchell S, Ferando I, Watanabe M, Allison TF, Kurdian A, Fotion NN, Gandal MJ, Golshani P, Plath K, Lowry WE, Parent JM, Mody I and Novitch BG (2021) Identification of neural oscillations and epileptiform changes in human brain organoids. Nat Neurosci 24(10):1488–1500. [PMC free article: PMC9070733] [PubMed: 34426698]
- Sarkar A, Mei A, Paquola ACM, Stern S, Bardy C, Klug JR, Kim S, Neshat N, Kim HJ, Ku M, Shokhirev MN, Adamowicz DH, Marchetto MC, Jappelli R, Erwin JA, Padmanabhan K, Shtrahman M, Jin X and Gage FH (2018) Efficient Generation of CA3 Neurons from Human Pluripotent Stem Cells Enables Modeling of Hippocampal Connectivity In Vitro. Cell Stem Cell 22:684-697 e689. [PMC free article: PMC6345574] [PubMed: 29727680]
- Schafer ST, Paquola ACM, Stern S, Gosselin D, Ku M, Pena M, Kuret TJM, Liyanage M, Mansour AA, Jaeger BN, Marchetto MC, Glass CK, Mertens J and Gage FH (2019) Pathological priming causes developmental gene network heterochronicity in autistic subject-derived neurons. Nat Neurosci 22:243–255. [PMC free article: PMC6402576] [PubMed: 30617258]
- Schuele SU, Bermeo AC, Alexopoulos AV, Locatelli ER, Burgess RC, Dinner DS and Foldvary-Schaefer N (2007a) Video-electrographic and clinical features in patients with ictal asystole. Neurology 69:434–441. [PubMed: 17664402]
- Schuele SU, Widdess-Walsh P, Bermeo A and Luders HO (2007b) Sudden unexplained death in epilepsy: the role of the heart. Cleve Clin J Med 74 Suppl 1:S121-127. [PubMed: 17455560]
- Schuster J, Laan L, Klar J, Jin Z, Huss M, Korol S, Noraddin FH, Sobol M, Birnir B and Dahl N (2019) Transcriptomes of Dravet syndrome iPSC derived GABAergic cells reveal dysregulated pathways for chromatin remodeling and neurodevelopment. Neurobiol Dis 132:104583. [PubMed: 31445158]
- Seto Y and Eiraku M (2019) Human brain development and its in vitro recapitulation. Neurosci Res 138:33–42. [PubMed: 30326251]
- Shi W, Dong P, Kuss MA, Gu L, Kievit F, Kim HJ and Duan B (2021) Design and Evaluation of an In Vitro Mild Traumatic Brain Injury Modeling System Using 3D Printed Mini Impact Device on the 3D Cultured Human iPSC Derived Neural Progenitor Cells. Adv Healthc Mater 10:e2100180. [PMC free article: PMC8222191] [PubMed: 33890428]
- Shi Y, Kirwan P and Livesey FJ (2012) Directed differentiation of human pluripotent stem cells to cerebral cortex neurons and neural networks. Nat Protoc 7:1836–1846. [PubMed: 22976355]
- Shiri Z, Levesque M, Etter G, Manseau F, Williams S and Avoli M (2017) Optogenetic Low-Frequency Stimulation of Specific Neuronal Populations Abates Ictogenesis. J Neurosci 37:2999–3008. [PMC free article: PMC6596724] [PubMed: 28209738]
- Shorvon S and Tomson T (2011) Sudden unexpected death in epilepsy. Lancet 378:2028–2038. [PubMed: 21737136]
- Skluzacek JV, Watts KP, Parsy O, Wical B and Camfield P (2011) Dravet syndrome and parent associations: the IDEA League experience with comorbid conditions, mortality, management, adaptation, and grief. Epilepsia 52 Suppl 2:95–101. [PubMed: 21463290]
- Stamberger H, Nikanorova M, Willemsen MH, Accorsi P, Angriman M, Baier H, Benkel-Herrenbrueck I, Benoit V, Budetta M, Caliebe A, Cantalupo G, Capovilla G, Casara G, Courage C, Deprez M, Destree A, Dilena R, Erasmus CE, Fannemel M, Fjaer R, Giordano L, Helbig KL, Heyne HO, Klepper J, Kluger GJ, Lederer D, Lodi M, Maier O, Merkenschlager A, Michelberger N, Minetti C, Muhle H, Phalin J, Ramsey K, Romeo A, Schallner J, Schanze I, Shinawi M, Sleegers K, Sterbova K, Syrbe S, Traverso M, Tzschach A, Uldall P, Van Coster R, Verhelst H, Viri M, Winter S, Wolff M, Zenker M, Zoccante L, De Jonghe P, Helbig I, Striano P, Lemke JR, Moller RS and Weckhuysen S (2016) STXBP1 encephalopathy: A neurodevelopmental disorder including epilepsy. Neurology 86:954–962. [PubMed: 26865513]
- Storace DA, Braubach OR, Jin L, Cohen LB and Sung U (2015) Monitoring brain activity with protein voltage and calcium sensors. Sci Rep 5:10212. [PMC free article: PMC4429559] [PubMed: 25970202]
- Sun AX, Yuan Q, Fukuda M, Yu W, Yan H, Lim GGY, Nai MH, D’Agostino GA, Tran HD, Itahana Y, Wang D, Lokman H, Itahana K, Lim SWL, Tang J, Chang YY, Zhang M, Cook SA, Rackham OJL, Lim CT, Tan EK, Ng HH, Lim KL, Jiang YH and Je HS (2019) Potassium channel dysfunction in human neuronal models of Angelman syndrome. Science 366:1486–1492. [PMC free article: PMC7735558] [PubMed: 31857479]
- Sun AX, Yuan Q, Tan S, Xiao Y, Wang D, Khoo AT, Sani L, Tran HD, Kim P, Chiew YS, Lee KJ, Yen YC, Ng HH, Lim B and Je HS (2016a) Direct Induction and Functional Maturation of Forebrain GABAergic Neurons from Human Pluripotent Stem Cells. Cell Rep 16:1942–1953. [PubMed: 27498872]
- 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 and Dolmetsch RE (2016b) A deleterious Nav1.1 mutation selectively impairs telencephalic inhibitory neurons derived from Dravet Syndrome patients. Elife 5:e13073. [PMC free article: PMC4961470] [PubMed: 27458797]
- Surges R, Adjei P, Kallis C, Erhuero J, Scott CA, Bell GS, Sander JW and Walker MC (2010) Pathologic cardiac repolarization in pharmacoresistant epilepsy and its potential role in sudden unexpected death in epilepsy: a case-control study. Epilepsia 51:233–242. [PubMed: 19817816]
- Surges R, Thijs RD, Tan HL and Sander JW (2009) Sudden unexpected death in epilepsy: risk factors and potential pathomechanisms. Nat Rev Neurol 5:492–504. [PubMed: 19668244]
- Tai C, Abe Y, Westenbroek RE, Scheuer T and Catterall WA (2014) Impaired excitability of somatostatin- and parvalbumin-expressing cortical interneurons in a mouse model of Dravet syndrome. Proc Natl Acad Sci U S A 111:E3139-3148. [PMC free article: PMC4121787] [PubMed: 25024183]
- Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K and Yamanaka S (2007) Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 131:861–872. [PubMed: 18035408]
- Takahashi K and Yamanaka S (2006) Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126:663–676. [PubMed: 16904174]
- Tan AY and Verrier RL (2013) The role of the autonomic nervous system in cardiac arrhythmias. Handbook of clinical neurology 117:135–145. [PubMed: 24095122]
- Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS and Jones JM (1998) Embryonic stem cell lines derived from human blastocysts. Science 282:1145–1147. [PubMed: 9804556]
- Tidball AM, Dang LT, Glenn TW, Kilbane EG, Klarr DJ, Margolis JL, Uhler MD and Parent JM (2017) Rapid Generation of Human Genetic Loss-of-Function iPSC Lines by Simultaneous Reprogramming and Gene Editing. Stem Cell Reports 9:725–731. [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 and Parent JM (2020) Variant-specific changes in persistent or resurgent sodium current in SCN8A-related epilepsy patient-derived neurons. Brain 143:3025–3040. [PMC free article: PMC7780473] [PubMed: 32968789]
- Trujillo CA, Gao R, Negraes PD, Gu J, Buchanan J, Preissl S, Wang A, Wu W, Haddad GG, Chaim IA, Domissy A, Vandenberghe M, Devor A, Yeo GW, Voytek B and Muotri AR (2019) Complex Oscillatory Waves Emerging from Cortical Organoids Model Early Human Brain Network Development. Cell Stem Cell 25:558-569 e557. [PMC free article: PMC6778040] [PubMed: 31474560]
- Upadhya D, Hattiangady B, Castro OW, Shuai B, Kodali M, Attaluri S, Bates A, Dong Y, Zhang SC, Prockop DJ and Shetty AK (2019) Human induced pluripotent stem cell-derived MGE cell grafting after status epilepticus attenuates chronic epilepsy and comorbidities via synaptic integration. Proc Natl Acad Sci U S A 116:287–296. [PMC free article: PMC6320542] [PubMed: 30559206]
- Vogelstein JT, Packer AM, Machado TA, Sippy T, Babadi B, Yuste R and Paninski L (2010) Fast nonnegative deconvolution for spike train inference from population calcium imaging. J Neurophysiol 104:3691–3704. [PMC free article: PMC3007657] [PubMed: 20554834]
- Watanabe M, Buth JE, Vishlaghi N, de la Torre-Ubieta L, Taxidis J, Khakh BS, Coppola G, Pearson CA, Yamauchi K, Gong D, Dai X, Damoiseaux R, Aliyari R, Liebscher S, Schenke-Layland K, Caneda C, Huang EJ, Zhang Y, Cheng G, Geschwind DH, Golshani P, Sun R and Novitch BG (2017) Self-Organized Cerebral Organoids with Human-Specific Features Predict Effective Drugs to Combat Zika Virus Infection. Cell Rep 21:517–532. [PMC free article: PMC5637483] [PubMed: 29020636]
- Williams EC, Zhong X, Mohamed A, Li R, Liu Y, Dong Q, Ananiev GE, Mok JC, Lin BR, Lu J, Chiao C, Cherney R, Li H, Zhang SC and Chang Q (2014) Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wild-type neurons. Hum Mol Genet 23:2968–2980. [PMC free article: PMC4014193] [PubMed: 24419315]
- Winden KD, Sundberg M, Yang C, Wafa SMA, Dwyer S, Chen PF, Buttermore ED and Sahin M (2019) Biallelic Mutations in TSC2 Lead to Abnormalities Associated with Cortical Tubers in Human iPSC-Derived Neurons. J Neurosci 39:9294–9305. [PMC free article: PMC6867816] [PubMed: 31591157]
- Xiang Y, Tanaka Y, Cakir B, Patterson B, Kim KY, Sun P, Kang YJ, Zhong M, Liu X, Patra P, Lee SH, Weissman SM and Park IH (2019) hESC-Derived Thalamic Organoids Form Reciprocal Projections When Fused with Cortical Organoids. Cell stem cell 24:487-497 e487. [PMC free article: PMC6853597] [PubMed: 30799279]
- Xie Y, Ng NN, Safrina OS, Ramos CM, Ess KC, Schwartz PH, Smith MA and O’Dowd DK (2020) Comparisons of dual isogenic human iPSC pairs identify functional alterations directly caused by an epilepsy associated SCN1A mutation. Neurobiol Dis 134:104627. [PubMed: 31786370]
- Xu R, Boreland AJ, Li X, Erickson C, Jin M, Atkins C, Pang ZP, Daniels BP and Jiang P (2021) Developing human pluripotent stem cell-based cerebral organoids with a controllable microglia ratio for modeling brain development and pathology. Stem Cell Reports 16:1923–1937. [PMC free article: PMC8365109] [PubMed: 34297942]
- Yamashita S, Chiyonobu T, Yoshida M, Maeda H, Zuiki M, Kidowaki S, Isoda K, Morimoto M, Kato M, Saitsu H, Matsumoto N, Nakahata T, Saito MK and Hosoi H (2016) Mislocalization of syntaxin-1 and impaired neurite growth observed in a human iPSC model for STXBP1-related epileptic encephalopathy. Epilepsia 57:e81-86. [PubMed: 26918652]
- Yang N, Chanda S, Marro S, Ng YH, Janas JA, Haag D, Ang CE, Tang Y, Flores Q, Mall M, Wapinski O, Li M, Ahlenius H, Rubenstein JL, Chang HY, Buylla AA, Sudhof TC and Wernig M (2017) Generation of pure GABAergic neurons by transcription factor programming. Nat Methods 14:621–628. [PMC free article: PMC5567689] [PubMed: 28504679]
- Yekhlef L, Breschi GL and Taverna S (2017) Optogenetic activation of VGLUT2-expressing excitatory neurons blocks epileptic seizure-like activity in the mouse entorhinal cortex. Sci Rep 7:43230. [PMC free article: PMC5322365] [PubMed: 28230208]
- Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA, Spain WJ, McKnight GS, Scheuer T and Catterall WA (2006) Reduced sodium current in GABAergic interneurons in a mouse model of severe myoclonic epilepsy in infancy. Nat Neurosci 9:1142–1149. [PubMed: 16921370]
- Zhang Y, Pak C, Han Y, Ahlenius H, Zhang Z, Chanda S, Marro S, Patzke C, Acuna C, Covy J, Xu W, Yang N, Danko T, Chen L, Wernig M and Sudhof TC (2013) Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron 78:785–798. [PMC free article: PMC3751803] [PubMed: 23764284]
- Zhou QG, Nemes AD, Lee D, Ro EJ, Zhang J, Nowacki AS, Dymecki SM, Najm IM and Suh H (2019) Chemogenetic silencing of hippocampal neurons suppresses epileptic neural circuits. J Clin Invest 129:310–323. [PMC free article: PMC6307945] [PubMed: 30507615]
- Zhou T, Benda C, Duzinger S, Huang Y, Li X, Li Y, Guo X, Cao G, Chen S, Hao L, Chan YC, Ng KM, Ho JC, Wieser M, Wu J, Redl H, Tse HF, Grillari J, Grillari-Voglauer R, Pei D and Esteban MA (2011) Generation of induced pluripotent stem cells from urine. J Am Soc Nephrol 22:1221–1228. [PMC free article: PMC3137570] [PubMed: 21636641]
- Zucco AJ, Pozzo VD, Afinogenova A, Hart RP, Devinsky O and D’Arcangelo G (2018) Neural progenitors derived from Tuberous Sclerosis Complex patients exhibit attenuated PI3K/AKT signaling and delayed neuronal differentiation. Mol Cell Neurosci 92:149–163. [PMC free article: PMC6250058] [PubMed: 30144504]
- Abstract
- Introduction
- Generating iPSCs
- Gene Editing of iPSCs
- Methods of 2D Neuronal Differentiation
- Physiological Assays for hPSC-Derived Neurons
- Genetic Epilepsy Modeling Using 2D hPSC Cultures
- Brain Organoid Models
- Modeling Genetic Epilepsies and Their Effects on Cortical Network Function With Brain Organoids
- Challenges and Future Directions
- Conclusions
- References
- Review Sodium Channelopathies in Human and Animal Models of Epilepsy and Neurodevelopmental Disorders.[Jasper's Basic Mechanisms of t...]Review Sodium Channelopathies in Human and Animal Models of Epilepsy and Neurodevelopmental Disorders.Yamakawa K, Meisler MH, Isom LL. Jasper's Basic Mechanisms of the Epilepsies. 2024
- Review Modeling genetic epilepsies in a dish.[Dev Dyn. 2020]Review Modeling genetic epilepsies in a dish.Niu W, Parent JM. Dev Dyn. 2020 Jan; 249(1):56-75. Epub 2019 Jul 11.
- Review Modeling Rett Syndrome with Human Pluripotent Stem Cells: Mechanistic Outcomes and Future Clinical Perspectives.[Int J Mol Sci. 2021]Review Modeling Rett Syndrome with Human Pluripotent Stem Cells: Mechanistic Outcomes and Future Clinical Perspectives.Gomes AR, Fernandes TG, Cabral JMS, Diogo MM. Int J Mol Sci. 2021 Apr 3; 22(7). Epub 2021 Apr 3.
- Review Application of induced pluripotent stem cells in epilepsy.[Mol Cell Neurosci. 2020]Review Application of induced pluripotent stem cells in epilepsy.Hirose S, Tanaka Y, Shibata M, Kimura Y, Ishikawa M, Higurashi N, Yamamoto T, Ichise E, Chiyonobu T, Ishii A. Mol Cell Neurosci. 2020 Oct; 108:103535. Epub 2020 Aug 4.
- A Robust Pipeline for the Multi-Stage Accelerated Differentiation of Functional 3D Cortical Organoids from Human Pluripotent Stem Cells.[Curr Protoc. 2023]A Robust Pipeline for the Multi-Stage Accelerated Differentiation of Functional 3D Cortical Organoids from Human Pluripotent Stem Cells.Whye D, Wood D, Saber WA, Norabuena EM, Makhortova NR, Sahin M, Buttermore ED. Curr Protoc. 2023 Jan; 3(1):e641.
- Functional Exploration of Epilepsy Genes in Patient-Derived Cells - Jasper's Bas...Functional Exploration of Epilepsy Genes in Patient-Derived Cells - Jasper's Basic Mechanisms of the Epilepsies
Your browsing activity is empty.
Activity recording is turned off.
See more...