Epilepsy & Behavior 104 (2020) 106848
Contents lists available at ScienceDirect
Epilepsy & Behavior journal homepage: www.elsevier.com/locate/yebeh
Review
Genetic and molecular basis of epilepsy-related cognitive dysfunction Lin Zhu a,1, Lu Chen a,1, Puying Xu a,1, Di Lu b, Shujuan Dai a, Lianmei Zhong a, Yanbing Han a, Mengqi Zhang c, Bo Xiao c, Lvhua Chang a,⁎,2, Qian Wu a,⁎,2 a b c
Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China Biomedicine Engineering Research Center, Kunming Medical University, 1168 Chun Rong West Road, Kunming, Yunnan 650500, PR China Department of Neurology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha, Hunan 410008, PR China
a r t i c l e
i n f o
Article history: Received 8 October 2019 Revised 6 December 2019 Accepted 6 December 2019 Available online xxxx Keywords: Epilepsy Seizure Cognitive dysfunction Gene Molecule Pathogenesis
a b s t r a c t Epilepsy is a common neurological disease characterized by recurrent seizures. About 70 million people were affected by epilepsy or epileptic seizures. Epilepsy is a complicated complex or symptomatic syndromes induced by structural, functional, and genetic causes. Meanwhile, several comorbidities are accompanied by epileptic seizures. Cognitive dysfunction is a long-standing complication associated with epileptic seizures, which severely impairs quality of life. Although the definitive pathogenic mechanisms underlying epilepsy-related cognitive dysfunction remain unclear, accumulating evidence indicates that multiple risk factors are probably involved in the development and progression of cognitive dysfunction in patients with epilepsy. These factors include the underlying etiology, recurrent seizures or status epilepticus, structural damage that induced secondary epilepsy, genetic variants, and molecular alterations. In this review, we summarize several theories that may explain the genetic and molecular basis of epilepsy-related cognitive dysfunction. © 2019 Elsevier Inc. All rights reserved.
1. Introduction Epilepsy is a common neurological disorder, which is characterized by recurrent seizures and affects approximately 70 million people [1]. Although epilepsy is generally considered to be caused by brain disorders like traumatic brain injury [2], intracranial tumors [3], cerebrovascular disease [4], neurodegenerative disorders [5], and genetic diseases or genetically related epileptic syndromes [6], about 20% of all epilepsy
syndromes are cryptogenic with an unknown etiology, leading to great diagnostic and therapeutic challenges [7]. Moreover, epilepsy is associated with an increased risk for neuropsychological comorbidities such as cognitive deficits, emotional disturbance, and psychiatric disorders, which severely impair quality of life. Clinical evaluation and therapeutic options for these comorbidities depend on the etiology of epilepsy and the age of onset [8]. Although the definitive pathogenic mechanisms underlying epilepsy-related cognitive dysfunction remain unclear,
Abbreviations: AEDs, antiepileptic drugs; GABA, gamma-aminobutyric acid; SAVAs, saporin-conjugated antivesicular GABA transporter antibodies; GABAAR, GABAA receptor; TLE, temporal lobe epilepsy; KCC2, potassium chloride cotransporter 2; CIC-2, chloride channel 2; GABABR, GABAB receptors; GAERS, Genetic Absence Epilepsy Rat from Strasbourg; NMDAR, N-methyl-D-aspartate receptor; LTP, long-term potentiation; GABRB3, γ-aminobutyric acid receptor subunit β-3; mEPSCs, miniature excitatory postsynaptic currents; PSD-95, postsynaptic density 95; APV, 2-amino-5-phosponovaleric acid; CaMKII, calmodulin dependent protein kinase II; PSD, postsynaptic density; HIP-1, Huntingtin interacting protein 1; iGluRs, ionotropic glutamate receptors; mGlu7, metabotropic glutamate receptor 7; PICK1, protein interacting with C kinase-1; A1R, adenosine A1 receptors; GlyR, glycine receptor; Ach, acetylcholine; SE, status epilepticus; ADNFLE, autosomal dominant nocturnal frontal lobe epilepsy; NREM, nonrapid eye movement; SNP, single-nucleotide polymorphism; 5-HT, 5-hydroxytryptamine; MECP2, methyl-CpG binding protein 2; SMEI, severe myoclonic epilepsy of infancy; ATP, adenosine triphosphate; LTD, long-term depression; TCA, tricarboxylic acid; PCDH19, protocadherin 19; TGF-beta1, transforming growth factor beta-1; BDNF, brain-derived neurotrophic factor; ApoE, apolipoprotein E; CDKL5, cyclin-dependent kinase-like 5; EEF1A2, elongation factor 1-α2; KCNH5, potassium channel Kv10.2; CLCN4, chloride channel 4; SUR1, sulfonylurea receptor; TBP, TATA-binding protein; STXBP1, syntaxin binding protein 1; TESC, tescalcin; DDR2, Discoidin domain-containing receptor 2; IQ, intelligence quotient; miRNAs, microRNAs; CREB, cAMP response element-binding protein; MEF2, myocyte enhancer factor-2; TrkB, tyrosine kinase receptor B; NGF, nerve growth factor; NT-3, neurotrophin-3; NT-4, neurotrophin-4; TrkA, tyrosine kinase receptor A; TrkC, tyrosine kinase receptor C; mTOR, mammalian target of rapamycin; TSC1, tuberous sclerosis complex 1; PTEN, phosphatase and tensin homolog on chromosome 10; NF1, neurofibromin 1; mTORC1, mTOR complex 1; PRSS 12, motopsin; SEZ-6, seizure-related gene 6; SNAP-25, synaptosomal-associated protein of 25 kDa; COX-2, cyclooxygenase-2; Nrf-2, nuclear factor erythroid 2-related factor 2; VGKC, voltage-gated potassium channel; BACE-1, β-site amyloid precursor protein cleaving enzyme 1; TLR-4, toll-like receptors 4; DBN, drebrin; PPARγ, peroxisome proliferator-activated receptor gamma; LIS-1, lissencephaly; IL-6, cytokine interleukin-6; ZnTs, zinc transporters; CL, cardiolipin; β2-Gpl, β2-glycoprotein; AR, α1-adrenergic receptors; Cdk5, cyclin-dependent kinase 5. ⁎ Corresponding authors at: Department of Neurology, First Affiliated Hospital, Kunming Medical University, 295 Xi Chang Road, Kunming, Yunnan 650032, PR China. E-mail addresses:
[email protected] (L. Chang),
[email protected] (Q. Wu). 1 First authors Lin Zhu, Lu Chen, and Puying Xu contributed equally to the work. 2 Lvhua Chang and Qian Wu contributed equally to the work.
https://doi.org/10.1016/j.yebeh.2019.106848 1525-5050/© 2019 Elsevier Inc. All rights reserved.
2
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
epilepsy is closely associated with cognitive alteration. Most causes of epilepsy that result in brain damage, such as, traumatic injury, structural change (focal cortical dysplasia, brain atrophy, etc.), genetic variants, molecular alterations, and some autoimmune and inflammatory encephalitis, can lead to cognitive comorbidities in epilepsy. Meanwhile, epilepsy treatments (such as antiepileptic drugs and surgery) are associated with epilepsy-related cognition and behavioral disorders. Especially, in utero exposure to some antiepileptic drugs (AEDs), such as valproate, carbamazepine, lamotrigine, and phenytoin may affect cognitive functions, such as, learning and memory in children. However, the doses and types of AEDs used in different periods of pregnancy require further study [9,10]. Moreover, recurrent seizures, chronic seizures, uncontrolled seizures, and even epileptic encephalopathies were considered intimately related to progressive cognitive decline, mental problems, and behavioral deficiencies [11,12], but these issues are still controversial and need more evidence. In summary, multiple factors result in epilepsy-related cognition comorbidities. In this review, we focused on the mainstream theories that may explain the genetic and molecular basis of epilepsy-related cognitive dysfunction and may provide new insight into the clinical treatment of this disease (Fig. 1; Table 1). 2. Neurotransmitter systems and their contributions to epilepsy and cognition 2.1. GABAergic system Gamma-aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system, which binds to specific transmembrane receptors in the plasma membrane of both pre- and postsynaptic neuronal processes; GABAergic interneurons contribute to epileptic seizures and neurobehavioral abnormalities by modulating hippocampal circuit function [13]. Saporin-conjugated antivesicular GABA transporter antibodies (SAVAs), which selectively disrupt the GABAergic pathway but have no impact on glutamatergic synapses, can reduce GABA release and thus trigger network hyperexcitability. This pathophysiological process may be caused by the progressive loss
of pyramidal neurons and the granule cell dispersion in the CA1 region [13]. Several molecules participate in the pathogenesis of epilepsy and epilepsy-related cognitive impairment via the GABAergic system. In mice, offspring whose mothers were exposed to A2AR antagonists during pregnancy and prior to weaning show abnormal migration of GABAergic neurons in the hippocampus, causing enhanced neuronal network excitability and seizure susceptibility as well as the loss of GABA neurons in the hippocampus and eventually leading to cognitive dysfunctions [14]. Point mutations in the gene encoding the GABAA receptor (GABAAR) [15] are associated with many types of epilepsy including catamenial epilepsy [16,17], absence epilepsy [18–20], and temporal lobe epilepsy (TLE) [21,22]. These mutations and alterations in tonic GABAAR-mediated conductance may disrupt neuronal network function [23–25]. Therefore, therapies targeting extrasynaptic GABAARs may be potentially effective to enhance recovery of cognitive function [23]. In addition, GABAARs regulate the transmembrane distribution of chloride, and GABAergic cells mediate neuronal inhibition by promoting chloride inflow via synaptic and extrasynaptic conductance in the hippocampus [26]. Both the potassium chloride cotransporter 2 (KCC2) [27–29] and voltage-gated chloride channel 2 (CIC-2) [30,31] are associated with epilepsy, including TLE and absence seizures. The ClC-2 levels in the pyramidal cells are upregulated in CA1 in pilocarpine-treated rat models, and the increase in ClC-2 currents can be reversed by L655708, a specific antagonist targeting α5 subunitcontaining GABAARs [26]. Therefore, some have speculated that CIC-2 is involved in GABAAR-mediated neuronal inhibition and thereby plays an important role in epilepsy and cognitive dysfunctions [26]. Seizures in absence epilepsy are associated with both GABAAR and GABAB receptors (GABABR), but cognitive impairment is mainly attributed to the dysfunction of GABABR. Using the Genetic Absence Epilepsy Rat from Strasbourg (GAERS), researchers have observed that seizures are enhanced by GABAAR and GABABR agonists and blocked by GABABR antagonists. Some scholars have also noted a similar phenomenon in the AY-9944 models and transgenic GABABR models of atypical absence seizures of Lennox–Gastaut syndrome [32]. Moreover, the administration of GABABR antagonists improves cognitive performance
Fig. 1. Genes and molecules in epilepsy and cognition. The Red parts indicate that those genes or molecules might have negative effect on epilepsy-related cognitive outcomes; The Blue parts indicate that those genes or molecules might have positive effect on epilepsy-related cognitive outcomes. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
3
Table 1 Molecules and genes in epilepsy-related cognitive impairment. Molecules/genes GABAAR GABABR GABAergic GABRβ-3
HIP1 Glutamatergic
mGlu7R GlyR α3 CaMKII
ACh
M1-mAChR M2 mAChR M4 mAChR nAChR
Histaminergic
H1, H2, H3, H4
Free radicals Peroxynitrite
Possible mechanism
Epilepsy/epilepsy syndromes
Point mutations, neuronal network disruption Regulates KCC2, Chloride and ClC-2 Neocortical hyperexcitability, Inhibition of NMDAR function, Impair hippocampal function Thalamocortical circuits, Heterozygous mutations, Homozygous mutations, Point mutations
Somatosensory disturbances, learning Catamenial epilepsy, absence epilepsy, TLE functions impairment, cognitive deficits Lennox–Gastaut syndrome, absence Learning and memory impairment epilepsy, atypical absence seizures Absence epilepsy, Rett syndrome, Sensory processing damage, Angelman syndrome somatosensory deficiency, impaired cognition, behavioral disorders Cognitive dysfunction, behavioral Focal and generalized epilepsies disorders Absence seizures Cognitive and emotional alteration
Defects in presynaptic function, as well as alterations in AMPA and NMDA receptor function Interact with PICK1 and reduce glutamate release of mGlu7 Exhibits an excitatory function via facilitating neurotransmitter release Thr286 phosphorylation Postsynaptic density Inhibits neuronal excitability Regulates GABAergic system Depression of excitatory postsynaptic potential (EPSP) via M4 Specific mutations (CHRNA4, CHRNB2 and CHRNA2) Modulating the homeostasis of neurotransmitter systems, such as AKT/GSK-3β, JNK and Ras-Raf-MEK Mitochondrial damage and activation of glutamate and NMDA receptors Inhibits LTP and induces GABA release Loss-of-function mutations Reduce excitatory synaptic transmission
MECP2
Cognitive outcome
TLE
Cognitive dysfunction and anxiety
TLE, Angelman syndrome, hyperthermia induced seizures TLE TLE TLE
Spatial learning and memory impairment Long-term cognitive deficits Cognitive dysfunction (animal) Cognitive dysfunction (animal) Cognitive dysfunction (animal)
ADNFLE
Executive deficits, visuospatial attention deficits, verbal memory impairment
TLE
Cognitive protection (possible)
Rett syndrome, secondary epilepsy, TLE, etc. TLE Rett syndrome, partial seizures, generalized seizures, Lennox–Gastaut syndrome, myoclonic status
Cognitive impairment Cognitive impairment Cognitive deficits Cognitive deficits Spatial memory impairment Memory dysfunction, cognitive impairment Intellectual disability Behavioral abnormalities Cognitive impairment
Truncation or exonic deletion mutations Impair sodium channel protein Nav1.1
Dravet syndrome
ATP
Modulate LTP and LTD
TLE neonatal seizures
PCDH19
Missense substitutions, premature termination (frameshift, nonsense, and splice-site mutations), in-frame deletion
Focal and generalized seizure, infantile seizures, early febrile seizures
Targets neuronal apoptosis
TLE
Cognitive, memory and behavioral protection
BDNF
Dendrite growth inhibition Neuronal Activity Neuronal and synaptic growth and differentiation
TLE, seizure disorders, Rett syndrome
Fear learning, memory and spatial cognition impairment
mTOR
Regulates TSC1, TSC2, PTEN, NF1 BDNF, etc.
Focal cortical dysplasia, infantile spasms, TLE
Cognitive, memory and behavioral protection
SCN
Bcl Family
SCN1A, SCN2B
Bcl-2, Bcl-xl, Bcl-w, Puma, Mcl-1, Bax, Bid
TLE, temporal lobe epilepsy; ADNFLE, autosomal dominant nocturnal frontal lobe epilepsy; LTP, long-term potentiation; LTD, long-term depression.
[32–34], whereas GABABR agonists impair learning and memory [35–38]. In the WAG/Rij rats, another model of absence seizure, a decrease in presynaptic GABABR function in the neocortex has been demonstrated to contribute to neocortical hyperexcitability [39]. In the lethargic models, GABABR antagonists suppress absence seizures by regulating GABABR-mediated inhibition on N-methyl-D-aspartate receptor (NMDAR) function, while GABABR agonists exhibit opposite effects [40]. In the AY-9944 models, cognitive dysfunction secondary to atypical absence seizures is GABABR-dependent. Additionally, hippocampal long-term potentiation (LTP) is reduced, leading to the impairment of hippocampal function, which is independent of seizure activities [41]. Han et al. have composed a comprehensive review concerning the correlation between GABABR function and absence epilepsy [42]. Furthermore, γ-aminobutyric acid receptor subunit β-3 (GABRB3) disruptions may be involved in the pathogenesis of epilepsy and influence cognitive outcomes. As GABRB3 is expressed in the reticular thalamic nucleus and cortex, it plays an important role in thalamocortical circuits that are essential for sensory processing. Heterozygous mutations of GABRB3 in mice have been found to cause somatosensory deficiencies [43–46] and increased epileptiform discharges [47]. Conversely, homozygous mutations are associated with absence seizures, cognitive dysfunction, disrupted coordination, and sensorimotor
deficits [47]. In human childhood absence epilepsy, point mutations in exon 1a and the N-terminal exon 2 of GABRB3 have been found to promote hyperglycosylation and reduced GABA currents. Moreover, reduced expression of GABRB3 has been observed in neurodevelopmental diseases, such as Rett syndrome, Angelman syndrome, and autism spectrum disorders [48,49]. Tanaka et al. have thoroughly discussed the relationship of GABRB3 with epilepsy and epilepsy-related cognitive outcomes [47]. 2.2. Glutamatergic system Several molecules participate in synaptic glutamatergic neurotransmission and their role in epilepsy. In chronic models induced by bicuculline, a competitive antagonist targeting the GABAAR, wholecell electrophysiological recordings on pyramidal neurons have demonstrated a reduction in the frequency and amplitude of miniature excitatory postsynaptic currents (mEPSCs), which is mediated by AMPA (2-amino-3-3-hydroxy-5-methyl-isoxazole-4-yl propionic acid) and NMDARs [50]. Additionally, expression levels of glutamatergic synaptic markers, such as, postsynaptic density 95 (PSD-95), are significantly decreased, indicating that seizures may induce inhibition of the growth of dendritic arbors that results in a reduced number of glutamatergic synapses on dendrites [50,51]. In the bicuculline model, administration of
4
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
2-amino-5-phosponovaleric acid (APV; an NMDAR antagonist) can abolish seizure-induced dendritic growth suppression as well as the alteration in the expression of the glutamatergic postsynaptic proteins, suggesting that seizure-induced dendritic growth suppression in hippocampal CA1 may be NMDAR-dependent. Through NMDARs, calcium signaling affects several physiological activities, including the transcription of genes related to dendritic growth and formation of dendrites [52]. Calmodulin dependent protein kinase II (CaMKII), a Ser/Thr protein kinase containing four isoforms (α, β, γ, δ), is essential for the formation of synapses, synthesis, the secretion of neurotransmitters, the function of receptors and ion channels, synaptic plasticity, and cognition [52–56]; CaMKII is associated with various types of epilepsy. Dong and Rosenberg found that the pentylenetetrazol-induced brief seizure can result in a remarkable decline in CaMKII Thr286 phosphorylation [57]. Ni et al. observed a decrease in CaMKII mRNA levels in penicillin-induced developmental epilepticus, which may explain long-term cognitive deficits in this model [58]. Moreover, changes in the expression level of total CaMKIIα and p-T286-CaMKIIα in the postsynaptic density (PSD) and translocation of total CaMKIIα from the PSD to the cytosol were noted, which may be potential mechanisms underlying spatial learning and memory impairment in hyperthermia-induced seizures [59]. Deletion mutations in the gene encoding Huntingtin interacting protein 1 (HIP-1) are associated with clinical epilepsy, including cognitive and neurobehavioral disorders [60]. Behavioral problems linked to deletion in the HIP-1 gene include inattention, hyperactivity, impulsivity, aggression, autism, depression, and self-abuse. Animal experiments have proven that both homozygous and heterozygous deletions in the HIP-1 gene may cause epilepsy. Additionally, depletion of HIP-1 can result in presynaptic dysfunction and functional abnormalities in the ratio of AMPA and NMDAR function [61,62]; HIP-1 colocalizes with NMDA ionotropic glutamate receptors (iGluRs) in hippocampal and cortical neurons. Relevant studies have found that a loss of HIP-1 can decrease NMDA-induced AMPA-type iGluR clathrin-mediated internalization [61,63]. Additionally, HIP1 insufficiency disrupts the maintenance of surface AMPA receptors that are required for synaptic potentiation [64]. In summary, because of the role of HIP1 in AMPA and NMDA iGluR functions, depletion of HIP1 can predispose the brain to epilepsy and relative cognitive disturbances. Metabotropic glutamate receptor 7 (mGlu7) receptors are members of the group-III metabotropic glutamate receptors [65,66]. Sansig et al. have found that reduction in mGlu7 receptors increases the risk of convulsive seizures [67]. Moreover, the interaction between mGlu7 receptors and protein interacting with C kinase-1 (PICK1) can cause absence seizures [68,69]. The mGlu7 receptors are located in the presynaptic area and reduce glutamate release of mGlu7 by acting with GABABR, adenosine A1 receptors (A1R), and N-type voltage-sensitive calcium channels in cortical synaptosomes [70]. Based on this evidence, mGlu7 receptors are considered an important target for cognitive and emotional functions. Changes in the glycine receptor (GlyR) α3 subunit are associated with the pathophysiology of epilepsy [71–73]. In patients with TLE, a P185L amino acid transition can elevate the expression of GlyR α3, resulting in the hyperactivity of neurotransmitter receptors. Activation of the presynaptic GlyR α3 L185L transition can promote the release of neurotransmitters, enhancing neuronal functions in the cortical network. Thus, GlyR variations can disrupt homeostatic maintenance of neural excitability and trigger neuropsychiatric disorders, such as, cognitive decline and anxiety [74]. 2.3. Acetylcholinergic system Acetylcholine (ACh) is a neurotransmitter involved both in cognitive processes [75] and in seizure generation [76–78]. A study on rat models of status epilepticus (SE) showed that a single and sustained generalized seizure can increase responsiveness to cholinergic stimuli [79,80].
The M1 mAChR, a metabotropic ACh receptor, colocalizes with NR1, a subunit of the NMDAR [81], and plays an important role in the development of epilepsy. Pilocarpine cannot trigger seizures in mouse models lacking M1 mAChR [82]. Additionally, the M1 mAChR is associated with the Kv7 channel-mediated M-current that inhibits neuronal excitability [83]. The M2 mAChR also participates in the development of epilepsy through a decrease in the release of GABA [84]. Under normal physiological conditions, ACh transiently inhibits GABA release and activates the M2 mAChR, facilitating the expression of LTP [85]. However, in patients with TLE, GABAergic transmission affects mAChR-mediated regulation of LTP and impairs the M2 mAChR-mediated regulation of the GABAergic system [84]. Suppression of cholinergic signaling also causes cognitive impairment in mice [86] as well as schizophrenia and Alzheimer's disease in humans [87,88]. Some scholars have proposed that mAChRs can regulate cognitive function by increasing neuronal activity via the M1 mAChR, suppressing the excitatory postsynaptic potential via M4 mAChR, and suppressing the generation of inhibitory postsynaptic potentials via M2 mAChR [84,89]. These findings highlight the complex role of muscarinic receptors in the pathogenesis of epilepsy and relevant cognitive dysfunction [84]. Alteration in one neurotransmitter system may affect the activity of other systems through synergistic actions or cross-activation. Both NMDARs and endogenous ACh participate in modulating interictal electrical discharges in vitro. Mikroulis and Psarropoulou found that mAChR positively contributes to the frequency of NMDAR-dependent epileptiform discharges, which is associated with an increased level of endogenous ACh. Additionally, the mAChRmediated increase in the frequency of interictal electrical discharges is more remarkable following early-life generalized sustained convulsion [75]. The hyperactivity of the cholinergic system in the hippocampus may impair cognitive function and may also lower the seizure threshold in adults [75]. Autosomal dominant nocturnal frontal lobe epilepsy (ADNFLE) is an inherited epilepsy syndrome. Several mutations in nicotinic receptor subunit genes have been identified [90,91]. These nucleotide variants can cause a special form of epilepsy manifesting as nocturnal seizures that occur mostly during nonrapid eye movement (NREM) sleep [92]. This inherited disease was first reported by Scheffer et al. in a family of 27 affected individuals in which there were no neuropsychiatric or cognitive symptoms [93]. However, recent studies have demonstrated that cognitive disorders, especially deficits in executive functions, are common in patients with ADNFLE [94]. Currently, the relationship between ADNFLE and cognitive impairment remains unknown. Interestingly, several mutations in the AchRs subunit genes CHRNA4, CHRNB2, and CHRNA2 have been reported in families with ADNFLE [95]. Moreover, Bertrand et al. discovered a mutation in the β2 nAChR in a pair of monozygotic twins who exhibited verbal memory disabilities [96]. Subsequently, Picard et al. identified three mutations in the CHRNA4 gene and one mutation in the CHRNB2 gene in 11 patients who presented with memory decline and executive dysfunction [94]. The CHRNA4 and CHRNB2 genes encode nAchR subunits, and an increasing number of studies showed that genetic variants of these subunits may be associated with different cognitive dysfunctions. For example, the single-nucleotide polymorphism (SNP) C1545T in the CHRNA4 gene is associated with visuospatial attention deficits [97,98], and mutations in the α4 subunit, including S252L, T265I, and S248F, may affect core executive functions [94,99]. Mutations in β2 nAChR may affect general intellectual skills and verbal functions [96], and the missense mutation I312M in the CHRNB2 gene is associated with cognitive impairments, especially verbal memory deficiencies [96,100]. Steinlein et al. noted that the nAChR mutation-related risk for cognitive disorders was lower than the ADNFLE mutation-related risk [92]. In summary, genetic variants in nicotinic receptors may lead to alterations in the cholinergic transmission system, which is associated with psychiatric and behavioral
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
symptoms [101–105], and the relationship between ADNFLE mutations and neuropsychiatric impairments deserves more attention [92]. 2.4. Histaminergic system The central histaminergic system is controlled by H1, H2, H3, and H4 receptors and is also involved in the pathogenesis of epilepsy. The histaminergic system modulates the homeostasis of neurotransmitter systems via H3 heteroreceptors, which regulate the secretion of neurotransmitters such as GABA, glutamate, dopamine, 5hydroxytryptamine (5-HT), noradrenaline, and acetylcholine [106]. Therefore, the histaminergic system is essential for homeostatic maintenance and cognitive function [104,106]. Some scholars have found that the overproduction of histamine can inhibit seizures, indicating that histamine has neuroprotective effects [107–109]. Additionally, histamine can protect hippocampal neurons from kainate-induced injury through H1 and H3 receptors [110]. Further studies have suggested that this protective effect may be mediated by AKT/GSK-3β [111], JNK, and Ras-RafMEK [109,112] signaling pathways and heat shock proteins [113]. Bhowmik et al. have performed a comprehensive review describing the relationship between histamine H3 receptor and epilepsy as well as epilepsy-related cognitive dysfunctions [108]. 3. Genetic epilepsy syndromes and cognition 3.1. Rett syndrome Rett syndrome is to a neurodevelopmental disease characterized by progressive motor deficits, cognitive dysfunction, social-behavioral problems, and seizures [114]. Approximately 70%~80% of all patients with Rett syndrome exhibit seizures at 2~5 years of age [115–118]. Epilepsy forms in the Rett syndrome include partial seizures, generalized seizures [117–119], Lennox–Gastaut syndrome, and myoclonic status [120]. Accumulating evidence shows that loss-of-function mutations in the gene encoding X-linked methyl-CpG binding protein 2 (MECP2) can cause Rett syndrome [121] in humans [122] and animal models [123–126]; MECP2 dysfunction results in a reduction of excitatory synaptic transmission in the hippocampus [125,127], impairment of LTP at excitatory synapses in the hippocampus [128–130], alterations in inhibitory transmission in the whole brain [131–133], and hyperexcitability in cortical neurons and networks [134–136]. These changes may be one plausible explanation for seizures and cognitive disabilities. Recent studies have found that knockout of MECP2 in excitatory neurons inhibits GABAergic transmission in pyramidal neurons in the somatosensory cortex [114]. Moreover, loss of MECP2 in excitatory neurons may reduce the number of GABAergic synapses and increase the excitability of pyramidal neurons [114]. Considering that MECP2 is important for maintaining GABAergic transmission and cortical circuits, we speculate that MECP2 dysfunction is a crucial contributor to cognitive deficits in patients with Rett syndrome. 3.2. Dravet syndrome Dravet syndrome, previously known as severe myoclonic epilepsy of infancy (SMEI) [137], is a rare form of epilepsy that manifests as seizures, developmental retardation, and cognitive dysfunction [138,139]. Dravet syndrome is caused by truncation or exonic deletion mutations in the SCN1A [140], SCN2A [141], and GABRG2 genes [142]. The exact mechanisms underlying epilepsy and cognitive disorders caused by GABRG2 mutations remain unclear; nevertheless, truncation of GABRG2 and its subunits may lead to the formation of protein aggregates, which is a pathological hallmark of neurodegenerative diseases [138]. The SCN1A gene variants include missense mutations, truncating mutations that generate a truncated protein [143], and genomic deletions and duplications [144]. The SCN1A mutations have been found
5
to be associated with both seizures and cognitive impairment in Dravet syndrome [145]. Some scholars have identified SCN1A mutations that impair the function of the sodium channel protein Nav1.1, leading to a decrease in sodium currents [146], and they may cause an outburst of action potentials in inhibitory interneurons [147]. In addition, SCN1A mutations can disrupt the overall excitation–inhibition balance and thus cause abnormal activation of hippocampal circuits, which lower the threshold for seizures [148]. N6-cyclopentyladenosine, an A1R agonist, can reduce presynaptic depolarization through the activation of delayed rectifying potassium channels and blocking voltage-gated calcium channels [149] producing the inhibition of aberrant neural circuit hyperexcitability in experimental Dravet syndrome models and reducing febrile seizure-like events without blocking excitatory synaptic transmission [148]. These results indicate that A1R may be involved in hippocampal circuits in Dravet syndrome by reducing excitability through a mechanism that differs from traditional agents that boost GABA [150]. In short, presynaptic or postsynaptic hyperexcitation and abnormal regulation of hippocampal circuits may be the mechanisms underlying early-life febrile seizures and cognitive impairment in Dravet syndrome [148]. Although cognitive disorders are common in pediatric patients with Dravet syndrome, antiepileptic therapies do not improve cognitive function [151]. Therefore, brain abnormalities in Dravet syndrome may contribute to cognitive dysfunction through mechanisms independent of epilepsy [151]. Some have hypothesized that impaired function of Nav1.1 may be a potential mechanism, which is also associated with autism [152,153] and Alzheimer's disease [154–157]. Nav1.1 dysfunction disrupts neural network function [158]. The selective knockdown of Nav1.1 in the Broca area results in spatial memory impairment through the dysregulation of hippocampal theta frequency [151]. 4. Oxidative stress 4.1. Free radicals Epileptic seizures and postictal periods are characterized by increased energy turnover in the brain and massive energy consumption [159]. These alterations lead to oxidative stress and the overproduction of free radicals, such as, hydroxyl and nitroxyl [160]. The generation of free radicals is mediated by two mechanisms: mitochondrial damage [160,161] and the activation of glutamate and NMDARs [162]. Mitochondrial damage is associated with several genetic forms of epilepsy [163,164]. Free radicals may aggravate epilepsy and cognitive dysfunction by directly influencing LTP [165,166], and the free-radical levels are correlated with epilepsy severity [167]. Additionally, pretreatment with melatonin (N-acetyl-5-methoxytrptamine), an indoleamine derivative of serotonin with potent antioxidant and free-radical scavenger properties [168], can reduce the level of hydroxyl and prevent cognitive impairment in rat models [159,169]. Another free-radical scavenger, Trolox, exhibits neuroprotective effects against epilepsy and cognitive dysfunction in a mouse model of Rett syndrome [170]. However, although there is evidence linking free radicals to epilepsy-related cognitive impairment, the exact mechanism underlying this association remains to be fully elucidated. 4.2. Peroxynitrite Peroxynitrite is a strong oxidant and cytotoxic mediator produced from the reaction of nitric oxide and the superoxide radical, which is the major contributor to seizures and hypoxic–ischemic brain damage [171–173]. Peroxynitrite can cause cognitive deficits by inhibiting LTP and inducing GABA release in the hippocampus [174]. Thus, we speculate that peroxynitrite may contribute to epileptic seizures and epilepsy-related cognitive dysfunctions through GABA signaling [175].
6
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
4.3. Adenosine triphosphate (ATP) Abnormal adenosine triphosphate (ATP) levels can lead to seizures and epilepsy-related cognitive impairment through energy failure and impairing LTP and long-term depression (LTD). The ATP levels reflect cell energy levels and are regulated by the tricarboxylic acid (TCA) cycle. Deficits of the TCA cycle can lead to seizures, and its supplementation can reverse seizures and seizure-induced cell death [176]. Both LTP and LTD are regulated by ATP and its dephosphorylated product adenosine [177]. In rat models, ATP hydrolysis is enhanced by kainate during the neonatal period, which is important for LTP induction. Cognato et al. hypothesized that enhanced ATP hydrolysis regulates cognitive impairment in adult rats that experienced a convulsive seizure during the postnatal period [177]. The correlation between purinergic metabolism and cognitive dysfunction following neonatal seizures may improve our understanding of seizure-induced memory deficits [177].
In patients with epilepsy, Bcl-2 levels are elevated in serum and brain tissues [192,206], and its high level is as a hallmark of activated apoptosis signaling in pediatric TLE [206]. Additionally, the serum Bcl2 levels are correlated with the clinical characteristics of epilepsy, such as, the disease duration, seizure frequency, intellectual ability, and disease severity [206]. Clusterin, a protein interacting with Bcl-xL, has proapoptotic effects and causes hippocampal neuronal death in CA3 in patients who experienced seizures [207]. Transforming growth factor beta-1 (TGF-beta1) has neuroprotective properties, which can protect neurons against seizure-induced damage and improve cognitive function [208]. In lithium-pilocarpine-induced status epilepticus rats, exogenous TGF-beta1 administration significantly attenuates the detrimental effects of hippocampal injury, which is mediated by the upregulation of Bcl-2 and downregulated Bax and caspase-3 [208]. Therapeutic strategies targeting apoptosis hold promise for treating seizureinduced brain damage and cognitive disorders. 6. Other molecules
5. Genes and epigenetics 5.1. Protocadherin Protocadherin 19 (PCDH19) is a major epileptogenic gene, and its mutations are associated with cognitive impairment induced by epilepsy [178,179]; PCDH19 may participate in the pathogenesis of focal and generalized seizures and infantile or early-onset febrile seizures [169,180]. Patients with PCDH19 mutations usually exhibit intellectual disabilities, and more than half of them present behavioral abnormalities [181] and cognitive impairment [169]. Clinical variations in the PCDH19 gene include missense substitutions, truncating mutations, and fragment deletions [169,180]. Until recently, the specific relationship between genotypes and phenotypes is still unclear [169]. Camacho et al. found that PCDH19 mutations have a female predominance, and PCDH19-related seizures tend to diminish or even disappear in adolescence. The authors also noted that mental disability and psychiatric symptoms are more severe in these patients [181]. Furthermore, Marini et al. demonstrated that patients carrying PCDH19 mutations usually have focal seizures accompanied by affective symptoms, indicating that the frontotemporal limbic system may be involved in PCDH19related epilepsy [169]. 5.2. Bcl gene family Neuronal loss may disrupt the excitation–inhibition balance within brain networks [182,183], and it can lead to epilepsy by affecting the minimal latency [184,185], minimal delayed spontaneous seizures, [186] or by disrupting the thalamocortical circuits [187]. Neuroprotective treatment targeting neuronal apoptosis can reduce epilepsy risk [188], reduce the number of spontaneous seizures [189], and improve memory and behavioral abnormalities [190]. Seizure-induced neuronal death is characterized by excitotoxic necrosis and the activation of apoptosis signal pathways. Bcl family proteins, such as Bcl-2, Bcl-xL, Puma, Bcl-w, and Mcl-1, are crucial molecules regulating the initiation, integration, and execution of the intrinsic apoptosis pathway [191–194]. Intrinsic apoptosis is initiated by DNA damage, hypoxia, calcium overload, growth factor withdrawal, oxidative stress, and misfolded proteins [195,196]. Seizures can downregulate the expression of antiapoptotic genes, such as, Bcl-2 [197], Bcl-w [198,199], and Mcl-1 [200], and upregulate the expression of proapoptotic genes, such as, Bax [201–203], Bid [137], Bad [202], Bim [204], Puma [137], and Bmf [203]. Relevant evidence showed that downregulation of proapoptotic genes, such as, Puma, Bak, and Bmf, can protect against seizure-induced neuronal death. Conversely, downregulated of antiapoptotic Mcl-1 and Bcl-w leads to neuronal injury in the hippocampus [205].
6.1. BDNF Brain-derived neurotrophic factor (BDNF), a 14-kDa secreted protein, is involved in neuronal activity, neuronal and synaptic growth and differentiation, and the growth of pyramidal cell dendrites [209,210]. The expression of BDNF is altered in epileptic animal models and in patients with TLE [211]. Vezzani et al. found that seizures can elevate the expression of BDNF mRNA and protein [212]. Additional evidence indicates that loss of BDNF can suppress hyperexcitability [213], and that BDNF administration within the hippocampus induces the epileptogenesis [214]. Furthermore, aberrant epigenetic regulation of BDNF plays an important role in the pathophysiology of seizureinduced cognitive deficiencies, Rett syndrome, Alzheimer's disease, and schizophrenia [209]. Transcription of BDNF is modulated by various transcription factors, such as, MECP2, myocyte enhancer factor-2 (MEF2), and cAMP response element-binding protein (CREB) [215], which may be involved in seizure-induced dendrite growth inhibition [216]. Brain-derived neurotrophic factor participates in the dynamic transcriptional mechanisms involved in cognition in the adult brain. In addition, BDNF can promote tyrosine phosphorylation in the cytoplasm by binding to tyrosine kinase receptor B (TrkB), which is a vital process for the activation of intracellular signaling cascades [217]. Brain-derived neurotrophic factor has three analogs, nerve growth factor (NGF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4), while TrkB has two analogs, tyrosine kinase receptor A (TrkA) and tyrosine kinase receptor C (TrkC). NGF binds to TrkA, BDNF and NT4 to TrkB, and NT3 to TrkC. In animal models and humans with epilepsy, seizures induce a striking increase in BDNF expression [218–221] and enhance the activation of TrkB in dentate granule cells and mossy fiber pathways in the hippocampus [222]. Additionally, BDNF enhances LTP [223] and synaptic plasticity in epileptic animal models [224]. Similarly, activation of TrkB in mossy fiber pathways can enhance LTP in the hippocampus in the kainic acid-induced epilepsy models [225]. Furthermore, activation of TrkB mediated by BDNF can compromise GABA-mediated inhibition [226]. Increasing the level of BDNF or TrkB via exogenous injection or transgenic techniques exacerbates seizure susceptibility and severity [227]. Conversely, the conditional knockout of TrkB prevents epilepsy progression, and the deletion of TrkB also reduces the risk for epilepsy in animal models [217]. Noteworthy, estrogen can elevate BDNF levels; hippocampal neurons and estrogen administration in female rats produce epileptiform responses of CA3 pyramidal cells, which are reversed by a Trk antagonist [228]. These findings strongly suggest that BDNF and Trk signaling may contribute to epileptogenesis [217,229]. Several studies have reported an association between BDNF and fear learning or spatial cognition [230–232]. The fear stimulus may elevate BDNF transcript including exons I and IV in the amygdala [233],
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
hippocampus, and prefrontal lobe cortex [230]. This process is regulated by chromatin modification, which is characterized by increased histone acetylation (prominently histone H4) and phosphorylation at the respective promoters [234]. Upregulated histone H4 acetylation at BDNF gene promoters is observed in the hippocampus in rat models of status epilepticus [235]. Tian et al. also noted that the BDNF transcription during long-term memory formation is mediated by NMDA glutamate receptor activation [236]. Despite this cumulative evidence, the role of BDNF in the development of epilepsy and related cognitive impairment requires further investigation. 6.2. Mammalian target of rapamycin (mTOR) In the brain, mammalian target of rapamycin (mTOR) is a kinase modulated by glutamate and dopamine receptors [237], and it is negatively regulated by tumor suppressor genes tuberous sclerosis complex 1 (TSC1), TSC2, phosphatase and tensin homolog on chromosome 10 (PTEN), and neurofibromin 1 (NF1) [20]. Dysregulation of mTOR may lead to “TORopathies” [238], a disease spectrum including focal cortical dysplasia, hemimegalencephaly, tuberous sclerosis, infantile spasms, and TLE [239]. In the infantile spasm model, rapamycin treatment effectively inhibits the overactivation of the mTOR signaling in the cortex, which suppresses spasms and partially improves cognitive deficits [239]. In epilepsy models, rapamycin ameliorates the epilepsy-related pathology and reduced the frequency of spontaneous seizures [240]. Further investigations showed that the protective effects of mTOR inhibitors are time- and dose-dependent [241]. In rats with acute hypoxia-induced neonatal seizures, seizures would activate mTOR complex 1 (mTORC1) and its downstream targets, such as, phospho-4E-BP1 (Thr37/46), phospho-p70S6K (Thr389), and phospho-S6 (Ser235/236). Upstream signaling molecules, including BDNF, phospho-Akt (Thr308), and phospho-ERK (Thr202/Tyr204), may also be activated [242]. Additionally, rapamycin, the specific mTORC1 inhibitor, can reverse the increased glutamatergic neurotransmission and seizure susceptibility, and it can attenuate epileptic seizures and neuropsychic disturbances [242]. Therefore, the mTORC1 signaling pathway may be a promising target for the treatment of seizures and related cognitive disorders [242]. The PTEN and mTOR coregulate the process of epileptogenesis and cognitive functions; PTEN negatively regulates the activity of the PI3K/ Akt/mTOR pathway, a signaling cascade critical to cell proliferation Table 2 Other genes and molecules possibly related to epilepsy and cognition with unclear mechanisms. Type
Other genes
Other molecules
SNP variants MicroRNAs
Identified molecules Histone demethylase KDM5C (JARID1C/SMCX) [244]; Cyclin-dependent kinase-like 5 (CDKL5); Elongation factor 1-α2 (EEF1A2); Voltage-gated potassium channel Kv10.2 (KCNH5); Chloride channel 4 (CLCN4); ARHGEF15 (an EC-specific guanine nucleotide exchange factor) [245]; Sulfonylurea receptor (SUR1) (Hernandez-Sanchez et al., 2001); TATA-binding protein (TBP) [246]; Syntaxin binding protein 1 (STXBP1) [247] Apolipoprotein E (ApoE) [248]; Motopsin (PRSS 12) [249]; seizure-related gene 6 (SEZ-6) [249]; Synaptosomal-associated protein of 25 kDa (SNAP-25) [250]; Cyclooxygenase-2 (COX-2) [251]; Nuclear factor erythroid 2-related factor 2 (Nrf-2) [252]; Voltage-gated potassium channel (VGKC) [253]; β-site amyloid precursor protein cleaving enzyme 1 (BACE-1) [254]; Toll-like receptors 4 (TLR-4) [255]; Drebrin (DBN) [256]; Kainite [257]; Peroxisome proliferator-activated receptor gamma (PPARγ) [258]; Lissencephaly (LIS-1) [259]; Cytokine interleukin-6 (IL-6) [260]; Autophagy and zinc transporters (ZnTs) [261]; Cardiolipin (CL) and β2-glycoprotein (β2-Gpl) antibodies [262]; α1-adrenergic receptors (AR) subtypes [263]; Cyclin-dependent kinase 5 (Cdk5); SnoNs (an embryonically regulated transcription factor) rs7294919 [264]; rs10784502 [265]; rs10494373 [266] miR-34a, miR-132, miR-184, miR-21, miR-29a, miR-34a, miR-125b, and miR-497 [267–270]
7
and cell growth. Also, mutations in the PTEN increase the risk for epilepsy and autism [243]. Consistent with this idea, loss of PTEN leads to seizures and cognitive dysfunctions in mice [243]. These findings indicate that PTEN is an essential molecule for neuronal development and excitability in the cerebral cortex [243]. 6.3. More genes, molecules, microRNA, and SNP variants Several other genes, molecules, miRNA, and SNP variants showed in Table 2 have been shown to be associated with different types of epileptic seizures and epilepsy-related cognitive deficiencies. Although the exact mechanisms remain unclear, we hypothesize that a complex molecular and genetic network is involved in epileptogenesis, and that the molecules and genes in this network are potential targets for improving the clinical prognosis of epilepsy and cognitive outcomes. 7. Conclusions There are a variety of molecules and pathways involved in the pathogenesis of epilepsy and related cognitive deficits. We hope that this review provides readers, researchers, and clinicians an overall perspective on the genetic and molecular factors that are possibly associated with seizure-induced cognitive decline. Acknowledgments None. Funding This work was supported by the National Natural Science Foundation of China (81601134, 81501025), Yunnan Applied Basic Research Projects (2017FE468(-144)), the Natural Science Foundation of Hunan Province (No. 2016JJ3174), Yunnan Provincial Key Projects (2017FA041), Program for Science and Technology Innovation Team in Kunming Medical University (CXTD201614) and funded by China Scholarship Council. Declaration of competing interest None. Rereferences [1] Thijs RD, Surges R, O'Brien TJ, Sander JW. Epilepsy in adults. Lancet 2019;393: 689–701. [2] Gazaryan LM, Selyanina NV, Karakulova YV, Sosnin DY. The level of neuregulin-1 after traumatic brain injury and formation of post-traumatic epilepsy. Bull Exp Biol Med 2019;167:207–9. [3] Robert-Boire V, Desnous B, Lortie A, Carmant L, Ellezam B, Weil AG, et al. Seizures in pediatric patients with primary brain tumors. Pediatr Neurol 2019;97:50–5. [4] Yang C, Nicholas VH, Zhao J, Wu B, Zhong H, Li Y, et al. A novel CCM1/KRIT1 heterozygous nonsense mutation (c.1864CNT) associated with familial cerebral cavernous malformation: a genetic insight from an 8-year continuous observational study. J Mol Neurosci 2017;61:511–23. [5] Lehmann-Horn F, D'Amico A, Bertini E, Lomonaco M, Merlini L, Nelson KR, et al. Myotonia permanens with Nav1.4-G1306E displays varied phenotypes during course of life. Acta Myol 2017;36:125–34. [6] Symonds JD, Zuberi SM, Johnson MR. Advances in epilepsy gene discovery and implications for epilepsy diagnosis and treatment. Curr Opin Neurol 2017;30:193–9. [7] Stommel EW, Seguin R, Thadani VM, Schwartzman JD, Gilbert K, Ryan KA, et al. Cryptogenic epilepsy: an infectious etiology? Epilepsia 2001;42:436–8. [8] Devinsky O, Vezzani A, O'Brien TJ, Jette N, Scheffer IE, de Curtis M, et al. Epilepsy. Nat Rev Dis Primers 2018;4:18024. [9] Cohen MJ, Meador KJ, May R, Loblein H, Conrad T, Baker GA, et al. Fetal antiepileptic drug exposure and learning and memory functioning at 6years of age: the NEAD prospective observational study. Epilepsy Behav 2019;92:154–64. [10] McCorry D, Bromley R. Does in utero exposure of antiepileptic drugs lead to failure to reach full cognitive potential? Seizure 2015;28:51–6. [11] Spatola M, Dalmau J. Seizures and risk of epilepsy in autoimmune and other inflammatory encephalitis. Curr Opin Neurol 2017;30:345–53. [12] Helmstaedter C, Witt JA. Epilepsy and cognition — a bidirectional relationship? Seizure 2017;49:83–9.
8
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
[13] Antonucci F, Alpar A, Kacza J, Caleo M, Verderio C, Giani A, et al. Cracking down on inhibition: selective removal of GABAergic interneurons from hippocampal networks. J Neurosci 2012;32:1989–2001. [14] Silva CG, Metin C, Fazeli W, Machado NJ, Darmopil S, Launay PS, et al. Adenosine receptor antagonists including caffeine alter fetal brain development in mice. Sci Transl Med 2013;5:197ra04. [15] Macdonald RL, Kang JQ, Gallagher MJ. Mutations in GABAA receptor subunits associated with genetic epilepsies. J Physiol 2010;588:1861–9. [16] Joshi S, Kapur J. Neurosteroid regulation of GABAA receptors: a role in catamenial epilepsy. Brain Res 2019;1703:31–40. [17] Maguire JL, Stell BM, Rafizadeh M, Mody I. Ovarian cycle-linked changes in GABA (A) receptors mediating tonic inhibition alter seizure susceptibility and anxiety. Nat Neurosci 2005;8:797–804. [18] Cope DW, Di Giovanni G, Fyson SJ, Orban G, Errington AC, Lorincz ML, et al. Enhanced tonic GABAA inhibition in typical absence epilepsy. Nat Med 2009;15: 1392–8. [19] Duveau V, Buhl DL, Evrard A, Ruggiero C, Mande-Niedergang B, Roucard C, et al. Pronounced antiepileptic activity of the subtype-selective GABAA-positive allosteric modulator PF-06372865 in the GAERS absence epilepsy model. CNS Neurosci Ther 2019;25:255–60. [20] Ehninger D, Silva AJ. Rapamycin for treating tuberous sclerosis and autism spectrum disorders. Trends Mol Med 2011;17:78–87. [21] Ge YX, Retracted Tian XZ. Clc-2 knockout attenuated experimental temporal lobe epilepsy in mice by tonic inhibition mediated by GABAA receptors. Brain Res Bull 2016;125:222. [22] Peng Z, Huang CS, Stell BM, Mody I, Houser CR. Altered expression of the delta subunit of the GABAA receptor in a mouse model of temporal lobe epilepsy. J Neurosci 2004;24:8629–39. [23] Brickley SG, Mody I. Extrasynaptic GABA(A) receptors: their function in the CNS and implications for disease. Neuron 2012;73:23–34. [24] Bright DP, Aller MI, Brickley SG. Synaptic release generates a tonic GABA (A) receptor-mediated conductance that modulates burst precision in thalamic relay neurons. J Neurosci 2007;27:2560–9. [25] Vida I, Bartos M, Jonas P. Shunting inhibition improves robustness of gamma oscillations in hippocampal interneuron networks by homogenizing firing rates. Neuron 2006;49:107–17. [26] Ge YX, Liu Y, Tang HY, Liu XG, Wang X. ClC-2 contributes to tonic inhibition mediated by alpha5 subunit-containing GABA(A) receptor in experimental temporal lobe epilepsy. Neuroscience 2011;186:120–7. [27] Fiumelli H, Cancedda L, Poo MM. Modulation of GABAergic transmission by activity via postsynaptic Ca2+-dependent regulation of KCC2 function. Neuron 2005;48: 773–86. [28] Karlocai MR, Wittner L, Toth K, Magloczky Z, Katarova Z, Rasonyi G, et al. Enhanced expression of potassium-chloride cotransporter KCC2 in human temporal lobe epilepsy. Brain Struct Funct 2016;221:3601–15. [29] Pathak HR, Weissinger F, Terunuma M, Carlson GC, Hsu FC, Moss SJ, et al. Disrupted dentate granule cell chloride regulation enhances synaptic excitability during development of temporal lobe epilepsy. J Neurosci 2007;27:14012–22. [30] D'Agostino D, Bertelli M, Gallo S, Cecchin S, Albiero E, Garofalo PG, et al. Mutations and polymorphisms of the CLCN2 gene in idiopathic epilepsy. Neurology 2004;63: 1500–2. [31] Everett K, Chioza B, Aicardi J, Aschauer H, Brouwer O, Callenbach P, et al. Linkage and mutational analysis of CLCN2 in childhood absence epilepsy. Epilepsy Res 2007;75:145–53. [32] Getova D, Bowery NG, Spassov V. Effects of GABAB receptor antagonists on learning and memory retention in a rat model of absence epilepsy. Eur J Pharmacol 1997; 320:9–13. [33] Froestl W, Gallagher M, Jenkins H, Madrid A, Melcher T, Teichman S, et al. SGS742: the first GABA(B) receptor antagonist in clinical trials. Biochem Pharmacol 2004; 68:1479–87. [34] Getova DP, Bowery NG. Effects of high-affinity GABAB receptor antagonists on active and passive avoidance responding in rodents with gammahydroxybutyrolactone-induced absence syndrome. Psychopharmacology (Berl) 2001;157:89–95. [35] Castellano C, Brioni JD, Nagahara AH, McGaugh JL. Post-training systemic and intraamygdala administration of the GABA-B agonist baclofen impairs retention. Behav Neural Biol 1989;52:170–9. [36] Castellano C, McGaugh JL. Oxotremorine attenuates retrograde amnesia induced by post-training administration of the GABAergic agonists muscimol and baclofen. Behav Neural Biol 1991;56:25–31. [37] McNamara RK, Skelton RW. Baclofen, a selective GABAB receptor agonist, dosedependently impairs spatial learning in rats. Pharmacol Biochem Behav 1996;53: 303–8. [38] Swartzwelder HS, Tilson HA, McLamb RL, Wilson WA. Baclofen disrupts passive avoidance retention in rats. Psychopharmacology (Berl) 1987;92:398–401. [39] Inaba Y, D'Antuono M, Bertazzoni G, Biagini G, Avoli M. Diminished presynaptic GABA(B) receptor function in the neocortex of a genetic model of absence epilepsy. Neurosignals 2009;17:121–31. [40] Hosford DA, Lin FH, Wang Y, Caddick SJ, Rees M, Parkinson NJ, et al. Studies of the lethargic (lh/lh) mouse model of absence seizures: regulatory mechanisms and identification of the lh gene. Adv Neurol 1999;79:239–52. [41] Wu Y, Chan KF, Eubanks JH, Guin Ting Wong C, Cortez MA, Shen L, et al. Transgenic mice over-expressing GABA(B)R1a receptors acquire an atypical absence epilepsylike phenotype. Neurobiol Dis 2007;26:439–51. [42] Noebels J, Avoli M, Rogawski M, Olsen R, Delgado-Escueta A. Jasper's basic mechanisms of the epilepsies. Oxford University Press; 2012.
[43] DeLorey TM, Sahbaie P, Hashemi E, Li WW, Salehi A, Clark DJ. Somatosensory and sensorimotor consequences associated with the heterozygous disruption of the autism candidate gene, Gabrb3. Behav Brain Res 2011;216:36–45. [44] Laurie DJ, Wisden W, Seeburg PH. The distribution of thirteen GABAA receptor subunit mRNAs in the rat brain. III. Embryonic and postnatal development. J Neurosci 1992;12:4151–72. [45] Pirker S, Schwarzer C, Wieselthaler A, Sieghart W, Sperk G. GABA(A) receptors: immunocytochemical distribution of 13 subunits in the adult rat brain. Neuroscience 2000;101:815–50. [46] Zhang JH, Sato M, Tohyama M. Different postnatal ontogenic profiles of neurons containing beta (beta 1, beta 2 and beta 3) subunit mRNAs of GABAA receptor in the rat thalamus. Brain Res Dev Brain Res 1991;58:289–92. [47] Tanaka M, DeLorey TM, Delgado-Escueta A. GABRB3, epilepsy, and neurodevelopment. th , In: Olsen RW, Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, editors. Jasper's basic mechanisms of the epilepsies. Bethesda (MD): National Center for Biotechnology Information (US); 2012 [Michael A Rogawski, Antonio V Delgado-Escueta, Jeffrey L Noebels, Massimo Avoli and Richard W Olsen]. [48] Hogart A, Nagarajan RP, Patzel KA, Yasui DH, Lasalle JM. 15q11-13 GABAA receptor genes are normally biallelically expressed in brain yet are subject to epigenetic dysregulation in autism-spectrum disorders. Hum Mol Genet 2007;16:691–703. [49] Samaco RC, Hogart A, LaSalle JM. Epigenetic overlap in autism-spectrum neurodevelopmental disorders: MECP2 deficiency causes reduced expression of UBE3A and GABRB3. Hum Mol Genet 2005;14:483–92. [50] Swann JW, Le JT, Lam TT, Owens J, Mayer AT. The impact of chronic network hyperexcitability on developing glutamatergic synapses. Eur J Neurosci 2007;26:975–91. [51] Swann JW, Le JT, Lee CL. Recurrent seizures and the molecular maturation of hippocampal and neocortical glutamatergic synapses. Dev Neurosci 2007;29:168–78. [52] Wayman GA, Lee YS, Tokumitsu H, Silva AJ, Soderling TR. Calmodulin-kinases: modulators of neuronal development and plasticity. Neuron 2008;59:914–31. [53] Petersen JD, Chen X, Vinade L, Dosemeci A, Lisman JE, Reese TS. Distribution of postsynaptic density (PSD)-95 and Ca2+/calmodulin-dependent protein kinase II at the PSD. J Neurosci 2003;23:11270–8. [54] Sacktor TC, Fenton AA. What does LTP tell us about the roles of CaMKII and PKMzeta in memory? Mol Brain 2018;11:77. [55] Shioda N, Fukunaga K. Physiological and pathological roles of CaMKII-PP1 signaling in the brain. Int J Mol Sci 2017;19. [56] Zalcman G, Federman N, Romano A. CaMKII isoforms in learning and memory: localization and function. Front Mol Neurosci 2018;11:445. [57] Dong Y, Rosenberg HC. Prolonged changes in Ca2+/calmodulin-dependent protein kinase II after a brief pentylenetetrazol seizure; potential role in kindling. Epilepsy Res 2004;58:107–17. [58] Ni H, Jiang YW, Tao LY, Cen JN, Wu XR. Effects of penicillin-induced developmental epilepticus on hippocampal regenerative sprouting, related gene expression and cognitive deficits in rats. Toxicol Lett 2009;188:161–6. [59] Xiong Y, Zhou H, Zhang L. Influences of hyperthermia-induced seizures on learning, memory and phosphorylative state of CaMKIIalpha in rat hippocampus. Brain Res 2014;1557:190–200. [60] Ramocki MB, Bartnik M, Szafranski P, Kolodziejska KE, Xia Z, Bravo J, et al. Recurrent distal 7q11.23 deletion including HIP1 and YWHAG identified in patients with intellectual disabilities, epilepsy, and neurobehavioral problems. Am J Hum Genet 2010;87:857–65. [61] Metzler M, Gan L, Wong TP, Liu L, Helm J, Liu L, et al. NMDA receptor function and NMDA receptor-dependent phosphorylation of huntingtin is altered by the endocytic protein HIP1. J Neurosci 2007;27:2298–308. [62] Parker JA, Metzler M, Georgiou J, Mage M, Roder JC, Rose AM, et al. Huntingtininteracting protein 1 influences worm and mouse presynaptic function and protects Caenorhabditis elegans neurons against mutant polyglutamine toxicity. J Neurosci 2007;27:11056–64. [63] Metzler M, Li B, Gan L, Georgiou J, Gutekunst CA, Wang Y, et al. Disruption of the endocytic protein HIP1 results in neurological deficits and decreased AMPA receptor trafficking. EMBO J 2003;22:3254–66. [64] Petrini EM, Lu J, Cognet L, Lounis B, Ehlers MD, Choquet D. Endocytic trafficking and recycling maintain a pool of mobile surface AMPA receptors required for synaptic potentiation. Neuron 2009;63:92–105. [65] Conn PJ, Niswender CM. mGluR7's lucky number. Proc Natl Acad Sci U S A 2006; 103:251–2. [66] Niswender CM, Conn PJ. Metabotropic glutamate receptors: physiology, pharmacology, and disease. Annu Rev Pharmacol Toxicol 2010;50:295–322. [67] Sansig G, Bushell TJ, Clarke VR, Rozov A, Burnashev N, Portet C, et al. Increased seizure susceptibility in mice lacking metabotropic glutamate receptor 7. J Neurosci 2001;21:8734–45. [68] Bertaso F, Zhang C, Scheschonka A, de Bock F, Fontanaud P, Marin P, et al. PICK1 uncoupling from mGluR7a causes absence-like seizures. Nat Neurosci 2008;11: 940–8. [69] Celli R, Santolini I, Van Luijtelaar G, Ngomba RT, Bruno V, Nicoletti F. Targeting metabotropic glutamate receptors in the treatment of epilepsy: rationale and current status. Expert Opin Ther Targets 2019;23:341–51. [70] Martin R, Ladera C, Bartolome-Martin D, Torres M, Sanchez-Prieto J. The inhibition of release by mGlu7 receptors is independent of the Ca2+ channel type but associated to GABAB and adenosine A1 receptors. Neuropharmacology 2008;55: 464–73. [71] Eichler SA, Kirischuk S, Juttner R, Schaefermeier PK, Legendre P, Lehmann TN, et al. Glycinergic tonic inhibition of hippocampal neurons with depolarizing GABAergic transmission elicits histopathological signs of temporal lobe epilepsy. J Cell Mol Med 2008;12:2848–66.
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848 [72] Kankowski S, Forstera B, Winkelmann A, Knauff P, Wanker EE, You XA, et al. A novel RNA editing sensor tool and a specific agonist determine neuronal protein expression of RNA-edited glycine receptors and identify a genomic APOBEC1 dimorphism as a new genetic risk factor of epilepsy. Front Mol Neurosci 2017;10: 439. [73] Meier JC, Henneberger C, Melnick I, Racca C, Harvey RJ, Heinemann U, et al. RNA editing produces glycine receptor alpha3(P185L), resulting in high agonist potency. Nat Neurosci 2005;8:736–44. [74] Winkelmann A, Maggio N, Eller J, Caliskan G, Semtner M, Haussler U, et al. Changes in neural network homeostasis trigger neuropsychiatric symptoms. J Clin Invest 2014;124:696–711. [75] Mikroulis AV, Psarropoulou C. Endogenous ACh effects on NMDA-induced interictal-like discharges along the septotemporal hippocampal axis of adult rats and their modulation by an early life generalized seizure. Epilepsia 2012;53: 879–87. [76] Li B, Duysen EG, Volpicelli-Daley LA, Levey AI, Lockridge O. Regulation of muscarinic acetylcholine receptor function in acetylcholinesterase knockout mice. Pharmacol Biochem Behav 2003;74:977–86. [77] Turski L, Ikonomidou C, Turski WA, Bortolotto ZA, Cavalheiro EA. Review: cholinergic mechanisms and epileptogenesis. The seizures induced by pilocarpine: a novel experimental model of intractable epilepsy. Synapse 1989;3:154–71. [78] Villa C, Colombo G, Meneghini S, Gotti C, Moretti M, Ferini-Strambi L, et al. CHRNA2 and nocturnal frontal lobe epilepsy: identification and characterization of a novel loss of function mutation. Front Mol Neurosci 2019;12:17. [79] Meilleur S, Aznavour N, Descarries L, Carmant L, Mamer OA, Psarropoulou C. Pentylenetetrazol-induced seizures in immature rats provoke long-term changes in adult hippocampal cholinergic excitability. Epilepsia 2003;44:507–17. [80] Meilleur S, Carmant L, Psarropoulou C. Immature rat convulsions and long-term effects on hippocampal cholinergic neurotransmission. Neuroreport 2000;11:521–4. [81] Marino MJ, Rouse ST, Levey AI, Potter LT, Conn PJ. Activation of the genetically defined m1 muscarinic receptor potentiates N-methyl-D-aspartate (NMDA) receptor currents in hippocampal pyramidal cells. Proc Natl Acad Sci U S A 1998;95: 11465–70. [82] Hamilton SE, Loose MD, Qi M, Levey AI, Hille B, McKnight GS, et al. Disruption of the m1 receptor gene ablates muscarinic receptor-dependent M current regulation and seizure activity in mice. Proc Natl Acad Sci U S A 1997;94:13311–6. [83] Delmas P, Brown DA. Pathways modulating neural KCNQ/M (Kv7) potassium channels. Nat Rev Neurosci 2005;6:850–62. [84] Gigout S, Wierschke S, Lehmann TN, Horn P, Dehnicke C, Deisz RA. Muscarinic acetylcholine receptor-mediated effects in slices from human epileptogenic cortex. Neuroscience 2012;223:399–411. [85] Bliss TV, Collingridge GL. A synaptic model of memory: long-term potentiation in the hippocampus. Nature 1993;361:31–9. [86] Wess J. Muscarinic acetylcholine receptor knockout mice: novel phenotypes and clinical implications. Annu Rev Pharmacol Toxicol 2004;44:423–50. [87] Harbaugh RE, Roberts DW, Coombs DW, Saunders RL, Reeder TM. Preliminary report: intracranial cholinergic drug infusion in patients with Alzheimer's disease. Neurosurgery 1984;15:514–8. [88] Minzenberg MJ, Poole JH, Benton C, Vinogradov S. Association of anticholinergic load with impairment of complex attention and memory in schizophrenia. Am J Psychiatry 2004;161:116–24. [89] Hasselmo ME, McGaughy J. High acetylcholine levels set circuit dynamics for attention and encoding and low acetylcholine levels set dynamics for consolidation. Prog Brain Res 2004;145:207–31. [90] De Fusco M, Becchetti A, Patrignani A, Annesi G, Gambardella A, Quattrone A, et al. The nicotinic receptor beta 2 subunit is mutant in nocturnal frontal lobe epilepsy. Nat Genet 2000;26:275–6. [91] Steinlein OK, Mulley JC, Propping P, Wallace RH, Phillips HA, Sutherland GR, et al. A missense mutation in the neuronal nicotinic acetylcholine receptor alpha 4 subunit is associated with autosomal dominant nocturnal frontal lobe epilepsy. Nat Genet 1995;11:201–3. [92] Steinlein OK, Hoda JC, Bertrand S, Bertrand D. Mutations in familial nocturnal frontal lobe epilepsy might be associated with distinct neurological phenotypes. Seizure 2012;21:118–23. [93] Scheffer IE, Bhatia KP, Lopes-Cendes I, Fish DR, Marsden CD, Andermann E, et al. Autosomal dominant nocturnal frontal lobe epilepsy. A distinctive clinical disorder. Brain 1995;118(Pt 1):61–73. [94] Picard F, Pegna AJ, Arntsberg V, Lucas N, Kaczmarek I, Todica O, et al. Neuropsychological disturbances in frontal lobe epilepsy due to mutated nicotinic receptors. Epilepsy Behav 2009;14:354–9. [95] Steinlein OK, Bertrand D. Neuronal nicotinic acetylcholine receptors: from the genetic analysis to neurological diseases. Biochem Pharmacol 2008;76:1175–83. [96] Bertrand D, Elmslie F, Hughes E, Trounce J, Sander T, Bertrand S, et al. The CHRNB2 mutation I312M is associated with epilepsy and distinct memory deficits. Neurobiol Dis 2005;20:799–804. [97] Greenwood PM, Fossella JA, Parasuraman R. Specificity of the effect of a nicotinic receptor polymorphism on individual differences in visuospatial attention. J Cogn Neurosci 2005;17:1611–20. [98] Parasuraman R, Greenwood PM, Kumar R, Fossella J. Beyond heritability: neurotransmitter genes differentially modulate visuospatial attention and working memory. Psychol Sci 2005;16:200–7. [99] Wood AG, Saling MM, Fedi M, Berkovic SF, Scheffer IE, Benjamin C, et al. Neuropsychological function in patients with a single gene mutation associated with autosomal dominant nocturnal frontal lobe epilepsy. Epilepsy Behav 2010;17:531–5. [100] Cho YW, Yi SD, Lim JG, Kim DK, Motamedi GK. Autosomal dominant nocturnal frontal lobe epilepsy and mild memory impairment associated with CHRNB2
[101]
[102]
[103]
[104]
[105]
[106] [107]
[108]
[109]
[110]
[111]
[112]
[113] [114]
[115]
[116]
[117] [118] [119]
[120] [121] [122] [123]
[124]
[125] [126]
[127] [128]
[129] [130]
[131]
9
mutation I312M in the neuronal nicotinic acetylcholine receptor. Epilepsy Behav 2008;13:361–5. ArunSundar M, Shanmugarajan TS, Ravichandiran V. 3,4-Dihydroxyphenylethanol assuages cognitive impulsivity in Alzheimer's disease by attuning HPA-Axis via differential crosstalk of alpha7 nAChR with MicroRNA-124 and HDAC6. ACS Chem Nerosci 2018;9:2904–16. Bartus RT. On neurodegenerative diseases, models, and treatment strategies: lessons learned and lessons forgotten a generation following the cholinergic hypothesis. Exp Neurol 2000;163:495–529. Elnagar MR, Walls AB, Helal GK, Hamada FM, Thomsen MS, Jensen AA. Probing the putative alpha7 nAChR/NMDAR complex in human and murine cortex and hippocampus: different degrees of complex formation in healthy and Alzheimer brain tissue. PLoS One 2017;12:e0189513. Haense C, Kalbe E, Herholz K, Hohmann C, Neumaier B, Krais R, et al. Cholinergic system function and cognition in mild cognitive impairment. Neurobiol Aging 2012;33:867–77. Winterer G, Musso F, Konrad A, Vucurevic G, Stoeter P, Sander T, et al. Association of attentional network function with exon 5 variations of the CHRNA4 gene. Hum Mol Genet 2007;16:2165–74. Passani MB, Giannoni P, Bucherelli C, Baldi E, Blandina P. Histamine in the brain: beyond sleep and memory. Biochem Pharmacol 2007;73:1113–22. Alachkar A, Lazewska D, Frank A, Kiec-Kononowicz K, Sadek B, Latacz G, et al. Studies on anticonvulsant effects of novel histamine H3R antagonists in electrically and chemically induced seizures in rats. Int J Mol Sci 2018;19. Bhowmik M, Khanam R, Vohora D. Histamine H3 receptor antagonists in relation to epilepsy and neurodegeneration: a systemic consideration of recent progress and perspectives. Br J Pharmacol 2012;167:1398–414. Lintunen M, Sallmen T, Karlstedt K, Panula P. Transient changes in the limbic histaminergic system after systemic kainic acid-induced seizures. Neurobiol Dis 2005; 20:155–69. Yawata I, Tanaka K, Nakagawa Y, Watanabe Y, Murashima YL, Nakano K. Role of histaminergic neurons in development of epileptic seizures in EL mice. Brain Res Mol Brain Res 2004;132:13–7. Bongers G, Sallmen T, Passani MB, Mariottini C, Wendelin D, Lozada A, et al. The Akt/GSK-3beta axis as a new signaling pathway of the histamine H(3) receptor. J Neurochem 2007;103:248–58. Jin C, Lintunen M, Panula P. Histamine H(1) and H(3) receptors in the rat thalamus and their modulation after systemic kainic acid administration. Exp Neurol 2005; 194:43–56. Turturici G, Sconzo G, Geraci F. Hsp70 and its molecular role in nervous system diseases. Biochem Res Int 2011;2011:618127. Zhang W, Peterson M, Beyer B, Frankel WN, Zhang ZW. Loss of MeCP2 from forebrain excitatory neurons leads to cortical hyperexcitation and seizures. J Neurosci 2014;34:2754–63. Cardoza B, Clarke A, Wilcox J, Gibbon F, Smith PE, Archer H, et al. Epilepsy in Rett syndrome: association between phenotype and genotype, and implications for practice. Seizure 2011;20:646–9. Jian L, Nagarajan L, de Klerk N, Ravine D, Christodoulou J, Leonard H. Seizures in Rett syndrome: an overview from a one-year calendar study. Eur J Paediatr Neurol 2007;11:310–7. Nissenkorn A, Gak E, Vecsler M, Reznik H, Menascu S, Ben Zeev B. Epilepsy in Rett syndrome—the experience of a National Rett Center. Epilepsia 2010;51:1252–8. Pintaudi M, Calevo MG, Vignoli A, Parodi E, Aiello F, Baglietto MG, et al. Epilepsy in Rett syndrome: clinical and genetic features. Epilepsy Behav 2010;19:296–300. Vignoli A, Fabio RA, La Briola F, Giannatiempo S, Antonietti A, Maggiolini S, et al. Correlations between neurophysiological, behavioral, and cognitive function in Rett syndrome. Epilepsy Behav 2010;17:489–96. Kim HJ, Kim SH, Kim HD, Lee JS, Lee YM, Koo KY, et al. Genetic and epileptic features in Rett syndrome. Yonsei Med J 2012;53:495–500. Chahrour M, Zoghbi HY. The story of Rett syndrome: from clinic to neurobiology. Neuron 2007;56:422–37. Armstrong DD. Neuropathology of Rett syndrome. J Child Neurol 2005;20:747–53. Chen RZ, Akbarian S, Tudor M, Jaenisch R. Deficiency of methyl-CpG binding protein-2 in CNS neurons results in a Rett-like phenotype in mice. Nat Genet 2001;27:327–31. Guy J, Hendrich B, Holmes M, Martin JE, Bird A. A mouse Mecp2-null mutation causes neurological symptoms that mimic Rett syndrome. Nat Genet 2001;27: 322–6. Nelson ED, Kavalali ET, Monteggia LM. MeCP2-dependent transcriptional repression regulates excitatory neurotransmission. Curr Biol 2006;16:710–6. Shahbazian M, Young J, Yuva-Paylor L, Spencer C, Antalffy B, Noebels J, et al. Mice with truncated MeCP2 recapitulate many Rett syndrome features and display hyperacetylation of histone H3. Neuron 2002;35:243–54. Chao HT, Zoghbi HY, Rosenmund C. MeCP2 controls excitatory synaptic strength by regulating glutamatergic synapse number. Neuron 2007;56:58–65. Asaka Y, Jugloff DG, Zhang L, Eubanks JH, Fitzsimonds RM. Hippocampal synaptic plasticity is impaired in the Mecp2-null mouse model of Rett syndrome. Neurobiol Dis 2006;21:217–27. Guy J, Gan J, Selfridge J, Cobb S, Bird A. Reversal of neurological defects in a mouse model of Rett syndrome. Science 2007;315:1143–7. Moretti P, Levenson JM, Battaglia F, Atkinson R, Teague R, Antalffy B, et al. Learning and memory and synaptic plasticity are impaired in a mouse model of Rett syndrome. J Neurosci 2006;26:319–27. Dani VS, Chang Q, Maffei A, Turrigiano GG, Jaenisch R, Nelson SB. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc Natl Acad Sci U S A 2005;102:12560–5.
10
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
[132] Zhang L, He J, Jugloff DG, Eubanks JH. The MeCP2-null mouse hippocampus displays altered basal inhibitory rhythms and is prone to hyperexcitability. Hippocampus 2008;18:294–309. [133] Zhang ZW, Zak JD, Liu H. MeCP2 is required for normal development of GABAergic circuits in the thalamus. J Neurophysiol 2010;103:2470–81. [134] Calfa G, Hablitz JJ, Pozzo-Miller L. Network hyperexcitability in hippocampal slices from Mecp2 mutant mice revealed by voltage-sensitive dye imaging. J Neurophysiol 2011;105:1768–84. [135] Chao HT, Chen H, Samaco RC, Xue M, Chahrour M, Yoo J, et al. Dysfunction in GABA signalling mediates autism-like stereotypies and Rett syndrome phenotypes. Nature 2010;468:263–9. [136] D'Cruz JA, Wu C, Zahid T, El-Hayek Y, Zhang L, Eubanks JH. Alterations of cortical and hippocampal EEG activity in MeCP2-deficient mice. Neurobiol Dis 2010;38: 8–16. [137] Engel T, Murphy BM, Hatazaki S, Jimenez-Mateos EM, Concannon CG, Woods I, et al. Reduced hippocampal damage and epileptic seizures after status epilepticus in mice lacking proapoptotic Puma. FASEB J 2010;24:853–61. [138] Kang JQ, Shen W, Lee M, Gallagher MJ, Macdonald RL. Slow degradation and aggregation in vitro of mutant GABAA receptor gamma2(Q351X) subunits associated with epilepsy. J Neurosci 2010;30:13895–905. [139] Wyers L, Van de Walle P, Hoornweg A, Tepes Bobescu I, Verheyen K, Ceulemans B, et al. Gait deviations in patients with dravet syndrome: a systematic review. Eur J Paediatr Neurol 2019;23:357–67. [140] Berkovic SF, Harkin L, McMahon JM, Pelekanos JT, Zuberi SM, Wirrell EC, et al. Denovo mutations of the sodium channel gene SCN1A in alleged vaccine encephalopathy: a retrospective study. Lancet Neurol 2006;5:488–92. [141] Kang JQ, Macdonald RL. Making sense of nonsense GABA(A) receptor mutations associated with genetic epilepsies. Trends Mol Med 2009;15:430–8. [142] Hirose S. A new paradigm of channelopathy in epilepsy syndromes: intracellular trafficking abnormality of channel molecules. Epilepsy Res 2006;70(Suppl. 1): S206–17. [143] Yang C, Zhao J, Wu B, Zhong H, Li Y, Xu Y. Identification of a novel deletion mutation (c.1780delG) and a novel splice-site mutation (c.1412-1GNA) in the CCM1/ KRIT1 gene associated with familial cerebral cavernous malformation in the Chinese population. J Mol Neurosci 2017;61:8–15. [144] Petrelli C, Passamonti C, Cesaroni E, Mei D, Guerrini R, Zamponi N, et al. Early clinical features in Dravet syndrome patients with and without SCN1A mutations. Epilepsy Res 2012;99:21–7. [145] Ritter-Makinson S, Clemente-Perez A, Higashikubo B, Cho FS, Holden SS, Bennett E, et al. Augmented reticular thalamic bursting and seizures in Scn1a-Dravet syndrome. Cell Rep 2019;26:54–64 [e6]. [146] Yu FH, Mantegazza M, Westenbroek RE, Robbins CA, Kalume F, Burton KA, et al. Reduced sodium current in GABAergic interneurons in a mouse model of severe myoclonic epilepsy in infancy. Nat Neurosci 2006;9:1142–9. [147] Ogiwara I, Miyamoto H, Morita N, Atapour N, Mazaki E, Inoue I, et al. 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 2007;27: 5903–14. [148] Gu F, Hazra A, Aulakh A, Ziburkus J. Purinergic control of hippocampal circuit hyperexcitability in Dravet syndrome. Epilepsia 2014;55:245–55. [149] Manita S, Kawamura Y, Sato K, Inoue M, Kudo Y, Miyakawa H. Adenosine a(1)-receptor-mediated tonic inhibition of glutamate release at rat hippocampal CA3-CA1 synapses is primarily due to inhibition of N-type CA(2+) channels. Eur J Pharmacol 2004;499:265–74. [150] Boison D. The adenosine kinase hypothesis of epileptogenesis. Prog Neurobiol 2008;84:249–62. [151] Bender AC, Natola H, Ndong C, Holmes GL, Scott RC, Lenck-Santini PP. Focal Scn1a knockdown induces cognitive impairment without seizures. Neurobiol Dis 2013; 54:297–307. [152] O'Roak BJ, Deriziotis P, Lee C, Vives L, Schwartz JJ, Girirajan S, et al. Exome sequencing in sporadic autism spectrum disorders identifies severe de novo mutations. Nat Genet 2011;43:585–9. [153] Weiss LA, Escayg A, Kearney JA, Trudeau M, MacDonald BT, Mori M, et al. Sodium channels SCN1A, SCN2A and SCN3A in familial autism. Mol Psychiatry 2003;8: 186–94. [154] Kim DY, Carey BW, Wang H, Ingano LA, Binshtok AM, Wertz MH, et al. BACE1 regulates voltage-gated sodium channels and neuronal activity. Nat Cell Biol 2007;9: 755–64. [155] Kovacs DM, Gersbacher MT, Kim DY. Alzheimer's secretases regulate voltage-gated sodium channels. Neurosci Lett 2010;486:68–72. [156] Martinez-Losa M, Tracy TE, Ma K, Verret L, Clemente-Perez A, Khan AS, et al. Nav1.1-overexpressing interneuron transplants restore brain rhythms and cognition in a mouse model of Alzheimer's disease. Neuron 2018;98:75–89.e5. [157] Verret L, Mann EO, Hang GB, Barth AM, Cobos I, Ho K, et al. Inhibitory interneuron deficit links altered network activity and cognitive dysfunction in Alzheimer model. Cell 2012;149:708–21. [158] Bender AC, Morse RP, Scott RC, Holmes GL, Lenck-Santini PP. SCN1A mutations in Dravet syndrome: impact of interneuron dysfunction on neural networks and cognitive outcome. Epilepsy Behav 2012;23:177–86. [159] Mares J, Stopka P, Nohejlova K, Rokyta R. Oxidative stress induced by epileptic seizure and its attenuation by melatonin. Physiol Res 2013;62(Suppl. 1):S67–74. [160] Folbergrova J, Kunz WS. Mitochondrial dysfunction in epilepsy. Mitochondrion 2012;12:35–40. [161] Langmeier M, Mares J. Changes in some ultrastructural parameters of cortical synapses in the initial phases of kindling. Physiol Bohemoslov 1984;33:367–75.
[162] Willmore LJ, Ueda Y. Posttraumatic epilepsy: hemorrhage, free radicals and the molecular regulation of glutamate. Neurochem Res 2009;34:688–97. [163] Feng J, Feng L, Zhang G. Mitochondrial damage in hippocampal neurons of rats with epileptic protein expression of Fas and caspase-3. Exp Ther Med 2018;16:2483–9. [164] Kunz WS, Bimpong-Buta NY, Kudin AP, Elger CE. The role of mitochondria in epilepsy: implications for neurodegenerative diseases. Toxicol Mech Methods 2004; 14:19–23. [165] Jacoby S, Sims RE, Hartell NA. Nitric oxide is required for the induction and heterosynaptic spread of long-term potentiation in rat cerebellar slices. J Physiol 2001;535:825–39. [166] Lee KY, Chung K, Chung JM. Involvement of reactive oxygen species in long-term potentiation in the spinal cord dorsal horn. J Neurophysiol 2010;103:382–91. [167] de Freitas RL, Santos IM, de Souza GF, Tome Ada R, Saldanha GB, de Freitas RM. Oxidative stress in rat hippocampus caused by pilocarpine-induced seizures is reversed by buspirone. Brain Res Bull 2010;81:505–9. [168] Reiter RJ. Oxidative damage in the central nervous system: protection by melatonin. Prog Neurobiol 1998;56:359–84. [169] Marini C, Darra F, Specchio N, Mei D, Terracciano A, Parmeggiani L, et al. Focal seizures with affective symptoms are a major feature of PCDH19 gene-related epilepsy. Epilepsia 2012;53:2111–9. [170] Janc OA, Muller M. The free radical scavenger Trolox dampens neuronal hyperexcitability, reinstates synaptic plasticity, and improves hypoxia tolerance in a mouse model of Rett syndrome. Front Cell Neurosci 2014;8:56. [171] Ikeno S, Nagata N, Yoshida S, Takahashi H, Kigawa J, Terakawa N. Immature brain injury via peroxynitrite production induced by inducible nitric oxide synthase after hypoxia-ischemia in rats. J Obstet Gynaecol Res 2000;26:227–34. [172] Radi R, Cassina A, Hodara R, Quijano C, Castro L. Peroxynitrite reactions and formation in mitochondria. Free Radic Biol Med 2002;33:1451–64. [173] Stowe DF, Camara AK. Mitochondrial reactive oxygen species production in excitable cells: modulators of mitochondrial and cell function. Antioxid Redox Signal 2009;11:1373–414. [174] Ohkuma S, Katsura M, Higo A, Shirotani K, Hara A, Tarumi C, et al. Peroxynitrite affects Ca2 + influx through voltage-dependent calcium channels. J Neurochem 2001;76:341–50. [175] Yang J, Liu Z, Xie Y, Yang Z, Zhang T. Peroxynitrite alters GABAergic synaptic transmission in immature rat hippocampal slices. Neurosci Res 2013;75:210–7. [176] Kovac S, Abramov AY, Walker MC. Energy depletion in seizures: anaplerosis as a strategy for future therapies. Neuropharmacology 2013;69:96–104. [177] Cognato GP, Vuaden FC, Savio LE, Bellaver B, Casali E, Bogo MR, et al. Nucleoside triphosphate diphosphohydrolases role in the pathophysiology of cognitive impairment induced by seizure in early age. Neuroscience 2011;180:191–200. [178] Gerosa L, Francolini M, Bassani S, Passafaro M. The role of protocadherin 19 (PCDH19) in neurodevelopment and in the pathophysiology of early infantile epileptic encephalopathy-9 (EIEE9). Dev Neurobiol 2019;79:75–84. [179] Niazi R, Fanning EA, Depienne C, Sarmady M, Abou Tayoun AN. A mutation update for the PCDH19 gene causing early-onset epilepsy in females with an unusual expression pattern. Hum Mutat 2019;40:243–57. [180] Specchio N, Marini C, Terracciano A, Mei D, Trivisano M, Sicca F, et al. Spectrum of phenotypes in female patients with epilepsy due to protocadherin 19 mutations. Epilepsia 2011;52:1251–7. [181] Camacho A, Simon R, Sanz R, Vinuela A, Martinez-Salio A, Mateos F. Cognitive and behavioral profile in females with epilepsy with PDCH19 mutation: two novel mutations and review of the literature. Epilepsy Behav 2012;24:134–7. [182] Pitkanen A. Drug-mediated neuroprotection and antiepileptogenesis: animal data. Neurology 2002;59:S27–33. [183] Sloviter RS. Progress on the issue of excitotoxic injury modification vs. real neuroprotection; implications for post-traumatic epilepsy. Neuropharmacology 2011;61: 1048–50. [184] Li T, Ren G, Lusardi T, Wilz A, Lan JQ, Iwasato T, et al. Adenosine kinase is a target for the prediction and prevention of epileptogenesis in mice. J Clin Invest 2008;118: 571–82. [185] Mouri G, Jimenez-Mateos E, Engel T, Dunleavy M, Hatazaki S, Paucard A, et al. Unilateral hippocampal CA3-predominant damage and short latency epileptogenesis after intra-amygdala microinjection of kainic acid in mice. Brain Res 2008;1213: 140–51. [186] Bumanglag AV, Sloviter RS. Minimal latency to hippocampal epileptogenesis and clinical epilepsy after perforant pathway stimulation-induced status epilepticus in awake rats. J Comp Neurol 2008;510:561–80. [187] Paz JT, Christian CA, Parada I, Prince DA, Huguenard JR. Focal cortical infarcts alter intrinsic excitability and synaptic excitation in the reticular thalamic nucleus. J Neurosci 2010;30:5465–79. [188] Narkilahti S, Nissinen J, Pitkanen A. Administration of caspase 3 inhibitor during and after status epilepticus in rat: effect on neuronal damage and epileptogenesis. Neuropharmacology 2003;44:1068–88. [189] Jimenez-Mateos EM, Hatazaki S, Johnson MB, Bellver-Estelles C, Mouri G, Bonner C, et al. Hippocampal transcriptome after status epilepticus in mice rendered seizure damage-tolerant by epileptic preconditioning features suppressed calcium and neuronal excitability pathways. Neurobiol Dis 2008;32:442–53. [190] Loscher W, Brandt C. Prevention or modification of epileptogenesis after brain insults: experimental approaches and translational research. Pharmacol Rev 2010; 62:668–700. [191] Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 family reunion. Mol Cell 2010;37:299–310. [192] D'Orsi B, Mateyka J, Prehn JHM. Control of mitochondrial physiology and cell death by the Bcl-2 family proteins Bax and Bok. Neurochem Int 2017;109:162–70.
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848 [193] Hotchkiss RS, Strasser A, McDunn JE, Swanson PE. Cell death. N Engl J Med 2009; 361:1570–83. [194] Youle RJ, Strasser A. The BCL-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol 2008;9:47–59. [195] Galluzzi L, Blomgren K, Kroemer G. Mitochondrial membrane permeabilization in neuronal injury. Nat Rev Neurosci 2009;10:481–94. [196] Tait SW, Green DR. Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol 2010;11:621–32. [197] Graham SH, Chen J, Stetler RA, Zhu RL, Jin KL, Simon RP. Expression of the protooncogene bcl-2 is increased in the rat brain following kainate-induced seizures. Restor Neurol Neurosci 1996;9:243–50. [198] Henshall DC, Skradski SL, Lan JQ, Ren T, Simon RP. Increased Bcl-w expression following focally evoked limbic seizures in the rat. Neurosci Lett 2001;305:153–6. [199] Murphy B, Dunleavy M, Shinoda S, Schindler C, Meller R, Bellver-Estelles C, et al. Bcl-w protects hippocampus during experimental status epilepticus. Am J Pathol 2007;171:1258–68. [200] Mori M, Burgess DL, Gefrides LA, Foreman PJ, Opferman JT, Korsmeyer SJ, et al. Expression of apoptosis inhibitor protein Mcl1 linked to neuroprotection in CNS neurons. Cell Death Differ 2004;11:1223–33. [201] Ananth C, Thameem Dheen S, Gopalakrishnakone P, Kaur C. Domoic acid-induced neuronal damage in the rat hippocampus: changes in apoptosis related genes (bcl-2, bax, caspase-3) and microglial response. J Neurosci Res 2001;66:177–90. [202] Henshall DC, Araki T, Schindler CK, Lan JQ, Tiekoter KL, Taki W, et al. Activation of Bcl-2-associated death protein and counter-response of Akt within cell populations during seizure-induced neuronal death. J Neurosci 2002;22:8458–65. [203] Moran C, Sanz-Rodriguez A, Jimenez-Pacheco A, Martinez-Villareal J, McKiernan RC, Jimenez-Mateos EM, et al. Bmf upregulation through the AMP-activated protein kinase pathway may protect the brain from seizure-induced cell death. Cell Death Dis 2013;4:e606. [204] Murphy BM, Engel T, Paucard A, Hatazaki S, Mouri G, Tanaka K, et al. Contrasting patterns of Bim induction and neuroprotection in Bim-deficient mice between hippocampus and neocortex after status epilepticus. Cell Death Differ 2010;17: 459–68. [205] Henshall DC, Engel T. Contribution of apoptosis-associated signaling pathways to epileptogenesis: lessons from Bcl-2 family knockouts. Front Cell Neurosci 2013;7: 110. [206] Kilany A, Raouf ER, Gaber AA, Aloush TK, Aref HA, Anwar M, et al. Elevated serum Bcl-2 in children with temporal lobe epilepsy. Seizure 2012;21:250–3. [207] Kim YS, Choi MY, Ryu JH, Lee DH, Jeon BT, Roh GS, et al. Clusterin interaction with Bcl-xL is associated with seizure-induced neuronal death. Epilepsy Res 2012;99: 240–51. [208] Li LY, Li JL, Zhang HM, Yang WM, Wang K, Fang Y, et al. TGFbeta1 treatment reduces hippocampal damage, spontaneous recurrent seizures, and learning memory deficits in pilocarpine-treated rats. J Mol Neurosci 2013;50:109–23. [209] Lubin FD. Epigenetic gene regulation in the adult mammalian brain: multiple roles in memory formation. Neurobiol Learn Mem 2011;96:68–78. [210] Porcher C, Medina I, Gaiarsa JL. Mechanism of BDNF modulation in GABAergic synaptic transmission in healthy and disease brains. Front Cell Neurosci 2018;12:273. [211] Binder DK. The role of BDNF in epilepsy and other diseases of the mature nervous system. Adv Exp Med Biol 2004;548:34–56. [212] Vezzani A, Ravizza T, Moneta D, Conti M, Borroni A, Rizzi M, et al. Brain-derived neurotrophic factor immunoreactivity in the limbic system of rats after acute seizures and during spontaneous convulsions: temporal evolution of changes as compared to neuropeptide Y. Neuroscience 1999;90:1445–61. [213] Binder DK, Routbort MJ, Ryan TE, Yancopoulos GD, McNamara JO. Selective inhibition of kindling development by intraventricular administration of TrkB receptor body. J Neurosci 1999;19:1424–36. [214] Koyama R, Ikegaya Y. To BDNF or not to BDNF: that is the epileptic hippocampus. Neuroscientist 2005;11:282–7. [215] Chen WG, Chang Q, Lin Y, Meissner A, West AE, Griffith EC, et al. Derepression of BDNF transcription involves calcium-dependent phosphorylation of MeCP2. Science 2003;302:885–9. [216] Nishimura M, Owens J, Swann JW. Effects of chronic network hyperexcitability on the growth of hippocampal dendrites. Neurobiol Dis 2008;29:267–77. [217] McNamara JO, Scharfman HE. Temporal lobe epilepsy and the BDNF receptor, TrkB. th , In: Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV, editors. Jasper's basic mechanisms of the epilepsies. Bethesda (MD): National Center for biotechnology information (US); 2012 Michael A Rogawski, Antonio V DelgadoEscueta, Jeffrey L Noebels, Massimo Avoli and Richard W Olsen. [218] Gall CM, Isackson PJ. Limbic seizures increase neuronal production of messenger RNA for nerve growth factor. Science 1989;245:758–61. [219] Murray KD, Isackson PJ, Eskin TA, King MA, Montesinos SP, Abraham LA, et al. Altered mRNA expression for brain-derived neurotrophic factor and type II calcium/calmodulin-dependent protein kinase in the hippocampus of patients with intractable temporal lobe epilepsy. J Comp Neurol 2000;418:411–22. [220] Wetmore C, Olson L, Bean AJ. Regulation of brain-derived neurotrophic factor (BDNF) expression and release from hippocampal neurons is mediated by nonNMDA type glutamate receptors. J Neurosci 1994;14:1688–700. [221] Yan Q, Rosenfeld RD, Matheson CR, Hawkins N, Lopez OT, Bennett L, et al. Expression of brain-derived neurotrophic factor protein in the adult rat central nervous system. Neuroscience 1997;78:431–48. [222] He XP, Pan E, Sciarretta C, Minichiello L, McNamara JO. Disruption of TrkBmediated phospholipase Cgamma signaling inhibits limbic epileptogenesis. J Neurosci 2010;30:6188–96. [223] Schinder AF, Poo M. The neurotrophin hypothesis for synaptic plasticity. Trends Neurosci 2000;23:639–45.
11
[224] Sutula T, Steward O. Facilitation of kindling by prior induction of long-term potentiation in the perforant path. Brain Res 1987;420:109–17. [225] Goussakov IV, Fink K, Elger CE, Beck H. Metaplasticity of mossy fiber synaptic transmission involves altered release probability. J Neurosci 2000;20:3434–41. [226] Tanaka T, Saito H, Matsuki N. Inhibition of GABAA synaptic responses by brainderived neurotrophic factor (BDNF) in rat hippocampus. J Neurosci 1997;17: 2959–66. [227] Xu B, Michalski B, Racine RJ, Fahnestock M. The effects of brain-derived neurotrophic factor (BDNF) administration on kindling induction, Trk expression and seizure-related morphological changes. Neuroscience 2004;126:521–31. [228] Scharfman HE. Hyperexcitability in combined entorhinal/hippocampal slices of adult rat after exposure to brain-derived neurotrophic factor. J Neurophysiol 1997;78:1082–95. [229] Huang YZ, He XP, Krishnamurthy K, McNamara JO. TrkB-Shc signaling protects against hippocampal injury following status epilepticus. J Neurosci 2019;39: 4624–30. [230] Bredy TW, Wu H, Crego C, Zellhoefer J, Sun YE, Barad M. Histone modifications around individual BDNF gene promoters in prefrontal cortex are associated with extinction of conditioned fear. Learn Mem 2007;14:268–76. [231] Levenson JM, Roth TL, Lubin FD, Miller CA, Huang IC, Desai P, et al. Evidence that DNA (cytosine-5) methyltransferase regulates synaptic plasticity in the hippocampus. J Biol Chem 2006;281:15763–73. [232] Wood MA, Attner MA, Oliveira AM, Brindle PK, Abel T. A transcription factorbinding domain of the coactivator CBP is essential for long-term memory and the expression of specific target genes. Learn Mem 2006;13:609–17. [233] Ou LC, Gean PW. Transcriptional regulation of brain-derived neurotrophic factor in the amygdala during consolidation of fear memory. Mol Pharmacol 2007;72: 350–8. [234] Berton O, McClung CA, Dileone RJ, Krishnan V, Renthal W, Russo SJ, et al. Essential role of BDNF in the mesolimbic dopamine pathway in social defeat stress. Science 2006;311:864–8. [235] Huang Y, Doherty JJ, Dingledine R. Altered histone acetylation at glutamate receptor 2 and brain-derived neurotrophic factor genes is an early event triggered by status epilepticus. J Neurosci 2002;22:8422–8. [236] Tian F, Marini AM, Lipsky RH. NMDA receptor activation induces differential epigenetic modification of Bdnf promoters in hippocampal neurons. Amino Acids 2010; 38:1067–74. [237] Hoeffer CA, Klann E. mTOR signaling: at the crossroads of plasticity, memory and disease. Trends Neurosci 2010;33:67–75. [238] Crino PB. Focal brain malformations: seizures, signaling, sequencing. Epilepsia 2009;50(Suppl. 9):3–8. [239] Raffo E, Coppola A, Ono T, Briggs SW, Galanopoulou AS. A pulse rapamycin therapy for infantile spasms and associated cognitive decline. Neurobiol Dis 2011;43: 322–9. [240] Goto J, Talos DM, Klein P, Qin W, Chekaluk YI, Anderl S, et al. Regulable neural progenitor-specific Tsc1 loss yields giant cells with organellar dysfunction in a model of tuberous sclerosis complex. Proc Natl Acad Sci U S A 2011;108:E1070–9. [241] Galanopoulou AS, Gorter JA, Cepeda C. Finding a better drug for epilepsy: the mTOR pathway as an antiepileptogenic target. Epilepsia 2012;53:1119–30. [242] Talos DM, Sun H, Zhou X, Fitzgerald EC, Jackson MC, Klein PM, et al. The interaction between early life epilepsy and autistic-like behavioral consequences: a role for the mammalian target of rapamycin (mTOR) pathway. PLoS One 2012;7:e35885. [243] Kazdoba TM, Sunnen CN, Crowell B, Lee GH, Anderson AE, D'Arcangelo G. Development and characterization of NEX- Pten, a novel forebrain excitatory neuronspecific knockout mouse. Dev Neurosci 2012;34:198–209. [244] Poeta L, Fusco F, Drongitis D, Shoubridge C, Manganelli G, Filosa S, et al. A regulatory path associated with X-linked intellectual disability and epilepsy links KDM5C to the polyalanine expansions in ARX. Am J Hum Genet 2013;92:114–25. [245] Veeramah KR, Johnstone L, Karafet TM, Wolf D, Sprissler R, Salogiannis J, et al. Exome sequencing reveals new causal mutations in children with epileptic encephalopathies. Epilepsia 2013;54:1270–81. [246] Tremolizzo L, Curto NA, Marzorati L, Lanzani F, Tarantino P, Annesi G, et al. Earlyonset SCA17 with 43 TBP repeats: expanding the phenotype? Neurol Sci 2011; 32:941–3. [247] Vatta M, Tennison MB, Aylsworth AS, Turcott CM, Guerra MP, Eng CM, et al. A novel STXBP1 mutation causes focal seizures with neonatal onset. J Child Neurol 2012; 27:811–4. [248] Palanisamy A, Rajendran NN, Narmadha MP, Ganesvaran RA. Association of apolipoprotein E epsilon4 allele with cognitive impairment in patients with epilepsy and interaction with phenytoin monotherapy. Epilepsy Behav 2013; 26:165–9. [249] Mitsui S, Hidaka C, Furihata M, Osako Y, Yuri K. A mental retardation gene, motopsin/prss12, modulates cell morphology by interaction with seizure-related gene 6. Biochem Biophys Res Commun 2013;436:638–44. [250] Corradini I, Donzelli A, Antonucci F, Welzl H, Loos M, Martucci R, et al. Epileptiform activity and cognitive deficits in SNAP-25(+/−) mice are normalized by antiepileptic drugs. Cereb Cortex 2014;24:364–76. [251] Rojas A, Jiang J, Ganesh T, Yang MS, Lelutiu N, Gueorguieva P, et al. Cyclooxygenase2 in epilepsy. Epilepsia 2014;55:17–25. [252] Wang W, Wu Y, Zhang G, Fang H, Wang H, Zang H, et al. Activation of Nrf2-ARE signal pathway protects the brain from damage induced by epileptic seizure. Brain Res 2014;1544:54–61. [253] Illingworth MA, Hanrahan D, Anderson CE, O'Kane K, Anderson J, Casey M, et al. Elevated VGKC-complex antibodies in a boy with fever-induced refractory epileptic encephalopathy in school-age children (FIRES). Dev Med Child Neurol 2011;53: 1053–7.
12
L. Zhu et al. / Epilepsy & Behavior 104 (2020) 106848
[254] Corbett BF, Leiser SC, Ling HP, Nagy R, Breysse N, Zhang X, et al. Sodium channel cleavage is associated with aberrant neuronal activity and cognitive deficits in a mouse model of Alzheimer's disease. J Neurosci 2013;33:7020–6. [255] Iori V, Maroso M, Rizzi M, Iyer AM, Vertemara R, Carli M, et al. Receptor for advanced glycation endproducts is upregulated in temporal lobe epilepsy and contributes to experimental seizures. Neurobiol Dis 2013;58:102–14. [256] Kreis P, Hendricusdottir R, Kay L, Papageorgiou IE, van Diepen M, Mack T, et al. Phosphorylation of the actin binding protein Drebrin at S647 is regulated by neuronal activity and PTEN. PLoS One 2013;8:e71957. [257] Chamberlain SE, Jane DE, Jones RS. Pre- and post-synaptic functions of kainate receptors at glutamate and GABA synapses in the rat entorhinal cortex. Hippocampus 2012;22:555–76. [258] Hong S, Xin Y, HaiQin W, GuiLian Z, Ru Z, ShuQin Z, et al. The PPARgamma agonist rosiglitazone prevents cognitive impairment by inhibiting astrocyte activation and oxidative stress following pilocarpine-induced status epilepticus. Neurol Sci 2012; 33:559–66. [259] Hunt RF, Dinday MT, Hindle-Katel W, Baraban SC. LIS1 deficiency promotes dysfunctional synaptic integration of granule cells generated in the developing and adult dentate gyrus. J Neurosci 2012;32:12862–75. [260] Nelson TE, Olde Engberink A, Hernandez R, Puro A, Huitron-Resendiz S, Hao C, et al. Altered synaptic transmission in the hippocampus of transgenic mice with enhanced central nervous systems expression of interleukin-6. Brain Behav Immun 2012;26:959–71. [261] McAuliffe JJ, Bronson SL, Hester MS, Murphy BL, Dahlquist-Topala R, Richards DA, et al. Altered patterning of dentate granule cell mossy fiber inputs onto CA3 pyramidal cells in limbic epilepsy. Hippocampus 2011;21:93–107.
[262] Lehtimaki KA, Peltola J, Liimatainen S, Haapala AM, Arvio M. Cardiolipin and beta (2)-glycoprotein I antibodies associate with cognitive impairment and seizure frequency in developmental disorders. Seizure 2011;20:438–41. [263] Kunieda T, Zuscik MJ, Boongird A, Perez DM, Luders HO, Najm IM. Systemic overexpression of the alpha 1B-adrenergic receptor in mice: an animal model of epilepsy. Epilepsia 2002;43:1324–9. [264] Bao Y, Hudson QJ, Perera EM, Akan L, Tobet SA, Smith CA, et al. Expression and evolutionary conservation of the tescalcin gene during development. Gene Expr Patterns 2009;9:273–81. [265] Fusco A, Fedele M. Roles of HMGA proteins in cancer. Nat Rev Cancer 2007;7: 899–910. [266] Stein JL, Medland SE, Vasquez AA, Hibar DP, Senstad RE, Winkler AM, et al. Identification of common variants associated with human hippocampal and intracranial volumes. Nat Genet 2012;44:552–61. [267] Amir S, Ma AH, Shi XB, Xue L, Kung HJ, Devere White RW. Oncomir miR-125b suppresses p14(ARF) to modulate p53-dependent and p53-independent apoptosis in prostate cancer. PLoS One 2013;8:e61064. [268] Bielefeld P, Mooney C, Henshall DC, Fitzsimons CP. miRNA-mediated regulation of adult hippocampal neurogenesis; implications for epilepsy. Brain Plast 2017;3:43–59. [269] Cava C, Manna I, Gambardella A, Bertoli G, Castiglioni I. Potential role of miRNAs as theranostic biomarkers of epilepsy. Mol Ther Nucleic Acids 2018; 13:275–90. [270] Henshall DC. MicroRNAs in the pathophysiology and treatment of status epilepticus. Front Mol Neurosci 2013;6:37.