Female patients with late-onset Alzheimer's disease (LOAD) exhibited a more pronounced decline in the ERRα-regulated bioenergetic network within neurons
Previous neuroimaging and blood biomarker studies have suggested that brain hypometabolism contributes to the sex-dimorphic effects observed in LOAD22,23. To examine this in detail, bulk transcriptomic analysis was performed on brain tissue samples from 613 individuals who participated in the ROSMAP study24 - including 133 non-dementia males, 219 non-dementia females, 88 LOAD males, and 173 LOAD females subjects (Fig. 1a-d). The phenotypic status of these samples (LOAD vs. non-dementia) was defined based on multiple clinicopathological features, including neuritic plaque load (CERAD), neurofibrillary tangle pathology (Braak stage), and cognitive status (Cogdx and DCFDX) (Fig. 1a). DEG analysis between sex-specific LOAD versus ND samples revealed a significantly higher number of DEGs in females than males (DEGLOAD vs. ND: female = 6,615 genes; male = 440 genes) (Fig. 1b, Supplementary Table 1). While the small number of differentially expressed genes (DEGs) identified in the male cohort failed to cluster into any meaningful pathways (Supplementary Table 1), the upregulated DEGs in the female LOAD group were enriched for pathways related to neuroinflammation, including TNFα signalling via NF-κB, IL-2/STAT5 signalling, TGFβ signalling, and IL-6/JAK/STAT3 signalling. In contrast, the pathways enriched by the set of downregulated DEGs in females were primarily metabolic, such as oxidative phosphorylation (OXPHOS) and mTORC1 signalling (Fig. 1c).
Among the significant pathways identified, oxidative phosphorylation (OXPHOS) was the highest ranked (smallest p-value). Further analysis of the most likely common transcriptional regulators of the enriched genes suggested estrogen-related receptor alpha (ERR1/ERRα) as a key regulator (Fig. 1d, Supplementary Table 2). ERRα, also known as NR3B1, is a nuclear receptor that shares substantial DNA sequence homology with estrogen receptor alpha (ERα), despite estradiol (E2) being an unlikely endogenous ligand for ERRα25. To further investigate the relevance of ERRα activity in different brain cell types, its relative gene expression level was evaluated in a single-nucleus transcriptomic dataset26, which indicated that neurons generally express the highest levels of the ESRRA transcript (Fig. 1e). The importance of ERRα activities in neurons was further validated by the similar enrichment of PPARGC1A transcript, which encodes its co-activator PGC1α in these cells (Fig. 1e). As compared to non-dementia (ND) controls, while no obvious differences in ESRRA transcript levels were found in LOAD (Supplementary Fig. 1, Note: although p < 0.05, the Log2FC values were all ≤ |0.07|, too small to be considered significant), immunohistochemistry analyses however revealed obvious reductions in nuclear signals of ERRα, particularly among affected female subjects (Fig. 1f). This suggests that the loss of ERRα nuclear activities at the post-translational level may serve as a link to the female-specific changes observed in the disease, potentially related to the dramatic hormonal changes that occur during the menopausal transition.
Cognitive and memory functions correlate with the residual estradiol levels after the induction of accelerated ovarian failure (AOF) which emulates menopause
Menopause in women is characterized by a sharp decline in estrogen (E2) levels within a relatively short period, whereas aging men do not experience a comparable change in testosterone levels27. To investigate how dramatic endocrine changes associated with menopause may affect brain function, menopause was artificially induced in laboratory mice using 4-vinylcyclohexene diepoxide (VCD). VCD is an occupational chemical that causes selective destruction of small pre-antral ovarian follicles by accelerating the natural, apoptotic process of follicular atresia, while leaving the overall ovarian anatomy intact in rodents28,29. Young adult mice (3 months old, postnatal day 90) were chosen for this study, such that any unpredictable confounding effects that could arise from chronological aging could be minimized (Fig. 2a). While the VCD treatment had no noticeable effect on body weight change (Fig. 2b), it resulted in elevated circulating levels of follicle-stimulating hormone (FSH) (Fig. 2b) but an opposite effect to estradiol (Fig. 2d) - the primary form of estrogen during reproductive years. These findings observed on experimental day 90 indicates a successful induction of accelerated ovarian failure, mimicking the menopausal transition in these test animals. To test whether the dramatic endocrine changes initiated in the periphery (i.e., loss of estradiol) may alter brain function, a battery of behavioural tests was performed on the animals. While animals subjected to VCD exposure showed no obvious changes in motor function (Supplementary Fig. 2a-b), they did exhibit declines in spatial learning and memory (Fig. 2e), as well as short-term working memory (Fig. 2f). The estradiol-deficient animals also spent significantly less time in the centre zone of the open-field test, indicating a mild anxiety-like behaviour (Fig. 2g). Notably, the degree of changes in these behavioural measures was correlated with the residual circulating estradiol levels after treatment (Fig. 2e-f). The neuroprotective effects of estradiol were further supported by immunohistochemistry analysis, which revealed obvious neurite degeneration in the hippocampal regions of VCD-treated animals (Fig. 2h). These behavioural and anatomical changes were correlated with declines in neuronal function, as evidenced by significant impairments in field excitatory postsynaptic potentials (fEPSPs) in the Schaffer collateral pathway (Fig. 2i) and long-term potentiation (LTP) (Fig. 2j).
Estradiol depletion impairs of the ERRα signalling network in the brain
The behavioural and electrophysiological findings demonstrated a functional connection between circulating estradiol and the brain. To delineate the details at the cellular and molecular levels, a bulk transcriptomic analysis was performed using total cerebral cortex tissues harvested from the animals. With the brain transcriptome profiles of the VCD-treated and vehicle-treated groups being clearly distinct from one another (Fig. 3a), a total of 342 upregulated and 542 downregulated transcripts were identified (Fig. 3b, Supplementary Table 3). While the upregulated differentially expressed genes (DEGs) were not cluster into any meaningful pathways, many of the significantly downregulated genes were implicated in mitochondrial energetics networks, including oxidative phosphorylation (OXPHOS), thermogenesis, and the citrate cycle (TCA cycle). Additionally, these downregulated genes were associated with a list of neurodegenerative disorders, such as Parkinson's disease, prion disease, Huntington's disease, Alzheimer's disease, and amyotrophic lateral sclerosis (Fig. 3c, Supplementary Table 4). Further gene overlapping analysis suggested that the majority of genes clustered in the OXPHOS pathway were indeed common to those clustered in various neurodegenerative disorders (Fig. 3d). Moreover, subsequent transcription factor analyses of all downregulated DEGs revealed again the ERR1/ERRα as the primary upstream regulator of these genes, with at least one occurrence of the highly conserved ERRα binding motif TGACCTY in the regions spanning 4 kb cantered on their transcription starting sites [-2kb, +2kb] (Fig. 3e). In agreement with the findings observed with human brain tissues (Fig. 1f), protein signals of ERRα, as well as that of its essential co-activator PGC1α were predominantly enriched in neuronal nuclei under normal conditions (Fig. 3f). However, systemic VCD administration triggered a loss of such signals (Fig. 3f). While this was unlikely due to changes at the transcription level (Supplementary Fig. 3a), significant reductions in the protein-protein interactions between ERRα and PGC1α were observed. This may also have promoted the default degradation of the intrinsically disordered PGC1α protein within the cells (Fig. 2g, Supplementary Fig. 3b).
Previous studies have revealed that the ligand-binding domain (LBD) and activation function-2 (AF2) domains of ERRα are involved in interacting with the 3rd LXXLL motif of PGC1α30. Additionally, previous research has shown that the cholesterol binding - the natural ERRα ligand - to the LBD may concurrently enhance the binding affinity between ERRα and PGC1α31, although the precise details remain unclear. By in silico simulation, we provided further structural evidence that helps explain this phenomenon. First, we simulated a complete, open structure of the ERRα ligand-binding domain (Open ERRα) by merging the previously reported, yet incomplete, open structures of the ERRα LBD extracted from the Protein Data Bank (PDB: 7E2E and 2PJL)32,33 (Supplementary Fig. 3c) with the simulated missing alpha-helix segment reconstructed by the AlphaFold234 (Supplementary Fig. 3d). The resulted combined structure then constituted a complete, open LBD structure that allows possible cholesterol binding. We then further docked this cholesterol-bound ERRα conformation against the AlphaFold2-simulated 3rd LXXLL motif (amino acids 208–216) of PGC1α34 using the HADDOCK 2.4 algorithm35. Compared to the docking result generated from the closed, yet complete ERRα LBD structure (PDB: 1XB7)33, which prohibits cholesterol ligand binding (Supplementary Fig. 3c), cholesterol binding to the complete, open LBD structure resulted in conformational remodelling of substructures near the cholesterol-binding pocket (Supplementary Fig. 3e). This remodelling is predicted to facilitate more hydrophobic interactions and hydrogen bond formation between ERRα and the 3rd LXXLL motif of PGC1α, thereby supporting their protein-protein interaction and downstream ERRα transcriptional activities (Fig. 3h). With brain tissues harvested from the AOF mouse model, we found that systematic VCD treatment resulted in diminished levels of cholesterol being bound to ERRα (Fig. 3i). These results hinted that the post-translational modulation on ERRα signalling is likely related to cholesterol availability in these cells.
ERα signalling activates ERRα signalling by sustaining cholesterol homeostasis
The diminished ERRα-bound cholesterol in the AOF mouse model suggested a broad metabolic reprogramming effect, likely which is likely initiated by the dramatic loss of estrogen receptor signalling. Unbiased metabolomics (Fig. 4a-b, Supplementary Table 5) and targeted differential metabolic gene expression analysis (Fig. 4c, Supplementary Table 6) consistently suggested an impairment of cholesterol biosynthesis, as accompanied by disruption of central carbon metabolism (i.e., glycolysis and TCA cycle) (Fig. 4d). In addition to serving as a major energy source, glucose may also contribute carbons to cholesterol biosynthesis in the brain36 through a series of reactions. These include glycolysis and the subsequent conversion of the glycolytic end product pyruvate to acetyl-CoA inside the mitochondria. The acetyl-CoA can then fuse with oxaloacetate to form citrate, which can be re-exported to the cytoplasm. There, citrate lyase can regenerate acetyl-CoA, which can then commit to the mevalonate pathway for cholesterol biosynthesis (Fig. 4d). In the adult brain, the two primary mechanisms for meeting neuronal cholesterol requirements are: (1) the de novo neuronal biosynthesis of cholesterol, and (2) the lipoprotein-mediated transfer of the metabolite from neighbouring glial cells37. To determine if specific ER (estrogen receptor) signalling is involved, the key dysregulated metabolites and genes belonging to the related metabolic steps (as labelled in red in Fig. 4d) were re-evaluated using a mature primary neuronal culture model, which was done with and without treatment using a long-lasting ester derivative of 17β-estradiol (i.e., 100 nM estradiol cypionate, E2) and specific ER antagonists (i.e., ERα: 100 nM MPP dihydrochloride; ERβ: 100 nM PTHPP; GPER: 100 nM G-15) (Fig. 4e). Targeted metabolite analyses revealed that ERα (estrogen receptor alpha) was primarily responsible for mediating the effect of estradiol on neurons (Fig. 4e). Furthermore, based on initial predictions made using the ENCODE, CHEA (Table S7), and ChIP-atlas databases (Supplementary Fig. 4a), chromatin immunoprecipitation analysis showed that 5 of the dysregulated metabolic genes identified from the AOF model were indeed ERα targets (Fig. 4e). These target genes were also having high expression in neurons relative to other brain cell types (Supplementary Fig. 5). These ERα target genes included Pdha1, which encodes a key component of the pyruvate dehydrogenase complex responsible for converting pyruvate to acetyl-CoA; so as the Dhcr24 and Cyp51 genes that respectively encode the 24-dehydrocholeserol reductase and lanosterol 14-alpha demethylase of the cholesterol biosynthesis pathway (Fig. 4e). In addition to the de novo cholesterol biosynthesis process, neurons can also directly uptake cholesterol from the circulating apolipoprotein E (ApoE) chaperones released by the neighbouring astrocytes38. Indeed, gene expression key neuronal ApoE receptors such as Lrp1 and Vldlr were also sensitive to ERα signalling (Fig. 4e, Supplementary Table 7). Further supporting this, ChIP-PCR revealed that estradiol treatment induced robust ERα binding to the predicted binding sites in the promoter regions of these receptor genes, and that such binding could be suppressed by MPP dihydrochloride, a small molecule antagonist that selectively inhibits estradiol binding to ERα (Fig. 4e, Supplementary Fig. 4)39. The selective decline in expression of these genes (except for LRP1) in female patients was validated using the ROSMAP brain transcriptomics data. Furthermore, a greater degree of this gene expression decline was associated with more severe cognitive impairment (Fig. 4g). These data not only revealed the existence of a glucose-cholesterol biosynthetic axis in neurons and its importance for maintaining cognitive function, but also highlighted the upstream regulatory role of ERα signalling in this process. To further validate these findings, a stable isotope tracing analysis was performed to trace down the immediate fates of glucose-13C6 in neurons under the chronic influence of estradiol (Fig. 4h). At 4 hours after the isotope exposure, isotopologue profiling revealed that estradiol pre-programmed the cells to have a robust flux of glucose-derived pyruvate towards mevalonate biosynthesis - the key precursor of cholesterol (Fig. 4h). The necessity of ERα signalling for this effect was further validated, as co-pretreatment of the cells with MPP dihydrochloride effectively prevented the increased flux of glucose-derived metabolites towards mevalonate biosynthesis (Fig. 4h). At the downstream levels of these events, the relative abundances of ERRα-bound cholesterol (Fig. 4i), the robustness of the ERRα interaction with PGC1α (Fig. 4j, Supplementary Fig. 6), and ERRα nuclear transcription activities (Fig. 4k) were all found to be regulated in a similar manner.
Loss of ERRα dysregulates the neuronal NAAG metabolic axis
ERRα is a master regulator of cellular energy metabolism40. However, the detailed metabolic reprogramming events and their impact on cellular resilience against stress, particularly in neurons, remained unclear following the loss of ERRα. To investigate so, adeno-associated virus (AAV) carrying a microRNA-30 (miR30)-based short hairpin (shRNA) to specifically knocked down ERRα specifically in neurons41 was stereotaxically injected into both the medial prefrontal cortex and hippocampal CA1-2 regions of the C57BL/6 mice (Fig. 5a-b, Supplementary Fig. 7a). A battery of behavioural assessments and brain tissue investigation were then conducted at approximately 3 months after the initial injection of the AAV which the lag time allow recovery and the shRNA effect to take place. The successful knockdown of Esrra was first validated by both transcript and protein levels analyses in the injected tissue region, which were dissected and harvested under a fluorescent microscope (Supplementary Fig. 7a-b). As expected, Esrra knockdown in these target brain regions resulted in poorer cognitive and memory performances (Supplementary Fig. 7c). Subsequent brain histological analysis, using the ectopically expressed GFP signals as visual guides, revealed significant neurite losses upon Esrra knockdown (Fig. 5b). This finding at least in part explained the behavioural functional changes observed in these animals. With the GFP + brain tissues enriched, transcriptomic (Fig. 5c-d, Supplementary Table 8) and metabolomic profiling (Fig. 5e, Supplementary Table 9) assays were conducted. The integrated transcriptomic and metabolomic analysis revealed that the TCA cycle was the top affected pathway upon Esrra knockdown in neurons (Fig. 5f). Further profiling of the TCA cycle's series of biochemical reactions revealed diminished abundances of metabolites (i.e., succinate and fumarase) beyond the step catalysed by succinyl-CoA ligase (SUCLG1) (Fig. 5g), hinting that the downstream reactions of the TCA cycle could be affected. As a possible homeostatic response to such challenges, metabolic reprogramming that utilizes aspartate aminotransferase to connect the segment between oxaloacetate (4C) and α-ketoglutarate (5C) was likely enhanced, as suggested by the concomitant reduction of aspartate levels in the system (Fig. 5g). To validate the increased reliance of aspartate to the TCA cycle, isotopologue tracing with labelled 13C4-aspartate was conducted in a primary neuronal culture model, such that the (1) aspartate aminotransferase facilitated transfer of amino group from aspartate (4C) to α-ketoglutarate (5C), (2) the conversion of α-ketoglutarate to glutamate as well as (3) its re-entrance to the TCA cycle via the action of glutamate dehydrogenase could all be detected [Fig. 4h(1)]. The results validated that this "mini version" of the TCA cycle was activated in neurons depleted of functional ERRα, either due to the silencing effect of Esrra shRNA or the treatment with the ERRα inverse agonist, thiadiazoleacrylamide XCT-79042. As these cells became more dependent on the 4-carbon backbone of aspartate to "complete the cycle", elevated levels of M + 4 citrate were observed as compared to neurons in the control treatment groups [Fig. 4h(2)]. Furthermore, when the assay time was prolonged, M + 4 citrate and M + 3 glutamate became the dominant forms of their respective species [Fig. 4h(2–3)]. This was because the labelled aspartate continued to feed the mini-TCA cycle, resulting in the de novo generation of these isotopologues [Fig. 4h(4)]. Intriguingly, the resulting loss in the levels of exogenously labelled M + 4 aspartate was not observed for its endogenous M + 0 isotopologue [Fig. 4h(4)]. Moreover, even though this rewired mini-TCA cycle predicts a 1:1 ratio of glutamate utilized versus glutamate regenerated in the reactions (i.e., no net loss), the resulting unlabelled M + 0 isotopologue [Fig. 4h(3)] as well as its total quantity [Fig. 4h(5)] were however elevated instead. These observations hinted that an unknown replenishment mechanism was turned on for these endogenous amino acids when Esrra was lost.
With reference to the unbiased metabolomics data (Table S9), it was speculated that an enhanced breakdown and/or reduced biosynthesis of N-acetyl-aspartyl-glutamate (NAAG) contributed to the observed homeostatic changes. NAAG is a neuron-specific dipeptide synthesized from an initial conjugation of an acetyl-CoA-derived acetyl group to aspartate (i.e., N-acetyl-aspartate, NAA), followed by a second conjugation reaction with glutamate43. Indeed, our 13C4-aspartate isotopologue tracing experiment revealed that the contributions of both aspartate and glutamate carbons to the formations of NAA and NAAG were significantly reduced in neurons deficient of functional ERRα, regardless of their sources of origin (i.e., endogenous [M + 0 glutamate and M + 0 aspartate] or exogenous [M + 3 glutamate and M + 4 aspartate]) (Fig. 5i). The diminished pre-existing unlabelled forms of NAA and NAAG indeed supported the notion of pro-catabolic shift, as evidenced by their reduced total content (Table S7). Additionally, the relatively lower levels of the newly synthesized versions of heavily labelled neuropeptides also indicated a diminished anabolic biosynthesis as well (Fig. 5i). Consequently, the net NAAG-NAA catabolic shift could have unleashed the unlabelled M + 0 forms of aspartate and glutamate, as detected in the isotopologue study [Fig. 5h(4)]. Concordantly, diminished ERRα signalling axis observed previously in the AOF mouse model (Fig. 3e-i) was also accompanied by reductions in the total levels of succinate, fumarate, NAA, and even NAAG, as evident from the metabolomics data (Supplementary Fig. 8, Supplementary Table 5).
In clinical studies, higher levels of NAAG have been associated with better brain functional outcomes in patients of various neurological conditions, such as psychosis44; schizophrenia45 and multiple sclerosis46. In this study, the relevance of NAAG to better brain function was further validated in human LOAD samples. According to the dorsolateral prefrontal cortex metabolomics profile data extracted from the ROSMAP cohort [N = 339 for disease-affected (AD + MCI), N = 153 for non-dementia]47, sex-specific analyses revealed more dramatic changes in metabolites among female affected subjects compared to males (Fig. 5j, Supplementary Table 10). Subsequent metabolite set enrichment analysis revealed that the majority of the differentially changed metabolites were likely part of the amino acid metabolism, including the metabolism of aspartic acid (Fig. 5k). Further analysis revealed that while no significant correlations between deregulated metabolites and cognitive impairment were found in males, a list of amino acid- and lipid-related metabolites were identified in the female group (Fig. 5l). Notably, the reduction in NAAG was the one most correlated with cognitive decline in female subjects (Fig. 5l-m), supporting the idea that the metabolic homeostasis of this neuron-specific metabolite is an important factor in determining the sex-biased functional outcome in LOAD. In contrast, the total levels of brain glutamate and aspartate, which could be contributed by various brain cell types, showed less consistent trends compared to the changes observed specifically in neurons (Fig. 5n-o). Nevertheless, brain glutamate levels were still induced and this was correlated with more severe cognitive decline in both sexes (Fig. 5o). This elevation in glutamate was more pronounced in females, so as the trend of changes across different cognitive scores was also more consistent in females (Fig. 5o).
Elevated spontaneous postsynaptic activity, coupled with bioenergetic incompetency due to loss of ERRα, confer heightened neuronal vulnerability to excitotoxic insults
Previous studies have shown that NAAG exhibits neuroprotective effects against NMDA-receptor-mediated excitotoxicity, likely through its partial antagonist action at the NMDA receptor48. The current data suggest that the acquired aspartate-dependence results in a net hydrolytic effect on NAAG, which would concurrently increase the free glutamate pools within neurons - the major excitatory neurotransmitter in the brain. Spontaneous synaptic vesicle fusion is a fundamental feature of all synapses, and these random release events typically activate receptors within a single postsynaptic site, giving rise to miniature postsynaptic currents49. To investigate whether the metabolic changes occurring during ERRα loss would affect the electrophysiological properties of neurons, whole-cell patch clamp recordings were performed. Compared to scrambled shRNA-treated controls, Esrra-knockdown resulted in a significant increase in the frequency of spontaneous miniature excitatory postsynaptic currents (mEPSCs), even without any obvious changes in their amplitude (Fig. 6a). Likewise, similar patterns of changes were observed in neurons chronically exposed to the ERRα antagonist XCT-790 (Fig. 6a). In a separate group of neurons, the spontaneous miniature inhibitory postsynaptic currents (mIPSCs) were evaluated. Both Esrra-knockdown and XCT-790 treatment did not change frequency nor the amplitude of mIPSCs, as they were recorded using patch pipettes with CsCl replacing K-gluconate and with excitatory postsynaptic currents blocked by DNQX (Fig. 6b). The solo increase in mEPSC frequency suggested presynaptic changes. Supporting this notion, the relative abundance of the presynaptic vesicular glutamate transporter VGLUT1 was elevated in the ERRα-deficient neurons. In contrast, the abundance of postsynaptic glutamatergic receptors was not significantly altered (Fig. 6c). Since VGLUT1 mediates the transport of intracellular glutamate into secretory vesicles50, its upregulation would likely increase the probability of stochastic, spontaneous glutamate release, thereby leading to the observed changes in miniature excitatory postsynaptic currents.
Postsynaptic currents are a major energy-consuming process in the brain51. The pronounced increase in mEPSC frequency suggested that a net increase in the number of spontaneous postsynaptic firing events per second could have occurred chronically in the ERRα-deficient neurons. This would impose a significant metabolic demand on these cells. While this minor increment in the spontaneous neuronal activity would unlikely affect the energy levels of a healthy neuron, as they have reserved respiratory capacity to compensate for such additional metabolic demands52; here even at the resting condition in the absence of any external stimulation, the total ATP levels in the ERRα-deficient neurons were already lower than that observed in the control neurons (Fig. 6d). As hinted by the TCA cycle analysis (Fig. 5g-h), the diminished commitment to reactions beyond succinyl-CoA suggests reduced activities of the succinate dehydrogenase (SDH) enzyme complex. According to the bulk transcriptomics data (Fig. 5c-d), expression levels of Sdha and Sdhd—two of the four major subunits of the enzyme—were significantly reduced (Table S8). The same patterns of changes were observed as well in primary neuronal cells subjected to Esrra knockdown and XCT-790 treatment (Supplementary Fig.S9b). According to the ENCODE (Supplementary Table 11) and ChIP-atlas (Supplementary Fig. 9a) databases, Sdha and Sdhd appear to be the downstream targets of ERRα, and was further validated by ChIP-PCR experiments (Supplementary Fig. 9c) and enzyme functional assays (Fig. 6f) in both Esrra knockdown or XCT-790 treatment models. More importantly, the ROSMAP study also indicated that the reduction in SDHA expression was unique and correlated with cognitive decline in females (Fig. 6e), highlighting that the fidelity of SDH function is one of the important factors contributing to female vulnerability in LOAD.
In addition to its central role in the TCA cycle, SDH also serves as Complex II of the electron transport chain (ETC). As the sole enzymatic complex that regulates the flux of both the TCA cycle and the ETC, the functional capacity of SDH is considered a source of the reserved respiratory capacity, and thus a key determinant of the ATP production rate in response to a sudden increase in energy demand53. To assess whether the recovery of ATP in ERRα-deficient neurons was suboptimal, dynamic changes in the mitochondrial oxygen consumption rate (OCR) was first evaluated (Fig. 6g). Both the basal respiration rate (Fig.S9d) and the reserved respiratory capacity (Fig. 6g) were significantly reduced in the ERRα-deficient groups. These respiratory inefficiencies, when coupled with the incresed spontaneous post-synaptic firing, likely explained the relatively lower levels of total ATP even when the neurons were at rest (Fig. 6d). These neurons were then synaptically challenged for 30 seconds with glutamate, which induces large fluxes of sodium and potassium and thus stimulates the accelerated hydrolysis of ATP by the Na+/K+-pump54. Real-time monitoring of mitochondrial ATP dynamics using the ATP-Red live cell dye revealed that while glutamate treatment resulted in similar magnitudes of ATP depletion relative to the corresponding baseline levels across all tested groups (Supplementary Fig. 9e), the subsequent 30-minute recovery of mitochondrial ATP was significantly less efficient in the ERRα-deficient groups (Fig. 6h). At 4 hours after the glutamate challenge, when the neurons were expected to have better recovered, nearly 16% of these neurons instead became apoptotic via the activation of caspase 3/7 (Fig. 6i). Together, these data revealed that neurons lacking active ERRα are more vulnerable to external excitatory stimulus or insults that lead to elevated energy demand and bioenergetic stresses.