bims-medebr Biomed News
on Metabolism of the developing brain
Issue of 2025–11–16
25 papers selected by
Regina F. Fernández, Johns Hopkins University



  1. Cell Death Discov. 2025 Nov 10. 11(1): 525
      Traumatic brain injury (TBI) is a critical condition caused by physical trauma to the head, leading to primary brain edema, hemorrhage, and swelling, along with secondary injuries such as oxidative damage, neuroinflammation, and mitochondrial dysfunction. Astrocytes play a vital role in the astrocyte-neuron lactate shuttle (ANLS), which facilitates the transfer of lactate from astrocytes to neurons as an energy source. This study investigates the role of the pyruvate kinase M2 (PKM2) gene in astrocytes and its impact on neuronal survival following TBI. We hypothesized that deletion of the PKM2 gene in astrocytes would result in increased neuronal death due to impaired lactate supply via the ANLS. Additionally, we hypothesized that lactate administration post-TBI would mitigate neuronal death and alleviate cognitive impairment. To test these hypotheses, we utilized tamoxifen to specifically delete the PKM2 gene in astrocytes of Aldh1l1-CreERT2; PKM2f/f mice. Following TBI induction, sodium L-lactate was administered, and the mice were sacrificed 24 h later. Our analysis included assessments of neuronal death, microtubule disruption, oxidative damage, and the activity of enzymes associated with the ANLS. The findings confirmed that PKM2 gene deletion in astrocytes exacerbated neuronal death and worsened cognitive impairment. Conversely, lactate administration post-TBI reduced neuronal death and improved cognitive outcomes. These results suggest that lactate administration could serve as a potential therapeutic strategy for preventing and treating neurological damage following TBI.
    DOI:  https://doi.org/10.1038/s41420-025-02829-7
  2. J Neurochem. 2025 Nov;169(11): e70294
      Alzheimer's disease (AD) is more than just amyloid and tau. While often described as a disease of metabolic dysfunction, AD can more accurately be described as a disorder of metabolic inflexibility that leads to bioenergetic failure. In the healthy brain, neurons, glia, and vascular cells dynamically share and switch between different fuel sources (e.g., glucose, lactate, ketones, and fatty acids) to match functional demand. In AD, this adaptability is progressively lost because cellular metabolism is actively reprogrammed to support neuroinflammatory and disease-associated processes at the cost of neuronal function. Microglia, in particular, upregulate glycolytic metabolism, alter lipid handling, and prioritize immune functions, which actively depletes the brain's energy supply. These adaptations are initially compensatory but ultimately trap the brain in a rigid metabolic program that deprioritizes neuronal support. This metabolic shift unfolds along a biphasic trajectory: early, glia-driven hypermetabolism aligned with inflammation, followed by late-stage brain hypometabolism and energy collapse that leads to neuronal dysfunction. System-level consequences include altered excitability, decreased network connectivity, sleep disruption, and cognitive decline. Critically, these changes feed forward to accelerate AD pathogenesis: glycolytically biased microglia and stressed neurons promote amyloid-β production, tau release, and protein aggregation, adding to metabolic rigidity. Evidence from human neuroimaging studies, brain/cerebral spinal fluid (CSF) multi-omic studies, and preclinical studies demonstrate that shifts in glycolytic flux, tricarboxylic acid cycle (TCA) intermediates, and lipid metabolism parallel amyloid and tau pathology and cognition decline. We hypothesize that these metabolic programs, while initially protective, are chronically maladaptive yet reversible. We propose that restoring metabolic flexibility can mitigate amyloid and tau pathology, neuronal loss, and functional decline. Ongoing preclinical studies and clinical trials are actively exploring metabolism as a therapeutic target in AD. Collectively, these findings define AD as a disorder of metabolic inflexibility, where adaptive shifts in cellular metabolism become pathologically rigid and drive disease progression, while offering a promising target for therapeutic intervention in AD.
    Keywords:  Alzheimer's disease; glia; metabolism
    DOI:  https://doi.org/10.1111/jnc.70294
  3. Int J Mol Med. 2026 Jan;pii: 20. [Epub ahead of print]57(1):
      Astrocytes, the predominant glial cells within the central nervous system, participate in a variety of processes, including metabolic homeostasis, regulation of blood‑brain barrier function, and the integration of neuronal function and structure. Lipids, which are critical components of astrocyte architecture and functionality, play a pivotal role in energy production, membrane fluidity, and the integration of astrocyte‑neuronal structure and function via lipid droplet storage and lipid metabolism. Research indicates that the proper storage of lipid droplets (LDs) in astrocytes is essential for maintaining normal physiological functions of the CNS. Fatty acids released from astrocyte LDs undergo β‑oxidation within mitochondria and are intricately linked to neuronal inflammatory signaling, oxidative stress and mitochondrial energy production. Furthermore, dysregulated lipid metabolism in astrocytes is strongly linked to the onset and progression of neurological disorders. The alteration of lipid metabolic profiles in astrocytes across various microenvironments, along with the identification and screening of critical lipid metabolites, has emerged as a focal point in current research. Nonetheless, the precise mechanisms through which aberrant lipid metabolism in astrocytes influences the onset and progression of neurodegenerative diseases require further elucidation. This article seeks to synthesize recent advancements in the study of LDs‑key organelles responsible for lipid homeostasis in astrocytes‑to elucidate the response characteristics and underlying mechanisms of lipid metabolism in these cells. Furthermore, it aims to investigate the therapeutic potential of inhibiting abnormal lipid secretion and excessive lipid accumulation in astrocytes in the context of neurodegenerative disease progression.
    Keywords:  astrocytes; lipid droplets; lipid metabolism; neurodegeneration; neurons; β‑oxidation
    DOI:  https://doi.org/10.3892/ijmm.2025.5691
  4. Neurochem Res. 2025 Nov 12. 50(6): 357
      Astrocytes are essential partners of neurons and have many important functions in the brain. Almost all of these astrocytic functions require energy that is provided by cellular adenosine triphosphate (ATP). Accordingly, astrocytes contain a millimolar concentration of cellular ATP that is maintained by continuous and rapid regeneration from adenosine diphosphate (ADP) and adenosine monophosphate (AMP), the main products of cellular energy-consuming reactions. In this article we describe the current knowledge on the cellular content, the consumption and the metabolic regeneration of ATP in astrocytes, explore the consequences of an application of metabolic inhibitors on astrocytic ATP metabolism and summarize the importance of endogenous energy stores and exogenous energy substrates for the maintenance of a high cellular ATP content. In addition, we give insight in recent studies on the visualization of ATP in astrocytes by genetically encoded ATP sensors, summarize the importance of astrocytic ATP release and extracellular ATP processing and discuss recent data on the restoration of ATP in ATP-deprived astrocytes. The current knowledge on the ATP metabolism of astrocytes clearly demonstrates the high potential of this important brain cell type to flexibly use different metabolic pathways and a broad range of endogenous and exogenous sources to maintain, regenerate and restore cellular ATP levels. These processes secure that ATP is continuously available for the many ATP consuming processes that enable astrocytes to perform their functions in the healthy brain.
    Keywords:  ATP; Astrocytes; Energy metabolism; Glycolysis; Oxidative phosphorylation
    DOI:  https://doi.org/10.1007/s11064-025-04604-7
  5. Neurosci Bull. 2025 Nov 15.
      There is a vicious cycle between brain metabolism and epileptic seizures that compounds the deleterious consequences of seizures. Human epilepsy samples implicate cholesterol 25-hydroxylase (CH25H) in linking lipid metabolism and immunity. CH25H expression increased in microglia after status epilepticus, with its product 25-hydroxycholesterol (25-HC) accumulating in the hippocampus and blood. Thus, we generated microglia-specific CH25H knockdown mice to study the role of CH25H specifically in epilepsy. CH25H knockdown inhibited the assembly and activation of NLRP3 inflammasome and restrained the loss of neurons in the hippocampal area in epileptic mice. More importantly, CH25H knockdown reduced the number of recurrent seizures and time in seizure by electroencephalogram recording, which was partly reversed after 25-HC treatment. Untargeted metabolomics showed that another lipid metabolite, arachidonic acid, might be a potential biomarker of CH25H-mediated epilepsy. These findings suggest that microglial CH25H regulated the status epilepticus in a hydroxylase-dependent mechanism.
    Keywords:  25-HC; CH25H; Epilepsy; Microglia; NLRP3; Neuronal loss
    DOI:  https://doi.org/10.1007/s12264-025-01526-w
  6. Metabolism. 2025 Nov 12. pii: S0026-0495(25)00305-1. [Epub ahead of print] 156436
      The human brain, despite accounting for only 2 % of total body weight, exhibits an exceptionally high lipid content (approximately 20 % of its mass), highlighting the critical role of lipid metabolism in maintaining neural homeostasis and function. Neurodegenerative diseases-including Alzheimer's disease (AD), Parkinson's disease (PD), stroke, Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS)-are characterized by progressive neuronal dysfunction and myelin degeneration. These conditions predominantly affect aging populations and represent a growing global health challenge. While aging remains the primary risk factor, compelling evidence now underscores the involvement of dysregulated lipid metabolism in their pathogenesis. However, the precise mechanisms linking dynamic lipid metabolic alterations to disease progression remain incompletely elucidated. This review systematically examines the multifaceted contributions of lipid metabolism to neurodegenerative processes and critically assesses emerging therapeutic strategies that target lipid pathways for the treatment of neurodegenerative disorders.
    Keywords:  Aging; Alzheimer's disease; Lipid metabolism; Neurodegenerative diseases; Parkinson's disease; Stroke
    DOI:  https://doi.org/10.1016/j.metabol.2025.156436
  7. Geroscience. 2025 Nov 14.
      The p38 mitogen-activated protein kinase has a well-characterized role in modulation of inflammatory processes throughout the body. In the central nervous system, p38 is primarily studied within neurons and microglia, most commonly in the context of neurological insult. The present study was designed to determine its function in astrocytes during non-pathological aging. We generated a conditional knockout model in which a tamoxifen-inducible Aldh1l1 promoter drives Cre recombinase expression in mice with exon 1 of the p38α gene flanked by loxP sites. Knockout of astrocyte p38α was achieved via tamoxifen administration in young sexually mature mice at 3-4 months old. Animals were subsequently aged to 21-24 months prior to performing electrophysiological, immunohistochemical, and biochemical analyses. We found that early loss of astrocyte p38α was associated with a reduction in hippocampal neuroinflammation and concomitant enhancement of synaptic strength in aged female mice. In subsequent experiments in younger animals, the knockout reduced peripheral GFAP levels and increased non-synaptic mitochondrial uncoupling. These findings indicate that astrocyte p38α has wide-ranging effects on brain metabolism, inflammation, and synaptic function during the course of normal aging, including release of GFAP from the central nervous system to the periphery. Follow-up studies exploring the role of astrocyte p38α in various age-associated neuropathological contexts are warranted.
    Keywords:  Aging; Astrocyte; Neuroinflammation; P38 MAPK
    DOI:  https://doi.org/10.1007/s11357-025-01948-4
  8. J Lipid Res. 2025 Nov 12. pii: S0022-2275(25)00207-X. [Epub ahead of print] 100944
      Fatty acids (FAs) play multifaceted roles in neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). This review systematically summarizes current understanding of fatty acid metabolism and its diverse implications in NDD pathology. Short-chain fatty acids (SCFAs), primarily generated by gut microbiota, regulate neuroinflammation, gut-brain communication, and blood-brain barrier (BBB) integrity via epigenetic modifications and immune modulation. Medium-chain fatty acids (MCFAs) exhibit therapeutic potential by improving energy metabolism and neuromuscular function, particularly in ALS models. Long-chain polyunsaturated fatty acids (PUFAs), notably docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), contribute to neuronal membrane integrity, synaptic plasticity, and antioxidant defense, mitigating oxidative stress and neuroinflammation. Conversely, saturated and certain n-6 fatty acids may exacerbate neurodegeneration through pro-inflammatory and oxidative pathways. Emerging evidence highlights fatty acid involvement in key pathological processes such as lipid peroxidation, mitochondrial dysfunction, ferroptosis, and BBB disruption. Therapeutically, targeted supplementation, dietary modification, microbiome manipulation, and advanced nanotechnology-based delivery systems are promising strategies. Nevertheless, precise therapeutic efficacy depends critically on disease stage, dosage, genetic background, and individual metabolic context. Integrating personalized medicine with precision nutritional strategies and novel drug-delivery platforms offers promising avenues to translate fatty acid-based interventions into clinical practice, potentially improving patient outcomes in the aging global population.
    Keywords:  Alzheimer’s disease; Fatty acids; Parkinson’s disease; amyotrophic lateral sclerosis; gut-brain axis; neurodegenerative diseases; neuroinflammation; oxidative stress; polyunsaturated fatty acids; short-chain fatty acids
    DOI:  https://doi.org/10.1016/j.jlr.2025.100944
  9. Brain. 2025 Nov 14. pii: awaf430. [Epub ahead of print]
    Kajus Merkevicius, Dmitrii Smirnov, Lea D Schlieben, Rebecca Ganetzky, René G Feichtinger, Huafang Jiang, Fang Fang, Tomohiro Ebihara, Kei Murayama, Giulia Ferrera, Anna Ardissone, Dariusz Rokicki, Dorota Wesol-Kucharska, Sabine Schröder, Peter Bauer, Aida Bertoli-Avella, Elsebeth Østergaard, Peter Freisinger, Mirian C H Janssen, Matias Wagner, Omar Abouyousef, Bader Alhaddad, Lama AlAbdi, Fowzan Alkuraya, Charlotte L Alston, Anna Baghdasaryan, Diana Barca, Ivo Barić, Marcello Bellusci, Andrea Bevot, Eugen Boltshauser, Ingo Borggraefe, Juliette Bouchereau, Claudio Bruno, Birute Burnyte, Amy Calhoun, Kari Casas, Mahmut Coker, Ellen Crushell, Pascal De Lonlay, Carlo Dionisi-Vici, Felix Distelmaier, Marni J Falk, Ana Cristina Ferreira, Carlos R Ferreira, Can Ficicioglu, Gulden Fatma Gokçay, Johannes Häberle, Oliver Heath, Albrecht Hellenschmidt, Julia Hoefele, Georg F Hoffmann, Tomas Honzik, Martina Huemer, Patrícia Janeiro, Amel Karaa, Çiğdem Seher Kasapkara, Ilse Kern, Joerg Klepper, Thomas Klopstock, Ina Knerr, Johannes Koch, Zita Krumina, Costanza Lamperti, Elise Lebigot, Zhimei Liu, Esther M Maier, Diego Martinelli, Robert McFarland, Bryce Mendelsohn, Maria Judits Molnar, Helen Mundy, Marie-Cecile Nassogne, Anabela Oliveira, Katrin Õunap, Chiara Panicucci, Sumit Parikh, Heidi Peters, Samia Pichard, Barbara Plecko, Danijela P Ramadža, Gabriela M Repetto, Isabel Rivera, Richard J Rodenburg, Alessandro Rossi, Manuel Schiff, Kathrin Seidemann, Wendy E Smith, Sérgia Soares, Barbara Siri, Katja Steinbrucker, Pasquale Striano, Jolanta Sykut-Cegielska, Galit Tal, Robert W Taylor, Kostas Tsiakas, Sema Kalkan Ucar, Eva Hoytema van Konijnenburg, Mathias Woidy, Joy Yaplito-Lee, Yilmaz Yildiz, Martin Zenker, Petra Zsidegh, Dominik Westphal, Wolfgang Sperl, Thomas Meitinger, Garry K Brown, Holger Prokisch, Johannes A Mayr, Saskia B Wortmann.
      This retrospective study on X-linked PDHA1-related pyruvate dehydrogenase complex (PDHc) deficiency combined a systematic literature review with a multicenter survey exploring genotypes, phenotypes, and survival. Data from 891 individuals (45% unpublished) were included. Of note, 53% of cases were females. Median age at last assessment was six years (range 0-80 years, n = 622). We detected 331 different (118 unpublished) PDHA1 variants of which 75% (305/405) had occurred de novo. Variants in this study were uploaded to ClinVar (SCV006297015 - SCV006297345). The 10 most frequent variants accounted for 36% of the diagnoses. Sixty-nine percent of the variants were private; missense (50%) and frameshift (20%) variants were most common. Frameshift/nonsense (FS/N) variants in males (44/401, 11%) were confined to regions escaping nonsense-mediated decay (NMD) and were significantly less frequent than in females (151/461, 33%). Neonatal or infantile (405/529, 77%) presentations were most frequent, with pre/perinatal abnormalities reported in 47% (159/342). FS/N variants in NMD-predicted region 3.9 (95% Confidence Interval (CI) 1.54-11.04) times increased the odds of fetal findings. Females presented significantly earlier (2 months, interquartile range (IQR) 7.0, n = 224) than males (8 months, IQR 16.6, n = 233), with increased risk of neonatal presentation (odds ratio (OR) 3.01 (95% CI 1.279-7.616) when harboring FS/N variants in NMD-predicted region. The overall (n = 242) mean survival time was 10.9 (95% CI 9.9-11.9) years. On average, females survived 4.5 (95% CI 2.62-6.40) years longer than males despite presenting more severe phenotypes. Poor survival was associated with male sex (hazard ratio (HR) 3.3 (95% CI 1.95-5.62)), neonatal presentation (HR 5.5 (95% CI 2.17-14.09)), FS/N variants in NMD-predicted region (HR 4.0 (95% CI 1.78, 9.16)), and splice variants (HR 2.3 (95% CI 1.15, 4.59)). More severe clinical phenotypes were predicted by neonatal or infantile presentations and by female sex. Developmental delay (DD), intellectual disability (ID), muscle hypotonia, abnormal movements, seizures, feeding difficulties, and microcephaly were the most frequent phenotypes, all occurring in more than half. Corpus callosum or basal ganglia alterations and cerebral atrophy were common. Four percent (36/891) were reported to have mild phenotypes with no DD nor ID (25/36 males). This is the largest dataset on a nuclear-encoded defect of mitochondrial energy metabolism. The genotypic and phenotypic details further defines disease landscape and can be used for variant interpretation. The correlations between genotypes, sex, phenotypes and survival, adds a substantial improvement to counselling.
    Keywords:  genotype-phenotype correlation; inborn errors of metabolism; inborn metabolic disease; ketogenic diet; mitochondrial disease; treatment
    DOI:  https://doi.org/10.1093/brain/awaf430
  10. Magn Reson Med. 2025 Nov 11.
       PURPOSE: To investigate whether varying delivery times of hyperpolarized [1-13C]pyruvate, enabled by the increased apparent T1 dissolved in deuterium oxide (D2O), affects the observed kinetics of glycolytic brain metabolism in vivo.
    METHODS: Eighteen healthy mice were injected with 300 μL of hyperpolarized [1-13C]pyruvate dissolved in D2O at increasing injection times (15 s, 60s, 90s, 120 s). After collecting T2-weighted scans, slab dynamic 13C MRS data were acquired. Time-course curves of [1-13C]pyruvate, [1-13C]lactate and [1-13C]lactate/total carbon ratio were calculated. Mean full-width-half-maximum (FWHM) and area-under-curve (AUC) values were compared across injection times. A simplified one-compartment model of pyruvate metabolism was fit using the conversion rate constant (kPL) and effective lactate decay rate (R1eff). Dynamic EPSI images, acquired using an injection time of 15 s and 60 s for comparison.
    RESULTS: The mean FWHM values of the time-course curves of [1-13C]pyruvate and [1-13C]lactate showed a significant increase (p < 0.01) with increasing injection times, while no statistical significance was found across the AUC values. The time-course curves of lactate/total carbon ratio showed elongated plateaus with increased injection times. Kinetic modeling showed good agreement between fitted and acquired lactate data, with AUC of normalized lactate profile remaining constant across infusion times. Dynamic EPSI images acquired with a longer infusion time (60 s) showed the ability to monitor brain metabolism as it approached pseudo-steady state.
    CONCLUSIONS: Increased delivery times of hyperpolarized [1-13C]pyruvate dissolved in D2O approaches pseudo-steady state metabolism in vivo and allowing for the potential to cater new acquisition and reconstruction approaches for enhanced imaging.
    Keywords:  deuterium oxide; hyperpolarized 13C‐pyruvate; injection time; murine brain
    DOI:  https://doi.org/10.1002/mrm.70175
  11. Brain Commun. 2025 ;7(6): fcaf397
      Pyridoxine-dependent epilepsy (PDE) due to recessive ALDH7A1 mutations is characterized by intractable epilepsy that is often unresponsive to antiseizure medications. Irrespective of pyridoxine (vitamin B6) supplementation and lysine reduction therapy, patients present severe residual neurocognitive deficits. We evaluated upstream inhibition of 2-aminoadipic semialdehyde synthase (AASS) as a novel therapeutic strategy to reduce the accumulating metabolites (α-aminoadipic semialdehyde, Δ1-piperideine-6-carboxylate, pipecolic acid, 6-oxo-pipecolic acid and 2S,6S-/2s,6R-oxopropylpiperidine-2-carboxylic acid) considered neurotoxic. We utilized an existing mouse knockout model of hyperlysinaemia (Aass-knockout) and generated a PDE model, a Aldh7a1 single knockout model via CRISPR/Cas (clustered regularly interspaced short palindromic repeats and CRISPR-associated protein) and generated the double-knockout Aass/Aldh7a1 mice. Next-generation metabolomics screening was performed to measure all known biomarkers in brain, liver and plasma of wild-type and mutant mice. Metabolomics confirmed the known metabolite markers for Aldh7a1-knockout and Aass knockout mice in all samples. The potentially neurotoxic metabolites (Δ1-piperideine-6-carboxylate, pipecolic acid, 6-oxo-pipecolic acid and 2S,6S-/2s,6R-oxopropylpiperidine-2-carboxylic acid) significantly decreased in double-knockout Aass/Aldh7a1 mice brain and liver tissues compared to Aldh7a1-knockout mice. Plasma analysis revealed a significant reduction of known biomarkers, suggesting a reliable monitoring option in human patients. We demonstrate the first mammalian evidence that AASS inhibition is a viable strategy to rescue abnormal brain metabolism associated with PDE. This may target the intellectual disability and neurologic deficits caused by persistent lysine catabolic-related neurotoxicity despite adequate vitamin B6 supplementation.
    Keywords:  2-aminoadipic semialdehyde synthase inhibition; lysine biochemistry; metabolic epilepsy; mouse model; therapy
    DOI:  https://doi.org/10.1093/braincomms/fcaf397
  12. Cell Rep. 2025 Nov 07. pii: S2211-1247(25)01292-6. [Epub ahead of print]44(11): 116521
      Traumatic brain injury (TBI) increases one's risk of developing Alzheimer's disease and tauopathy. Yet, the mechanisms linking TBI to neurodegenerative disease remain poorly defined. Mounting recent evidence indicates that defects in brain lymphatic drainage contribute to multiple neurodegenerative diseases. Here, we investigated whether promoting brain lymphatic drainage recuperation following TBI via treatment with the lymphangiogenic factor vessel endothelial growth factor C (VEGFC) mitigates the ability of TBI to exacerbate tauopathy. In this study, we show that a single mild TBI leads to worsened neuropathology, brain macrophage activation, and neurodegeneration in the PS19 mouse model of tauopathy. Moreover, we find that viral-vector-based delivery of VEGFC into the meningeal compartment 24 h post-TBI ameliorates tau-mediated neurodegenerative disease pathogenesis. Findings from these studies offer new insights into how TBI leads to the development of tauopathy later in life and suggest that VEGFC-based treatments might offer a therapeutic strategy to limit tauopathy after sustaining a head injury.
    Keywords:  Alzheimer's Disease; CP: neuroscience; Vegfc; brain lymphatics; microglia; neurodegenerative disease; tauopathy; traumatic brain injury
    DOI:  https://doi.org/10.1016/j.celrep.2025.116521
  13. Mol Neurobiol. 2025 Nov 13. 63(1): 37
      Neuronal differentiation requires extensive metabolic remodeling to support increased energetic and biosynthetic demands. Here, we present an integrated multi-omics and functional characterization of metabolic transitions during early differentiation of human induced pluripotent stem cells (iPSCs) into excitatory cortical neurons using doxycycline-inducible overexpression of neurogenin-2 (NGN2). We analyzed parental iPSCs and induced neurons (iNs) at days 7 and 14 of differentiation, integrating gene expression profiling, label-free quantitative proteomics, high-resolution respirometry, fluorescence lifetime imaging microscopy (FLIM), and 13C₆-glucose metabolic flux analysis. Our data reveal progressive metabolic remodeling associated with neuronal maturation, including enhanced oxidative phosphorylation, increased mitochondrial content, and respiratory capacity. Proteomic analyses showed upregulation of mitochondrial and antioxidant pathways, while FLIM indicated a progressive increase in enzyme-bound NAD(P)H, consistent with a shift toward oxidative metabolism. Notably, 13C₆-glucose tracing revealed delayed labeling of the intracellular pool of fully labeled glucose and tricarboxylic acid cycle metabolites, together with enhanced labeling of pentose phosphate pathway intermediates and glutathione in iNs, indicating a shift toward biosynthetic and antioxidant glucose utilization during differentiation. Despite this enhancement in mitochondrial function, differentiated neurons maintained glycolytic activity, suggesting metabolic flexibility. Our results define the first week of differentiation as a critical window of metabolic specialization and establish NGN2-iPSC-derived cortical neurons as a versatile and well-characterized model system for investigating bioenergetic remodeling during early human neurodevelopment. It provides a robust foundation for mechanistic insights and high-throughput evaluation of metabolic pathways relevant to human disease.
    Keywords:  Cellular bioenergetics; Human iPSCs; Metabolic flux analysis; Neuronal differentiation; Proteomics; Respirometry
    DOI:  https://doi.org/10.1007/s12035-025-05284-8
  14. STAR Protoc. 2025 Nov 07. pii: S2666-1667(25)00598-2. [Epub ahead of print]6(4): 104192
      Metabolic disturbances are common in motor neuron disease (MND), and elucidating their mechanisms may reveal therapies. Here, we present a protocol to assess glucose homeostasis in mice, including glucose tolerance, insulin tolerance, and glucagon challenge tests. We describe steps for fasting, intraperitoneal injections, and serial blood glucose measurements. The protocol also includes plasma collection for catecholamine analysis using matrix-assisted laser desorption ionization (MALDI) mass spectrometry imaging and immunofluorescence of pancreatic hormones, enabling comprehensive metabolic profiling in aged and neurodegenerative mouse models. For complete details on the use and execution of this protocol, please refer to McDonald et al.1 and McDonald et al.2.
    Keywords:  Metabolism; Metabolomics; Neuroscience
    DOI:  https://doi.org/10.1016/j.xpro.2025.104192
  15. Stem Cells Transl Med. 2025 Nov 14. pii: szaf060. [Epub ahead of print]14(11):
      Within the central nervous system (CNS), mitochondria serve as vital energy sources for neurons, glial cells, and vascular functions, maintaining intracellular metabolic balance. Recent studies involving cellular models, rodents, and humans reveal that metabolically active mitochondria can be released into the extracellular space, playing roles in intercellular communication within the CNS. When taken up by neurons, these extracellular mitochondria may provide neuroprotective effects. Conversely, damaged mitochondria and their released components during severe tissue injury or inflammation can contribute to neurodegenerative processes. Thus, mitochondria secreted under pathological conditions in the CNS hold promise as biomarkers indicative of recovery. Additionally, transplantation of external mitochondria shows potential as a therapeutic approach for various CNS disorders. This mini review focuses on recent advances in the transfer of mitochondria between cells, the use of extracellular mitochondria as biomarkers, and the prospects of mitochondria transplantation from experimental research to clinical application, particularly in diseases like stroke.
    Keywords:  biomarkers; central nervous system; extracellular mitochondria; mitochondria transplantation; mitochondrial modification; stroke
    DOI:  https://doi.org/10.1093/stcltm/szaf060
  16. Neurotherapeutics. 2025 Nov 13. pii: S1878-7479(25)00267-3. [Epub ahead of print] e00789
      Traumatic brain injury (TBI) constitutes a major global public health concern associated with substantial mortality and long-term disability, while its diagnostic approaches and underlying pathophysiological mechanisms remain to be fully elucidated. In this study, we conducted longitudinal metabolomic profiling of cerebrospinal fluid (CSF) from 23 TBI patients (9 mild, 14 severe) and 5 uninjured controls using high-resolution mass spectrometry. Comprehensive quantification of metabolites was performed at three critical post-injury time points (days 1, 3, and 7), revealing distinct temporal metabolic patterns. Our results demonstrated significant alterations in the CSF metabolome following TBI. Early-phase changes (day 1) predominantly involved energy-related metabolites, including sphingosine, glucose, and dl-lactate. More pronounced metabolic shifts were observed by day 3, characterized by marked variations in amino acids (l-glutamine, l-histidine) and medium-chain fatty acids (caprylic acid, octanoic acid), suggesting the transition from primary to secondary injury mechanisms. The day 7 profile revealed accumulation of repair-associated metabolites such as 2'-deoxyuridine 5'-monophosphate and 1,2-dihexadecanoyl-sn-glycerol, potentially indicative of processes occurring in the chronic phase, which may include both reparative mechanisms and ongoing pathology. Notably, we identified significant alterations in established biomarkers (trimethylamine N-oxide) and novel small peptides (e.g., Gly-His-Lys), with distinct metabolic signatures differentiating mild versus severe TBI cases. These findings delineate temporally dynamic and severity-dependent metabolic reprogramming in TBI, providing mechanistic insights into the progression from acute injury through secondary pathogenesis to chronic recovery phases. The identified metabolic signatures may serve as potential biomarkers for injury staging and therapeutic monitoring.
    Keywords:  Biomarker; Cerebrospinal fluid; Metabolomics; Traumatic brain injury
    DOI:  https://doi.org/10.1016/j.neurot.2025.e00789
  17. Front Pharmacol. 2025 ;16 1557612
      The ketogenic diet is a therapeutic strategy applied to reduce brain hyperexcitability in conditions such as epilepsy, Parkinson's and Alzheimer's disease, migraines, and autism. This diet reduces circulating glucose levels and increases ketone bodies, with β-hydroxybutyrate (BHB) being one of the leading promoters of the beneficial effects. BHB was previously reported as a mediator of cognitive restoration and memory formation. Herein, we investigate the effect of exogenous BHB on hippocampal neuronal excitability and synaptic plasticity mechanisms, regardless of the pathological or neurodegenerative conditions. Electrophysiological experiments were conducted to explore both passive and active neuronal properties, including action potential firing and spontaneous and evoked postsynaptic responses. Electrical stimulation along the CA3-CA1 pathway enabled the assessment of both short- and long-term synaptic plasticity, as well as the mechanisms mediated by AMPA and NMDA receptors. Experiments were conducted in hippocampal slices treated with 3-β-hydroxybutyrate glycerides (DHB) and niacin (HCAR2 agonist). Although DHB incubation did not alter passive membrane properties, it significantly increased neuronal excitability, reflected in an elevated firing rate upon depolarizing stimulation and enhanced spontaneous excitatory postsynaptic currents in CA1 pyramidal neurons, which were dependent on synaptic inputs. DHB treatment led to a reduction in long-term potentiation (LTP) in CA1 neurons, suggesting a metaplastic effect independent of NMDA receptor activation. Importantly, these DHB-induced neuronal alterations were found to be independent of HCAR2 receptor activation, supporting the involvement of distinct intracellular pathways and long-term modulatory mechanisms. Our findings indicate that DHB exerts a modulatory effect on hippocampal neural activity by enhancing excitability and concurrently promoting a compensatory reduction in LTP, suggesting a homeostatic balancing mechanism.
    Keywords:  brain slice; electrophysiology; hippocampus; ketone; long-term potentiation (LTP); niacin; β-hydroxybutyrate (BHB)
    DOI:  https://doi.org/10.3389/fphar.2025.1557612
  18. EMBO Rep. 2025 Nov 10.
      Sphingolipids govern diverse cellular processes; their dysregulation underlies numerous diseases. Despite extensive characterizations, understanding the orchestration of the sphingolipid network within living organisms remains challenging. We established a versatile genetic platform of CRISPR-engineered reporters of 52 sphingolipid regulators, recapitulating endogenous gene activity and protein distribution. This platform further allows conditional protein degradation for functional characterization. In addition, we developed the biosensor OlyAw to detect ceramide phosphoethanolamine and visualize membrane raft dynamics in vivo. Using this platform, we established comprehensive profiles of the sphingolipid metabolic network in the brain at the transcriptional and translational levels. The highly heterogeneous patterns indicate extensive coordination between distinct cell types and regions, suggesting the brain functions as a coherent unit to execute specific steps of sphingolipid metabolism. As a proof-of-concept application, we showed cell type-specific requirements of sphingomyelinases, including CG6962/dSMPD4 and CG3376/aSMase, degrading distinct subcellular pools of ceramide phosphoethanolamine to maintain brain function. These findings establish a foundation for future studies on brain sphingolipid metabolism and showcase the utilization of this genetic platform in elucidating in vivo mechanisms of sphingolipid metabolism.
    Keywords:  Brain; Cell-type Specificity; Spatial Heterogeneity; Sphingolipids; Systemic Profiling
    DOI:  https://doi.org/10.1038/s44319-025-00632-0
  19. Exp Neurobiol. 2025 Nov 11.
      Mitochondrial bioenergetics plays a fundamental role in neuronal development and function. Prenatal exposure to corticosterone in rats (Corti. Pup) has previously been shown to cause delayed neurodevelopment and synaptic plasticity deficits, showing attention deficit hyperactivity disorder (ADHD) - like behaviors. However, the underlying mitochondrial metabolic adaptations remain unclear. This study investigated mitochondrial function and metabolic remodeling in prefrontal cortex neurons of Corti.Pups, focusing on oxidative phosphorylation, calcium handling, and redox balance. We assessed neuronal viability, reactive oxygen species (ROS) production, and oxygen consumption rate (OCR) through experiments conducted in both neuron-glia co-culture and neuron-only conditions. Furthermore, we evaluated electron transport chain (ETC) activity, mitochondrial membrane potential (MMP), and mitochondrial Ca2+ uptake in purified isolated mitochondria. In results, Corti.Pup neurons exhibited increased vulnerability to glutamate-induced excitotoxicity in the absence of glial support. Despite reduced ROS production, these neurons showed elevated mitochondrial OCR and proton leak, coupled with decreased non-mitochondrial OCR and ETC complex activity. Surprisingly, MMP remained elevated despite ETC dysfunction, and mitochondrial Ca2+ uptake was suppressed. These features indicate mitochondrial metabolic reprogramming, prioritizing MMP maintenance over ATP synthesis. The observed mitochondrial inefficiency and compensatory adaptations may impair energy production, contributing to delayed neuronal development in Corti.Pups. These findings suggest that mitochondrial dysfunction and metabolic remodeling play central roles in the pathogenesis of neurodevelopmental disorders such as ADHD.
    Keywords:  ADHD; Corticosterone; Mitochondria; Neurodevelopment; OCR
    DOI:  https://doi.org/10.5607/en25025
  20. Sci Adv. 2025 Nov 14. 11(46): eadu0726
      Mitochondrial dysfunction is a hallmark of Parkinson's disease (PD), but the mechanisms by which it drives autosomal dominant and idiopathic forms of PD remain unclear. To investigate this, we generated and performed a comprehensive phenotypic analysis of a knock-in mouse model carrying the T61I mutation in the mitochondrial protein CHCHD2 (coiled-coil-helix-coiled-coil-helix domain-containing 2), which causes late-onset symptoms indistinguishable from idiopathic PD. We observed pronounced mitochondrial disruption in substantia nigra dopaminergic neurons, including distorted ultrastructure and CHCHD2 aggregation, as well as disrupted mitochondrial protein-protein interactions in brain lysates. These abnormalities were associated with a whole-body metabolic shift toward glycolysis, elevated mitochondrial reactive oxygen species (ROS), and progressive accumulation of aggregated α-synuclein. In idiopathic PD, CHCHD2 gene expression also correlated with α-synuclein levels in vulnerable dopaminergic neurons, and CHCHD2 protein accumulated in early Lewy aggregates. These findings delineate a pathogenic cascade in which CHCHD2 accumulation impairs mitochondrial respiration and increases ROS production, driving α-synuclein aggregation and neurodegeneration.
    DOI:  https://doi.org/10.1126/sciadv.adu0726
  21. J Neurochem. 2025 Nov;169(11): e70292
      Protein-tyrosine phosphatase receptor-type D (PTPRD) is an adhesion-coupled phosphatase that translates extracellular binding codes into intracellular phosphotyrosine signaling from embryogenesis through adulthood. Alternative inclusion of the Ig-domain mini-exons meA and meB tailors the ectodomain surface, thereby dictating high-affinity engagement with IL1RAPL1, IL1RAP, Slitrks, LRFN4/5 (SALM3/5), neuroligin-3, and other postsynaptic partners. Intracellularly, the catalytically active D1 domain and scaffold-like D2 module, anchored to liprin-α, coordinate presynaptic vesicle release, postsynaptic receptor composition, and synaptic plasticity. Beyond synapses, PTPRD restrains embryonic neurogenesis, promotes STAT3-dependent gliogenesis, accelerates oligodendrocyte myelination, and guides Sema3a/Fyn-mediated axon and dendrite patterning. In the adult brain it serves as the high-affinity hypothalamic and cerebellar receptor for asprosin, thereby coupling systemic energy and hydration states to feeding and drinking behavior. Human genetic studies and mouse models link these molecular activities to a spectrum of conditions-including restless legs syndrome, addiction, Alzheimer's disease, ADHD, OCD, autism spectrum disorder, and metabolic syndrome. Because PTPRD functions are pathway-specific and shaped by mini-exon usage or redundancy with other family members (PTPRS/PTPRF), domain- or ligand-selective interventions represent plausible therapeutic strategies. Elucidating its full ligand repertoire, substrate landscape, and structural basis for allosteric regulation will be critical for converting this versatile receptor from a mechanistic curiosity into a tractable target for neurodevelopmental, neuropsychiatric, and metabolic disorders.
    Keywords:  Neuropsychiatric and metabolic disorders; PTPRD; synaptic adhesion molecule
    DOI:  https://doi.org/10.1111/jnc.70292
  22. Tissue Cell. 2025 Nov 13. pii: S0040-8166(25)00512-9. [Epub ahead of print]98 103230
      The kynurenine pathway is the principal route of tryptophan metabolism in the brain, generating several neuroactive metabolites, including kynurenic acid (KYNA). KYNA functions as both a neuromodulator and a neuroprotective compound, and its dysregulation has been associated with numerous neurological and psychiatric disorders. Kynurenine aminotransferase-2 (KAT-2) is the key enzyme responsible for KYNA synthesis, yet its precise cellular localization in the mouse brain remains insufficiently characterized. In this study, we systematically compared KAT-2 expression in primary astrocytic, microglial, and neuronal cultures derived from mouse brain, complemented by in situ immunolabeling of brain sections. Immunocytochemistry combined with quantitative colocalization analysis revealed that KAT-2 is expressed in all three major brain cell types, with significant overlap with cell type-specific markers. Furthermore, KAT-2 immunoreactivity was largely restricted to the soma, showing a perinuclear distribution in glial cells and partial extension into dendritic compartments in neurons. These findings provide the first parallel characterization of KAT-2 distribution across astrocytes, microglia, and neurons in the mouse brain. Overall, our results indicate that KAT-2 is widely expressed in neural cells, a finding that supports the hypothesis that KAT-2 contributes broadly to kynurenine metabolism. Taken together, our findings provide a foundation for future studies aimed at defining the cell type-specific functional roles of KAT-2.
    Keywords:  Cell culture; Immunocytochemistry; Kynurenic acid; Kynurenine aminotransferase
    DOI:  https://doi.org/10.1016/j.tice.2025.103230
  23. J Neuroinflammation. 2025 Nov 11. 22(1): 265
      Emerging evidence underscores the critical role of neuroinflammation and metabolism in the pathophysiology of epilepsy. Metabolic dysregulation and neuroinflammation exacerbate each other, creating a vicious cycle. Although this interplay offers novel insights into the mechanisms underlying neurological disorders and potential therapeutic approaches, its role in epilepsy remains unclear. In this study, we review the interplay between metabolism and neuroinflammation, focusing on glucose, glutamate /γ-aminobutyric acid (GABA), tryptophan-kynurenine, adenosine, and lipid metabolism, aiming to contribute to the development of innovative therapeutic strategies for epilepsy. We also discuss the potential anti-neuroinflammatory and antiepileptic effects of metabolic therapies.
    Keywords:  Adenosine; Epilepsy; Glucose; Glutamate; Lipid; Metabolic dysfunction; Metabolic therapy; Neuroinflammation; Tryptophan; Γ-aminobutyric acid
    DOI:  https://doi.org/10.1186/s12974-025-03553-w
  24. Hum Mol Genet. 2025 Nov 13. pii: ddaf166. [Epub ahead of print]
      Bi-allelic mutations in GBA1, a gene that encodes the lysosomal enzyme β-glucocerebrosidase (GCase), cause Gaucher disease (GD). Although GD carriers do not exhibit clinical manifestations, GBA1 mutations are the highest risk factor for Parkinson's disease (PD) in GD patients and carriers of the disease [1-5]. GCase breaks down glucosylceramide (GluCer), a sphingolipid that accumulates in GD. GluCer is deacylated by the lysosomal enzyme acid ceramidase (ACDase) to glucosylsphingosine (GluSph) [6-8]. GluSph is neurotoxic and accumulates to high levels in neuronopathic GD brains [9, 10]. However, whether this metabolic pathway involving ACDase plays a role in GBA1-associated PD (GBA1/PD) is not known. In this report we used induced pluripotent stem cells (hiPSCs) from PD patients harboring heterozygote GBA1 mutations to examine the role of ACDase in promoting α-synuclein accumulation and aggregation, a hallmark of PD. Compared to isogenic controls, hiPSC-derived PD dopamine (DA) neurons had elevated levels of pathogenic α-synuclein species. There was also reduced nuclear localization of transcription factor EB (TFEB), impaired autophagy, and decreased levels of cathepsin D (CathD), a lysosomal protease involved in α-synuclein degradation [11]. Treatment of the mutant DA neurons with a number of different ACDase inhibitors, or CRISPR/Cas9 knockdown (KD) of the ASAH1 gene, reversed all the phenotypic abnormalities of the mutant DA neurons. We conclude that in GBA1/PD-DA neurons, ACDase contributes to deregulation of key nodes of the autophagy/lysosomal pathway (ALP) involved in α-synuclein clearance. Our results suggest that ACDase is a potential therapeutic target for treating GBA1-associated PD.
    Keywords:  GBA1; Parkinson’s disease; TFEB; acid ceramidase; ASAH1; α-synuclein
    DOI:  https://doi.org/10.1093/hmg/ddaf166
  25. Sci Adv. 2025 Nov 14. 11(46): eadz3889
      Continuous glucose monitors have proven invaluable for monitoring blood glucose levels for diabetics, but they are of limited use for observing glucose dynamics at the cellular (or subcellular) level. We have developed a second generation, genetically encoded intensity-based glucose sensing fluorescent reporter (iGlucoSnFR2). We show that when it is targeted to the cytosol, it reports intracellular glucose consumption and gluconeogenesis in cell culture, along with efflux from the endoplasmic reticulum. It outperforms the original iGlucoSnFR in vivo when observed by fiber photometry in mouse brain and reports transient increase in glucose concentration when stimulated by noradrenaline or electrical stimulation. Last, we demonstrate that membrane localized iGlucoSnFR2 can be calibrated in vivo to indicate absolute changes in extracellular glucose concentration in awake mice. We anticipate iGlucoSnFR2 facilitating previously unobservable measurements of glucose dynamics with high spatial and temporal resolution in living mammals and other experimental organisms.
    DOI:  https://doi.org/10.1126/sciadv.adz3889