Skip to main content
Brain Communications logoLink to Brain Communications
. 2021 Mar 9;3(2):fcab021. doi: 10.1093/braincomms/fcab021

Proteomic differences in the hippocampus and cortex of epilepsy brain tissue

Geoffrey Pires 1,2,3,#, Dominique Leitner 1,#, Eleanor Drummond 2,4, Evgeny Kanshin 5, Shruti Nayak 5, Manor Askenazi 6,7, Arline Faustin 2, Daniel Friedman 1, Ludovic Debure 2, Beatrix Ueberheide 2,5,7, Thomas Wisniewski 2,8,9,, Orrin Devinsky 1,
PMCID: PMC8214864  PMID: 34159317

Abstract

Epilepsy is a common neurological disorder affecting over 70 million people worldwide, with a high rate of pharmaco-resistance, diverse comorbidities including progressive cognitive and behavioural disorders, and increased mortality from direct (e.g. sudden unexpected death in epilepsy, accidents, drowning) or indirect effects of seizures and therapies. Extensive research with animal models and human studies provides limited insights into the mechanisms underlying seizures and epileptogenesis, and these have not translated into significant reductions in pharmaco-resistance, morbidities or mortality. To help define changes in molecular signalling networks associated with seizures in epilepsy with a broad range of aetiologies, we examined the proteome of brain samples from epilepsy and control cases. Label-free quantitative mass spectrometry was performed on the hippocampal cornu ammonis 1–3 region (CA1–3), frontal cortex and dentate gyrus microdissected from epilepsy and control cases (n = 14/group). Epilepsy cases had significant differences in the expression of 777 proteins in the hippocampal CA1 − 3 region, 296 proteins in the frontal cortex and 49 proteins in the dentate gyrus in comparison to control cases. Network analysis showed that proteins involved in protein synthesis, mitochondrial function, G-protein signalling and synaptic plasticity were particularly altered in epilepsy. While protein differences were most pronounced in the hippocampus, similar changes were observed in other brain regions indicating broad proteomic abnormalities in epilepsy. Among the most significantly altered proteins, G-protein subunit beta 1 (GNB1) was one of the most significantly decreased proteins in epilepsy in all regions studied, highlighting the importance of G-protein subunit signalling and G-protein-coupled receptors in epilepsy. Our results provide insights into common molecular mechanisms underlying epilepsy across various aetiologies, which may allow for novel targeted therapeutic strategies.

Keywords: epilepsy, seizures, proteomics, synaptophysin


Pires, Leitner et al. present a comprehensive characterization of the human epileptic proteome. The results reveal extensive protein alterations common across diverse epilepsies, providing insights on pathogenesis and potential novel drug targets.

Graphical Abstract

Graphical Abstract.

Graphical Abstract

Introduction

Epilepsy is a spectrum disorder with diverse aetiologies and outcomes affecting over 70 million people worldwide.1–4 Aetiologies include ion channel and many other genetic mutations, infection, structural lesions, metabolic abnormalities and autoimmune disorders, although the cause remains unknown in many cases.5–9 The mechanisms underlying ictogenesis and epileptogenesis alter the balance of excitation and inhibition.10 Anti-seizure drugs (ASDs) target multiple mechanisms, including ion channels (e.g. sodium, calcium, potassium), neurotransmitter receptors and metabolism (e.g. GABA-A receptors, GABA transporter 1, GABA transaminase), synaptic vesicles (e.g. SV2A) and regulators of cell growth (e.g. mTOR inhibition). However, since one-third of patients have an incomplete response to ASDs, dietary, neuromodulatory and resective or disconnective surgical procedures, a broader understanding of the cellular and molecular drivers of ictogenesis and epileptogenesis is essential to identify new therapeutic targets.4,11,12

Human and animal epilepsy studies reveal that seizures can arise from neocortical or limbic lesions, including focal cortical dysplasia (FCD), hippocampal sclerosis, tumours, inflammation and vascular lesions.13–15 The hippocampus is often involved in focal epilepsy as a primary (e.g. mesial temporal lobe epilepsy [MTLE]) or secondary seizure focus (e.g. areas robustly connected with entorhinal cortex and hippocampus). The hippocampal cornu ammonis (CA) CA14 region and dentate gyrus are involved in seizure generation and in hippocampal sclerosis.16

To date, our knowledge of brain protein changes in epilepsy comes from experimental MTLE models and human brain tissue. The most robust changes in epilepsy brain tissue involve proteins related to neurotransmitters, cell signalling, glucose metabolism and protein synthesis.17,18 Genomic, epigenetic and transcriptomic studies have revealed insights into mechanisms underlying ictogenesis and epileptogenesis by identifying novel genetic loci and profound changes in gene expression in humans and animal models.19–24 However, the poor correlation between RNA expression and protein levels limits the value of isolated transcriptomic or genomic studies.25 Unbiased, mass spectrometry-based proteomic studies of human epilepsy tissue can complement these studies, particularly since proteins are direct drug targets. Proteomics provides a global view of protein changes at a functional or network level. The majority of previous epilepsy proteomics studies have examined animal model tissue, rather than human brain tissue.26,27 Two previous studies have performed proteomics on human epilepsy brain tissue, however, these were limited by either small numbers, analysis of single regions or low sensitivity proteomics approaches. These studies identified 77 proteins differentially expressed in MTLE hippocampi compared with controls28 and 18 differentially expressed proteins between high- and low-spiking regions in six epilepsy patients.29 We hypothesized that we would identify more extensive protein differences in epilepsy by using a more sensitive proteomic approach, and that we would generate novel insight into the pathophysiology of epilepsy by examining protein changes in multiple brain regions that are differentially affected in epilepsy.

Recent advances in proteomics methods have shown that proteomics can be successfully performed using formalin fixed paraffin embedded (FFPE) tissue.30,31 We recently developed a new method combining laser capture microdissection (LCM) and mass spectrometry that allows localized proteomics using microdissected FFPE tissue to more precisely evaluate protein differences in microscopic brain regions or pathological lesions rather than bulk tissue.30,31 We specifically optimized this technique to allow the use of FFPE tissue so that archived human tissue specimens collected at autopsy could be used. This is a particular advantage of our approach, as the vast majority of autopsy human tissue specimens are FFPE blocks, which are currently an underutilized, but exceptionally valuable resource for research.

Therefore, to advance our knowledge of proteomics that is common to human epilepsy syndromes, we applied this method to epilepsy and control human brains across three regions: CA13 of the hippocampus, the dentate gyrus, and the frontal cortex.

Materials and methods

Ethics statement

All studies were approved by the Institutional Review Board (IRB) at New York University (NYU). In all cases, written informed consent was obtained from the legal guardian or patient, and material used had ethical approval. All patient data and samples were deidentified and stored according to NIH guidelines.

Human brain tissue

All human tissue samples were collected at autopsy. Epilepsy and control cohorts were studied. The epilepsy cases (n = 14) were acquired through the North American SUDEP Registry (NASR) and included cases from multiple sites around the country whose cause of death (COD) was not SUDEP. The control cases (n = 14) were acquired through the NYU autopsy service. All control and epilepsy neuropathology reports were performed and finalized at the NYU Pathology Department. Histological evaluation of all cases was performed by neuropathologists and findings were classified using the international consensus criteria.2,13,32 Storage, as well as sample processing (sample collection, embedding, archival and sectioning) was performed for all control and epilepsy cases per the NYU Pathology Department protocol. For proteomics analyses, specimens were retrieved from the Center for Biospecimen Research and Development (CBRD). The NYU Experimental Pathology Research Laboratory sectioned all cases for LCM and CBRD sectioned all cases for immunohistological validation. Six cases did not have seizure frequency and ten cases seizure type information, which came from a population with health care disparities that were not under regular medical care and therefore had limited medical records. Regarding epilepsy syndrome, there was a broad representation of various epilepsies in the cases with known clinical information, the majority of which excluded known frontal lobe involvement. All cases were matched for sex and age (46 ± 14 years) to the best of our ability. Individual patient information [sex, age, COD, post-mortem interval (PMI) and seizure classification if applicable] is included in Tables 1 and 2.

Table 1.

Control case information

Case Age Sex COD PMI (hr) Significant neuropathology Brain region
1 55 M Cardiac arrest 64 HP, DG, FC
2 57 M Cardiac arrest 142 HP, DG, FC
3 56 F Cardiac arrest 46 HP, DG, FC
4 49 F Cardiac arrest 144 HP, DG, FC
5 59 F Cardiac arrest 57 HP, DG, FC
6 59 M Possible cardiac arrest 116 HP, DG, FC
7 57 M Cardiac arrest 91 HP, DG, FC
8 55 M Pulmonary fibrosis 57 Subacute, right temporal white matter microinfarct HP, DG, FC
9 59 M Cardiac arrest 60 HP, DG, FC
10 38 M Anoxic encephalopathy secondary to cardiac arrest 44 Acute and chronic hypoxic-ischaemic changes, diffuse HP, DG, FC
11 59 M Supraglottic squamous cell carcinoma <120 Ischaemic infarct, left frontal MCA/ACA HP, DG, FC
12 49 M Cardiac arrest 48 Meningeal fibrosis HP, DG, FC
13 50 M Possible cardiac arrest (contributing—acute gastroenteritis) 50 Hypoxic-ischaemic changes, diffuse HP, DG, FC
14 54 M Cardiac arrest 66 Hypoxic-ischaemic injury, acute, multifocal HP, DG, FC

COD = cause of death; PMI = post-mortem interval; h = hour; HP = hippocampus; DG = dentate gyrus; FC = frontal cortex; ACA = anterior cerebral artery; MCA = middle cerebral artery.

Table 2.

Epilepsy case information

Case Age Sex COD PMI (hr) Significant neuropathology Disease duration (yr) Seizure type ASDs at time of death Brain region
1 50 M Choking on foreign object 15 49.5 Focal none FC
2 34 F Pulmonary embolism 13 FCD-IIA 32 Focal none HP, DG, FC
3 45 M Suicide 27 DDG 20 Focal FBM, LAM, LOR, VPA HP, DG
4 36 M Drowning 48 HS 12 Focal PHT, LAM, CBZ, ESL HP, DG, FC
5 36 M Overdose/intoxication 20 8 ND PHT, LEV HP, DG, FC
6 54 M Accident/trauma <24 Mild gliosis, contusion, focal cortical disorganization 1 ND PHT HP, DG
7 64 F Overdose 18 ND ND PHT HP, DG
8 9 F Drowning 30 FCD-IIA 8 ND LAM, PHT HP, DG
9 45 M Suicide <48 43 ND CLB, GBP HP, DG, FC
10 24 F Drowning <48 DDG ND ND LEV, VPA, PER, CBZ, CLO, LOR HP, DG, FC
11 28 M Accident/trauma <48 DDG 22 ND CBZ, LAC HP, DG, FC
12 23 M Drowning <48 FCD-IA ND ND VPA, TPM, CLO HP, DG, FC
13 32 M Ethanol intoxication and clobazam overdose 19 FCD-IIA, Wernicke’s encephalopathy 10 ND LAM, LOR, PHT HP, DG, FC
14 49 M Aspiration 43 DDG, HS, gliosis, hemisphere atrophy 48.4 ND fPHT HP, DG, FC

COD = cause of death; PMI = post-mortem interval; h = hour; yr = year; HP = hippocampus; DG = dentate gyrus; FC = frontal cortex; FCD = focal cortical dysplasia; HS = hippocampal sclerosis; DDG = dysgenesis of the dentate gyrus; ND = not determined; ASD = anti-seizure drug; CBZ = carbamazepine; CLB = clobazam; CLO = clonazepam; ESL = eslicarbazepinel; FBM = felbamate; fPHT = fosphenytoin; GBP = gabapentin; LAC = lacosamide; LAM = lamotrigine; LEV = levetiracetam; LOR = lorazepam; PER = perampanel; PHT = phenytoin; TPM = topiramate; VPA = valproate.

Laser capture microdissection

We sampled brain tissue from FFPE blocks of hippocampus or superior frontal gyrus collected and processed for autopsy neuropathology. LCM was performed using our published protocol.30,31,33 Briefly, 8 µm-thick FFPE sections containing hippocampus or superior frontal gyrus were collected onto LCM PET Frame Slides (Leica). Sections were stained with Cresyl violet to visualize neurons and regions of interest (ROIs). For each sample, 10 mm2 of hippocampus (including CA13) and superior frontal gyrus (including layers I-IV) and 4 mm2 of dentate gyrus (granule cell layer) were microdissected in separate tubes using a LMD6500 microscope at 5× magnification (Leica) (Fig. 1). ROIs were collected into ultrapure Pierce Water, LC−MS Grade (ThermoScientific). After collection, samples were centrifuged at 14 000 g for 2 min and stored at −80°C until peptide extraction.

Figure 1.

Figure 1

Quantitative proteomics analysis of epilepsy and non-epilepsy control cases. (A) Experimental workflow. Specific protein regions were excised by laser capture microdissection (LCM) from FFPE autopsy brain tissue from control and epilepsy cases (n = 14 in each group) and proteins quantified by label-free quantitative mass spectrometry (MS). Statistical, pathway and network analyses were performed to identify proteins differing in these regions. Three regions were analysed [hippocampus CA13 (n = 14 control, n = 13 epilepsy), frontal cortex (n = 14 control, n = 11 epilepsy) and dentate gyrus (n = 14 control, n = 13 epilepsy)]. Validation of protein differences was performed using immunohistochemistry (IHC). (B) LC−MS/MS computational analysis. Proteins groups were filtered and quantified using MaxQuant. (C) Venn diagram of proteins identified in each region. A total of 3216 protein groups were detected across all brain regions by MS, with 2440 overlapping protein groups between the three regions and 2861 overlapping protein groups between the hippocampus and the frontal cortex.

Label-free quantitative mass spectrometry proteomics

The study included six experimental groups based on two states (epilepsy, control) and three brain regions. Samples were processed and analysed using label-free quantitative mass spectra (MS) in three batches: hippocampus (n = 13 epilepsy, n = 14 controls), frontal cortex (n = 11 epilepsy, n = 14 control) and dentate gyrus (n = 13 epilepsy, n = 14 control). The same epilepsy and control cases were used in the three batches. For each ROI analysed, samples were prepared for label-free quantitative MS.30,31 Samples were thawed at room temperature (RT) and resuspended by adding 50 μl of 100 mM ammonium bicarbonate and 20% acetonitrile solution to the cap and the tube spun down. This step was repeated three more times to ensure thorough cap washing. Samples were then incubated at 95°C for 1 h followed by a 65°C incubation for 2 h. Reduction was performed using dithiothreitol at 57°C for 1 h (2 µl of 0.2 M) followed by alkylation with iodoacetamide at RT in the dark for 45 min (2 µl of 0.5 M). The samples were subsequently digested with 300 ng of sequencing grade modified trypsin (Promega) overnight at RT with gentle shaking. After digestion samples were acidified with trifluoroacetic acid to a pH of 2 and peptides were desalted using POROS R2 C18 beads as previously described.30,31,33 The samples were resuspended in 0.5% acetic acid and stored at −80°C until further analysis. Aliquots of the sample were loaded onto an Acclaim PepMap 100 (75 µm × 2 cm) precolumn connected to a PepMap RSLC C18 (2 µm, 100 A × 50 cm) analytical column using the autosampler of an EASY-nLC 1200 (ThermoScientific). The samples were gradient eluted directly into an Orbitrap Fusion Lumos mass spectrometer (ThermoScientific) using a 165 min gradient (Solvent A consisting of 2% acetonitrile in 0.5% acetic acid and Solvent B of 80% acetonitrile in 0.5% acetic acid). High resolution full MS were acquired with a resolution of 240,000, an automatic gain control (AGC) target of 1e6, a maximum ion time of 50 ms, and a scan range of 400–1500 m/z. After each full MS, MS/MS HCD spectra were acquired in the ion trap. All MS/MS spectra were collected using the following: ion trap scan rate Rapid, AGC target of 2e4, maximum ion time of 18 ms, one microscan, 2 m/z isolation window, fixed first mass of 110 m/z and NCE of 32.

Proteomics computational analysis

MS data were analysed using MaxQuant34,35 software version 1.6.3.4 and searched against the SwissProt subset of the H. sapiens Uniprot database (http://www.uniprot.org/) containing 20,365 entries (February 2019 release) to which 248 common laboratory contaminants were added. The enzyme specificity was set to trypsin with a maximum number of missed cleavages set to 2. Peptide identification was performed with an initial precursor mass deviation of 7 ppm and a fragment mass deviation of 20 ppm with subsequent non-linear mass recalibration.36 Oxidation of methionine residues was searched as variable modification; carbamidomethylation of cysteines was searched as a fixed modification. The false discovery rate (FDR) for peptide, protein and site identification was set to 1% and was calculated using a decoy database approach. The minimum peptide length was set to 6. The option match between runs (1 min time tolerance) was enabled to correlate identification and quantitation results across different runs. Protein quantification was performed with built-in MaxQuant LFQ algorithm37 with the following settings: minimum ratio count of 2, ‘fastLFQ’ option enabled, minimum/average number of neighbours 3/6. LFQ normalization was performed within sample groups that belong to the same brain region.

For subsequent statistical analyses we limited our data set to proteins quantified in at least eight samples in at least one of the conditions/brain region (5772% of microdissected samples, depending on the region analysed). All common contaminant entries were removed, resulting in a matrix with 3216 protein groups. Data analysis used Perseus v. 1.6.2.3.38 (http://www.perseus-framework.org/) using R environment for statistical computing and graphics (http://www.r-project.org/). Protein interaction networks and enrichment analyses were done in STRING39,40 v. 11.0 (https://string-db.org/).

Statistical analyses and interpretation of results

For principal component analysis (PCA) missing values were imputed from a normal distribution with width 0.3 and downshift 1.8 using Perseus. Hierarchical clustering was used to visualize sample metadata in the context of a clustering of those proteins associated with disease (whether positively or negatively, with a 5% FDR), following a preliminary kmeans clustering using k = 2 and the Hartigan and Wong algorithm using the ComplexHeatmap package in the R environment.41 For the hierarchical clustering, Euclidean distance was used as the distance measure and complete-linkage was used to combine clusters. Two-sample Student’s t-test was used to find differentially expressed proteins between epilepsy and control groups (per each ROI). We corrected for multiple hypothesis testing by calculating the permutation-based FDR (n = 250). Only protein groups with FDR <5% were deemed significantly different between groups (Supplementary Tables 5–11). For each ROI, protein groups absent in one of the disease groups but present in at least eight cases in the other disease group were by definition considered as differentially expressed (i.e. protein group absent in all control cases but present in at least eight epilepsy cases in the hippocampus was considered as increased in epilepsy in the hippocampus). These significant protein groups are included in Supplementary Table 12.

Pathway and network analysis

The genes encoding protein groups significantly altered in epilepsy and control were analysed using Ingenuity Pathway Analysis (IPA, Qiagen). We analysed all differentially expressed proteins in the hippocampus (n = 777), frontal cortex (249) or dentate gyrus (n = 49). We also analysed differentially expressed proteins in both hippocampus and frontal cortex (n = 134) and differentially expressed proteins specific to hippocampus or frontal cortex. All pathway analyses are included in Supplementary Tables 16–21. Network analysis was performed using STRING (v.11). Visualization and analysis of the network was conducted via Cytoscape.

Cell-type specificity

For each ROI, genes encoding all significantly altered proteins in epilepsy and controls were assigned cell-type specificity derived from Lake et al.,42 in which each gene had only one cell type annotation from any brain region excluding the cerebellum with 1066 possible annotations. For each ROI, cell type enrichment analysis included the following annotations: neurons, excitatory and inhibitory neurons, astrocytes, endothelial, microglia, and oligodendrocytes. The ‘neurons’ annotation was given to proteins for which the gene in Lake et al. had both ‘excitatory neuron’ and ‘inhibitory neuron’ annotations, with no other cell type associated annotations. For the hippocampus, synaptic proteins were assigned based on the classification of ‘Synaptic protein’ in Lleo et al.43 Proteins with a neuron cell type annotation and assigned a synaptic protein were referred to as ‘neuro-synaptic’.

Immunohistochemistry

To validate proteomics data, we immunostained FFPE tissue sections as described.31 Sections were incubated overnight at 4°C with either anti-SYP (synaptic protein synaptophysin) antibody (1:50, abcam, catalogue #ab8049) or anti-TUJ1 antibody (1:300, Biolegend, catalogue #802001). Sections were then incubated for 2 h at RT with fluorescent secondary antibodies (all diluted 1:500, Jackson Immunoresearch) and coverslipped (ProLongDiamond Antifade Mountant, Invitrogen). All immunostained sections from one immunostaining combination were scanned at 20× magnification on a NanoZoomer HT2 (Hamamatsu) microscope using the same settings. Fiji was used to analyse images with the same threshold for area positive for pixels relative to total area and reported as a percentage. Statistical analyses used unpaired t-tests. Representative images were obtained by confocal imaging using a Zeiss 700 confocal microscope. For each immunostaining combination, z-stacked images were collected with the same settings at 20× and 63× magnification with a 13 and 11 μm z-step and are depicted as a maximum projection image.

Data availability

The mass spectrometry raw files are accessible under MassIVE ID: MSV000085517.

Results

Quantitative proteomic analysis of epilepsy and non-epilepsy control cases

To identify proteomic alterations as a result of various aetiologies and seizures in the epilepsy brain, regions of the hippocampus (CA13 region) and frontal cortex were microdissected from all epilepsy and control cases (Fig. 1A). Cases were matched for sex, age (46 ± 14 years) and formalin archival time (2 ± 1 years) to the best of our ability (Tables 1 and 2). Histological evaluation of all epilepsy cases identified FCD in 4/14 cases, dysgenesis of the dentate gyrus (DDG) in 4/14 cases and hippocampal sclerosis (HS) in 2/14 cases (Tables 1 and 2). Control cases had no significant neuropathology (Tables 1 and 2). Regarding antiepileptic drug treatment, 12 epilepsy cases were prescribed at least 1 ASD (Tables 1 and 2). Regarding ASD adherence, 2/12 cases were known to be taking medications as prescribed, while 3/12 were unknown, and 7/12 were non-adherent for at least one ASD.

Given the role of the dentate gyrus in epileptogenesis and epileptic neuropathology,44,45 we microdissected and analysed the dentate gyrus proteome separately from CA13. Given the reduced size of the dentate gyrus, these samples were smaller than hippocampus and frontal cortex samples (4 mm2 versus 10 mm2; Fig. 1A) and may explain the lower number of protein groups identified and higher coefficient of variation in dentate samples.

Combined, we analysed 79 samples from 28 cases using label-free quantitative MS and identified 59 903 peptides mapping to 4,894 protein groups (Fig. 1B). Additional filtering based on a minimum of 8 cases with quantification per group in an ROI resulted in 3,216 proteins groups being included in our analyses: frontal cortex—3014, hippocampus—2989 and dentate gyrus—2652 protein groups. The proteins identified overlapped extensively across regions (Fig. 1C).

Proteomic differences in epilepsy and control cases in hippocampus (CA1−3) and frontal cortex

Protein expression differences between epilepsy and controls were identified in the hippocampus and frontal cortex. PCA showed a striking separation of epilepsy and control groups in hippocampus (permutation-based test, P = 2.1×10−5, Fig. 2A), and to a lesser extent, in frontal cortex (P = 0.002; Fig. 2A) in PC1, confirming that the hippocampus is a more impacted region than the frontal cortex. Presence of FCD, DDG and HS did not account for this separation (Fig. 2A). Supervised hierarchical clustering revealed robust clustering for all hippocampi and frontal cortices from epilepsy cases, consistent with the PCA (Fig. 2B). Variables such as age, sex and neuropathological findings did not drive the clustering (Fig. 2B). Only age of epilepsy onset separated different epilepsy cases in the heatmap: cases with an age of onset below or above 20 years old clustered separately. However, several cases did not have comprehensive clinical information. Those cases came from a population with health care disparities that were not under regular medical care and therefore had limited medical records.

Figure 2.

Figure 2

Proteomic differences in epilepsy and control cases in hippocampus (CA1−3) and frontal cortex. (A) PCAs of overall data set for each region. PCAs of epilepsy (purple) and control (grey) cases in the hippocampus (left panel) and frontal cortex (right panel) showing that epilepsy hippocampus (P = 5.1×10−5) and frontal cortex (P = 0.002782) contained significantly different protein expression. Each point represents an individual case. Neuropathology (DDG, Dysgenesis of the Dentate Gyrus, FCD; Focal Cortical Dysplasia; HS, Hippocampal Sclerosis) is indicated on the PCA. Significance was determined using a non-parametric permutation-based t-test. Error bars represent PC1 min and max. (B) Supervised hierarchical clustering. Dendrogram showing hierarchical clustering of brain samples based on protein expression levels by region (left panel, hippocampus; right panel, frontal cortex). Information on diagnosis, sex, age, age of disease onset (OnsetAge), disease duration (Duration), seizure type (SeizureType), and frequency of lifetime seizures (SeizureFreq) is indicated according to the legend at the bottom of the graph. Heatmaps on the bottom show scaled (protein z-score) expression values (colour-coded according to the legend on the right) for proteins used for clustering.

Differentially expressed proteins between epilepsy and controls were determined using Student’s t-test and Permutation based 5% FDR. There were significant alterations in the expression of 777 proteins in the hippocampus and 296 proteins in the frontal cortex (Fig. 3A and B; Supplementary Tables 7 and 9). A total of 134 proteins were significantly altered in both regions (Supplementary Table 13). The top 20 most significantly altered proteins in epilepsy by region are presented in Tables 3 and 4. Of the 40 top altered proteins, 30 (75%) have been associated with epilepsy in humans or animal models (Tables 3 and 4; Supplementary Tables 1 and 2). We observed that several altered proteins were encoded by genes in which mutations cause epilepsy,7 including proteins SCN2A, SCN8A, STXBP1, GABBR1, GABBR2, GABRA1, SLC25A12, SLC25A22, SLC6A1 and SCARB2 (Fig. 3C).

Figure 3.

Figure 3

Hippocampal and cortical differences in the epilepsy proteome. Volcano plots show protein expression differences of all proteins identified in the hippocampus (A) and frontal cortex (B). All colour-coded proteins had significantly altered expression in epilepsy and are indicated above the designated line showing 5% FDR. Proteins were assigned to a specific cell-type (Neu, Neuron [blue]; In, Inhibitory Neuron [dark green]; Ex, Excitatory Neuron [light green]; Oli, Oligodendroglia [yellow]; Ast, Astrocyte [orange]; Mic, Microglia [red]; End, Endothelial cell [brown]). Significance was determined using a Student’s t-test with Permutation based false discovery rate (FDR) set to 5% (exact P-values for each protein group are provided in Supplementary Table 5). Proteins with higher expression in epilepsy are on the right of the plot and proteins with lower expression are on the left. The 20 top most significantly altered proteins are identified by gene abbreviations. (C) Proteins of interest with significantly altered levels in epilepsy in hippocampus and frontal cortex. Individual points show protein expression in each individual case. Data show mean ± SEM; ****P < 0.0001; ***P < 0.001; **P < 0.01. HIPP, hippocampus; FC, frontal cortex.

Table 3.

Top 20 significant proteins in the hippocampal CA1 − 3 region of epilepsy cases

Gene ID Protein Name UniProt ID P Value Fold Change Human Animal Model References
Decreased
ATP6V1A V-type proton ATPase catalytic subunit A P38606 1.202E−10 1.591 46–49
GNB2 Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-2 P62879 2.951E−10 1.853 49
ATP6V1C1 V-type proton ATPase subunit C 1 P21283 6.918E−10 1.840 49
GNAI2 Guanine nucleotide-binding protein G(i) subunit alpha-2 P04899 1.622E−09 1.569 50 , 49
ACTC1 Actin, alpha cardiac muscle 1 P68032 5.754E−09 2.990
GNB1 Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1 P62873 8.128E−09 1.959 7 , 50 , 51 , 52
NAP1L4 Nucleosome assembly protein 1-like 4 Q99733 9.333E−09 1.840 50 , 49
TUBB4B Tubulin beta-4B chain P68371 1.023E−08 1.682
ABI1 Abl interactor 1 Q8IZP0 1.288E−08 1.464 49
GLO1 Lactoylglutathione lyase Q04760 1.66E−08 1.485 49
COX6C Cytochrome c oxidase subunit 6 C P09669 2.399E−08 2.000
COX6B1 Cytochrome c oxidase subunit VIb isoform 1 P14854 2.512E−08 2.949 7 , 50 , 49
SLC25A4 ADP/ATP translocase 1 P12235 2.692E−08 2.479 50 , 49
SYP Synaptophysin P08247 3.236E−08 2.428 7 , 50 , 49 , 53–55
NDUFB9 NADH dehydrogenase [ubiquinone] 1 beta subcomplex subunit 9 Q9Y6M9 3.467E−08 1.790 50 , 49
PPP2R1A Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A alpha isoform P30153 3.548E−08 1.558 7 , 48
DNM1 Dynamin-1 Q05193 4.898E−08 1.385 7 , 49 , 56 , 57
Increased
NCL Nucleolin P19338 2.692E−10 1.828 50 , 58 , 59
RPS11 40S ribosomal protein S11 P62280 6.026E−09 1.959
SLC16A2 Monocarboxylate transporter 8 P36021 2.188E−08 2.732 50 , 49

Table 4.

Top 20 significant proteins in the frontal cortex of epilepsy cases

Gene ID Protein Name UniProt ID P Value Fold change Human Animal model References
Decreased
LANCL1 Glutathione S-transferase LANCL1 O43813 9.12E−08 1.464 49
MBP Myelin basic protein P02686 1.445E−06 3.891 49 , 50 , 60 , 61
FBXO2 F-box only protein 2 Q9UK22 6.457E−06 1.366 48 50
GLUL Glutamine synthetase P15104 1.288E−05 1.485 7,48,62,64
GNB1 Guanine nucleotide-binding protein G(I)/G(S)/G(T) subunit beta-1 P62873 1.549E−05 1.753 7,50,52
COX6B1 Cytochrome c oxidase subunit 6B1 P14854 2.042E−05 2.085 7 , 49 , 50
SLC25A6 ADP/ATP translocase 3 P12236 2.951E−05 1.636
SLC25A4 ADP/ATP translocase 1 P12235 3.311E−05 2.114 49 , 50
TPP1 Tripeptidyl-peptidase 1 O14773 3.802E−05 1.741 7 , 49 , 50
ASAH1 Acid ceramidase Q13510 4.169E−05 1.853 7 , 48–50 , 65
Increased
PRR36 Proline-rich protein 36 Q9H6K5 3.548E−07 1.919
GRM2 Metabotropic glutamate receptor 2 Q14416 1.778E−06 1.580 66–69
GPHN Gephyrin Q9NQX3 5.888E−06 1.569 7 , 70–73
GRIN1 Glutamate receptor ionotropic, NMDA 1 Q05586 7.943E−06 1.580 7 , 49 , 74
PSMC2 26S proteasome regulatory subunit 7 P35998 9.333E−06 1.347
RTN4RL2 Reticulon-4 receptor-like 2 Q7M6Z0 1.66E−05 1.516
KIF3A Kinesin-like protein KIF3A Q9Y496 1.862E−05 1.526 50
NRXN3 Neurexin-3-beta Q9HDB5 3.311E−05 1.366 75
ARHGAP23 Rho GTPase Activating Protein 23 Q9P227 3.981E−05 1.444 49
PSMC5 26S proteasome regulatory subunit 8 P62195 4.571E−05 1.257 50

We sought to determine whether protein differences in epilepsy were cell type specific. Of the 777 altered proteins in the epilepsy hippocampus, neuronal proteins were significantly enriched (Fisher’s exact test, P = 6.49×10−7), particularly excitatory neuron proteins (P = 7.128×10−6) (Fig. 4A). There was no enrichment of inhibitory neuronal, oligodendroglial, astrocytic, microglial or endothelial proteins (Fig. 4A). These enrichments were not observed in the frontal cortex (Fig. 4B), suggesting more extensive protein alterations in hippocampal neurons. All neuron-specific proteins (‘neuron’, ‘inhibitory neuron’ or ‘excitatory neuron’ proteins) were predominantly decreased in the epileptic hippocampus compared with controls (Fisher’s exact test, P = 0.0013, Fig. 4C).

Figure 4.

Figure 4

Cell-type enrichment and pathway analysis of differentially expressed proteins in the hippocampus and frontal cortex in epilepsy. For each brain region [Hippocampus (A), frontal cortex (B)], cell-type enrichment analysis was performed for each cell type using a Fisher’s exact test. Significance threshold is represented by a dotted red line (P < 0.05); ****P < 0.0001. (C) Cell-type-specific protein expression differences in epilepsy. Each graph plots all significant proteins quantified in the epilepsy hippocampus (top panels) or frontal cortex (bottom panels). The further the point is away from the dashed line at 0, the greater the difference in expression in epilepsy and controls. Proteins with a positive log ratio have greater expression in epilepsy, and proteins with a negative log ratio have a lower expression in epilepsy. All color-coded proteins correspond to neuronal proteins (Neuron, blue; Inhibitory Neuron, dark green; Excitatory Neuron, dark green), oligodendrocyte proteins (yellow) and astrocyte proteins (orange). The cumulative cell type specific protein differences showed that there were significantly less neuronal proteins in the hippocampus in epilepsy (P = 0.001) but not in the frontal cortex (P = 0.371). No differences in the amount of oligodendroglial (hippocampus, P = 0.096; frontal cortex, P = 1), astrocytic (hippocampus, P = 0.565; frontal cortex, P = 0.108), microglial (not shown) or endothelial proteins (data not shown) were observed. Significance was determined using a Fisher’s exact test comparing cumulative cell type specific protein expression between epilepsy and control cases. Pathway analysis of the altered proteome in the hippocampus (D) and frontal cortex (E). IPA was used to identify the top 10 signalling pathways associated with the genes encoding protein groups that were significantly altered in both the hippocampus (pink) and the frontal cortex (blue). The red line corresponds to FDR corrected P-value thresholds (hippocampus, P = 0.0001; frontal cortex, P = 0.001). All pathways and proteins are shown in Supplementary Tables 16 and 15.

Pathway analysis showed that altered hippocampal and frontal cortex proteins were significantly associated with four pathways: protein biogenesis, mitochondrial function, synaptic plasticity and G-protein signalling (Fig. 4D and E; Supplementary Tables 18 and 19). Proteins associated with EIF2 signalling were particularly enriched. Most (37/53) were ribosomal proteins and 96.9% were upregulated (19/19 in frontal cortex, 31/33 in the hippocampus), indicating increased translational machinery and ribosome biogenesis. Other proteins involved in this pathway included translation initiation factors (EIF2AK2, EIF3E, EIF3J, EIF4E, EIF4G1) and protein kinases (MAPK1, MAPK3). The mTOR signalling pathway, as well as the regulation of eIF4 and p70S6K signalling pathway—which is part of the mTORC1-S6K signalling axis—were also significantly represented, suggesting that mTOR pathway may drive this increase in proteins related to protein synthesis. The mTOR signalling pathway is involved in protein synthesis, synaptic plasticity, autophagy and other functions which may influence neuronal excitability and epileptogenesis.76,77 Oxidative phosphorylation proteins were also enriched in both regions, with broader evidence of altered mitochondrial function including pathways such as mitochondrial dysfunction, sirtuin signalling, glycolysis and TCA cycle, highlighting impaired neuronal energy regulation in epilepsy. The synaptogenesis signalling pathway was also significantly enriched, confirming synaptic dysfunction resulting from epilepsy. Finally, G-protein signalling was also significantly represented in both regions (G Beta Gamma Signalling/Ephrin B signalling; Fig. 4D and E; Supplementary Tables 16 and 17). Notably, we observed a unique enrichment of glutamate receptor signalling pathway proteins in the frontal cortex and in the remodelling of epithelial adherens junction pathway in the hippocampus. Combined, these results suggest that although changes were more prominent in the hippocampus, similar pathways were enriched in frontal cortex, identifying widespread brain protein alterations in epilepsy.

Proteomic differences in epilepsy and control cases in the dentate gyrus

Proteomic analysis of the dentate gyrus in epilepsy was limited by smaller tissue area and increased variation. PCA only showed a modest separation between epilepsy and control cases (P = 0.246) in the dentate gyrus (Supplementary Fig. 1A). However, quantification of individual proteins showed that 49 proteins were significantly altered in the dentate gyrus in epilepsy (Supplementary Fig. 1B and Table 11). Notably, 24 proteins were uniquely altered in the dentate gyrus, including 2 identified only in dentate gyrus (CPSF1, FAM120C) and 2 identified only in dentate gyrus and frontal cortex (CDC5L, AMER2) (Supplementary Table 5). Hierarchical clustering induced by pre-selection of these differently expressed proteins showed that 10/13 epilepsy cases clustered, supporting dentate gyrus dysfunction in epilepsy (Supplementary Fig. 1C). Supplementary Table 3 lists the 20 most significantly altered proteins; 16 (80%) are associated with epilepsy in humans or animal models, while 4 (20%) were novel (AMER2, ATP5BP, RPL15, TPI1), adding to the list of potential new epilepsy-associated proteins (Tables 3 and 4; Supplementary Tables 1 and 2). The G-Protein Subunit Beta 1 (GNB1) was the only protein to rank among the top 20 significantly altered proteins in all regions (hippocampus, P = 8.19×10−9; frontal cortex; P = 1.55×10−5, dentate gyrus; P = 6.96×10−6; Supplementary Fig. 1D). Despite the epileptic encephalopathies that GNB1 mutations can cause,78 the underlying disease mechanisms are still largely unknown and the specific role of GNB1 in epileptogenesis has not yet been studied. Pathway analysis of altered proteins in the dentate gyrus showed the most enrichment in proteins involved in mitochondrial dysfunction, revealing a common epilepsy proteomic signature in all regions (Supplementary Fig. 1E and Table 18).

Inter-regional epilepsy proteomic signature

The common enriched pathways in the hippocampus and frontal cortex (Fig. 4D and E), as well as the overlapping altered proteins (Fig. 5A), indicate shared protein changes across both regions. We performed correlation analyses of the directional changes in hippocampus and frontal cortex for the (i) 939 significantly altered proteins and (ii) 134 common proteins altered in both regions (Fig. 5B and C; Supplementary Table 13). We found a modest positive correlation between protein levels in the hippocampus and frontal cortex when all significant proteins were examined (Pearson’s correlation, r2=0.32, Fig. 5B), suggesting that the hippocampus was more severely and/or differently impacted compared with the frontal cortex. The correlation improved when analysing the changes of the 134 significantly different proteins shared by both the hippocampus and frontal cortex (r2=0.73, Fig. 5C), confirming broad similar protein alterations throughout the epileptic brain. Interestingly, this correlation was also present in the dentate gyrus (hippocampus/dentate gyrus, r2=0.64; frontal cortex/dentate gyrus, r2=0.72, Fig. 5D and E), despite only 13/134 of these proteins being significantly altered in the dentate gyrus (Fig. 5A, D and E; Supplementary Table 13), supporting an epilepsy-associated proteomic signature. Hierarchical clustering of these specific proteins shows almost complete clustering of all epilepsy cases throughout the three regions analysed (Fig. 5F). These 134 enriched proteins relate to EIF2 and mTOR signalling pathways, mitochondrial dysfunction and other related metabolic pathways such as oxidative phosphorylation, glycolysis, TCA cycle and gluconeogenesis (Fig. 6A and B; Supplementary Table 19). Proteins involved in G-protein signalling were also significantly enriched (Ephrin B signalling; Fig. 6A and B; Supplementary Table 19).

Figure 5.

Figure 5

Inter-regional epilepsy proteomic signature. (A) Venn diagram of significant proteins in each region. The Venn diagram shows the 963 altered proteins in epilepsy across all regions analysed. A total of 939 proteins were altered in hippocampus and frontal cortex altogether. There were 134 overlapping proteins between the hippocampus and the frontal cortex (purple), and 13 overlapping proteins across the three regions (dark blue). (B−E) Correlation between the protein fold changes in the hippocampus and the frontal cortex. Graphs represent correlations using the 939 significantly altered proteins (top left panel, Pearson’s correlation, r2=0.32), and the overlapping 134 proteins altered in both hippocampus and frontal cortex [top right panel, Pearson’s correlation (r2=0.73)]. The proteomic signature was also preserved in the dentate gyrus, despite only 13/134 proteins being significant [bottom panels, Pearson’s correlation; hippocampus/dentate gyrus (bottom left panel), r2=0.64; frontal cortex/dentate gyrus (bottom right panel), r2=0.72]. (F) Hierarchical clustering of the 134 proteins showing similar directional changes in hippocampus CA13, frontal cortex and dentate gyrus. The dendrogram shows hierarchical clustering of brain samples based on protein expression levels by region. The heatmap on the bottom show scaled (protein z-score) expression values (color-coded according to the legend on the right) for proteins used for clustering.

Figure 6.

Figure 6

Pathway and network analysis of the epilepsy proteomic signature. (A) Pathway analysis of the epilepsy proteomic signature. IPA was performed on genes encoding for protein groups significantly altered in the proteomic signature. The red line corresponds to FDR corrected P-value threshold (P = 0.05). (B) Epilepsy proteomic signature protein network. The network depicts altered proteins in both hippocampus and frontal cortex. Visualization and analysis of the network was conducted via Cytoscape. Color-coding represents directional changes (upregulated shown in red, downregulated shown in blue). Node sizes correspond to degree of interaction. Disconnected nodes in the network were removed. Black rectangles correspond to proteins involved in pathways of interest.

Together, these results support a broad impact of epilepsy on mitochondrial function, protein biogenesis and G-protein signalling. Notably, synaptic plasticity-related signalling pathways were not among shared significant proteins changes across hippocampus and frontal cortex, suggesting that the synaptic-associated mechanisms differ between these regions.

Intra-regional epilepsy proteomic changes

We found that 635 proteins reached significance in the hippocampus and not in the frontal cortex. These highly significant hippocampal differences were enriched in proteins involved in the remodelling of epithelial junction (P = 7.71×10−18), with prominent involvement of cytoskeleton-associated proteins (actin-related [ACTC1, ACTG1, ACTN1, ACTR2, ACTR3, ARPC4, ARPC1A], dynamins [DNM1, DNM2. DNM3, DNM1L] and tubulins [TUBA1A, TUBA1B, TUBA4A, TUBB3, TUBB, TUBB2A, TUBB4A]) and clathrin-mediated endocytosis signalling (P = 7.92×10−16), suggesting a reorganization of the cortical microarchitecture and a remodelling of neuronal circuitries, or an impact on the brain vasculature (Supplementary Table 20). A significant alteration in synaptogenesis signalling pathway proteins was also observed (P = 6.26×10−14; Supplementary Table 20). These findings, together with the significant decrease of neuronal proteins observed previously (Fig. 4C) suggest neuronal synaptic dysfunction in the hippocampus. Neuronal loss is another possible explanation but was not seen histologically. Using a publicly available data set of the most updated synaptic proteome,43 we confirmed that the significant proteins in the hippocampus were highly enriched in neuro-synaptic proteins (33/63 neuro-synaptic proteins, Fisher’s exact test, P = 3.939×10−6; Supplementary Table 7). The expression of significant synaptic proteins in epilepsy was dramatically decreased (55/58 proteins, Fisher’s exact test, 8.133×10−10, Fig. 7A), suggesting extensive synaptic alteration in the hippocampus. These proteomic changes were not significant in the frontal cortex (Fisher’s exact test, P = 0.089; Fig. 7B; Supplementary Table 7). We validated the decrease of SYP in the epileptic hippocampus using IHC (Fig. 7C, E and G). To determine whether these protein changes were unique to this region or simply less pronounced in the frontal cortex than in the hippocampus, we examined the directional changes of the significantly altered hippocampal proteins and their respective directional change in the frontal cortex. When we closely examined these differences, we discovered that the vast majority of these proteins were showing trends for similar changes in the frontal cortex (462/635 proteins in agreement [72%], Fisher’s exact test, P < 2.2×10−16, Fig. 7I), suggesting that overall the same pathological protein changes were happening in the frontal cortex, but not to the same extent. IPA confirmed that the proteins that were in agreement were involved in the remodelling of epithelial adherens junctions (P = 2.14×10−12), and synaptogenesis (P = 1.52×10−10). The significantly decreased SYP immunoreactivity in the frontal cortex also supported this observation (Fig. 7D, F and H).

Figure 7.

Figure 7

Altered synaptic transmission in epilepsy. (A) The graph depicts all significant proteins identified in the hippocampus. The further the point is away from the dashed line at 0, the greater the difference in expression in epilepsy and controls. Proteins with a positive log ratio have greater expression in epilepsy and proteins with a negative log ratio have a greater expression in controls. All proteins highlighted in green correspond to synaptic proteins. (B) The graph shows all significant proteins identified in the frontal cortex. All proteins highlighted in green correspond to synaptic proteins. (C) Synaptophysin (SYP) and Tubulin-β 3 (TUJ1) double fluorescent immunohistochemistry of n = 5 epilepsy and n = 5 control cases in the hippocampus showed a major decrease in SYP immunoreactivity in the epileptic hippocampus [panels show CA1 region, images taken at ×20 (top panels) and ×63 magnification (bottom panels)]. (D) SYP and TUJ1 double fluorescent immunohistochemistry of n = 5 epilepsy and n = 5 control cases in the frontal cortex also showing major decrease in SYP immunoreactivity in the epileptic cortex [images taken at ×20 (top panels) and ×63 magnification (bottom panels)]. (E−H) Quantification of SYP immunohistochemistry (E−G) and proteomic expression levels (F−H) in the hippocampus CA13 (E, F) and frontal cortex (G, H) (n = 5/group for IHC and n = 14/group for proteomics) epilepsy. For IHC quantification, individual points show SYP % load in each individual case (E−G). Significance was determined using a two-tailed unpaired t-test. Data show mean ± SD; **P < 0.01. For proteomics, individual points show protein expression in each individual case (F−H). Data show mean ± SEM; ****P < 0.0001. HIPP, hippocampus; FC, frontal cortex. (I, J) Topographical changes in epilepsy support the idea of selective vulnerability in hippocampus. The left panel shows a contingency table of the protein directional changes between epilepsy and control in the hippocampus and frontal cortex using the significant proteins in hippocampus only. The right panel shows a contingency table of the protein directional changes between epilepsy and control in the hippocampus and frontal cortex using the significant proteins in frontal cortex only. Proteins were annotated as either increased or decreased in epilepsy. Only proteins present in both regions were included in the analysis. Significance was determined using Fisher’s exact test.

Similarly, we found that 156 proteins reached significance in the frontal cortex and not in the hippocampus. While these frontal cortex protein changes were also suggestive of altered synaptogenesis, these changes were mostly enriched in proteins involved in increased glutamate receptor signalling (8/57 proteins, P = 2.61×10−7, z-score = 2.236; Supplementary Table 21), which occurs in glutamatergic synapses, and plays a critical role in excitotoxic damage in epilepsy.79 Proteins in this pathway included protein Glutamine Synthetase (GLUL), Excitatory amino acid transporters 1 and 2 (SLC1A2, SLC1A3), Glutamate receptor ionotropic NMDA 1 and 2B (GRIN1, GRIN2B), Glutamate receptor ionotropic AMPA 2 (GRIA2) and Glutamate metabotropic receptor 2 (GRM2). Interestingly, GLUL downregulation was associated with reduced astrocyte protection against glutamate excitotoxicity to neurons.80 Here again, we found that the majority of these significant changes which seemed specific to the frontal cortex were showing trends for similar directional changes in the hippocampus (110/156 proteins in agreement [72%], P = 1.753×10−7, Fig. 7J), suggesting that most of these changes were also occurring in the hippocampus, yet to a lesser extent. IPA confirmed that the proteins in agreement were most enriched in the glutamatergic signalling pathway (P = 7.58×10−5).

Combined, our results indicate that 696/939 (74%) significant proteins had similar directional changes in both the hippocampus and frontal cortex in epilepsy (Supplementary Table 21). These findings suggest that most pathological protein changes in epilepsy occurred in both regions but to a different degree, supporting greater vulnerability or a primary role of the hippocampus compared with the frontal cortex.

Discussion

In this study, we have generated the first comprehensive characterization of the human epileptic proteome. We identified extensive protein alterations common to various epilepsy syndromes in multiple brain regions. The epileptic hippocampus and frontal cortex showed dysregulation in pathways associated with mitochondrial function, protein synthesis, synaptic transmission and remodelling of the neuronal network architecture. These protein alterations were present despite the lack of major neurodegeneration. Our proteomics analysis provides a network-level analysis of protein changes in epilepsy and identifies hundreds of new potential drug targets. Given the paucity of comprehensive proteomics studies, it was difficult to cross-validate our findings. However, Wang et al. presented the most updated summary of the genes associated with epilepsy, grouping these genes according to the manifestation of epilepsy in specific phenotypes. We compared our list of 939 altered proteins (hippocampus and frontal cortex) with this list and found that 20/939 proteins were encoded by genes in which mutations cause epilepsy.7 Importantly, 98/939 proteins were encoded by neurodevelopment-associated epilepsy genes, epilepsy-related genes, or potentially epilepsy-associated genes. In addition, we compared the top 20 most significant proteins in each region with the few other human and animal studies available. While there were some differences compared with previous literature, the majority of the proteins in Supplementary Tables 1–3 had similar directional changes to those previously reported. However, some of these discrepancies may be related to the evaluation of various epilepsies, brain regions and techniques. Our results also suggest that the G-protein heterotrimeric complexes may have a particularly important role in epilepsy and specifically highlighted the widespread decrease in GNB1 expression levels in epilepsy.

Our results showed that the hippocampus was particularly vulnerable in epilepsy and suggested that protein changes in epilepsy may progressively extend through the brain in a topographic manner. The increased vulnerability of the hippocampus was evident in the broad range of epilepsy syndromes included in this study. The hippocampus is an epileptogenic brain region that is highly susceptible to damage from seizures. As a result, hippocampal sclerosis is a common neuropathological feature of epilepsy and its incidence varies from 30 to 45% of all epilepsy syndromes and 56% of MTLE neuropathologically.81 Hippocampal sclerosis is the most common pathology associated with drug-resistant epilepsy in older adults.82 Our cohort only included two cases with hippocampal sclerosis, which alone was not enough to explain such extensive hippocampal neuronal alterations. In contrast to earlier studies, we included a heterogeneous case cohort to identify protein alterations across a spectrum of epilepsy syndromes. Our study suggests that hippocampal vulnerability is a consistent feature across all types of epilepsy. It remains unknown why the hippocampus is so vulnerable in epilepsy as well as in ischaemia, limbic encephalitis, multiple sclerosis, and neurodegenerative diseases such as Alzheimer’s disease.83–87 The cognitive deficits in temporal lobe epilepsy are associated with Alzheimer-like amyloid β and tau protein changes.88 Hippocampus vulnerability in epilepsy and other neurological disorders may result from a low threshold for glutamatergic excitotoxicity or for synaptic remodelling. Some studies show vulnerability correlates with increased neuroplasticity, which could reflect different energy production mechanisms and requirements, making it highly sensitive to metabolic and oxidative stress, which can occur in epilepsy.89

Our results suggest that epilepsy affects brain areas in parallel yet to a different degree, supporting that epilepsy may be a progressive disease.90 Hippocampal protein alterations reflected pathologic changes of degeneration, such as neuronal and synaptic remodelling, which were also observed in the frontal cortex without reaching a significant threshold of 5% FDR (Supplementary Fig. 2). While decreased synaptophysin immunoreactivity was found in temporal lobe epilepsy-associated HS,91 our study is the first to report significantly decreased synaptophysin levels in epilepsy cases in the frontal cortex and hippocampus without hippocampal sclerosis. In contrast, frontal cortex protein changes seemed to mainly reflect earlier pathological changes such as an altered glutamatergic system, with non-significant but similar directional changes in the hippocampus. However, both regions showed mitochondrial dysfunction and altered protein synthesis, although these changes were more prominent in the hippocampus (Supplementary Fig. 2). The shared regional changes in mitochondrial function and protein biosynthesis, as well as the more localized frontal changes in the glutamatergic system suggest that these may be relatively earlier events in the disease process. The lack of significant glutamatergic changes in the hippocampus may reflect the statistical noise of the large protein changes, or that as the disease progresses, these neurons may be selectively lost or compensatory changes wash out any effect. Glutamatergic hyperactivity may be an early insult, driving or resulting from epileptic activity, and leading to oxidative stress and mitochondrial dysfunction; although, this remains unproven. Mitochondrial dysfunction lowers intracellular levels of ATP and alters calcium homeostasis, and these two pathways may be linked in a feed-forward pathologic cycle.92 Whether mitochondrial dysfunction is a cause or a consequence of epilepsy—or both—our study shows it is a widespread change in the epilepsy brain. Many metabolic and mitochondrial disorders cause seizures and disorders such as myoclonic epilepsy with ragged red fibres.93,94 Seizures occur in 35–60% of individuals with primary mitochondrial diseases.95 Mitochondrial oxidative stress occurs in temporal lobe epilepsy,96–98 but it remains uncertain if this is part of epileptogenesis or a secondary effect of seizures, ASDs or other factors. Mitochondrial functions can affect neuronal hyperexcitability via generation of ATP, metabolite/neurotransmitter biosynthesis, calcium homeostasis, reactive oxygen species production and apoptosis. Our study suggests that mitochondrial dysfunction may both contribute to and result from epilepsy. Our study also identified significantly increased protein biosynthesis in epilepsy. While this increase could be a result of loss of neuronal homeostasis due to mitochondrial dysfunction,99 increased protein synthesis may be driven by altered mTOR signalling, which was a prominent finding in our results. mTOR signalling pathways are implicated in a number of epileptogenic processes.100 Our results suggest increased ribosome biogenesis and protein production in chronic epilepsy. Protein synthesis may shift towards greater production of both pro-epileptic factors and compensatory anti-epileptic factors.101 Also, dysregulated mTOR signalling, part of the proteomic signature of epilepsy, may stimulate excessive synthesis of ion channels and receptors, leading to hyperexcitability.102

Our findings identified significant G-protein subunit alterations in epilepsy (Supplementary Fig. 2). Specifically, we detected 20 protein G-subunits in our study, with 13 of them being differentially expressed in at least one region (GNG7, GNAO1, GNG2, GNG4, GNB1, GNB2, GNAI1, GNAI2, GNAQ, GNAZ, GNB4, GNA11 and GNL1). Heterotrimeric G-proteins consist of two functional units, an α-subunit (Gα) and a tightly associated βγ complex. After activation, GPCRs change their conformation and transduce this into intracellular signals involved in diverse signalling pathways including the cAMP/PKA pathway, calcium/protein kinase C (PKC) pathway, IP3/DAG/phospholipase C pathway, β-arrestin pathway, protein tyrosine kinase pathway, ERK/MAPK pathway, PI3K/AKT pathway, Rho pathway and G-protein-gated calcium channels inwardly rectifying potassium channels.103 Animal studies suggest that GPCRs are important mediators of neuronal excitability, supporting a potential therapeutic role in epilepsy.104 However, no FDA-approved ASD suppresses epileptic seizures by directly acting on GPCRs. GNB1 may be central to GPCR dysfunction in epilepsy.104 We found that one G-protein subunit, GNB1, was significantly decreased in all the regions. Missense, splice-site and frameshift variants in the GNB1 gene are associated with multiple neurological phenotypes, including seizures in most cases.51,52,105–108 Mice harbouring the GNB1 K78R human pathogenic recapitulate many clinical features, including developmental delay, motor and cognitive deficits, and absence-like generalized seizures, and that cellular models displayed extended bursts of firing followed by extensive recovery periods.109 Given the therapeutic potential of targeting G-proteins, and the novelty of our finding, it would be interesting to further investigate the role of GNB1 in epilepsy. The involvement of GNB1 variants in human epilepsies and our data suggest this gene may play an important role in many epilepsies. Among the 134 currently available drugs targeting GPCRs,110 there are several drugs targeting GPCRs involved in pathways significantly impacted in our epilepsy data set. GNB1 is involved in 28 pathways that are significantly impacted in the hippocampus of epilepsy cases (27 inhibited, 1 activated), and 15 of these pathways include GPCRs with an FDA approved drug for non-epilepsy related indications. Future studies should evaluate whether any of these drugs or novel derivatives may be useful in ameliorating seizures in specific epilepsy syndromes. This may especially be of interest for the CREB signalling in neurons and endocannabinoid developing neuron pathways that were significantly decreased in our data set, as well as drugs targeting GPCRs that have been specifically identified as affected in some epilepsy syndromes like ADRA2 in the adrenergic signalling pathway7 in addition to glutamatergic and GABAergic targets.

Our study provides a rich resource for epilepsy research, adding to a recent transcriptomic analysis of human MTLE brain.50 Combining different ‘-omics’ may provide complimentary facets to a more comprehensive understanding of epilepsy pathogenesis and help identify novel therapeutic targets. A limitation of this study is the heterogeneity of our cohort with diverse epilepsies, therapies and genetic backgrounds. However, our study was designed to identify common protein changes in epilepsy across heterogeneous cases, which increased variation, making the robust protein changes we observed more significant. Future studies should examine specific epilepsy syndromes as well as the role of ASDs. Our study was not powered to assess the impact of treatments, which could bias this study by altering protein levels and altering non-epilepsy pathways. For example, altered GABAergic receptors or mitochondrial function could reflect ASDs such as clobazam, clonazepam or valproic acid.111

In conclusion, our study is the first comprehensive proteomic study in the human epilepsy brain and identifies novel proteins in critical pathways that may be considered as potential drug targets. These pathways mainly include protein biogenesis, mitochondrial function, and synaptogenesis signalling. Combined, our findings provide a valuable resource for epilepsy research.

Supplementary material

Supplementary material is available at Brain Communications online.

Funding

This work was supported by Finding a Cure for Epilepsy and Seizures and the National Institute of Health grants U01NS090415, 1RF1AG058267, P30AG066512 and P01AG060882. This work was supported by funding from the Bluesand Foundation to ED, Philippe Chatrier Foundation to G.P. The proteomics work was in part supported by the New York University School of Medicine and a shared instrumentation grant from the National Institute of Health, 1S10OD010582-01A1 for the purchase of an Orbitrap Fusion Lumos.

Competing interests

The authors report no competing interests.

Supplementary Material

fcab021_Supplementary_Data

Glossary

ASD =

anti-seizure drug

CBZ =

carbamazepine

CLB =

clobazam

CLO =

clonazepam

COD =

cause of death

CSF =

Cerebrospinal fluid

DDG =

dysgenesis of the dentate gyrus

DG =

dentate gyrus

ESL =

eslicarbazepine

FBM =

felbamate

FC =

frontal cortex

FCD =

focal cortical dysplasia

FDR =

false discovery rate

FFPE =

formalin fixed paraffin embedded

fPHT =

fosphenytoin

GBP =

gabapentin

HIPP =

hippocampus

HP =

hippocampus

Hr =

hour(s)

HS =

hippocampal sclerosis

IPA =

Ingenuity Pathway Analysis

LAC =

lacosamide

LCM =

laser capture microdissection

LAM =

lamotrigine

LEV =

levetiracetam

LOR =

lorazepam

MCA/ACA =

middle cerebral artery/anterior cerebral artery

MTLE =

mesial temporal lobe epilepsy

MTLE + HS =

mesial temporal lobe epilepsy with hippocampal sclerosis

MS =

mass spectrometry

ND =

not determined

PCA =

principal component analysis

PER =

perampanel

PHT =

phenytoin

PMI =

post-mortem interval

TPM =

topiramate

VPA =

valproate

Yr =

year(s)

References

  • 1.Ngugi AK, Bottomley C, Kleinschmidt I, Sander JW, Newton CR.. Estimation of the burden of active and life-time epilepsy: a meta-analytic approach. Epilepsia. 2010;51(5):883−–890.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Aronica E, Mühlebner A.. Neuropathology of epilepsy. Handb Clin Neurol. 2017;145:193−216. [DOI] [PubMed] [Google Scholar]
  • 3.Engel J.Epileptogenesis, traumatic brain injury, and biomarkers. Neurobiol Dis. 2019;123:3−7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Thijs RD, Surges R, O'Brien TJ, Sander JW.. Epilepsy in adults. Lancet (London, England). 2019;393(10172):689−701. [DOI] [PubMed] [Google Scholar]
  • 5.Halvorsen M, Petrovski S, Shellhaas R, et al. Mosaic mutations in early-onset genetic diseases. Genet Med. 2016;18(7):746−749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Scheffer IE, Berkovic S, Capovilla G, et al. ILAE classification of the epilepsies: position paper of the ILAE Commission for Classification and Terminology. Epilepsia. 2017;58(4):512−521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Wang J, Lin ZJ, Liu L, et al. Epilepsy-associated genes. Seizure. 2017;44:11−20. [DOI] [PubMed] [Google Scholar]
  • 8.D'Gama AM, Walsh CA.. Somatic mosaicism and neurodevelopmental disease. Nat Neurosci. 2018;21(11):1504−1514. [DOI] [PubMed] [Google Scholar]
  • 9.Dunn P, Albury CL, Maksemous N, et al. Next generation sequencing methods for diagnosis of epilepsy syndromes. Front Genet. 2018;9:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Staley K.Molecular mechanisms of epilepsy. Nat Neurosci. Mar. 2015;18(3):367−372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hesdorffer DC, Crandall LA, Friedman D, Devinsky O.. Sudden unexplained death in childhood: a comparison of cases with and without a febrile seizure history. Epilepsia. 2015;56(8):1294−1300. [DOI] [PubMed] [Google Scholar]
  • 12.Devinsky O, Hesdorffer DC, Thurman DJ, Lhatoo S, Richerson G.. Sudden unexpected death in epilepsy: epidemiology, mechanisms, and prevention. Lancet Neurol. 2016;15(10):1075−1088. [DOI] [PubMed] [Google Scholar]
  • 13.Blumcke I, Thom M, Aronica E, et al. The clinicopathologic spectrum of focal cortical dysplasias: a consensus classification proposed by an ad hoc Task Force of the ILAE Diagnostic Methods Commission. Epilepsia. 2011;52(1):158−174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kandratavicius L, Balista PA, Lopes-Aguiar C, et al. Animal models of epilepsy: use and limitations. Neuropsychiatr Dis Treat. 2014;10:1693−1705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Wong M, Roper SN.. Genetic animal models of malformations of cortical development and epilepsy. J Neurosci Methods. 2016;260:73−82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Reyes-Garcia SZ, Scorza CA, Araújo NS, et al. Different patterns of epileptiform-like activity are generated in the sclerotic hippocampus from patients with drug-resistant temporal lobe epilepsy. Sci Rep. 2018;8(1):7116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fukata Y, Fukata M.. Epilepsy and synaptic proteins. Curr Opin Neurobiol. 2017;45:1−8. [DOI] [PubMed] [Google Scholar]
  • 18.Mahoney JM, Mills JD, Muhlebner A, et al. 2017 WONOEP appraisal: studying epilepsy as a network disease using systems biology approaches. Epilepsia. 2019;60(6):1045−1053. [DOI] [PubMed] [Google Scholar]
  • 19.Genetic determinants of common epilepsies: a meta-analysis of genome-wide association studies. Lancet Neurol. 2014;13(9):893−903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Miller-Delaney SF, Bryan K, Das S, et al. Differential DNA methylation profiles of coding and non-coding genes define hippocampal sclerosis in human temporal lobe epilepsy. Brain. 2015;138(Pt 3):616−631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Dębski KJ, Pitkanen A, Puhakka N, et al. Etiology matters - Genomic DNA methylation patterns in three rat models of acquired epilepsy. Sci Rep. 2016;6:25668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Casillas-Espinosa PM, Powell KL, Zhu M, et al. Evaluating whole genome sequence data from the Genetic Absence Epilepsy Rat from Strasbourg and its related non-epileptic strain. PloS One. 2017;12(7):e0179924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Genome-wide mega-analysis identifies 16 loci and highlights diverse biological mechanisms in the common epilepsies. Nat Commun. 2018;9(1):5269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Ellis CA, Petrovski S, Berkovic SF.. Epilepsy genetics: clinical impacts and biological insights. The Lancet Neurology. 2020;19(1):93−100. [DOI] [PubMed] [Google Scholar]
  • 25.Seyfried NT, Dammer EB, Swarup V, et al. A multi-network approach identifies protein-specific co-expression in asymptomatic and symptomatic Alzheimer's disease. Cell Syst. 2017;4(1):60−72.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bitsika V, Duveau V, Simon-Areces J, et al. High-throughput LC-MS/MS proteomic analysis of a mouse model of mesiotemporal lobe epilepsy predicts microglial activation underlying disease development. J Proteome Res. 2016;15(5):1546−1562. [DOI] [PubMed] [Google Scholar]
  • 27.Schouten M, Bielefeld P, Fratantoni SA, et al. Multi-omics profile of the mouse dentate gyrus after kainic acid-induced status epilepticus. Sci. Data. 2016;3:160068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Yang JW, Czech T, Felizardo M, Baumgartner C, Lubec G.. Aberrant expression of cytoskeleton proteins in hippocampus from patients with mesial temporal lobe epilepsy. Amino Acids. 2006;30(4):477−493. [DOI] [PubMed] [Google Scholar]
  • 29.Keren-Aviram G, Dachet F, Bagla S, Balan K, Loeb JA, Dratz EA.. Proteomic analysis of human epileptic neocortex predicts vascular and glial changes in epileptic regions. PloS One. 2018;13(4):e0195639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Drummond ES, Nayak S, Ueberheide B, Wisniewski T.. Proteomic analysis of neurons microdissected from formalin-fixed, paraffin-embedded Alzheimer's disease brain tissue. Sci Rep. 2015;5:15456. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Drummond E, Nayak S, Faustin A, et al. Proteomic differences in amyloid plaques in rapidly progressive and sporadic Alzheimer's disease. Acta Neuropathologica. 2017;133(6):933−954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.David Ellison SL, Chimelli L, Harding BN. et al. Neuropathology: a reference text of CNS pathology, 3rd ed. MOSBY an imprint of Elsevier Limited: Maryland Heights, MO; 2013. [Google Scholar]
  • 33.Drummond E, Nayak S, Pires G, Ueberheide B, Wisniewski T.. Isolation of Amyloid Plaques and Neurofibrillary Tangles from Archived Alzheimer's Disease Tissue Using Laser-Capture Microdissection for Downstream Proteomics. Methods Mol Biol (Clifton, NJ). 2018;1723:319−334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cox J, Mann M.. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26(12):1367−1372. [DOI] [PubMed] [Google Scholar]
  • 35.Cox J, Matic I, Hilger M, et al. A practical guide to the MaxQuant computational platform for SILAC-based quantitative proteomics. Nat Protoc. 2009;4(5):698−705. [DOI] [PubMed] [Google Scholar]
  • 36.Cox J, Michalski A, Mann M.. Software lock mass by two-dimensional minimization of peptide mass errors. J Am Soc Mass Spectrom. 2011;22(8):1373–1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Cox J, Hein MY, Luber CA, Paron I, Nagaraj N, Mann M.. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol Cell Proteomics. 2014;13(9):2513−2526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tyanova S, Temu T, Sinitcyn P, et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods. 2016;13(9):731−740. [DOI] [PubMed] [Google Scholar]
  • 39.Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43(Database)D447–D452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45(D1):D362–D368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gu Z, Eils R, Schlesner M.. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics. 2016;32(18):2847−2849. [DOI] [PubMed] [Google Scholar]
  • 42.Lake BB, Chen S, Sos BC, et al. Integrative single-cell analysis of transcriptional and epigenetic states in the human adult brain. Nat Biotechnol. 2018;36(1):70−80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lleo A, Nunez-Llaves R, Alcolea D, et al. Changes in synaptic proteins precede neurodegeneration markers in preclinical Alzheimer's disease cerebrospinal fluid. Mol Cell Proteomics. 2019;18(3):546−560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Dengler CG, Coulter DA.. Normal and epilepsy-associated pathologic function of the dentate gyrus. Prog Brain Res. 2016;226:155−178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McGuone D, Leitner D, William C, et al. Neuropathologic changes in sudden unexplained death in childhood. J Neuropathol Exp Neurol. 2020;79(3):336−346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Fassio A, Esposito A, Kato M, et al. ; C4RCD Research Group. De novo mutations of the ATP6V1A gene cause developmental encephalopathy with epilepsy. Brain. 2018;141(6):1703−1718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Persike DS, Marques-Carneiro JE, Stein MLL, et al. Altered proteins in the hippocampus of patients with mesial temporal lobe epilepsy. Pharmaceuticals (Basel). 2018;11(4):95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Qin L, Liu X, Liu S, et al. Differentially expressed proteins underlying childhood cortical dysplasia with epilepsy identified by iTRAQ proteomic profiling. PLoS One. 2017;12(2):e0172214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hansen KF, Sakamoto K, Pelz C, Impey S, Obrietan K.. Profiling status epilepticus-induced changes in hippocampal RNA expression using high-throughput RNA sequencing. Sci Rep. 2014;4:6930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Guelfi S, Botia JA, Thom M, et al. Transcriptomic and genetic analyses reveal potential causal drivers for intractable partial epilepsy. Brain. 2019;142(6):1616−1630. [DOI] [PubMed] [Google Scholar]
  • 51.Petrovski S, Kury S, Myers CT, et al. ; University of Washington Center for Mendelian Genomics. Germline de novo mutations in GNB1 cause severe neurodevelopmental disability, hypotonia, and seizures. Am J Hum Genet. 2016;98(5):1001−1010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hemati P, Revah-Politi A, Bassan H, et al. DDD study. Refining the phenotype associated with GNB1 mutations: clinical data on 18 newly identified patients and review of the literature. Am J Med Genet A. 2018;176(11):2259−2275. [DOI] [PubMed] [Google Scholar]
  • 53.Tarpey PS, Smith R, Pleasance E, et al. A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation. Nat Genet. 2009;41(5):535−543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang FX, Sun QJ, Zheng XY, et al. Abnormal expression of synaptophysin, SNAP-25, and synaptotagmin 1 in the hippocampus of kainic acid-exposed rats with behavioral deficits. Cell Mol Neurobiol. 2014;34(6):813−824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang MY, Zheng CY, Zou MM, et al. Lamotrigine attenuates deficits in synaptic plasticity and accumulation of amyloid plaques in APP/PS1 transgenic mice. Neurobiol Aging. 2014;35(12):2713−2725. [DOI] [PubMed] [Google Scholar]
  • 56.Kang JQ.Defects at the crossroads of GABAergic signaling in generalized genetic epilepsies. Epilepsy Res. 2017;137:9−18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.von Spiczak S, Helbig KL, Shinde DN, et al. EuroEPINOMICS-RES NLES Working Group. encephalopathy: a new disease of vesicle fission. Neurology. 2017;89(4):385−394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Walker A, Russmann V, Deeg CA, et al. Proteomic profiling of epileptogenesis in a rat model: focus on inflammation. Brain Behav Immun. 2016;53:138−158. [DOI] [PubMed] [Google Scholar]
  • 59.Keck M, van Dijk RM, Deeg CA, et al. Proteomic profiling of epileptogenesis in a rat model: focus on cell stress, extracellular matrix and angiogenesis. Neurobiol Dis. 2018;112:119−135. [DOI] [PubMed] [Google Scholar]
  • 60.You Y, Bai H, Wang C, et al. Myelin damage of hippocampus and cerebral cortex in rat pentylenetetrazol model. Brain Res. 2011;1381:208−216. [DOI] [PubMed] [Google Scholar]
  • 61.Ye Y, Xiong J, Hu J, et al. Altered hippocampal myelinated fiber integrity in a lithium-pilocarpine model of temporal lobe epilepsy: a histopathological and stereological investigation. Brain Res. 2013;1522:76−87. [DOI] [PubMed] [Google Scholar]
  • 62.Häberle J, Shahbeck N, Ibrahim K, Hoffmann GF, Ben-Omran T.. Natural course of glutamine synthetase deficiency in a 3 year old patient. Mol Genet Metab. 2011;103(1):89−91. [DOI] [PubMed] [Google Scholar]
  • 63.Eid T, Thomas MJ, Spencer DD, et al. Loss of glutamine synthetase in the human epileptogenic hippocampus: possible mechanism for raised extracellular glutamate in mesial temporal lobe epilepsy. Lancet. 2004;363(9402):28−37. [DOI] [PubMed] [Google Scholar]
  • 64.Zhou Y, Dhaher R, Parent M, et al. Selective deletion of glutamine synthetase in the mouse cerebral cortex induces glial dysfunction and vascular impairment that precede epilepsy and neurodegeneration. Neurochem Int. 2019;123:22−33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhou J, Tawk M, Tiziano FD, et al. Spinal muscular atrophy associated with progressive myoclonic epilepsy is caused by mutations in ASAH1. Am J Hum Genet. Jul. 2012;91(1):5−14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Aronica EM, Gorter JA, Paupard MC, Grooms SY, Bennett MV, Zukin RS.. Status epilepticus-induced alterations in metabotropic glutamate receptor expression in young and adult rats. J Neurosci. 1997;17(21):8588−8595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pacheco Otalora LF, Couoh J, Shigamoto R, Zarei MM, Garrido Sanabria ER.. Abnormal mGluR2/3 expression in the perforant path termination zones and mossy fibers of chronically epileptic rats. Brain Res. 2006;1098(1):170−185. [DOI] [PubMed] [Google Scholar]
  • 68.Corti C, Battaglia G, Molinaro G, et al. The use of knock-out mice unravels distinct roles for mGlu2 and mGlu3 metabotropic glutamate receptors in mechanisms of neurodegeneration/neuroprotection. J Neurosci. 2007;27(31):8297−8308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Griffin NG, Wang Y, Hulette CM, et al. Differential gene expression in dentate granule cells in mesial temporal lobe epilepsy with and without hippocampal sclerosis. Epilepsia. 2016;57(3):376−385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Lionel AC, Vaags AK, Sato D, et al. Rare exonic deletions implicate the synaptic organizer Gephyrin (GPHN) in risk for autism, schizophrenia and seizures. Hum Mol Genet. 2013;22(10):2055−2066. [DOI] [PubMed] [Google Scholar]
  • 71.Dejanovic B, Lal D, Catarino CB, et al. Exonic microdeletions of the gephyrin gene impair GABAergic synaptic inhibition in patients with idiopathic generalized epilepsy. Neurobiol Dis. 2014;67:88−96. [DOI] [PubMed] [Google Scholar]
  • 72.Förstera B, Belaidi AA, Jüttner R, et al. Irregular RNA splicing curtails postsynaptic gephyrin in the cornu ammonis of patients with epilepsy. Brain. 2010;133(Pt 12):3778−3794. [DOI] [PubMed] [Google Scholar]
  • 73.Thind KK, Yamawaki R, Phanwar I, Zhang G, Wen X, Buckmaster PS.. Initial loss but later excess of GABAergic synapses with dentate granule cells in a rat model of temporal lobe epilepsy. J Comp Neurol. 2010;518(5):647−667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.de Moura JC, Tirapelli DP, Neder L, et al. Amygdala gene expression of NMDA and GABA(A) receptors in patients with mesial temporal lobe epilepsy. Hippocampus. 2012;22(1):92−97. [DOI] [PubMed] [Google Scholar]
  • 75.Vlaskamp DRM, Callenbach PMC, Rump P, et al. Copy number variation in a hospital-based cohort of children with epilepsy. Epilepsia Open. 2017;22(2):44−254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Meng XF, Yu JT, Song JH, Chi S, Tan L.. Role of the mTOR signaling pathway in epilepsy. J Neurol Sci. 2013;332(1−2):4−15. [DOI] [PubMed] [Google Scholar]
  • 77.Ribierre T, Baulac S.. mTOR pathway in familial focal epilepsies. Oncotarget. 2017;8(4):5674−5675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Revah-Politi A, Sands TT, Colombo S, et al. GNB1 encephalopathy. In: Adam MP, Ardinger HH, Pagon RA, et al. eds. GeneReviews [Internet]. Seattle (WA); 2020. [Google Scholar]
  • 79.Barker-Haliski M, White HS.. Glutamatergic mechanisms associated with seizures and epilepsy. Cold Spring Harbor Perspect Med. 2015;5(8):a022863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zou J, Wang YX, Dou FF, et al. Glutamine synthetase down-regulation reduces astrocyte protection against glutamate excitotoxicity to neurons. Neurochem Int. 2010;56(4):577−584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Thom M.Review: hippocampal sclerosis in epilepsy: a neuropathology review. Neuropathol Appl Neurobiol. 2014;40(5):520−543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Punia V, Bena J, Gonzalez-Martinez J, et al. Histopathologic substrate of drug-resistant epilepsy in older adults and the elderly undergoing surgery. Epilepsia Open. 2019;4(2):328−333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Nikonenko AG, Radenovic L, Andjus PR, Skibo GG.. Structural features of ischemic damage in the hippocampus. Anat Rec (Hoboken, NJ: 2007). 2009;292(12):1914−1921. [DOI] [PubMed] [Google Scholar]
  • 84.Hatanpaa KJ, Raisanen JM, Herndon E, et al. Hippocampal sclerosis in dementia, epilepsy, and ischemic injury: differential vulnerability of hippocampal subfields. J Neuropathol Exp Neurol. 2014;73(2):136−142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Halliday G.Pathology and hippocampal atrophy in Alzheimer's disease. Lancet Neurol. 2017;16(11):862−864. [DOI] [PubMed] [Google Scholar]
  • 86.Rocca MA, Barkhof F, De Luca J, et al. ; MAGNIMS Study Group. The hippocampus in multiple sclerosis. Lancet Neurol. 2018;17(10):918−926. [DOI] [PubMed] [Google Scholar]
  • 87.Hansen N.Long-term memory dysfunction in limbic encephalitis. Front Neurol. 2019;10:330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Gourmaud S, Shou H, Irwin DJ, et al. Alzheimer-like amyloid and tau alterations associated with cognitive deficit in temporal lobe epilepsy. Brain. 2020;143(1):191−209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Bartsch T, Wulff P.. The hippocampus in aging and disease: from plasticity to vulnerability. Neuroscience. 2015;309:1−16. [DOI] [PubMed] [Google Scholar]
  • 90.Cole AJ.Is epilepsy a progressive disease? The neurobiological consequences of epilepsy. Epilepsia. 2000;41(Suppl 2):S13−S22. [DOI] [PubMed] [Google Scholar]
  • 91.Looney MR, Dohan FC, Davies KG, Seidenberg M, Hermann BP, Schweitzer JB.. Synaptophysin immunoreactivity in temporal lobe epilepsy-associated hippocampal sclerosis. Acta Neuropathol. 1999;98(2):179−185. [DOI] [PubMed] [Google Scholar]
  • 92.Kang HC, Lee YM, Kim HD.. Mitochondrial disease and epilepsy. Brain Dev. 2013;35(8):757−761. [DOI] [PubMed] [Google Scholar]
  • 93.DiMauro S, Hirano M, Kaufmann P, et al. Clinical features and genetics of myoclonic epilepsy with ragged red fibers. Adv Neurol. 2002;89:217−229. [PubMed] [Google Scholar]
  • 94.Tryoen-Toth P, Richert S, Sohm B, et al. Proteomic consequences of a human mitochondrial tRNA mutation beyond the frame of mitochondrial translation. J Biol Chem. 2003;278(27):24314−24323. [DOI] [PubMed] [Google Scholar]
  • 95.Rahman S.Mitochondrial disease and epilepsy. Dev Med Child Neurol. 2012;54(5):397−406. [DOI] [PubMed] [Google Scholar]
  • 96.Kudin AP, Zsurka G, Elger CE, Kunz WS.. Mitochondrial involvement in temporal lobe epilepsy. Exp Neurol. 2009;218(2):326−332. [DOI] [PubMed] [Google Scholar]
  • 97.Rowley S, Patel M.. Mitochondrial involvement and oxidative stress in temporal lobe epilepsy. Free Radic Biol Med. 2013;62:121−131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Volmering E, Niehusmann P, Peeva V, et al. Neuropathological signs of inflammation correlate with mitochondrial DNA deletions in mesial temporal lobe epilepsy. Acta Neuropathol. 2016;132(2):277−288. [DOI] [PubMed] [Google Scholar]
  • 99.Spaulding EL, Burgess RW.. Accumulating evidence for axonal translation in neuronal homeostasis. Front Neurosci. 2017;11:312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hodges SL, Lugo JN.. Therapeutic role of targeting mTOR signaling and neuroinflammation in epilepsy. Epilepsy Res. 2020;161:106282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kim JK, Cho J, Kim SH, et al. Brain somatic mutations in MTOR reveal translational dysregulations underlying intractable focal epilepsy. J Clin Invest. 2019;129(10):4207−4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Pernice HF, Schieweck R, Kiebler MA, Popper B.. mTOR and MAPK: from localized translation control to epilepsy. BMC Neurosci. 2016;17(1):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Marinissen MJ, Gutkind JS.. G-protein-coupled receptors and signaling networks: emerging paradigms. Trends Pharmacol Sci. 2001;22(7):368−376. [DOI] [PubMed] [Google Scholar]
  • 104.Yu Y, Nguyen DT, Jiang J.. G protein-coupled receptors in acquired epilepsy: druggability and translatability. Prog Neurobiol. 2019;183:101682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Steinrucke S, Lohmann K, Domingo A, et al. Novel GNB1 missense mutation in a patient with generalized dystonia, hypotonia, and intellectual disability. Neurol Genet. 2016;2(5):e106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Lohmann K, Masuho I, Patil DN, et al. Novel GNB1 mutations disrupt assembly and function of G protein heterotrimers and cause global developmental delay in humans. Hum Mol Genet. 2017;26(6):1078−1086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jones HF, Morales-Briceno H, Barwick K, et al. Myoclonus-dystonia caused by GNB1 mutation responsive to deep brain stimulation. Mov Dis. 2019;34(7):1079−1080. [DOI] [PubMed] [Google Scholar]
  • 108.Endo W, Ikemoto S, Togashi N, et al. Phenotype-genotype correlations in patients with GNB1 gene variants, including the first three reported Japanese patients to exhibit spastic diplegia, dyskinetic quadriplegia, and infantile spasms. Brain Dev. 2020;42(2):199−204. [DOI] [PubMed] [Google Scholar]
  • 109.Colombo S, Petri S, Shalomov B, et al. G Protein-Coupled Potassium Channels Implicated in Mouse and Cellular Models of GNB1 Encephalopathy. 2019;697235, doi:10.1101/697235%JbioRxiv [Google Scholar]
  • 110.Sriram K, Insel PA.. G protein-coupled receptors as targets for approved drugs: how many targets and how many drugs? Mol Pharmacol. 2018;93(4):251−258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Czapiński P, Blaszczyk B, Czuczwar SJ.. Mechanisms of action of antiepileptic drugs. Curr Topics Med Chem. 2005;5(1):3−14. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

fcab021_Supplementary_Data

Data Availability Statement

The mass spectrometry raw files are accessible under MassIVE ID: MSV000085517.


Articles from Brain Communications are provided here courtesy of Oxford University Press

RESOURCES