Abstract

Approximately 22% of Alzheimer’s disease (AD) patients suffer from seizures, and the co-occurrence of seizures and epileptiform activity exacerbates AD pathology and related cognitive deficits, suggesting that seizures may be a targetable component of AD progression. Given that alterations in neuronal excitatory:inhibitory (E:I) balance occur in epilepsy, we hypothesized that decreased markers of inhibition relative to those of excitation would be present in AD patients. We similarly hypothesized that in 5XFAD mice, the E:I imbalance would progress from an early stage (prodromal) to later symptomatic stages and be further exacerbated by pentylenetetrazol (PTZ) kindling.

Post-mortem AD temporal cortical tissues from patients with or without seizure history were examined for changes in several markers of E:I balance, including levels of the inhibitory GABAA receptor, the sodium potassium chloride cotransporter 1 (NKCC1) and potassium chloride cotransporter 2 (KCC2) and the excitatory NMDA and AMPA type glutamate receptors. We performed patch-clamp electrophysiological recordings from CA1 neurons in hippocampal slices and examined the same markers of E:I balance in prodromal 5XFAD mice. We next examined 5XFAD mice at chronic stages, after PTZ or control protocols, and in response to chronic mTORC1 inhibitor rapamycin, administered following kindled seizures, for markers of E:I balance.

We found that AD patients with comorbid seizures had worsened cognitive and functional scores and decreased GABAA receptor subunit expression, as well as increased NKCC1/KCC2 ratios, indicative of depolarizing GABA responses. Patch clamp recordings of prodromal 5XFAD CA1 neurons showed increased intrinsic excitability, along with decreased GABAergic inhibitory transmission and altered glutamatergic neurotransmission, indicating that E:I imbalance may occur in early disease stages. Furthermore, seizure induction in prodromal 5XFAD mice led to later dysregulation of NKCC1/KCC2 and a reduction in GluA2 AMPA glutamate receptor subunit expression, indicative of depolarizing GABA receptors and calcium permeable AMPA receptors. Finally, we found that chronic treatment with the mTORC1 inhibitor, rapamycin, at doses we have previously shown to attenuate seizure-induced amyloid-β pathology and cognitive deficits, could also reverse elevations of the NKCC1/KCC2 ratio in these mice.

Our data demonstrate novel mechanisms of interaction between AD and epilepsy and indicate that targeting E:I balance, potentially with US Food and Drug Administration-approved mTOR inhibitors, hold therapeutic promise for AD patients with a seizure history.

See Blum et al. (https://doi.org/10.1093/brain/awae146) for a scientific commentary on this article.

Introduction

Seizures can be a comorbidity in patients with Alzheimer’s disease (AD), with 10%–22% showing clinically identifiable seizures and up to 64% displaying subclinical epileptiform activity.1 Notably, the co-occurrence of seizures and epileptiform activity in AD patients enhances disease progression and worsens cognitive performance.1-5 We and others have demonstrated bidirectional interactions, with AD and epilepsy sharing similar pathology and convergence upon common underlying cellular mechanisms. Human temporal lobe epilepsy brain tissue shows increased amyloid-β (Aβ) plaques and increased hyperphosphorylated tau (pTau),6-8 and the accumulation of these neuropathological proteins in AD animal models is associated with elevated neuronal hyperexcitability and seizure susceptibility.9-14 Furthermore, we recently demonstrated that a history of seizures is associated with enhanced Aβ and pTau pathology in human AD.14

While mechanisms of seizure generation in AD are not fully elucidated, evidence points to a synaptic excitatory:inhibitory (E:I) imbalance in chronic epilepsy, including altered expression and function of certain neurotransmitter receptors and ion cotransporters.15-17 Chronic epilepsy human and animal model studies show evidence of reduced inhibitory neurotransmission, with decreases in the ratio of α1/α3 inhibitory gamma-aminobutyric acid type A receptor (GABAAR) subunits that diminish inhibitory postsynaptic currents (IPSCs).18-22 Inhibitory GABAergic transmission is further attenuated by paradoxical depolarizing GABAAR currents due to reversal of the chloride (Cl) gradient caused by increases in the ratio of the sodium (Na+) potassium (K+) Cl co-transporter 1 (NKCC1), which imports Cl, to that of the potassium Cl cotransporter 2 (KCC2), which exports Cl.23-26 Commensurate with decreased inhibitory drive in epilepsy, there is an increase in glutamate-receptor mediated excitability, in part due to alterations in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) and N-methyl-D-aspartate receptor (NMDAR) subunit expression, with increases in the ratios of GluA1/GluA2 AMPAR subunits and GluN2B/GluN2A NMDAR subunits resulting in GluA2-lacking, Ca2+-permeable AMPAR27-29 and extended decay times and elevated Ca2+ influx, respectively.30-32

Pathological E:I alterations are not unique to epilepsy, as evidence for neuronal hyperexcitability has been seen in other neurodevelopmental and neurodegenerative disorders.33-36 In AD patients, altered GABAAR subunit regulation has been observed,37 including decreased α1/α3 expression.38 Furthermore, evidence for paradoxically depolarizing GABA currents has been found, with elevations of NKCC1 after Aβ1–42 hippocampal injection39 and reduced KCC2 in AD mouse models, with behavioural rescue after restoration of membrane KCC2.40,411–42 has also been shown to increase Ca2+-permeable AMPARs which can result in memory deficits via reduced long-term potentiation and synapse loss.42-45 However, given the prevalence of network excitability and seizures in AD, and the evidence that seizures can alter E:I balance in epilepsy, understanding the impact of seizures and the evolution of neuronal hyperexcitability in AD progression may lead to new mitigable therapeutic E:I targets. To understand the progression of E:I balance, we examined the 5XFAD mouse model at two stages: prodromal and symptomatic. To understand seizure impact on E:I imbalance in AD, we examined human AD with known seizure history and 5XFAD mice at a symptomatic stage after seizure induction during the prodromal stage.

Preclinical and clinical epilepsy research has revealed several candidate signalling pathways that might link E:I imbalance and network hyperexcitability in epilepsy to neurodegeneration. One example is the substantial evidence for the activation of the mammalian target of rapamycin complex 1 (mTORC1) as a key common element between epilepsy and neurodegeneration. mTORC1 mediates pathological tau and APP processing46-50 and can regulate excitatory and inhibitory receptors and network function.51,52 Models of tuberous sclerosis complex, characterized by constitutive mTORC1 overactivation commonly resulting in epilepsy,53,54 have demonstrated that blockade of mTORC1 with rapamycin can rescue GABAergic neurotransmission and epilepsy.55-58 Rapamycin treatment has also rescued neuronal hyperexcitability in models of autism spectrum disorder59 and neuropathic pain.60 Together, these results indicate that targeting mTORC1 activity might restore E:I balance across multiple disease models, including AD.

Considering these data and that rapamycin and other rapalogues are approved by the US Food and Drug Administration for other indications, mTORC1 is a promising target for therapeutic intervention for patients with AD and co-morbid epilepsy. Indeed, we recently found that AD patients with a history of seizure display enhanced tau and Aβ pathology, associated with increased mTORC1 activity.14 In addition, we observed that the induction of seizures in the 5XFAD mice exacerbates Aβ pathology and cognitive deficits, and that these changes are reversible with chronic low-dose rapamycin treatment.14

Given that E:I imbalance may contribute to the cognitive impairments observed in AD, we hypothesized that key components of E:I balance may be dysregulated in AD and that these alterations emerge early in the disease course and may be exacerbated by seizures. First, we found worsened cognitive and functional performance in AD patients with a seizure history compared with those without and found seizures were associated with greater alterations in several components of the GABAergic system in largely the same human control and AD temporal neocortex from patients with and without known seizure history as our previous study.14 Next, we examined the 5XFAD mouse model for protein and electrophysiological indications of hyperexcitability at the prodromal stage (4 months of age) and directly assessed the effects of chemoconvulsant [pentylenetetrazol (PTZ)]-induced seizures at this stage on E:I markers at a later, symptomatic time point (7 months of age). Finally, given that we found protective effects on pathology and cognition with rapamycin in this model,14 we used the tissue from our prior study to examine whether these therapeutic benefits extend to markers of E:I balance. Overall, we found that E:I imbalance began at a prodromal stage in 5XFAD mice and that seizures significantly exacerbated excitatory and inhibitory alterations, while chronic rapamycin treatment significantly attenuated these effects.

Materials and methods

Human subjects

All protocols and procedures were approved under the ethical standards of the Institutional Review Board of the University of Pennsylvania (Philadelphia, PA). The AD cognitive study population consisted of 105 patients, selected by querying the integrated neurodegenerative disease database61 of the Center for Neurodegenerative Disease and Research (CNDR) at the University of Pennsylvania for available global Clinical Dementia Rating (CDR) score and CDR Sum of Boxes (CDR-SOB) closest to death. All patients were enrolled in observational research at the University of Pennsylvania, with standardized assessments and medical history that included assessment of seizure history.62,63 Of the AD cases, 18 (6 males and 12 females; mean age at death = 77.38 years) had a reported clinical seizure history (AD+Sz). The remaining 87 AD patients (43 males and 44 females; mean age at death = 79.7 years) had no known seizure history (AD−Sz). AD clinical diagnosis was established during life based on the clinical history, neurological and neuropsychological assessment, and confirmed by post-mortem histopathological staging of AD neuropathological markers (Aβ42 and tau pathology), according to the National Institute on Aging-Alzheimer’s Association (NIA-AA) guidelines.62

For a subset of the population detailed above, post-mortem brain specimens and region matched controls were acquired from CNDR and consisted of superior/mid-temporal cortex frozen samples (control: n = 13, AD−Sz: n = 19, AD+Sz: n = 11) and formalin-fixed paraffin-embedded sections (control: n = 5, AD−Sz: n = 7, AD+Sz: n = 8).

Detailed methods for neurocognitive assessments and clinical characteristics for subjects included in both neurocognitive and biochemical studies can be found in the Supplementary material and Supplementary Table 1.

Mice

All animal procedures were approved and performed in accordance with the guidelines of the Institutional Animal Care and Use Committee (IACUC) Office of Animal Welfare of the University of Pennsylvania (Philadelphia, PA). Male and female 5XFAD and wild-type (WT) mice on a mixed B6SJL background second generation (F2) generated from mice originally purchased from Jackson Laboratory (MMRC stock #3840) were used in these studies. 5XFAD mice harbour the human APP and PSEN1 genes under the Thy1 promoter containing five mutations for aggressive amyloid development (APP: Swedish K670N/M671L, Florida I7516V and London V717I; PSEN1: M146L and L286V). Genotyping was conducted as specified by Jackson Laboratory. Mice were allocated randomly to treatment groups, which were counterbalanced for sex and litter. Additional methodology, including PTZ kindling, rapamycin treatment, electrophysiology and western blot are provided in the Supplementary material.

Statistics

For cognitive and functional performances, comparisons between AD+Sz and AD−Sz were made using the Mann–Whitney test. For global CDR, we determined the potential effects of clinical characteristics by separate ordinal regressions with seizure history and age at test, disease duration, sex or education as independent variables. For the CDR-SOB and Dementia Severity Rating Scale (DSRS), we examined these same clinical characteristics by separate analysis of covariance (ANCOVA), with seizure history as the independent variable and age at test, disease duration, sex or education as a covariate. For the human tissue studies, all AD to control comparisons were made with unpaired t-tests or Mann–Whitney tests for non-normal data. Control, AD−Sz and AD+Sz comparisons were made using ANOVA or the Kruskal–Wallis test for non-normal data. For human biochemical analyses, age at death, disease duration and sex were examined by multiple linear regression. For the prodromal mouse studies, 5XFAD to WT comparisons were made using unpaired t-tests and Kolmogorov–Smirnov (K-S) tests for cumulative probability comparisons. For the non-therapeutic PTZ study, comparisons were made using two-way ANOVA, with genotype and kindling as independent variables. For the therapeutic PTZ study, 5XFAD and WT mice were analysed separately using two-way ANOVAs, with PTZ and rapamycin treatment as independent variables. All ANOVA analyses were followed by Tukey’s post hoc test to examine group differences when main effects or interactions were found. Multiple linear regressions were performed for mouse biochemical studies to determine potential sex effects. Where sex effects were present, data-points are displayed as squares (female) or triangles (male) to visualize these differences, and histograms separated by sex can be found in the Supplementary material. Correlations were calculated by Pearson correlation coefficients. ANCOVA and ordinal regressions were performed in R (version 4.0), and the remaining statistical analyses were performed in GraphPad Prism 9 software (San Diego, CA). Histogram error bars represent the standard error of the mean. We considered the results to be significant at P < 0.05.

Results

Alzheimer’s disease patients with seizure history have worsened cognitive and functional performance

To establish potential associations between seizures and cognitive and functional deficits in AD, we examined scores from clinical (global CDR and CDR-SOB) and caretaker questionnaires (DSRS) for subjects with known seizure history. No significant differences between AD−Sz and AD+Sz were found in years since onset to final CDR assessment (mean AD−Sz = 8.3 years; mean AD+Sz = 9.9 years), nor in age at final CDR assessment (mean AD−Sz = 77.1 years; mean AD+Sz = 76.0 years) (Supplementary Table 1). The AD+Sz group showed elevated final global CDR compared with AD−Sz (P < 0.01, n = 18–87; Fig. 1A). Separate ordinal regressions revealed that age at test [Sz+age: age P < 0.05 (0.0034, 0.078)] and disease duration [Sz+duration: duration P < 0.001 (0.18, 0.44)] were both significant predictors of CDR, while no significant effects of sex or education were found (Supplementary Table 2). Importantly, seizures were significant predictors of CDR in all models [Sz+age: Sz P < 0.01 (0.59, 3.32); Sz+duration: Sz P < 0.05 (0.87, 5.57); Sz+sex: Sz P < 0.05 (0.48, 3.19); Sz+education: Sz P < 0.05 (0.53, 3.25)] (Supplementary Table 2). AD+Sz also showed worsened CDR-SOB compared with the AD−Sz group (P < 0.05, n = 18–87; Fig. 1B). Separate ANCOVAs revealed that seizures significantly impact CDR-SOB when age [F(1,102) = 4.81, P < 0.05], disease duration [F(1,94) = 6.23, P < 0.05] and education [F(1,102) = 4.1, P < 0.05] are adjusted for, and there was a significant interaction between seizures and sex [F(1,102) = 355.17, P < 0.0001] (Supplementary Fig. 1 and Supplementary Table 2). AD+Sz patients also showed significantly worsened overall functional rating (DSRS) compared with those in the AD−Sz group (P < 0.01, n = 14–19) (Fig. 1C), including significantly worse ratings for memory, recognition of family members, mobility, orientation to place, social and community activities and incontinence (P < 0.05, n = 14–19) (Supplementary Fig. 2). Separate ANCOVAs revealed that seizures significantly impact DSRS when age [F(1,30) = 6.68, P < 0.05], sex [F(1,30) = 6.78, P < 0.05] and education [F(1,30) = 5.95, P < 0.05] were adjusted for, and we found a significant interaction between seizures and disease duration [F(1,27) = 62.39, P < 0.0001] (Supplementary Table 2). Overall, these data indicated that seizures are associated with worsened cognitive performance and overall function, corroborating previous studies examining epileptiform activity in AD2-5 and suggesting heightened neuronal dysfunction.

Seizure history is associated with worse cognitive and functional performance in patients with Alzheimer’s disease. Comparisons between Alzheimer’s disease (AD) patients without seizure history (AD−Sz) and with seizure history (AD+Sz) in final global Clinical Dementia Rating (CDR) (A), CDR-Sum of Boxes (SOB) (B) and Dementia Severity Rating Scale (DSRS) totals (C). CDR: n = 18 (AD+Sz), n = 87 (AD−Sz); DSRS: n = 14 (AD+Sz), n = 19 (AD−Sz). *P < 0.05, **P < 0.01.
Figure 1

Seizure history is associated with worse cognitive and functional performance in patients with Alzheimer’s disease. Comparisons between Alzheimer’s disease (AD) patients without seizure history (AD−Sz) and with seizure history (AD+Sz) in final global Clinical Dementia Rating (CDR) (A), CDR-Sum of Boxes (SOB) (B) and Dementia Severity Rating Scale (DSRS) totals (C). CDR: n = 18 (AD+Sz), n = 87 (AD−Sz); DSRS: n = 14 (AD+Sz), n = 19 (AD−Sz). *P < 0.05, **P < 0.01.

Alzheimer’s disease with seizure history shows worse dysregulation of proteins that mediate E:I balance

We hypothesized that the worsened functional and cognitive deficits found in AD+Sz patients would be associated with worsening of markers of E:I imbalance. Thus, we examined the ratios of α1/α3 GABAAR subunits19 and of Cl cotransporters, NKCC1/KCC2.23,24,64 We found a significant decrease in GABAARα1 expression in AD+Sz compared with controls (F = 3.52, Tukey’s post hoc: P < 0.05) (Fig. 2B) that was not found in AD−Sz and which drove the reduction in GABAARα1 found across all AD patients (P < 0.05) (Fig. 2B). This alteration also resulted in a significantly decreased GABAARα1/GABAARα3 ratio in all AD patients when compared with controls (P < 0.01) (Fig. 2B). The ratio of NKCC1/KCC2 was also significantly increased in the AD+Sz group when compared with the AD−Sz group (Tukey’s post hoc: P < 0.05) (Fig. 2C). This difference was largely attributable to downregulation of KCC2 in the AD+Sz group compared with the AD−Sz group (Tukey’s post hoc: P < 0.05) (Fig. 2C). In addition, sex was a significant predictor of KCC2 dysregulation, with decreased levels in males [P < 0.05, (0.031, 0.230)] (Fig. 2C, Supplementary Fig. 3 and Supplementary Table 3). Taken together these results suggest AD+Sz patients had greater GABAergic dysfunction due to reduced tonic inhibition and indications of depolarizing GABAAR-mediated signals.

Dysregulation of proteins involved in excitatory:inhibitory balance in Alzheimer’s disease patients with and without seizure history. (A) Representative western blot images for B–E, showing non-adjacent bands originating from the same blot of the temporal cortex from control cases (Con; n = 13) and Alzheimer’s disease patients (AD; n = 30) split into subgroups: those without (AD−Sz; n = 19) or with (AD+Sz; n = 11) known seizure history. Semi-quantitative analysis of (B) GABAAR subunits GABAARα1 and GABAARα3, and corresponding ratio GABAARα1/GABAARα3; (C) Cl− cotransporters NKCC1 and KCC2, and corresponding ratio NKCC1/KCC2; (D) AMPAR subunits GluA1 and GluA2, and corresponding ratio GluA1/GluA2; (E) NMDAR subunits GluN2A and GluN2B, and corresponding ratio GluN2B/GluN2A. *P < 0.05, **P < 0.01. (F) Pearson correlations for all AD samples, showing the relationship between GluN2A and Clinical Dementia Rating-Sum of Boxes (CDR-SOB) score and (G) the relationship between GABAARα1 expression and brain weight at death in grams in AD+Sz patients. Grey areas indicate 95% confidence interval for the two means.
Figure 2

Dysregulation of proteins involved in excitatory:inhibitory balance in Alzheimer’s disease patients with and without seizure history. (A) Representative western blot images for BE, showing non-adjacent bands originating from the same blot of the temporal cortex from control cases (Con; n = 13) and Alzheimer’s disease patients (AD; n = 30) split into subgroups: those without (AD−Sz; n = 19) or with (AD+Sz; n = 11) known seizure history. Semi-quantitative analysis of (B) GABAAR subunits GABAARα1 and GABAARα3, and corresponding ratio GABAARα1/GABAARα3; (C) Cl cotransporters NKCC1 and KCC2, and corresponding ratio NKCC1/KCC2; (D) AMPAR subunits GluA1 and GluA2, and corresponding ratio GluA1/GluA2; (E) NMDAR subunits GluN2A and GluN2B, and corresponding ratio GluN2B/GluN2A. *P < 0.05, **P < 0.01. (F) Pearson correlations for all AD samples, showing the relationship between GluN2A and Clinical Dementia Rating-Sum of Boxes (CDR-SOB) score and (G) the relationship between GABAARα1 expression and brain weight at death in grams in AD+Sz patients. Grey areas indicate 95% confidence interval for the two means.

To explore inhibitory dysfunction further, we compared levels of the predominant, parvalbumin (PV)-expressing interneuron subtype65 in tissues from AD+Sz and AD−Sz, given that there is a well-established association between PV cell loss, cognition66 and epilepsy.67,68 Immunohistochemistry revealed that the AD+Sz group had significantly decreased PV+ cells compared with controls (Tukey’s post hoc: P < 0.01) (Supplementary Fig. 4A and B) that was not found in AD−Sz, which drove the significant decrease in PV+ cells found between all AD and control samples (P < 0.01) (Supplementary Fig. 4A and B).

To determine if neuronal excitation may be heightened in AD and whether seizures cause further alterations, we examined the expression of AMPA and NMDA glutamate receptor subunit proteins, namely ratios of GluA1/GluA2 and GluN2B/GluN2A subunits27,28,30 (Fig. 2D and E). While no differences were found between AD+Sz and AD−Sz in AMPAR subunits or subunit ratios (Fig. 2D), we found increased expression of GluA2 subunits in AD temporal cortex overall compared with controls (P < 0.05) (Fig. 2D), and the corresponding GluA1/GluA2 ratio was decreased in the AD cases compared with controls (Tukey’s post hoc: P < 0.05) (Fig. 2D). In examining NMDAR subunits, there was no difference in GluN2B or GluN2A levels nor in the ratio of GluN2B/GluN2A between the AD+Sz and AD−Sz groups (Fig. 2E). However, GluN2A and GluN2B subunits were both expressed at significantly higher levels in AD temporal cortex when compared with controls (P < 0.05) (Fig. 2E).

Finally, we determined correlations between clinical outcomes of AD and markers of E:I balance. We found that CDR-SOB was inversely correlated with GluN2A levels across all AD subjects (r = −0.39, P < 0.05) (Fig. 2F), while gross brain atrophy ratings at autopsy were associated with decreased levels of GABAARα1 in AD+Sz (r = 0.90, P < 0.01) (Fig. 2G), indicating an interaction between these markers of E:I balance and cognitive decline and AD severity, respectively. No correlations were found between markers of E:I balance and pathology previously measured in these samples.14 While AD patients were significantly older than controls at time of death [AD: mean = 75.6, standard deviation (SD) = 9.7; control: mean = 62.4, SD = 8.0; P < 0.001], the post-mortem interval (PMI) did not differ between the control and AD groups, and multiple linear regression revealed no significant relationships between age at death, disease duration or PMI and any marker of E:I balance (Supplementary Table 3). These results provided strong evidence of decreased inhibition and increased excitation in AD patients overall, regardless of seizure history, and an exacerbation of selective regulators to worsen GABAergic dysfunction in the AD+Sz group.

Excitatory:inhibitory imbalance in prodromal stage 5XFAD mice

Given that our human tissue samples are limited by the fact they are from patients with late-stage AD (Supplementary Table 1), we used the 5XFAD mouse model to examine earlier stages. Indeed, there is evidence of E:I imbalance in AD mouse models at early stages.69-71 We determined whether baseline E:I imbalance is present in prodromal stage (4 months of age) 5XFAD mice, when mild behavioural impairment,72 AD pathology,73 subclinical epileptiform electroencephalographic activity74 and decreased thresholds for chemoconvulsant-induced seizures14 are present.

To examine inhibitory synaptic function, we measured miniature inhibitory postsynaptic currents (mIPSCs) in CA1 pyramidal neurons in hippocampal slices from prodromal 5XFAD mice using whole-cell patch clamp recordings. We found decreased mIPSC amplitudes [P < 0.0001, n = 11 694 (WT) and 10 948 (5XFAD) events, 20 cells/group, 3 mice/group] (Fig. 3A–C) and increased mIPSC decay time [P < 0.001, n = 7477 (WT) and 6857 (5XFAD) events, 20 neurons/group, 3 mice/group] (Fig. 3A, D and E) in 5XFAD mice compared with WT mice, suggesting that decreased GABAARα1/GABAARα3 ratios21,22 underlie inhibitory synaptic dysfunction. In addition, we found increased inter-mIPSC intervals (reduced frequency) in 5XFAD compared with WT [P < 0.0001, n = 11 673 (WT) and 10 929 (5XFAD) events, 20 neurons/group, 3 mice/group] (Fig. 3A, F and G), indicating reduced inhibitory presynaptic inputs. We also examined the excitatory synaptic function of CA1 neurons by recording miniature excitatory postsynaptic currents (mEPSCs). mEPSC amplitudes were significantly increased in 5XFAD [P < 0.0001, n = 15 619 (WT) and 11 082 (5XFAD) events, 19 (WT) and 21 (5XFAD) neurons, 3 mice/group] (Fig. 3H–J), suggesting increased postsynaptic glutamate receptor densities. mEPSCs frequencies were significantly decreased in 5XFAD, as demonstrated by increased inter-mEPSC intervals [P < 0.0001, n = 15 600 (WT) and 11 060 (5XFAD), 19 (WT) and 21 (5XFAD) neurons, 3 mice/group] (Fig. 3H, K and L), consistent with decreased excitatory presynaptic drive. These changes in synaptic function were also associated with increased intrinsic excitability of CA1 pyramidal neurons in 5XFAD mice as measured by recording evoked action potentials (APs) in current clamp mode using depolarizing step currents (Supplementary Fig. 5). Together, these data demonstrated reduced inhibitory activity and overall hyperexcitability, which may underlie the increased seizure susceptibility we previously found in prodromal 5XFAD mice.14

Excitation:inhibition dysfunction in CA1 pyramidal neurons from prodromal 5XFAD mice. (A) Representative miniature inhibitory postsynaptic current (mIPSC) traces from wild-type (WT) and 5XFAD CA1 neurons. (B and C) Quantifications of mIPSC amplitudes. (D and E) Quantifications of mIPSC decay times. (F and G) Quantifications of inter-mIPSC intervals. n = 20 cells from three WT mice, 20 cells from three 5XFAD mice. ****P < 0.0001. (H) Representative miniature excitatory postsynaptic current (mEPSC) traces from WT and 5XFAD CA1 neurons. (I and J) Quantifications of mEPSC amplitudes. (K and L) Quantifications of inter-mEPSC intervals. n = 19 cells from three WT mice, 21 cells from three 5XFAD mice, ****P < 0.0001.
Figure 3

Excitation:inhibition dysfunction in CA1 pyramidal neurons from prodromal 5XFAD mice. (A) Representative miniature inhibitory postsynaptic current (mIPSC) traces from wild-type (WT) and 5XFAD CA1 neurons. (B and C) Quantifications of mIPSC amplitudes. (D and E) Quantifications of mIPSC decay times. (F and G) Quantifications of inter-mIPSC intervals. n = 20 cells from three WT mice, 20 cells from three 5XFAD mice. ****P < 0.0001. (H) Representative miniature excitatory postsynaptic current (mEPSC) traces from WT and 5XFAD CA1 neurons. (I and J) Quantifications of mEPSC amplitudes. (K and L) Quantifications of inter-mEPSC intervals. n = 19 cells from three WT mice, 21 cells from three 5XFAD mice, ****P < 0.0001.

To investigate the protein changes involved in E:I balance, we performed western blot on hippocampi and cortices from prodromal 5XFAD and WT mice (Fig. 4). Consistent with reduced mIPSC amplitude and increased decay times, we found decreased GABAARα1/GABAARα3 ratios in the hippocampus of 5XFAD compared with WT mice (P < 0.05, n = 8/group) (Fig. 4B). We also found a trend towards decreased KCC2 in the cortex (P = 0.09, n = 14–15) (Fig. 4G). Notably, excitatory glutamate AMPAR and NMDAR subunits in hippocampus and cortex remained unchanged at this stage (Fig. 4).

Excitatory:inhibitory (E:I) markers in prodromal 5XFAD mice. (A) Representative western blot images for B–I, showing non-adjacent bands originating from the same blot. Analysis of bands from western blots for E:I markers from the hippocampus (B–E) and cortex (F–I) of 5XFAD and wild-type (WT) mice. (B and F) GABAAR subunits GABAARα1 and GABAARα3 and corresponding GABAARα1/GABAARα3 ratio; (C and G) Cl− cotransporters NKCC1 and KCC2 and corresponding NKCC1/KCC2 ratio; (D and H) AMPAR subunits GluA1 and GluA2 and corresponding GluA1/GluA2 ratio; (E and I) NMDAR subunits GluN2A, GluN2B and corresponding GluN2B/GluN2A ratio. Error bars represent the standard error of the mean. n = 8–15 per group. *P < 0.05.
Figure 4

Excitatory:inhibitory (E:I) markers in prodromal 5XFAD mice. (A) Representative western blot images for BI, showing non-adjacent bands originating from the same blot. Analysis of bands from western blots for E:I markers from the hippocampus (BE) and cortex (FI) of 5XFAD and wild-type (WT) mice. (B and F) GABAAR subunits GABAARα1 and GABAARα3 and corresponding GABAARα1/GABAARα3 ratio; (C and G) Cl cotransporters NKCC1 and KCC2 and corresponding NKCC1/KCC2 ratio; (D and H) AMPAR subunits GluA1 and GluA2 and corresponding GluA1/GluA2 ratio; (E and I) NMDAR subunits GluN2A, GluN2B and corresponding GluN2B/GluN2A ratio. Error bars represent the standard error of the mean. n = 8–15 per group. *P < 0.05.

Kindling exacerbates the progressive dysregulation of proteins involved in E:I balance in 5XFAD mice

Given the findings of enhanced E:I imbalance in human tissue from AD patients with seizures, we sought to determine how E:I balance changes with disease progression in 5XFAD mice and whether chronic seizures are associated with further dysregulation of E:I imbalance. We performed PTZ seizure kindling or control protocols in prodromal (3.5-month-old) 5XFAD and littermate WT mice and analysed the expression levels of markers of E:I imbalance in the symptomatic (7-month-old) hippocampus (Fig. 5) and cortex (Supplementary Fig. 6). 5XFAD mice had decreased hippocampal GABAARα1/α3 ratios compared with WT mice, regardless of treatment group [genotype effect: F(1,61) = 22.76, P < 0.0001] (Fig. 5A), with significant decreases in both non-kindled (Tukey’s post hoc: P < 0.05) and PTZ-kindled 5XFAD mice (Tukey’s post hoc: P < 0.01). No differences in GABAARα1/α3 ratios were found due to kindling in either 5XFAD or WT mice. The changes in the hippocampal GABAAR ratio seem to be driven largely by decreases in the GABAARα1 subunit in 5XFAD mice [genotype effect: F(1,61) = 40.09, P < 0.0001] (Fig. 5A). In addition, sex was a significant predictor of hippocampal GABAARα1/α3 with greater decreases found in males [P < 0.05 (0.0156, 0.1591)] (Supplementary Fig. 7). Decreased GABAARα1/α3 ratios were also found in the cortex [interaction: F(1,43) = 4.25, P < 0.05], with a significant reduction in non-kindled 5XFAD mice compared with WT (Tukey’s post hoc: P < 0.05) (Supplementary Fig. 6A). These GABAAR changes were similar to patterns seen in human AD brain (Fig. 2) and prodromal (4-month) 5XFAD tissue (Fig. 4), demonstrating persistent GABAAR alterations into symptomatic disease stages in 5XFAD mice.

Excitatory:inhibitory imbalance in the hippocampus of 5XFAD mice following induced seizures. (A–E) Quantification of (A) GABAAR subunits GABAARα1 and GABAARα3 and corresponding ratio GABAARα1/GABAARα3; (B) Cl−cotransporters NKCC1 and KCC2 and corresponding ratio NKCC1/KCC2; (C) AMPAR subunits GluA1 and GluA2 and corresponding ratio GluA1/GluA2; and (D) NMDAR subunits GluN2A and GluN2B and corresponding ratio GluN2B/GluN2A. (E) Representative western blot images for A–D showing non-adjacent bands originating from the same blot. (F) Pearson correlation of pentylenetetrazol (PTZ)-kindled 5XFAD mice showing the relationship between NKCC1/KCC2 and Aβ normalized to 5XFAD-vehicle (Veh), (G) pTau AT100 (Thr212, Ser14)/Tau normalized to wild-type (WT)-Veh (H) and percentage of spontaneous alternations in the Y-maze. Group comparisons for Aβ ELISA, pTau western blots and the Y-maze have been published previously14 (H). Grey areas indicate 95% confidence interval for the two means. n = 12–19 WT-vehicle, 12–17 WT-PTZ, 12 5XFAD-vehicle and 17 5XFAD-PTZ. *P < 0.05, **P < 0.01. Females and males are designated by square and triangle data-points, respectively, where sex effects were found.
Figure 5

Excitatory:inhibitory imbalance in the hippocampus of 5XFAD mice following induced seizures. (A–E) Quantification of (A) GABAAR subunits GABAARα1 and GABAARα3 and corresponding ratio GABAARα1/GABAARα3; (B) Clcotransporters NKCC1 and KCC2 and corresponding ratio NKCC1/KCC2; (C) AMPAR subunits GluA1 and GluA2 and corresponding ratio GluA1/GluA2; and (D) NMDAR subunits GluN2A and GluN2B and corresponding ratio GluN2B/GluN2A. (E) Representative western blot images for AD showing non-adjacent bands originating from the same blot. (F) Pearson correlation of pentylenetetrazol (PTZ)-kindled 5XFAD mice showing the relationship between NKCC1/KCC2 and Aβ normalized to 5XFAD-vehicle (Veh), (G) pTau AT100 (Thr212, Ser14)/Tau normalized to wild-type (WT)-Veh (H) and percentage of spontaneous alternations in the Y-maze. Group comparisons for Aβ ELISA, pTau western blots and the Y-maze have been published previously14 (H). Grey areas indicate 95% confidence interval for the two means. n = 12–19 WT-vehicle, 12–17 WT-PTZ, 12 5XFAD-vehicle and 17 5XFAD-PTZ. *P < 0.05, **P < 0.01. Females and males are designated by square and triangle data-points, respectively, where sex effects were found.

We next tested whether there were age-related and/or kindling-induced alterations to Cl cotransporters in 5XFAD mice, given their critical role in regulating GABAergic inhibition. Kindling resulted in an increased NKCC1/KCC2 ratio in the hippocampus of symptomatic 5XFAD mice [interaction: F(1,61) = 5.78, P < 0.05] compared with WT and non-kindled 5XFAD mice (Tukey’s post hoc: P < 0.01, 0.05) (Fig. 5B), due to a significant increase in expression of the Cl importer, NKCC1 [interaction: F(1,61) = 7.93, P < 0.01, Tukey’s post hoc: P < 0.01] (Fig. 5B). 5XFAD mice showed decreased hippocampal expression of the Cl exporter, KCC2, regardless of PTZ treatment [genotype effect: F(1,61) = 12.35, P < 0.001], which was not seen in prodromal 5XFAD mice, suggesting worsened inhibitory deficits commensurate with pathological progression (Table 1). In the cortex (Supplementary Fig. 6), we also found a significant increase in NKCC1/KCC2 ratios due to PTZ kindling [kindling effect: F(1,43) = 4.49, P < 0.05], which was largely driven by a reduction of KCC2 in kindled WT mice [interaction: F(1,43) = 6.75, P < 0.05]. Interestingly, while cortical NKCC1/KCC2 ratios were not significantly different, there was a decrease in NKCC1 expression [genotype effect: F(1,43) = 5.2, P < 0.05] in non-kindled 5XFAD compared with non-kindled WT mice (Tukey’s post hoc: P < 0.05) (Supplementary Fig. 6B), which may indicate a compensatory mechanism. These data suggest that depolarizing GABA75 may contribute to the exacerbation of GABAergic dysfunction in the kindled 5XFAD hippocampus.

Table 1

Effects of Alzheimer’s disease and 5XFAD genotype on excitation:inhibition imbalance

Molecular markerBrain regionProdromal 5XFADSymptomatic 5XFADHuman AD
GABAARHippocampus−GABAARα1↓GABAARα1
−GABAARα3−GABAARα3
↓GABAARα1/α3↓GABAARα1/α3
Cortex−GABAARα1↓GABAARα1↓GABAARα1
−GABAARα3↓GABAARα3−GABAARα3
−GABAARα1/α3↓GABAARα1/α3↓GABAARα1/α3
Cl cotransportersHippocampus−NKCC1−NKCC1
−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2
Cortex−NKCC1↓NKCC1−NKCC1
−KCC2−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2−NKCC1/KCC2
AMPARHippocampus−GluA1−GluA1
−GluA2↓GluA2
−GluA1/GluA2−GluA1/GluA2
Cortex−GluA1−GluA1−GluA1
−GluA2↓GluA2↑GluA2
−GluA1/GluA2−GluA1/GluA2↓GluA1/GluA2
NMDARHippocampus−GluN2A↓GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Cortex−GluN2A↓GluN2A↑GluN2A
−GluN2B−GluN2B↑GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A−GluN2B/GluN2A
Molecular markerBrain regionProdromal 5XFADSymptomatic 5XFADHuman AD
GABAARHippocampus−GABAARα1↓GABAARα1
−GABAARα3−GABAARα3
↓GABAARα1/α3↓GABAARα1/α3
Cortex−GABAARα1↓GABAARα1↓GABAARα1
−GABAARα3↓GABAARα3−GABAARα3
−GABAARα1/α3↓GABAARα1/α3↓GABAARα1/α3
Cl cotransportersHippocampus−NKCC1−NKCC1
−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2
Cortex−NKCC1↓NKCC1−NKCC1
−KCC2−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2−NKCC1/KCC2
AMPARHippocampus−GluA1−GluA1
−GluA2↓GluA2
−GluA1/GluA2−GluA1/GluA2
Cortex−GluA1−GluA1−GluA1
−GluA2↓GluA2↑GluA2
−GluA1/GluA2−GluA1/GluA2↓GluA1/GluA2
NMDARHippocampus−GluN2A↓GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Cortex−GluN2A↓GluN2A↑GluN2A
−GluN2B−GluN2B↑GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A−GluN2B/GluN2A

Prodromal = 4 months old; symptomatic = 7 months old. AD = Alzheimer’s disease.

Table 1

Effects of Alzheimer’s disease and 5XFAD genotype on excitation:inhibition imbalance

Molecular markerBrain regionProdromal 5XFADSymptomatic 5XFADHuman AD
GABAARHippocampus−GABAARα1↓GABAARα1
−GABAARα3−GABAARα3
↓GABAARα1/α3↓GABAARα1/α3
Cortex−GABAARα1↓GABAARα1↓GABAARα1
−GABAARα3↓GABAARα3−GABAARα3
−GABAARα1/α3↓GABAARα1/α3↓GABAARα1/α3
Cl cotransportersHippocampus−NKCC1−NKCC1
−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2
Cortex−NKCC1↓NKCC1−NKCC1
−KCC2−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2−NKCC1/KCC2
AMPARHippocampus−GluA1−GluA1
−GluA2↓GluA2
−GluA1/GluA2−GluA1/GluA2
Cortex−GluA1−GluA1−GluA1
−GluA2↓GluA2↑GluA2
−GluA1/GluA2−GluA1/GluA2↓GluA1/GluA2
NMDARHippocampus−GluN2A↓GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Cortex−GluN2A↓GluN2A↑GluN2A
−GluN2B−GluN2B↑GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A−GluN2B/GluN2A
Molecular markerBrain regionProdromal 5XFADSymptomatic 5XFADHuman AD
GABAARHippocampus−GABAARα1↓GABAARα1
−GABAARα3−GABAARα3
↓GABAARα1/α3↓GABAARα1/α3
Cortex−GABAARα1↓GABAARα1↓GABAARα1
−GABAARα3↓GABAARα3−GABAARα3
−GABAARα1/α3↓GABAARα1/α3↓GABAARα1/α3
Cl cotransportersHippocampus−NKCC1−NKCC1
−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2
Cortex−NKCC1↓NKCC1−NKCC1
−KCC2−KCC2−KCC2
−NKCC1/KCC2−NKCC1/KCC2−NKCC1/KCC2
AMPARHippocampus−GluA1−GluA1
−GluA2↓GluA2
−GluA1/GluA2−GluA1/GluA2
Cortex−GluA1−GluA1−GluA1
−GluA2↓GluA2↑GluA2
−GluA1/GluA2−GluA1/GluA2↓GluA1/GluA2
NMDARHippocampus−GluN2A↓GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Cortex−GluN2A↓GluN2A↑GluN2A
−GluN2B−GluN2B↑GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A−GluN2B/GluN2A

Prodromal = 4 months old; symptomatic = 7 months old. AD = Alzheimer’s disease.

We next determined whether glutamate receptors may be dysregulated as a function of disease progression and by kindled seizures in 5XFAD mice. In the hippocampus, we found a significant effect to increase GluA1/GluA2 ratios in PTZ-kindled hippocampus [Genotype × Kindling: F(1,61) = 4.622, P < 0.05, Tukey’s post hoc: P < 0.01] (Fig. 5C), which contributes to E:I imbalance in epilepsy.30 In addition, both subunits were decreased in the hippocampus of 5XFAD mice regardless of kindling status [GluA1 genotype effect: F(1,61) = 7.722, P < 0.01; GluA2 genotype effect: F(1,61) = 30.28, P < 0.0001] (Fig. 5C), and similar alterations in AMPAR subunit expression and ratios were found in the cortex (Supplementary Fig. 6C). The NMDAR GluN2A subunit was also decreased in 5XFAD hippocampus [genotype effect: F(1,61) = 6.31, P < 0.05] (Fig. 5D) and cortex [genotype effect: F(1,43) = 9.0, P < 0.01, Tukey’s post hoc: P < 0.05] (Supplementary Fig. 6D) without change GluN2B and subunit ratios. These data demonstrate dysregulation to glutamate receptor expression beginning by 7 months of age in 5XFAD mice and an exacerbation of this effect due to PTZ kindling.

In addition, we used our retrospective measures of disease severity in this same cohort of mice14 and found that NKCC1/KCC2 was positively correlated with Aβ42 (r = 0.57, P < 0.05) and pTau AT100 (Thr212, Ser214)/Tau (r = 0.64, P < 0.05) in the hippocampus of PTZ-kindled 5XFAD mice and with performance on the Y-maze (r = −0.61, P < 0.01) (Fig. 5F–H), which we previously demonstrated was worsened by PTZ kindling.14 No correlations were found in non-kindled mice nor between GABAAR, AMPAR or NMDAR in kindled mice. These findings suggest a relationship between inhibitory dysregulation, AD pathology and cognitive decline.

In summary, 5XFAD mice showed a worsening of markers of inhibitory imbalance, which was associated with worsened pathology and cognitive decline, and the emergence of dysregulated glutamate receptors as pathology progresses from prodromal to later stages (Table 1). Furthermore, PTZ kindling exacerbated imbalance in both excitatory and inhibitory markers (Table 2).

Table 2

Effects of seizures in Alzheimer’s disease and 5XFAD mice

Molecular markerBrain region5XFAD + KindlingHuman AD + Seizures
GABAARHippocampus−GABAAR α1
−GABAAR α 3
−GABAAR α1/α3
Cortex−GABAAR α1↓GABAARα1
−GABAAR α 3↓GABAARα3
−GABAAR α1/α3−GABAAR α1/α3
Cl cotransportersHippocampus↑NKCC1
−KCC2
↑NKCC1/KCC2
Cortex−NKCC1−NKCC1
−KCC2↓KCC2
−NKCC1/KCC2↑NKCC1/KCC2
AMPARHippocampus−GluA1
−GluA2
↑GluA1/GluA2
Cortex−GluA1−GluA1
−GluA2−GluA2
↑GluA1/GluA2−GluA1/GluA2
NMDARHippocampus−GluN2A
−GluN2B
−GluN2B/GluN2A
Cortex−GluN2A−GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Molecular markerBrain region5XFAD + KindlingHuman AD + Seizures
GABAARHippocampus−GABAAR α1
−GABAAR α 3
−GABAAR α1/α3
Cortex−GABAAR α1↓GABAARα1
−GABAAR α 3↓GABAARα3
−GABAAR α1/α3−GABAAR α1/α3
Cl cotransportersHippocampus↑NKCC1
−KCC2
↑NKCC1/KCC2
Cortex−NKCC1−NKCC1
−KCC2↓KCC2
−NKCC1/KCC2↑NKCC1/KCC2
AMPARHippocampus−GluA1
−GluA2
↑GluA1/GluA2
Cortex−GluA1−GluA1
−GluA2−GluA2
↑GluA1/GluA2−GluA1/GluA2
NMDARHippocampus−GluN2A
−GluN2B
−GluN2B/GluN2A
Cortex−GluN2A−GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A

AD = Alzheimer’s disease.

Table 2

Effects of seizures in Alzheimer’s disease and 5XFAD mice

Molecular markerBrain region5XFAD + KindlingHuman AD + Seizures
GABAARHippocampus−GABAAR α1
−GABAAR α 3
−GABAAR α1/α3
Cortex−GABAAR α1↓GABAARα1
−GABAAR α 3↓GABAARα3
−GABAAR α1/α3−GABAAR α1/α3
Cl cotransportersHippocampus↑NKCC1
−KCC2
↑NKCC1/KCC2
Cortex−NKCC1−NKCC1
−KCC2↓KCC2
−NKCC1/KCC2↑NKCC1/KCC2
AMPARHippocampus−GluA1
−GluA2
↑GluA1/GluA2
Cortex−GluA1−GluA1
−GluA2−GluA2
↑GluA1/GluA2−GluA1/GluA2
NMDARHippocampus−GluN2A
−GluN2B
−GluN2B/GluN2A
Cortex−GluN2A−GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A
Molecular markerBrain region5XFAD + KindlingHuman AD + Seizures
GABAARHippocampus−GABAAR α1
−GABAAR α 3
−GABAAR α1/α3
Cortex−GABAAR α1↓GABAARα1
−GABAAR α 3↓GABAARα3
−GABAAR α1/α3−GABAAR α1/α3
Cl cotransportersHippocampus↑NKCC1
−KCC2
↑NKCC1/KCC2
Cortex−NKCC1−NKCC1
−KCC2↓KCC2
−NKCC1/KCC2↑NKCC1/KCC2
AMPARHippocampus−GluA1
−GluA2
↑GluA1/GluA2
Cortex−GluA1−GluA1
−GluA2−GluA2
↑GluA1/GluA2−GluA1/GluA2
NMDARHippocampus−GluN2A
−GluN2B
−GluN2B/GluN2A
Cortex−GluN2A−GluN2A
−GluN2B−GluN2B
−GluN2B/GluN2A−GluN2B/GluN2A

AD = Alzheimer’s disease.

mTORC1 inhibition modifies select seizure-induced E:I markers in the hippocampus of 5XFAD mice

Rapamycin treatment has rescued neuronal hyperexcitability in various disease models.55-60 Thus, we sought to determine whether chronic low-dose rapamycin treatment beginning at the cessation of PTZ kindling (prodromal stage ∼4 months, 2.24 mg/kg daily), previously used to rescue seizure-induced neuropathology and cognitive deficits in 5XFAD mice,14 could ameliorate PTZ-exacerbated E:I imbalance. The 5XFAD (Fig. 6) and WT (Supplementary Fig. 8) mice used in our previous report were analysed separately by two-way ANOVA, with PTZ kindling and rapamycin treatment as independent variables.

Differential effects of mTORC1 inhibition on seizure-induced excitation:inhibition imbalance in 5XFAD mice. (A–D) Quantification of (A) GABAARα1 and GABAARα3 and corresponding GABAARα1/GABAARα3 ratio; (B) Cl− cotransporters NKCC1 and KCC2 and corresponding NKCC1/KCC2 ratio; (C) AMPAR subunits GluA1 and GluA2 and corresponding ratio GluA1/GluA2; and (D) NMDAR subunits GluN2A and GluN2B and corresponding GluN2B/GluN2A ratio. (E) Representative western blot images for A–D showing non-adjacent bands originating from the same blot. n = 12–13 for each group. *P < 0.05, **P < 0.01.
Figure 6

Differential effects of mTORC1 inhibition on seizure-induced excitation:inhibition imbalance in 5XFAD mice. (AD) Quantification of (A) GABAARα1 and GABAARα3 and corresponding GABAARα1/GABAARα3 ratio; (B) Cl cotransporters NKCC1 and KCC2 and corresponding NKCC1/KCC2 ratio; (C) AMPAR subunits GluA1 and GluA2 and corresponding ratio GluA1/GluA2; and (D) NMDAR subunits GluN2A and GluN2B and corresponding GluN2B/GluN2A ratio. (E) Representative western blot images for AD showing non-adjacent bands originating from the same blot. n = 12–13 for each group. *P < 0.05, **P < 0.01.

In 5XFAD mice, rapamycin showed a strong trend (Tukey’s post hoc P = 0.058) to increase GABAARα1/GABAARα3 in non-kindled mice compared with the non-kindled, vehicle-treated group [kindling effect: F(1,45) = 6.1, P < 0.05; rapamycin effect: F(1,45) = 4.1, P < 0.05] (Fig. 6A). These shifts corresponded with GABAARα1 subunit expression [kindling effect: F(1,45) = 4.2, P < 0.05; rapamycin effect: F(1,45) = 6.1, P < 0.05] (Fig. 6A). Rapamycin also reversed the elevation of the NKCC1/KCC2 ratio found in PTZ-kindled 5XFAD mice [interaction: F(1,45) = 9.8, P < 0.01] compared with control 5XFAD mice (Tukey’s post hoc: P < 0.05) (Fig. 6B). The changes in Cl cotransporter ratio was largely driven by changes in the Cl importer, NKCC1 [interaction: F(1,45) = 7.95, P < 0.01] (Fig. 6B). These results likely reflect a rescue of GABAAR function in kindled 5XFAD mice by rapamycin.

The analysis of AMPAR subunits showed that PTZ kindling increased GluA1/GluA2 ratios in symptomatic 5XFAD mice (Tukey’s post hoc: P < 0.01) (Fig. 6C), consistent with the non-therapeutic cohort, which was reversed by rapamycin treatment [interaction: F(1,45) = 19.83, P < 0.01] (Fig. 6C). Rapamycin increased GluA1/GluA2 ratio in non-kindled 5XFAD mice (Tukey’s post hoc: P < 0.05), largely driven by increased GluA1 expression [interaction: F(1,45) = 4.3, P < 0.05] (Fig. 6C). Additionally, rapamycin treatment reduced GluN2A and GluN2B levels in untreated and PTZ-kindled mice [GluN2A rapamycin effect: F(1,45) = 5.2, P < 0.05; GluN2B rapamycin effect: F(1,45) = 4.1, P < 0.05] (Fig. 6D). PTZ-kindled, rapamycin treated 5XFAD mice had elevated GluN2B/GluN2A ratio compared with control 5XFAD mice [kindling effect: F(1,45) = 5.66, P < 0.05; Tukey’s post hoc: P < 0.05] (Fig. 6D).

In WT mice, no significant differences were found due to PTZ kindling or rapamycin treatment for GABAARs, AMPARs or NMDARs (Supplementary Fig. 8), although kindling did increase NKCC1 expression in WT mice across rapamycin and vehicle treatments [kindling effect: F(1,47) = 5.2, P < 0.05] (Supplementary Fig. 8B). Overall, these data suggested that rapamycin can ameliorate PTZ-exacerbated E:I imbalance in 5XFAD mice.

Discussion

Here we identified novel mechanisms of neuronal dysfunction that may underlie seizure-exacerbated cognitive and functional deficits in AD. Human AD temporal cortex showed dysregulation of E:I balance proteins, which was worsened in those with seizure history. Similarly, physiological and protein indications of neuronal E:I imbalance were found in prodromal 5XFAD mice, and further perturbations to synaptic protein regulators were seen at symptomatic stages, with additional exacerbations by PTZ-kindling (Tables 1 and 2). Together with our recent study demonstrating enhanced AD pathology in AD+Sz patients and PTZ-kindled mice,14 our data suggest that E:I imbalance begins at prodromal stages and may play a role in seizure-induced exacerbation of neuropathology and cognitive decline in AD. The most consistent E:I alterations include decreased GABAARα1 subunit expression and shifts in the balance between the Cl cotransporters NKCC1/KCC2 and the AMPAR GluA1/GluA2 subunit ratios as factors in this interaction between AD and seizures. Furthermore, our data indicate that using mTORC1 inhibitors such as rapamycin to therapeutically target the E:I imbalance may prove to be a promising therapy to alleviate AD progression due to neuronal hyperexcitability.

Our rare autopsy-confirmed AD cases with prospective collection of seizure history data suggest that seizures are associated with worsened cognitive function and E:I imbalance. We also found that markers of E:I balance correlated with clinical measures of decline and disease severity in these tissue samples. Importantly, with a limited sample size, we found seizures were significant predictors of clinical outcomes across separate statistical models when either sex, age at test, disease duration or education level were accounted for, justifying follow-up studies to further examine clinical and biochemical outcomes associated with seizure history in AD with more comprehensive statistical modelling that was not possible in the dataset here. It is important to note that the reports of seizure history for patients in this study did not contain information on the aetiology of seizures, age of onset (whether seizures began before or after AD diagnosis), extent of seizure severity, nor whether the seizures were controlled. Even with these limitations, when taken with our prior reports in these patients,14 our data suggest that seizures significantly worsen AD disease progression, indicating that AD patients should be more closely monitored for seizures, thus providing more comprehensive descriptions of seizure activity and history that were not available for the current study.

Here we found a specific decrease in GABAARα1/GABAARα3 ratios in AD patients compared with controls, supporting previous reports of reduced GABA sensitivity in membranes isolated from the temporal cortex of AD brains.38 Indeed, previous studies have found marked alterations in synaptic GABAergic signalling in AD, including downregulation of GABAAR subunits37,38,76,77 and decreased expression of inhibitory synapses overall.78,79 We show that AD patients with seizure history expressed significantly lower GABAARα1 and GABAARα3 subunits with GABAARα1 levels correlating with brain atrophy in AD patients with seizures. In addition, NKCC1/KCC2 ratios were increased in AD patients with a history of seizures as compared with those without, consistent with collapse of the neuronal Cl gradient. We also found a loss of PV-containing interneurons in the temporal cortex of AD patients compared with controls, consistent with the reported reduction of PV cells within the AD parahippocampal gyrus,80 which was driven by the AD+Sz group. Fast-spiking PV interneurons are major contributors to generation of gamma oscillations, which are disrupted in AD patients.81,82 Taken in the context of prior studies, these data suggest that a history of seizures in AD is associated with diminished GABAergic transmission via reduced GABAAR synapses, loss of interneurons and possibly a depolarizing GABA response at those synapses that remain due to elevated intracellular Cl. Greater loss of these GABAergic interneurons in AD patients with epilepsy may promote network hyperexcitability and contribute to the worsened cognitive and functional performance found in these subjects (Fig. 1).

Electrophysiological recordings in AD models, and 5XFAD mice in particular, are sparse, especially at stages in which plaque pathology has developed. Our electrophysiological data from the CA1 of prodromal 5XFAD mice demonstrated significantly increased network excitability as well as deficits in inhibitory synaptic transmission as evidenced by reductions in mIPSC amplitude and frequency as well as increased decay times. Consistent with the functional inhibitory changes, we found decreased GABAARα1/GABAARα3 ratios in the hippocampus of prodromal 5XFAD mice, GABAAR configurations previously shown to alter IPSC amplitude21 and decay time.22 These results align with prior studies showing reductions in GABARα1 and α5 transcripts in patients with mild cognitive impairment, suggesting GABAAR vulnerability at initial disease stages.83

When taken with the prodromal 5XFAD data, non-kindled 7-month-old 5XFAD mice showed a worsening of GABAAR dysregulation with reduced GABAARα1/GABAARα3 in the cortex in addition to the hippocampus (Table 1). We and others have also found decreased PV immunoreactivity,14,84-87 suggesting a broader deficit of inhibition and reflecting our results in AD temporal cortex. Indeed, GABAAR loss has been linked to seizures and cognitive impairment in mouse models of AD.88-90 This mechanism may be responsible for such changes in the 5XFAD model. Consistent with our human data, we found that induced seizures may exaggerate GABAergic dysfunction through increased NKCC1/KCC2 ratios, which were reversed in rapamycin-treated mice.

With respect to glutamatergic neurotransmission, studies of AD hippocampus highlighted decreased expression of GluA2 compared with controls,91-93 a configuration known to increase AMPAR Ca2+ permeability.27,28,30,94,95 Notably, the human AD temporal cortex we examined was late stage (Supplementary Table 1) and we instead found an increase in GluA2 with a decrease in corresponding GluA1/GluA2 ratio in AD compared with controls (Fig. 1E). These changes may represent a compensatory mechanism to neuronal hyperexcitability, but the significant GluA2 elevation may actually result in further impairments, given that Ca2+-permeable AMPAR is critical for the induction of late-phase long-term potentiation, essential for the formation of long-term memory,96,97 as seen in a neurodevelopmental disorder model.98 We also found an increase in NMDAR subunits in AD patients, which is consistent with studies of the AD parietal cortex demonstrating an increased excitatory to inhibitory synaptic ratio.99 Elevated GluN2A in AD patients may again represent a compensatory mechanism, as lower GluN2A was associated with worsened CDR-SOB scores across all AD patients in our dataset, consistent with correlations in working memory performance and GluN2A in aged rats.100 Elevated GluN2B is associated with epileptogenesis15,30 and extrasynaptic excitotoxic signalling in AD.101 Furthermore, elevations in AMPAR and NMDAR subunits are suggestive of elevated glutamatergic synapses overall, which may further tip E:I balance towards excitation.

While changes in AMPAR and NMDAR were not found in prodromal 5XFAD mice, we found elevated EPSC amplitude and increased intrinsic excitability of CA1 neurons, which are the primary outputs of hippocampus. Together with decreased GABAergic signals, this suggests increased excitability of the hippocampal network overall. At later, symptomatic stages we found increased expression of GluA1/GluA2 AMPAR subunit in PTZ kindled 5XFAD mice which was ameliorated with rapamycin treatment. Overall, these results suggest that AMPAR and NMDAR subunit composition is dynamic through AD progression and certain changes could be compensatory or neuroprotective from excitotoxicity, as often seen in acute epilepsy models.102,103

Recent epidemiologic research has underscored strong associations between epilepsy, late-onset seizures and AD.1,104-108 We and others have provided increasing preclinical and clinical evidence that seizures significantly contribute to neuropathology and cognitive decline and may also be a treatable component of this complex disease. Indeed, in a recent phase 2a clinical trial, levetiracetam, an antiepileptic drug, was shown to improve memory and executive function in AD patients with epileptiform activity.109 Beyond seizure suppression, our data suggest that targeting specific E:I imbalances also holds therapeutic potential. Notably, recent studies have identified neuronal Cl cotransporters as candidates to treat AD. Our data demonstrated significant exacerbation of NKCC1/KCC2 in both AD+Sz and seizure-kindled 5XFAD mice, although the contributing factor differed between these datasets, with AD+Sz showing significantly decreased KCC2, while seizure-kindled 5XFAD mice had elevated NKCC1. These discrepancies may be due to the brain region examined or disease stage, with our post-mortem tissue being late disease stage (Supplementary Table 1) and symptomatic, 7-month 5XFAD mice being an intermediate stage of neurodegeneration. The therapeutic strategy may depend on which alteration is found: the clinically available agent, bumetanide, antagonizes NKCC1, while CLP290 has been shown to stabilize membrane KCC2 in mouse models.41,110 Of note, both have been used to restore physiological neuronal activity and rescue cognitive deficits in AD models.41,111 Furthermore, NKCC1/KCC2 ratios were positively correlated with AD pathology and negatively correlated with performance related to spatial working memory in kindled 5XFAD mice, supporting previous literature linking neuronal excitability and AD progression.112 Overall, these data suggest that restoration of the Cl gradient may be therapeutically relevant, particularly for AD patients with seizure history.

We recently demonstrated that seizures exacerbate mTORC1 in AD patients and 5XFAD mice and that chronic low-dose rapamycin treatment is sufficient to ameliorate AD pathology and cognitive dysfunction in PTZ-kindled 5XFAD mice.14 Rapamycin is FDA-approved and has been proven to benefit both AD and epilepsy models.14,113,114 While rapalogues may affect numerous signalling pathways downstream of mTORC1 to attenuate the disease course, including autophagy and neuroinflammation,115,116 growing evidence demonstrates efficacy in regulating neuronal hyperexcitability.55-60 In the studies presented here, rapamycin reduced NKCC1/KCC2 and GluA1/GluA2 ratios in PTZ-kindled 5XFAD mice and showed a trend towards increased GABAARα1/GABAARα3 in non-kindled 5XFAD mice, suggesting a reversal of inhibitory dysfunction, which may also indicate restoration of the proper circuitry involved in memory rescue in these mice.14 However, the chronic rapamycin treatment induced small but significant elevations in GluN2B/GluN2A in PTZ-kindled 5XFAD mice and increased GluA1/GluA2 in non-kindled 5XFAD mice. In contrast, studies that used acute rapamycin treatments demonstrated that mTORC1 blockade decreases GluN2B and GluA1.117-120 Thus, the relative increases that we found in GluN2B and GluA1 in later stage mice may represent a compensatory mechanism in response to chronic rapamycin administration. Indeed, one prior study demonstrated that chronic rapamycin induces increased surface GluN2B, and that these changes are associated with amelioration of age-dependent cognitive decline.121 These data indicate that targeting E:I balance with rapamycin should be explored for clinical efficacy in AD patients with seizure history.

In summary, the data presented here identify novel mechanisms of neuronal dysfunction in AD that are amplified by seizures and may play a role in worsened cognitive outcomes associated with seizures in AD. Importantly, our data indicate the importance of taking a seizure history at a minimum in patients with AD. In addition, these data suggest that targeting E:I imbalance, perhaps with rapamycin as well as other antiepileptic agents, may hold therapeutic promise in AD patients with comorbid seizures.

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

Acknowledgements

The authors thank the CNDR at the University of Pennsylvania for supplying the human tissue used in these studies as well as the University of Maryland Brain and Tissue Bank, part of the NIH Neuro Biobank for additional control brain samples. We would also like to thank Dr Robert Vassar for supplying some of the 4-month 5XFAD mouse tissue.

Funding

These studies were supported by the National Institutes of Health (NIH) National Institute of Neurological Disorders and Stroke (NINDS): R21NS105437 (F.E.J.), R37NS115439 (F.E.J.), R01NS101156 (D.M.T.), National Institute on Aging (NIA): R01AG077692 (F.E.J. and D.M.T.) and T32AG000255 (A.J.B.), Alzheimer’s Association AARF-22-972333 (A.J.B.). Clinical and post-mortem tissue and data from Alzheimer’s disease patients were obtained from National Institutes of Health studies, including P30-AG-072979 (formerly AG010124), U19 AG062418 (formerly P50 NS053488), P01-AG-066597 and R01-AG-054519.

Competing interests

The authors report no competing interests.

Supplementary material

Supplementary material is available at Brain online.

References

1

Vossel
KA
,
Tartaglia
MC
,
Nygaard
HB
,
Zeman
AZ
,
Miller
BL
.
Epileptic activity in Alzheimer’s disease: Causes and clinical relevance
.
Lancet Neurol
.
2017
;
16
:
311
322
.

2

Vossel
KA
,
Ranasinghe
KG
,
Beagle
AJ
, et al.
Incidence and impact of subclinical epileptiform activity in Alzheimer’s disease
.
Ann Neurol
.
2016
;
80
:
858
870
.

3

Horvath
AA
,
Papp
A
,
Zsuffa
J
, et al.
Subclinical epileptiform activity accelerates the progression of Alzheimer’s disease: A long-term EEG study
.
Clin Neurophysiol
.
2021
;
132
:
1982
1989
.

4

Baker
J
,
Libretto
T
,
Henley
W
,
Zeman
A
.
A longitudinal study of epileptic seizures in Alzheimer’s disease
.
Front Neurol
.
2019
;
10
:
1266
.

5

Voglein
J
,
Ricard
I
,
Noachtar
S
, et al.
Seizures in Alzheimer's disease are highly recurrent and associated with a poor disease course
.
J Neurol
.
2020
;
267
:
2941
2948
.

6

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
:
191
209
.

7

Thom
M
,
Liu
JY
,
Thompson
P
, et al.
Neurofibrillary tangle pathology and Braak staging in chronic epilepsy in relation to traumatic brain injury and hippocampal sclerosis: A post-mortem study
.
Brain
.
2011
;
134
:
2969
2981
.

8

Tai
XY
,
Koepp
M
,
Duncan
JS
, et al.
Hyperphosphorylated tau in patients with refractory epilepsy correlates with cognitive decline: A study of temporal lobe resections
.
Brain
.
2016
;
139
(
Pt 9
):
2441
2455
.

9

Palop
JJ
,
Mucke
L
.
Epilepsy and cognitive impairments in Alzheimer disease
.
Arch Neurol
.
2009
;
66
:
435
440
.

10

Westmark
CJ
,
Westmark
PR
,
Beard
AM
,
Hildebrandt
SM
,
Malter
JS
.
Seizure susceptibility and mortality in mice that over-express amyloid precursor protein
.
Int J Clin Exp Pathol
.
2008
;
1
:
157
168
.

11

DeVos
SL
,
Goncharoff
DK
,
Chen
G
, et al.
Antisense reduction of tau in adult mice protects against seizures
.
J Neurosci
.
2013
;
33
:
12887
12897
.

12

Johnson
ECB
,
Ho
K
,
Yu
GQ
, et al.
Behavioral and neural network abnormalities in human APP transgenic mice resemble those of App knock-in mice and are modulated by familial Alzheimer’s disease mutations but not by inhibition of BACE1
.
Mol Neurodegener
.
2020
;
15
:
53
.

13

Chang
CW
,
Evans
MD
,
Yu
X
,
Yu
GQ
,
Mucke
L
.
Tau reduction affects excitatory and inhibitory neurons differently, reduces excitation/inhibition ratios, and counteracts network hypersynchrony
.
Cell Rep
.
2021
;
37
:
109855
.

14

Gourmaud
S
,
Stewart
DA
,
Irwin
DJ
, et al.
The role of mTORC1 activation in seizure-induced exacerbation of Alzheimer’s disease
.
Brain
.
2022
;
145
:
324
339
.

15

Rakhade
SN
,
Jensen
FE
.
Epileptogenesis in the immature brain: Emerging mechanisms
.
Nat Rev Neurol
.
2009
;
5
:
380
391
.

16

Bakker
J
,
Basedow
FJ
,
Dekker
AD
,
Papantoniou
C
.
Phosphorylation of AMPA-type glutamate receptors: The trigger of epileptogenesis?
J Neurosci
.
2013
;
33
:
5879
5880
.

17

Liu
R
,
Wang
J
,
Liang
S
,
Zhang
G
,
Yang
X
.
Role of NKCC1 and KCC2 in epilepsy: From expression to function
.
Front Neurol
.
2019
;
10
:
1407
.

18

Blair
RE
,
Sombati
S
,
Lawrence
DC
,
McCay
BD
,
DeLorenzo
RJ
.
Epileptogenesis causes acute and chronic increases in GABAA receptor endocytosis that contributes to the induction and maintenance of seizures in the hippocampal culture model of acquired epilepsy
.
J Pharmacol Exp Ther
.
2004
;
310
:
871
880
.

19

Noebels
JL
,
Avoli
M
,
Rogawski
M
,
Olsen
R
,
Delgado-Escueta
AV
.
“Jasper's Basic Mechanisms of the Epilepsies” workshop
.
Epilepsia
.
2010
;
51
:
1
5
.

20

Brooks-Kayal
AR
,
Shumate
MD
,
Jin
H
,
Rikhter
TY
,
Coulter
DA
.
Selective changes in single cell GABA(A) receptor subunit expression and function in temporal lobe epilepsy
.
Nat Med
.
1998
;
4
:
1166
1172
.

21

Zhou
C
,
Huang
Z
,
Ding
L
, et al.
Altered cortical GABAA receptor composition, physiology, and endocytosis in a mouse model of a human genetic absence epilepsy syndrome
.
J Biol Chem
.
2013
;
288
:
21458
21472
.

22

Eyre
MD
,
Renzi
M
,
Farrant
M
,
Nusser
Z
.
Setting the time course of inhibitory synaptic currents by mixing multiple GABA(A) receptor alpha subunit isoforms
.
J Neurosci
.
2012
;
32
:
5853
5867
.

23

Talos
DM
,
Sun
H
,
Kosaras
B
, et al.
Altered inhibition in tuberous sclerosis and type IIb cortical dysplasia
.
Ann Neurol
.
2012
;
71
:
539
551
.

24

Palma
E
,
Amici
M
,
Sobrero
F
, et al.
Anomalous levels of Cl- transporters in the hippocampal subiculum from temporal lobe epilepsy patients make GABA excitatory
.
Proc Natl Acad Sci U S A
.
2006
;
103
:
8465
8468
.

25

Barmashenko
G
,
Hefft
S
,
Aertsen
A
,
Kirschstein
T
,
Kohling
R
.
Positive shifts of the GABAA receptor reversal potential due to altered chloride homeostasis is widespread after status epilepticus
.
Epilepsia
.
2011
;
52
:
1570
1578
.

26

Kourdougli
N
,
Pellegrino
C
,
Renko
JM
, et al.
Depolarizing gamma-aminobutyric acid contributes to glutamatergic network rewiring in epilepsy
.
Ann Neurol
.
2017
;
81
:
251
265
.

27

Lippman-Bell
JJ
,
Zhou
C
,
Sun
H
,
Feske
JS
,
Jensen
FE
.
Early-life seizures alter synaptic calcium-permeable AMPA receptor function and plasticity
.
Mol Cell Neurosci
.
2016
;
76
:
11
20
.

28

Rakhade
SN
,
Zhou
C
,
Aujla
PK
,
Fishman
R
,
Sucher
NJ
,
Jensen
FE
.
Early alterations of AMPA receptors mediate synaptic potentiation induced by neonatal seizures
.
J Neurosci
.
2008
;
28
:
7979
7990
.

29

Egbenya
DL
,
Hussain
S
,
Lai
YC
,
Xia
J
,
Anderson
AE
,
Davanger
S
.
Changes in synaptic AMPA receptor concentration and composition in chronic temporal lobe epilepsy
.
Mol Cell Neurosci
.
2018
;
92
:
93
103
.

30

Loddenkemper
T
,
Talos
DM
,
Cleary
RT
, et al.
Subunit composition of glutamate and gamma-aminobutyric acid receptors in status epilepticus
.
Epilepsy Res
.
2014
;
108
:
605
615
.

31

Di Maio
R
,
Mastroberardino
PG
,
Hu
X
,
Montero
L
,
Greenamyre
JT
.
Pilocapine alters NMDA receptor expression and function in hippocampal neurons: NADPH oxidase and ERK1/2 mechanisms
.
Neurobiol Dis
.
2011
;
42
:
482
495
.

32

Di Maio
R
,
Mastroberardino
PG
,
Hu
X
,
Montero
LM
,
Greenamyre
JT
.
Thiol oxidation and altered NR2B/NMDA receptor functions in in vitro and in vivo pilocarpine models: Implications for epileptogenesis
.
Neurobiol Dis
.
2013
;
49
:
87
98
.

33

Canitano
R
,
Palumbi
R
.
Excitation/inhibition modulators in autism spectrum disorder: Current clinical research
.
Front Neurosci
.
2021
;
15
:
753274
.

34

Grigoras
IF
,
Stagg
CJ
.
Recent advances in the role of excitation-inhibition balance in motor recovery post-stroke
.
Fac Rev
.
2021
;
10
:
58
.

35

Lopatina
OL
,
Malinovskaya
NA
,
Komleva
YK
, et al.
Excitation/inhibition imbalance and impaired neurogenesis in neurodevelopmental and neurodegenerative disorders
.
Rev Neurosci
.
2019
;
30
:
807
820
.

36

Palop
JJ
,
Mucke
L
.
Network abnormalities and interneuron dysfunction in Alzheimer disease
.
Nat Rev Neurosci
.
2016
;
17
:
777
792
.

37

Kwakowsky
A
,
Calvo-Flores Guzman
B
,
Pandya
M
,
Turner
C
,
Waldvogel
HJ
,
Faull
RL
.
GABAA receptor subunit expression changes in the human Alzheimer’s disease hippocampus, subiculum, entorhinal cortex and superior temporal gyrus
.
J Neurochem
.
2018
;
145
:
374
392
.

38

Limon
A
,
Reyes-Ruiz
JM
,
Miledi
R
.
Loss of functional GABA(A) receptors in the Alzheimer diseased brain
.
Proc Natl Acad Sci U S A
.
2012
;
109
:
10071
10076
.

39

Lam
P
,
Vinnakota
C
,
Guzman
BC
, et al.
Beta-amyloid (Abeta1-42) increases the expression of NKCC1 in the mouse hippocampus
.
Molecules
.
2022
;
27
:
2440
.

40

Lagostena
L
,
Rosato-Siri
M
,
D’Onofrio
M
, et al.
In the adult hippocampus, chronic nerve growth factor deprivation shifts GABAergic signaling from the hyperpolarizing to the depolarizing direction
.
J Neurosci
.
2010
;
30
:
885
893
.

41

Keramidis
I
,
McAllister
BB
,
Bourbonnais
J
, et al.
Restoring neuronal chloride extrusion reverses cognitive decline linked to Alzheimer’s disease mutations
.
Brain
.
2023
;
146
:
4903
4915
.

42

Hara
Y
,
Punsoni
M
,
Yuk
F
, et al.
Synaptic distributions of GluA2 and PKMzeta in the monkey dentate gyrus and their relationships with aging and memory
.
J Neurosci
.
2012
;
32
:
7336
7344
.

43

Guntupalli
S
,
Widagdo
J
,
Anggono
V
.
Amyloid-beta-induced dysregulation of AMPA receptor trafficking
.
Neural Plast
.
2016
;
2016
:
3204519
.

44

Sanderson
JL
,
Freund
RK
,
Gorski
JA
,
Dell'Acqua
ML
.
β-Amyloid disruption of LTP/LTD balance is mediated by AKAP150-anchored PKA and Calcineurin regulation of Ca2+-permeable AMPA receptors
.
Cell Rep
.
2021
;
37
:
109786
.

45

Sun
H
,
Takesian
AE
,
Wang
TT
,
Lippman-Bell
JJ
,
Hensch
TK
,
Jensen
FE
.
Early seizures prematurely unsilence auditory synapses to disrupt thalamocortical critical period plasticity
.
Cell Rep
.
2018
;
23
:
2533
2540
.

46

Caccamo
A
,
Magri
A
,
Medina
DX
, et al.
mTOR regulates tau phosphorylation and degradation: Implications for Alzheimer’s disease and other tauopathies
.
Aging Cell
.
2013
;
12
:
370
380
.

47

Pei
JJ
,
An
WL
,
Zhou
XW
, et al.
P70 S6 kinase mediates tau phosphorylation and synthesis
.
FEBS Lett
.
2006
;
580
:
107
114
.

48

Oddo
S
.
The role of mTOR signaling in Alzheimer disease
.
Front Biosci (Schol Ed)
.
2012
;
4
:
941
952
.

49

Cai
Z
,
Chen
G
,
He
W
,
Xiao
M
,
Yan
LJ
.
Activation of mTOR: A culprit of Alzheimer’s disease?
Neuropsychiatr Dis Treat
.
2015
;
11
:
1015
1030
.

50

Wang
JZ
,
Xia
YY
,
Grundke-Iqbal
I
,
Iqbal
K
.
Abnormal hyperphosphorylation of tau: Sites, regulation, and molecular mechanism of neurofibrillary degeneration
.
J Alzheimers Dis
.
2013
;
33 Suppl 1
:
S123
S139
.

51

McCabe
MP
,
Cullen
ER
,
Barrows
CM
, et al.
Genetic inactivation of mTORC1 or mTORC2 in neurons reveals distinct functions in glutamatergic synaptic transmission
.
Elife
.
2020
;
9
:
e51440
.

52

Kim
SG
,
Lee
S
,
Kim
Y
, et al.
Tanc2-mediated mTOR inhibition balances mTORC1/2 signaling in the developing mouse brain and human neurons
.
Nat Commun
.
2021
;
12
:
2695
.

53

Talos
DM
,
Kwiatkowski
DJ
,
Cordero
K
,
Black
PM
,
Jensen
FE
.
Cell-specific alterations of glutamate receptor expression in tuberous sclerosis complex cortical tubers
.
Ann Neurol
.
2008
;
63
:
454
465
.

54

Wu
X
,
Sosunov
AA
,
Lado
W
, et al.
Synaptic hyperexcitability of cytomegalic pyramidal neurons contributes to epileptogenesis in tuberous sclerosis complex
.
Cell Rep
.
2022
;
40
:
111085
.

55

Amegandjin
CA
,
Choudhury
M
,
Jadhav
V
, et al.
Sensitive period for rescuing parvalbumin interneurons connectivity and social behavior deficits caused by TSC1 loss
.
Nat Commun
.
2021
;
12
:
3653
.

56

Bateup
HS
,
Johnson
CA
,
Denefrio
CL
,
Saulnier
JL
,
Kornacker
K
,
Sabatini
BL
.
Excitatory/inhibitory synaptic imbalance leads to hippocampal hyperexcitability in mouse models of tuberous sclerosis
.
Neuron
.
2013
;
78
:
510
522
.

57

Koene
LM
,
Niggl
E
,
Wallaard
I
,
Proietti-Onori
M
,
Rotaru
DC
,
Elgersma
Y
.
Identifying the temporal electrophysiological and molecular changes that contribute to TSC-associated epileptogenesis
.
JCI Insight
.
2021
;
6
:
e150120
.

58

Goto
J
,
Talos
DM
,
Klein
P
, 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
E1079
.

59

Iezzi
D
,
Curti
L
,
Ranieri
G
, et al.
Acute rapamycin rescues the hyperexcitable phenotype of accumbal medium spiny neurons in the valproic acid rat model of autism spectrum disorder
.
Pharmacol Res
.
2022
;
183
:
106401
.

60

Ma
X
,
Du
W
,
Wang
W
, et al.
Persistent Rheb-induced mTORC1 activation in spinal cord neurons induces hypersensitivity in neuropathic pain
.
Cell Death Dis
.
2020
;
11
:
747
.

61

Xie
SX
,
Baek
Y
,
Grossman
M
, et al.
Building an integrated neurodegenerative disease database at an academic health center
.
Alzheimers Dement
.
2011
;
7
:
e84
e93
.

62

Hyman
BT
,
Phelps
CH
,
Beach
TG
, et al.
National Institute on Aging-Alzheimer’s Association guidelines for the neuropathologic assessment of Alzheimer’s disease
.
Alzheimers Dement
.
2012
;
8
:
1
13
.

63

Besser
L
,
Kukull
W
,
Knopman
DS
, et al.
Version 3 of the National Alzheimer's Coordinating Center’s Uniform Data Set
.
Alzheimer Dis Assoc Disord
.
2018
;
32
:
351
358
.

64

Galanopoulou
AS
.
GABA(a) receptors in normal development and seizures: Friends or foes?
Curr Neuropharmacol
.
2008
;
6
:
1
20
.

65

Rudy
B
,
Fishell
G
,
Lee
S
,
Hjerling-Leffler
J
.
Three groups of interneurons account for nearly 100% of neocortical GABAergic neurons
.
Dev Neurobiol
.
2011
;
71
:
45
61
.

66

Murray
AJ
,
Woloszynowska-Fraser
MU
,
Ansel-Bollepalli
L
, et al.
Parvalbumin-positive interneurons of the prefrontal cortex support working memory and cognitive flexibility
.
Sci Rep
.
2015
;
5
:
16778
.

67

Drexel
M
,
Preidt
AP
,
Kirchmair
E
,
Sperk
G
.
Parvalbumin interneurons and calretinin fibers arising from the thalamic nucleus reuniens degenerate in the subiculum after kainic acid-induced seizures
.
Neuroscience
.
2011
;
189
:
316
329
.

68

Zamecnik
J
,
Krsek
P
,
Druga
R
, et al.
Densities of parvalbumin-immunoreactive neurons in non-malformed hippocampal sclerosis-temporal neocortex and in cortical dysplasias
.
Brain Res Bull
.
2006
;
68
:
474
481
.

69

Kim
KR
,
Kim
Y
,
Jeong
HJ
, et al.
Impaired pattern separation in Tg2576 mice is associated with hyperexcitable dentate gyrus caused by Kv4.1 downregulation
.
Mol Brain
.
2021
;
14
:
62
.

70

Kazim
SF
,
Chuang
SC
,
Zhao
W
,
Wong
RK
,
Bianchi
R
,
Iqbal
K
.
Early-onset network hyperexcitability in presymptomatic Alzheimer’s disease transgenic mice is suppressed by passive immunization with anti-human APP/abeta antibody and by mGluR5 blockade
.
Front Aging Neurosci
.
2017
;
9
:
71
.

71

Davis
KE
,
Fox
S
,
Gigg
J
.
Increased hippocampal excitability in the 3xTgAD mouse model for Alzheimer's disease in vivo
.
PLoS One
.
2014
;
9
:
e91203
.

72

Kimura
R
,
Ohno
M
.
Impairments in remote memory stabilization precede hippocampal synaptic and cognitive failures in 5XFAD Alzheimer mouse model
.
Neurobiol Dis
.
2009
;
33
:
229
235
.

73

Oakley
H
,
Cole
SL
,
Logan
S
, et al.
Intraneuronal beta-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer’s disease mutations: Potential factors in amyloid plaque formation
.
J Neurosci
.
2006
;
26
:
10129
10140
.

74

Abe
Y
,
Ikegawa
N
,
Yoshida
K
, et al.
Behavioral and electrophysiological evidence for a neuroprotective role of aquaporin-4 in the 5xFAD transgenic mice model
.
Acta Neuropathol Commun
.
2020
;
8
:
67
.

75

Schulte
JT
,
Wierenga
CJ
,
Bruining
H
.
Chloride transporters and GABA polarity in developmental, neurological and psychiatric conditions
.
Neurosci Biobehav Rev
.
2018
;
90
:
260
271
.

76

Govindpani
K
,
Turner
C
,
Waldvogel
HJ
,
Faull
RLM
,
Kwakowsky
A
.
Impaired expression of GABA signaling components in the Alzheimer’s disease middle temporal gyrus
.
Int J Mol Sci
.
2020
;
21
:
8704
.

77

Rissman
RA
,
Mishizen-Eberz
AJ
,
Carter
TL
, et al.
Biochemical analysis of GABA(A) receptor subunits alpha 1, alpha 5, beta 1, beta 2 in the hippocampus of patients with Alzheimer's disease neuropathology
.
Neuroscience
.
2003
;
120
:
695
704
.

78

Garcia-Marin
V
,
Blazquez-Llorca
L
,
Rodriguez
JR
, et al.
Diminished perisomatic GABAergic terminals on cortical neurons adjacent to amyloid plaques
.
Front Neuroanat
.
2009
;
3
:
28
.

79

Agarwal
S
,
Tannenberg
RK
,
Dodd
PR
.
Reduced expression of the inhibitory synapse scaffolding protein gephyrin in Alzheimer's disease
.
J Alzheimers Dis
.
2008
;
14
:
313
321
.

80

Sanchez-Mejias
E
,
Nunez-Diaz
C
,
Sanchez-Varo
R
, et al.
Distinct disease-sensitive GABAergic neurons in the perirhinal cortex of Alzheimer’s mice and patients
.
Brain Pathol
.
2020
;
30
:
345
363
.

81

Stam
CJ
,
van Cappellen van Walsum
AM
,
Pijnenburg
YA
, et al.
Generalized synchronization of MEG recordings in Alzheimer’s disease: Evidence for involvement of the gamma band
.
J Clin Neurophysiol
.
2002
;
19
:
562
574
.

82

Klein
AS
,
Donoso
JR
,
Kempter
R
,
Schmitz
D
,
Beed
P
.
Early cortical changes in gamma oscillations in Alzheimer’s disease
.
Front Syst Neurosci
.
2016
;
10
:
83
.

83

Rissman
RA
,
Bennett
DA
,
Armstrong
DM
.
Subregional analysis of GABA(A) receptor subunit mRNAs in the hippocampus of older persons with and without cognitive impairment
.
J Chem Neuroanat
.
2004
;
28
:
17
25
.

84

Ali
F
,
Baringer
SL
,
Neal
A
,
Choi
EY
,
Kwan
AC
.
Parvalbumin-positive neuron loss and amyloid-beta deposits in the frontal cortex of Alzheimer's disease-related mice
.
J Alzheimers Dis
.
2019
;
72
:
1323
1339
.

85

Flanigan
TJ
,
Xue
Y
,
Kishan Rao
S
,
Dhanushkodi
A
,
McDonald
MP
.
Abnormal vibrissa-related behavior and loss of barrel field inhibitory neurons in 5xFAD transgenics
.
Genes Brain Behav
.
2014
;
13
:
488
500
.

86

Giesers
NK
,
Wirths
O
.
Loss of hippocampal calretinin and parvalbumin interneurons in the 5XFAD mouse model of Alzheimer's disease
.
ASN Neuro
.
2020
;
12
:
1759091420925356
.

87

Caccavano
A
,
Bozzelli
PL
,
Forcelli
PA
, et al.
Inhibitory parvalbumin basket cell activity is selectively reduced during hippocampal sharp wave ripples in a mouse model of familial Alzheimer’s disease
.
J Neurosci
.
2020
;
40
:
5116
5136
.

88

Verret
L
,
Mann
EO
,
Hang
GB
, et al.
Inhibitory interneuron deficit links altered network activity and cognitive dysfunction in Alzheimer model
.
Cell
.
2012
;
149
:
708
721
.

89

Li
Y
,
Zhu
K
,
Li
N
, et al.
Reversible GABAergic dysfunction involved in hippocampal hyperactivity predicts early-stage Alzheimer disease in a mouse model
.
Alzheimers Res Ther
.
2021
;
13
:
114
.

90

Ulrich
D
.
Amyloid-beta impairs synaptic inhibition via GABA(A) receptor endocytosis
.
J Neurosci
.
2015
;
35
:
9205
9210
.

91

Carter
TL
,
Rissman
RA
,
Mishizen-Eberz
AJ
, et al.
Differential preservation of AMPA receptor subunits in the hippocampi of Alzheimer’s disease patients according to Braak stage
.
Exp Neurol
.
2004
;
187
:
299
309
.

92

Ikonomovic
MD
,
Mizukami
K
,
Davies
P
,
Hamilton
R
,
Sheffield
R
,
Armstrong
DM
.
The loss of GluR2(3) immunoreactivity precedes neurofibrillary tangle formation in the entorhinal cortex and hippocampus of Alzheimer brains
.
J Neuropathol Exp Neurol
.
1997
;
56
:
1018
1027
.

93

Mohamed
NE
,
Zhao
Y
,
Lee
JH
, et al.
Upregulation of AMPA receptor GluR2 (GluA2) subunits in subcortical ischemic vascular dementia is repressed in the presence of Alzheimer’s disease
.
Neurochem Int
.
2011
;
58
:
820
825
.

94

Burnashev
N
,
Monyer
H
,
Seeburg
PH
,
Sakmann
B
.
Divalent ion permeability of AMPA receptor channels is dominated by the edited form of a single subunit
.
Neuron
.
1992
;
8
:
189
198
.

95

Rajasekaran
K
,
Todorovic
M
,
Kapur
J
.
Calcium-permeable AMPA receptors are expressed in a rodent model of status epilepticus
.
Ann Neurol
.
2012
;
72
:
91
102
.

96

Park
P
,
Kang
H
,
Sanderson
TM
, et al.
The role of calcium-permeable AMPARs in long-term potentiation at principal neurons in the rodent hippocampus
.
Front Synaptic Neurosci
.
2018
;
10
:
42
.

97

Pang
PT
,
Lu
B
.
Regulation of late-phase LTP and long-term memory in normal and aging hippocampus: Role of secreted proteins tPA and BDNF
.
Ageing Res Rev
.
2004
;
3
:
407
430
.

98

Banke
TG
,
Barria
A
.
Transient enhanced GluA2 expression in young hippocampal neurons of a fragile X mouse model
.
Front Synaptic Neurosci
.
2020
;
12
:
588295
.

99

Lauterborn
JC
,
Scaduto
P
,
Cox
CD
, et al.
Increased excitatory to inhibitory synaptic ratio in parietal cortex samples from individuals with Alzheimer’s disease
.
Nat Commun
.
2021
;
12
:
2603
.

100

McQuail
JA
,
Beas
BS
,
Kelly
KB
, et al.
NR2A-containing NMDARs in the prefrontal cortex are required for working memory and associated with age-related cognitive decline
.
J Neurosci
.
2016
;
36
:
12537
12548
.

101

Babaei
P
.
NMDA and AMPA receptors dysregulation in Alzheimer’s disease
.
Eur J Pharmacol
.
2021
;
908
:
174310
.

102

Russo
I
,
Bonini
D
,
Via
LL
,
Barlati
S
,
Barbon
A
.
AMPA receptor properties are modulated in the early stages following pilocarpine-induced status epilepticus
.
Neuromolecular Med
.
2013
;
15
:
324
338
.

103

Leo
A
,
Giovannini
G
,
Russo
E
,
Meletti
S
.
The role of AMPA receptors and their antagonists in status epilepticus
.
Epilepsia
.
2018
;
59
:
1098
1108
.

104

Zhang
D
,
Chen
S
,
Xu
S
, et al.
The clinical correlation between Alzheimer’s disease and epilepsy
.
Front Neurol
.
2022
;
13
:
922535
.

105

Lam
AD
,
Sarkis
RA
,
Pellerin
KR
, et al.
Association of epileptiform abnormalities and seizures in Alzheimer disease
.
Neurology
.
2020
;
95
:
e2259
e2270
.

106

Lam
AD
,
Deck
G
,
Goldman
A
,
Eskandar
EN
,
Noebels
J
,
Cole
AJ
.
Silent hippocampal seizures and spikes identified by foramen ovale electrodes in Alzheimer’s disease
.
Nat Med
.
2017
;
23
:
678
680
.

107

Johnson
EL
,
Krauss
GL
,
Kucharska-Newton
A
, et al.
Dementia in late-onset epilepsy: The Atherosclerosis Risk in Communities study
.
Neurology
.
2020
;
95
:
e3248
e3256
.

108

Kawakami
O
,
Koike
Y
,
Ando
T
, et al.
Incidence of dementia in patients with adult-onset epilepsy of unknown causes
.
J Neurol Sci
.
2018
;
395
:
71
76
.

109

Vossel
K
,
Ranasinghe
KG
,
Beagle
AJ
, et al.
Effect of levetiracetam on cognition in patients with Alzheimer disease with and without epileptiform activity: A randomized clinical trial
.
JAMA Neurol
.
2021
;
78
:
1345
1354
.

110

Barbour
AJ
,
Nass
SR
,
Hahn
YK
,
Hauser
KF
,
Knapp
PE
.
Restoration of KCC2 membrane localization in striatal dopamine D2 receptor-expressing medium spiny neurons rescues locomotor deficits in HIV tat-transgenic mice
.
ASN Neuro
.
2021
;
13
:
17590914211022089
.

111

Taubes
A
,
Nova
P
,
Zalocusky
KA
, et al.
Experimental and real-world evidence supporting the computational repurposing of bumetanide for APOE4-related Alzheimer’s disease
.
Nature Aging
.
2021
;
1
:
932
947
.

112

Targa Dias Anastacio
H
,
Matosin
N
,
Ooi
L
.
Neuronal hyperexcitability in Alzheimer’s disease: What are the drivers behind this aberrant phenotype?
Transl Psychiatry
.
2022
;
12
:
257
.

113

Kim
JK
,
Lee
JH
.
Mechanistic target of rapamycin pathway in epileptic disorders
.
J Korean Neurosurg Soc
.
2019
;
62
:
272
287
.

114

Kaeberlein
M
,
Galvan
V
.
Rapamycin and Alzheimer’s disease: Time for a clinical trial?
Sci Transl Med
.
2019
;
11
:
eaar4289
.

115

Kodali
M
,
Attaluri
S
,
Madhu
LN
, et al.
Metformin treatment in late middle age improves cognitive function with alleviation of microglial activation and enhancement of autophagy in the hippocampus
.
Aging Cell
.
2021
;
20
:
e13277
.

116

Zeng
LH
,
Xu
L
,
Gutmann
DH
,
Wong
M
.
Rapamycin prevents epilepsy in a mouse model of tuberous sclerosis complex
.
Ann Neurol
.
2008
;
63
:
444
453
.

117

James
MH
,
Quinn
RK
,
Ong
LK
, et al.
mTORC1 inhibition in the nucleus accumbens ‘protects’ against the expression of drug seeking and ‘relapse’ and is associated with reductions in GluA1 AMPAR and CAMKIIalpha levels
.
Neuropsychopharmacology
.
2014
;
39
:
1694
1702
.

118

James
MH
,
Quinn
RK
,
Ong
LK
, et al.
Rapamycin reduces motivated responding for cocaine and alters GluA1 expression in the ventral but not dorsal striatum
.
Eur J Pharmacol
.
2016
;
784
:
147
154
.

119

Calabrese
V
,
Di Maio
A
,
Marino
G
, et al.
Rapamycin, by inhibiting mTORC1 signaling, prevents the loss of striatal bidirectional synaptic plasticity in a rat model of L-DOPA-induced dyskinesia
.
Front Aging Neurosci
.
2020
;
12
:
230
.

120

Zhang
Y
,
Ji
F
,
Wang
G
,
He
D
,
Yang
L
,
Zhang
M
.
BDNF activates mTOR to upregulate NR2B expression in the rostral anterior cingulate cortex required for inflammatory pain-related aversion in rats
.
Neurochem Res
.
2018
;
43
:
681
691
.

121

Majumder
S
,
Caccamo
A
,
Medina
DX
, et al.
Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1beta and enhancing NMDA signaling
.
Aging Cell
.
2012
;
11
:
326
335
.

Author notes

Aaron J Barbour and Sarah Gourmaud contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)

Supplementary data