bims-medebr Biomed News
on Metabolism of the developing brain
Issue of 2025–07–20
twenty-one papers selected by
Regina F. Fernández, Johns Hopkins University



  1. J Neural Eng. 2025 Jul 14.
       OBJECTIVE: The ketogenic diet is a well-known treatment for epilepsy. Despite decades of research, it is not yet known how the diet accomplishes its anti-seizure efficacy. One of the earliest proposed mechanisms was that the ketogenic diet is able to replenish cellular energy stores in the brain. Although several mechanisms have been suggested for how energy depletion may contribute to seizure generation and epileptogenesis, how the dynamics of energy depletion actually leads to abnormal electrical activity is not known.
Approach: In this work, we investigated the behavior of the tripartite synapse using a recently developed neurochemical model, which was modified to include ketone chemistry. We ran transient, non-steady-state simulations mimicking normoglycemia and ketosis for metabolic conditions known to be clinically treated with the ketogenic diet, as well as a condition for which the ketogenic diet was not effective clinically. 
Main Results: We found that reduction in glucose, as well as pathological decreases in the activity of glucose transporter 1, pyruvate dehydrogenase complex, monocarboxylate transporter 1 (MCT1), and mitochondrial complex I, all led to functioning of the tripartite synapse in a rapid burst-firing mode suggestive of epileptiform activity. This was rescued by the addition of the ketone D-β-hydroxybutyrate in the glucose deficit, glucose transporter 1 deficiency, and pyruvate dehydrogenase complex deficiency, but not in MCT1 deficiency or mitochondrial complex I deficiency.
Significance: We demonstrated that replenishment of cellular energy stores is a feasible mechanism for the efficacy of the ketogenic diet. Although we do not rule out other proposed mechanisms, our work suggests that cellular energy repletion may be the primary action of the ketogenic diet. Further study of the contribution of energy deficits to seizure onset and even epileptogenesis may yield novel therapies for epilepsy in the future.&#xD.
    Keywords:  ATP; cell metabolism; energy metabolism; epilepsy; ketogenic diet; ketone
    DOI:  https://doi.org/10.1088/1741-2552/adef7f
  2. bioRxiv. 2025 May 09. pii: 2025.05.05.652246. [Epub ahead of print]
      The importance of NAD + homeostasis for neuronal health has been emphasized by studies on nicotinamide mononucleotide adenylyl transferase 2 (NMNAT2), a NAD + -synthesizing enzyme, and sterile alpha and TIR motif-containing protein 1 (SARM1), a NAD + hydrolase. NMNAT2 declines caused by neurodegenerative insults activate SARM1 to degenerate axons. To elucidate the impact of the NMNAT2-SARM1 axis on brain energy metabolism, we employed multi-omics approaches to investigate the metabolic effects caused by neuronal NMNAT2 loss. The loss of NMNAT2 in glutamatergic neurons results in a striking metabolic shift in the cerebral cortex from glucose to lipid catabolism, reduced lipid abundance, and pronounced neurodegenerative phenotypes. Proteomic analysis found that neuronal NMNAT2 loss altered levels of glial enzymes central to glucose and lipid metabolism. Genetic deletion of SARM1 in NMNAT2-deficient mice restores lipid metabolism and mitigates neurodegeneration. Taken together, we show that neuronal NAD + reduction leads to SARM1-dependent maladaptive adaptations in both neurons and glia.
    DOI:  https://doi.org/10.1101/2025.05.05.652246
  3. bioRxiv. 2025 Jun 16. pii: 2025.06.15.659811. [Epub ahead of print]
      Apolipoprotein E ( APOE ) genetic variation is the strongest genetic risk factor for late onset Alzheimer's disease (LOAD). Studies on APOE genotype dependent changes have largely focused on amyloid beta (Aβ) aggregation, disease pathology, and lipid metabolism. Recently, there has been increased interest in the relationship between metabolic function and APOE genetic variation. In this study, we examined how APOE genotype can alter metabolism in the brains of young male and female APOE3 and APOE4 targeted replacement (TR) mice. In combination with this, we also examined cell type-specific differences using induced pluripotent stem cell (iPSC) derived astrocytes and neurons. We found sex and genotype dependent changes to metabolism in the brains of young APOE TR mice. Specifically, APOE4 mice show signs of metabolic stress and compensatory mechanisms in the brain. Using proteomics and stable isotope tracing metabolomics, we found that APOE4 iAstrocytes and iNeurons exhibit signs of inflammation, mitochondrial dysfunction, altered TCA cycle and malate-aspartate shuttle activity, and a metabolic shift toward glycolysis. Taken together, this data indicates APOE4 causes early changes to metabolism within the central nervous system. While this study establishes a relationship between APOE genotype and alterations in bioenergetics, additional studies are needed to investigate underlying mechanisms.
    DOI:  https://doi.org/10.1101/2025.06.15.659811
  4. Neurobiol Dis. 2025 Jul 11. pii: S0969-9961(25)00245-1. [Epub ahead of print] 107029
      Multiple findings underline a link between altered brain cholesterol metabolism and Alzheimer's disease (AD) pathogenesis. Physiologically, excess brain cholesterol is mainly converted into 24-hydroxycholesterol (24-OHC) by the neuron-specific enzyme CYP46A1. Of note, we previously observed in autopsy specimens from human AD brains that 24-OHC and, in parallel, CYP46A1 expression decreases at advanced stages, suggesting a possible cause-effect between these reductions and AD progression. In the present study, we aimed to investigate whether maintaining high levels of 24-OHC, by its exogenous administration or CYP46A1 overexpression, can counteract tau hyperphosphorylation and accumulation of prefibrillar tau oligomers. To create an AD-like in vitro model exhibiting tauopathy, we utilized okadaic acid (OKA), a chemical compound that induces tau hyperphosphorylation. Our data show that in 24-OHC-treated primary neurons derived from wild type mice, and in neurons from CYP46A1 overexpressing mice (CYP46Tg), elevated oxysterol levels effectively prevented tau hyperphosphorylation and oligomerization. Furthermore, the dendritic arborization decrease induced by OKA was prevented, maintaining the organization and stability of the neuronal cytoskeleton. While hypothesized underlying molecular mechanisms (GSK3β, CDK5, ERK1/2, and PP2A) seem not to be involved, the protective effect of 24-OHC remains evident. The data highlight the positive effects of 24-OHC and the need to prevent its reduction in the brain. This can be achieved either through the exogenous administration of 24-OHC using suitable technologies or by maintaining elevated levels and the activity of the enzyme CYP46A1. These therapeutic approaches could be useful to prevent or slow AD progression.
    Keywords:  24-hydroxycholesterol; Alzheimer's disease; CYP46A1; Dendritic arborization; Tau hyperphosphorylation; Tau oligomers
    DOI:  https://doi.org/10.1016/j.nbd.2025.107029
  5. J Biol Chem. 2025 Jul 15. pii: S0021-9258(25)02329-4. [Epub ahead of print] 110479
      Apolipoprotein E4 (ApoE4), the major genetic risk factor for Alzheimer's disease (AD), is vital for understanding cellular processes involved in AD pathogenesis. Evidence implicates endosomes as a central player in AD, where endosomal enlargement in neurons is among the earliest changes in AD. This enlargement was reported to be enhanced in APOE4 carriers. Cells internalize ApoE into endosomes for lipid delivery, and previous studies indicate that ApoE4 influences endosomes. However, the effect of ApoE4 on endosome function seems different depending on cell type, and our understanding of how ApoE4 influences endosomes in mature neurons, the cell type degenerating in AD, remains limited. We aimed to increase understanding of the impact ApoE4 has on endosomal dynamics in primary neurons and whether external triggers, such as time-in-culture/aging, synaptic activity, and cholesterol influence these endosomal changes. We show that without external triggers mature primary neurons from ApoE knockout (KO), ApoE3 and ApoE4 mice show no major differences in endosomal appearance and function and adapt similarly to increased synaptic activity. However, with prolonged time in culture, neurons with ApoE4 show reduced degradative ability, along with decreased number of active lysosomal compartments. Moreover, when supplying aged cultures with cholesterol, ApoE4 neurons have a predisposition to accumulate cholesterol in the endolysosomal system. Taken together, we show that ApoE4 impacts endolysosome function in primary neurons, but that changes emerge only after prolonged time in culture. A better understanding of how ApoE4 impacts neurons could provide important insights into ApoE4 directed therapy for AD.
    Keywords:  Apolipoprotein E (ApoE); cholesterol; endosome; lysosome; neuron
    DOI:  https://doi.org/10.1016/j.jbc.2025.110479
  6. Open Life Sci. 2025 ;20(1): 20251101
      Alzheimer's disease (AD) is characterized by amyloid-beta plaques and tau tangles in the brain, but these markers alone do not predict disease progression. The intersection of these pathologies with other processes including metabolic changes may contribute to disease progression. Brain glucose metabolism changes are among the earliest detectable events in AD. Pyruvate kinase (PKM) has been implicated as a potential biomarker to track these metabolic changes. We have developed an enzyme-linked immunosorbent assay (ELISA) to assess PKM levels in cerebrospinal fluid (CSF). First, we verified the relationship of CSF PKM levels with cognitive decline, revealing a correlation between elevated CSF PKM levels and accelerated cognitive decline in preclinical AD patients in a tau-dependent manner. We developed the ELISA using two PKM-specific antibodies and validated it through quality control steps, indicating robust quantification of PKM. We showed that ELISA measurements of PKM correlate with mass spectrometry values in matching samples. When tested on an independent cohort, the assay confirmed elevation of PKM in AD. These findings support the use of PKM as a potential biomarker for tracking early metabolic changes in AD, offering a novel tool for investigating metabolic alterations and their intersection with other underlying pathologies in AD progression.
    Keywords:  Alzheimer’s disease; biomarker; glucose metabolism; immunoassay; pyruvate kinase
    DOI:  https://doi.org/10.1515/biol-2025-1101
  7. Nucl Med Biol. 2025 Jul 15. pii: S0969-8051(25)00061-7. [Epub ahead of print]148-149 109052
       PURPOSE: This work aimed to monitor the Down Syndrome Ts65Dn animal model across lifespan to detect time-dependent in vivo molecular alterations that may be associated with neurodegeneration and neuroinflammation in this model.
    METHODS: Euploid and trisomic Ts65Dn animals were longitudinally evaluated at 2, 5, 14, 20, and 24 months of age using brain [18F]FDG PET and behavioral tasks (open field and novel object recognition). VOI-based SUV, Voxel-wise, and metabolic network analyses were performed. Cross-sectional post-mortem brain analysis was carried out at the same ages to measure neuronal loss and microglia activation.
    RESULTS: There was an increase in brain metabolism at 14 months of age in both genotypes, when compared to the other analyzed ages. [18F]FDG uptake correlated with microglia activation measured by Iba-1 immunohistochemistry, suggesting that the tracer increased due to a neuroinflammation process. In addition, metabolic network analysis showed more accentuated desynchronization in [18F]FDG uptake in older trisomic mice, than in euploid animals. At 24 months of age, trisomic animals presented a worse long-term memory recognition index and an age-dependent decrease in NeuN staining, which was not associated with [18F]FDG uptake.
    CONCLUSIONS: This is the first study to monitor the Ts65Dn mouse model throughout life - from 2 to 24 months of age - using [18F]FDG PET imaging and metabolic network analysis. Our results collectively highlight that trisomic mice experience early disruption in brain network organization, likely contributing to functional impairments associated with aging, neurodegeneration, and neuroinflammation.
    Keywords:  Brain connectivity; Down syndrome; Glucose metabolism; Neuroinflammation; Ts65Dn; [(18)F]FDG PET
    DOI:  https://doi.org/10.1016/j.nucmedbio.2025.109052
  8. Front Mol Neurosci. 2025 ;18 1602343
      The absence or dysfunction of the peroxisomal membrane protein Acyl-CoA Binding Domain-Containing Protein 5 (ACBD5) is the cause of the most recently discovered peroxisomal disorder "Retinal Dystrophy with Leukodystrophy" (RDLKD). ACBD5 is a tail-anchored protein, anchored by its C-terminus into the peroxisomal membrane; hence, the bulk of its amino acid sequence faces the cytosol. With respect to ACBD5's molecular functions, RDLKD is unique since it is not only an accessory protein for the import of very-long-chain fatty acids (VLCFAs) into peroxisomes but also the first identified peroxisomal tethering protein facilitating membrane contacts with the endoplasmic reticulum (ER). Consequently, RDLKD is neither a peroxisomal biogenesis disorder nor single enzyme deficiency, since a deficiency in ACBD5 likely affects several aspects of peroxisomal function including VLCFA degradation, ether lipid synthesis, docosahexaenoic acid synthesis but also the transfer of membrane lipids from the ER to peroxisomes. Hence, RDLKD appears to be a multifactorial disorder leading to a mosaic pathology, combining symptoms caused by the disruption of several pathways. In this review, we will highlight recent findings obtained from case reports of RDLKD patients as well as insights from ACBD5-deficient mouse models to better understand its complex retinal and brain pathology. Moreover, we will discuss the possible contribution of the different dysregulated metabolites in the neurological pathogenesis of this latest peroxisomal disorder.
    Keywords:  ACBD5; RDLKD; VAP; fatty acid metabolism; membrane contact sites; peroxisomes
    DOI:  https://doi.org/10.3389/fnmol.2025.1602343
  9. J Neuroinflammation. 2025 Jul 11. 22(1): 180
      
    Keywords:  Cholesterol; Lipid recycling; Neurological disease; Remyelination; Therapeutics
    DOI:  https://doi.org/10.1186/s12974-025-03490-8
  10. Res Sq. 2025 Jun 24. pii: rs.3.rs-6355361. [Epub ahead of print]
      Enhanced lipid metabolism, which involves the active import, storage, and utilization of fatty acids from the tumor microenvironment, plays a contributory role in malignant glioma transformation; thereby, serving as an important gain of function. In this work, through studies initially designed to understand and reconcile possible mechanisms underlying the anti-tumor activity of a high-fat ketogenic diet, we discovered that this phenotype of enhanced lipid metabolism observed in glioblastoma may also serve as a metabolic vulnerability to diet modification. Specifically, exogenous polyunsaturated fatty acids (PUFA) demonstrate the unique ability of short-circuiting lipid homeostasis in glioblastoma cells. This leads to lipolysis-mediated lipid droplet breakdown, an accumulation of intracellular free fatty acids, and lipid peroxidation-mediated cytotoxicity, which was potentiated when combined with radiation therapy. Leveraging this data, we formulated a PUFA-rich modified diet that does not require carbohydrate restriction, which would likely improve long-term adherence when compared to a ketogenic diet. The modified PUFA-rich diet demonstrated both anti-tumor activity and potent synergy when combined with radiation therapy in mouse glioblastoma models. Collectively, this work offers both a mechanistic understanding and novel approach of targeting this metabolic phenotype in glioblastoma through diet modification and/or nutritional supplementation that may be readily translated into clinical application.
    DOI:  https://doi.org/10.21203/rs.3.rs-6355361/v1
  11. bioRxiv. 2025 Jun 25. pii: 2025.06.23.661129. [Epub ahead of print]
      Peroxisomes are critical organelles that detoxify wastes while also catabolizing and anabolizing lipids. How peroxisomes coordinate protein import and support metabolic functions across complex tissues and timescales remains poorly understood in vivo . Using the Drosophila brain, we discover a striking enrichment of peroxisomes in the neuronal soma and the cortex glia that enwrap them. Unexpectedly, import of peroxisomal proteins into cortex glia, but not neurons, dramatically oscillated across time and peaked in the early morning. Rhythmic peroxisomal import in cortex glia autonomously required the circadian clock and Peroxin 5 (Pex5; peroxisomal biogenesis factor 5 homolog), with endogenous Pex5 protein peaking in the morning. Notably, removing Pex5 in cortex glia severely reduced sleep while concomitantly causing aberrant lipid metabolism characterized by ectopic lipid droplets and increases across multiple lipid families. Thus, the circadian import of peroxisomal proteins via Pex5 in cortex glia is essential for lipid homeostasis and organismal behavior.
    Highlights: Peroxisomal membrane and importer proteins are enriched in cortex glia (CG).The circadian clock autonomously decreases peroxisomal import in CG.The cytosolic importer Pex5 controls circadian peroxisomal import in CG. Loss of Pex5 in CG disrupts brain lipid metabolism and sleep behavior.
    DOI:  https://doi.org/10.1101/2025.06.23.661129
  12. iScience. 2025 Jul 18. 28(7): 112880
      Complex lipid metabolism plays a crucial role in regulating aging. We recently discovered that the phospholipid bis(monoacylglycero)phosphate (BMP) increases in aged human muscles and many mouse tissues. The phospholipase PLA2G15 is reportedly involved in BMP synthesis, however, its specific role in aging remains unknown. To elucidate the role of PLA2G15 in aging, we used Caenorhabditis elegans as a model. When silencing plag-15, the predicted worm orthologue of PLA2G15, we observed improved healthspan and lifespan extension. Semi-targeted lipidomics highlighted that instead of changes related to BMP, plag-15 RNAi led to lower levels of lysophosphatidic acid, lysophosphatidylcholine, and lysophosphatidylethanolamine. Transcriptome-guided epistasis experiments identified that the lifespan extension of plag-15 RNAi worms is regulated by transcription factors hlh-30 and elt-3, and lysosomal vitamin B12 transporter pmp-5 (human TFEB, GATA, and ABCD4 respectively). Overall, we conclude that targeting phospholipid remodeling through plag-15 could be a promising strategy to promote healthy aging.
    Keywords:  Lipidomics; Molecular physiology; Transcriptomics
    DOI:  https://doi.org/10.1016/j.isci.2025.112880
  13. ACS Pharmacol Transl Sci. 2025 Jul 11. 8(7): 1891-1918
      Lipids, mainly composed of cholesterol, phospholipids, sphingolipids, triacylglycerides, and fatty acids, have vital functions within cells. Some lipids function as signaling molecules or secondary messengers and are cellular membranes' energy sources and structural elements. More research is being conducted on metabolic reprogramming as a hallmark of cancer. However, compared with the metabolism of glucose or glutamine, lipid metabolism in cancer has received less attention. There is increasing evidence that certain parts of the lipid metabolism are altered in cancer cells. The alterations could influence the quantity of lipids involved in signaling functions, affect the synthesis and breakdown of lipids necessary for maintaining energy homeostasis, and modify the availability of structural lipids critical for membrane formation. The term "lipid metabolic reprogramming" refers to modifications in the lipid metabolism that can impact cellular processes such as cell division, growth, proliferation, and the cell cycle, ultimately resulting in cancer. Furthermore, interactions between cancer cells and nearby immune cells via an altered lipid metabolism promote the development and spread of tumors. The most recent studies on the involvement of lipid metabolism in different cancers and associated hallmarks and lipids in various aspects of cancer therapeutics, which affect multiple facets of tumorigenesis, are described in this review.
    Keywords:  and; cancer hallmarks; lipid droplets and rafts; lipid reprogramming; lipogenic factors; therapeutics; treatment resistance
    DOI:  https://doi.org/10.1021/acsptsci.5c00170
  14. Cell Commun Signal. 2025 Jul 16. 23(1): 341
       BACKGROUND: Deficits in mitochondrial bioenergetics and dynamics are strongly implicated in the selective vulnerability of striatal neurons in Huntington´s disease. Beyond these neuron-intrinsic factor, increasing evidence suggest that non-neuronal mechanisms, particularly astrocytic dysfunction involving disrupted homeostasis and metabolic support also contribute to disease progression. These findings underscore the critical role of metabolic crosstalk between neurons and astrocytes in maintaining striatal integrity. However, it remains unclear whether this impaired communication affects the transfer of mitochondria from astrocytes to striatal neurons, a potential metabolic support mechanism that may be compromised in Huntington´s Disease.
    METHODS: Primary striatal astrocytes were obtained from wild-type and R6/1 mice to investigate mitochondrial dynamics. Expression levels of key mitochondrial fusion and fission proteins were quantified by Western blotting and RT-PCR. Mitochondria morphology, oxidative stress and membrane potential were assessed using confocal microscopy following staining with mitochondria-specific dyes. Mitochondrial respiration was measured using the Oxygraph-2k respirometer system (Oroboros Instruments). Transmitophagy was evaluated by confocal imaging after labeling astrocytic mitochondria with Mitotracker dyes. To assess the functional impact of mitochondrial transfer on neurons, Sholl analysis, neuronal death and oxidative stress levels were quantified using specific fluorogenic probes.
    RESULTS: Striatal astrocytes from HD mice exhibited a significant increase in mitochondrial fission, and mitochondrial oxidative stress, mirroring alterations previously reported in striatal neurons. Analysis of mitochondrial oxygen consumption rate (OCR) revealed elevated respiration activity and enhanced ATP-linked respiration, indicative of a hypermetabolic state. Concurrently, increased lactate production suggested a shift toward dysregulated astrocytic energy metabolism. These mitochondrial alterations were functionally detrimental: astrocytic mitochondria derived from HD mice when taken up by striatal neurons via transmitophagy, led to reduced neuronal branching and disrupted oxidative homeostasis.
    CONCLUSIONS: Our findings demonstrate that striatal astrocytes from HD mice exhibit a hypermetabolic phenotype, characterized by increased mitochondrial respiration, disrupted mitochondrial dynamics, and elevated mitochondrial oxidative stress. Importantly, we identify a novel mechanism of astrocyte-neuron interaction involving the transfer of dysfunctional mitochondria from astrocytes to neurons. The uptake of these compromised mitochondria by striatal neurons results in reduced neuronal branching and increased reactive oxygen species (ROS) production. Collectively, these results highlight the pathological relevance of impaired astrocyte-to-neuron mitochondrial transfer and emphasize the contributory role of astrocytic dysfunction in Huntington´s disease progression.
    Keywords:  Astrocytes; Huntingtin; Mitochondria transfer; Neuroglial communication; R6/1 mice; Striatum
    DOI:  https://doi.org/10.1186/s12964-025-02341-6
  15. FEBS J. 2025 Jul 18.
      Phosphoserine phosphatase (PSP) catalyzes the dephosphorylation of 3-phosphoserine, which is the final step in the de novo biosynthesis of l-serine (l-Ser) via the phosphorylated pathway in human astrocytes. Individuals who are homozygous or compound heterozygous for functionally defective PSP variants exhibit reduced cerebrospinal fluid l-Ser levels and severe neurological symptoms. In the present study, single nucleotide polymorphisms in PSP were identified in hippocampal samples from Alzheimer's disease (AD) patients. Two single nucleotide polymorphisms, likely forming a haplotype (chr7:56088825 T>A and chr7:56088811 T>C, encoding R27S and D32G PSP variants, respectively), were detected exclusively in AD patients (three females and one male). Biochemical characterization of the recombinant R27S/D32G PSP enzyme revealed a slight decrease in thermostability, a 38-fold reduction in catalytic efficiency and a two-fold increase in IC50 for l-Ser, with the D32G substitution being the primary contributor to these effects. Despite its reduced enzyme activity, the R27S/D32G variant did not impair l-Ser biosynthesis either in an in vitro reconstructed pathway or in U251 human glioblastoma cells ectopically expressing the variant under heterozygous conditions. In these cells, PSP colocalized extensively with the other two phosphorylated pathway enzymes, namely phosphoglycerate dehydrogenase and phosphoserine aminotransferase, suggesting that they assemble into a functional complex, known as the serinosome. Notably, the R27S/D32G PSP variant exhibited increased nuclear localization compared to the wild-type enzyme. This mislocalization raises the intriguing possibility that PSP's moonlighting functions, including its putative role as a protein phosphatase, may be affected, potentially implicating it in pathways beyond l-Ser biosynthesis.
    Keywords:   l‐serine; moonlighting function; phosphorylated pathway; serine metabolism; serinosome
    DOI:  https://doi.org/10.1111/febs.70169
  16. JCI Insight. 2025 Jul 15. pii: e188459. [Epub ahead of print]
      Maladaptive integrated stress response (ISR) activation is observed in human diseases of the brain. Genetic mutations of eIF2B, a critical mediator of protein synthesis, cause chronic pathway activation resulting in a leukodystrophy but the precise mechanism is unknown. We generated N208Y eIF2Bα mice and found that this metabolite binding mutation leads to destabilization of eIF2Bα, a systemic ISR, and neonatal lethality. 2BAct, an eIF2B activator, rescued lethality and significantly extended the lifespan of this severe model, underscoring its therapeutic potential in pediatric disease. Continuous treatment was required for survival, as withdrawal led to ISR induction in all tissues and rapid deterioration, thereby providing a model to assess the impact of the ISR in vivo by tuning drug availability. Single nuclei RNA-sequencing of the CNS identified astrocytes, oligodendrocytes, and ependymal cells as the cell types most susceptible to eIF2B dysfunction and revealed dysfunctional maturation of oligodendrocytes. Moreover, ISR activation decreased cholesterol biosynthesis, a process critical for myelin formation and maintenance. As such, persistent ISR engagement may contribute to pathology in other demyelinating diseases.
    Keywords:  Cell biology; Cholesterol; Demyelinating disorders; Metabolism; Mouse models; Neuroscience
    DOI:  https://doi.org/10.1172/jci.insight.188459
  17. Biol Sex Differ. 2025 Jul 17. 16(1): 54
       BACKGROUND: Cognitive deficits and brain glucose hypometabolism, lipid peroxidation and mitochondrial dysfunction are early pathological events in murine models and patients with Alzheimer's disease (AD). Data from our previous research indicate that transgenic mice of the APP23 line, a murine AD model, exhibited higher energy expenditure and mitochondrial dysregulation in the liver as early as 3 months of age, which is considered the preplaque stage. Since women have a higher risk and mortality rate for AD, with potential sex-specific confounders as longevity, biological, genetic, and social factors also needing to be considered, sex differences in energy metabolism in AD remain insufficiently investigated.
    METHODS: Here, we investigated sex-specific differences in mitochondrial respiration and metabolic profiles of 3-4-month-old, preplaque APP23 transgenic mice, in which we did not detect inflammatory signals and pathological amyloid-beta (Aß) plaques in brain or liver. Their mitochondrial respiration was assessed measuring oxygen consumption rates in isolated primary hepatocytes, stromal vascular cells (SVCs) and re-differentiated adipocytes. Furthermore, we analyzed energy balance, including food intake, locomotor activity, energy expenditure and fecal calorie loss.
    RESULTS: We observed an upregulation of hepatic mitochondrial respiration in preplaque APP23 females. Female-derived SVCs and differentiated adipocytes improved mitochondrial flexibility with palmitate loading in vitro, which was in line with decreased plasma triglycerides in preplaque APP23 females in vivo. However, no differences in mitochondrial respiration were detected in hepatocytes and re-differentiated adipocytes derived from male APP23 mice. Furthermore, we corroborated an increased mortality during the preplaque stage, particularly in females, which exhibited reduced hyperactivity and caloric intake before death compared to survivors.
    CONCLUSIONS: Our data demonstrate that preplaque APP23 female mice have disequilibrated mitochondrial oxidation in hepatocytes and adipocytes as well as higher energy expenditure due to increased activity before AD manifestation. In contrast, male APP23 mice did not exhibit such metabolic changes. Constant excessive energy loss and limited calorie supply potentially contribute to the higher risk of mortality, especially in APP23 females during young adulthood. Alzheimer's disease (AD) affects men and women differently, with women at higher risk and mortality. This study explored sex differences in energy metabolism using APP23 transgenic mice, a model of AD, at young age (3-4 months) - before pathological amyloid-beta (Aß) plaques develop in the brain and liver. Female APP23 mice showed increased mitochondrial activity in liver and fat cells, higher energy expenditure, and more movement while eating less. They also excreted more energy in their feces. Notably, female APP23 mice had a lower survival rate than males. Before death, they became less active and ate even less, suggesting an inability to maintain energy balance. These findings indicate that female APP23 mice experience excessive energy loss, which may contribute to early mortality. Understanding these sex-specific metabolic differences could provide new insights into AD progression and highlight the need for targeted treatments.
    DOI:  https://doi.org/10.1186/s13293-025-00737-0
  18. Aging Cell. 2025 Jul 16. e70175
      The misclassification of functional genomic loci as pseudogenes has long obscured critical regulators of cellular homeostasis, particularly in aging-related pathways. One such locus, originally annotated as RPL29P31, encodes a 17-kDa protein now redefined as PERMIT (Protein that Mediates ER-Mitochondria Trafficking). Through rigorous experimental validation-including antibody development, gene editing, lipidomics, and translational models-p17/PERMIT has emerged as a previously unrecognized mitochondrial trafficking chaperone. Under aging or injury-induced stress, p17 mediates the ER-to-mitochondria translocation of Ceramide Synthase 1 (CerS1), facilitating localized C18-ceramide synthesis and autophagosome recruitment to initiate mitophagy. Loss of p17 impairs mitochondrial quality control, accelerating neurodegeneration, and sensorimotor decline in both injury and aging models. This Perspective highlights p17 as a paradigm-shifting discovery at the intersection of lipid signaling, mitochondrial biology, and genome reannotation, and calls for a broader reassessment of the "noncoding" genome in aging research. We summarize a rigorous multi-platform validation pipeline-including gene editing, antibody generation, lipidomics, proteomics, and functional rescue assays-that reclassified p17 as a bona fide mitochondrial trafficking protein. Positioned at the intersection of lipid metabolism, organelle dynamics, and genome reannotation, p17 exemplifies a growing class of overlooked proteins emerging from loci historically labeled as pseudogenes, urging a systematic reevaluation of the "noncoding" genome in aging research.
    DOI:  https://doi.org/10.1111/acel.70175
  19. bioRxiv. 2025 Jul 12. pii: 2025.07.08.663730. [Epub ahead of print]
      Visualizing signaling systems in the brain with high spatial resolution is critical to understand brain function and to develop therapeutics. Especially enzymes are often regulated on the post-translational level, resulting in a disconnect between protein levels and activity. Conventional antibody-based methods have limitations, including potential cross reactivity and the inability of antibodies to discriminate between active and inactive enzyme states. Monoacylglycerol lipase (MAGL), an enzyme degrading the neuroprotective endocannabinoid 2-arachidonoylglycerol, is the target of inhibitors currently in clinical trials for the treatment of several neurological disorders. To support translational and (pre)clinical studies and fully realize the therapeutic opportunities of MAGL inhibitors, it is essential to map the spatial distribution of MAGL activity throughout the brain in both health and disease. Here, we introduce selective fluorescent activity-based probes for MAGL enabling direct visualization of its enzymatic activity in lysates, cultured cells and tissue sections. We show that oxidative stress, which inactivates MAGL through the oxidation of regulatory cysteines, reduces probe labeling, thereby validating the probes activity-dependence. Extending this approach, we developed an activity-based histology protocol to visualize MAGL activity in fresh-frozen mouse and human brain tissues. This approach revealed robust MAGL activity in astrocytes and presynaptic terminals within the mouse hippocampus, and further allows detection of MAGL activity in the human cerebral cortex. Collectively, these findings establish selective activity-based probes as powerful tools mapping MAGL activity with high spatial resolution across mammalian brain tissue.
    DOI:  https://doi.org/10.1101/2025.07.08.663730
  20. Int J Mol Sci. 2025 Jul 03. pii: 6404. [Epub ahead of print]26(13):
      Microglia-mediated neuroinflammation is a key driver of Alzheimer's disease (AD). In AD, microglia are activated and trigger an increased secretion of pro-inflammatory factors. Rhein, an anthraquinone compound extracted from rhubarb, has been shown to reduce the secretion of pro-inflammatory cytokines including TNF-α and IL-1β in activated microglia. However, the mechanism of rhein on microglia-mediated neuroinflammation and neuronal damage in AD remains unclear. In this study, we found that rhein improved behavioral abnormalities in AD rats and reduced the levels of inflammatory factors such as IL-1β, iNOS, and NO in the brain of AD rats. In the LPS-induced microglial model, rhein significantly reduced the levels of inflammatory factors to improve neuroinflammation. Untargeted metabolomics showed that the reprogramming of glutamine metabolism occurred in M1 microglia. Targeted metabolomics and 13C, 15N isotope tracing experiments demonstrated that rhein regulated the metabolite levels in the glutamine-aspartate-arginine metabolic pathway. Meanwhile, the upregulated expression of proteins such as GLS1 and GOT1 within this pathway was reversed by rhein. Furthermore, we found that the glutamine-aspartate-arginine metabolic pathway regulates the production of nitric oxide (NO, a neuroinflammatory mediator). Rhein alleviates neuronal damage by inhibiting the glutamine-aspartate-arginine-NO metabolic pathway. In conclusion, our study shows that rhein may inhibits NO production by regulating the glutamine-aspartate-arginine metabolic pathway in activated microglia, thereby inhibiting the neuroinflammation and neuronal damage in AD.
    Keywords:  Alzheimer’s disease; Rhein; metabolic flux; metabolomics; microglia; neuroinflammation; neuron
    DOI:  https://doi.org/10.3390/ijms26136404
  21. Neurosci Lett. 2025 Jul 14. pii: S0304-3940(25)00206-X. [Epub ahead of print]864 138318
      Across a range of neurological disorders, there is a growing appreciation for how the gut influences brain health, but few ways of monitoring its effects. Although nutrition influences traumatic brain injury (TBI) recovery, its influence on biomarkers-whether as an intervention or confounder-is poorly understood. Beyond specialized diets, standard rodent diets may also affect brain function. Neuron-derived extracellular vesicles (NDEVs) offer a brain-specific complement to circulating biomarkers, but their sensitivity to diet is unknown. In this study, we isolated miRNAs from NDEVs from the serum of healthy and mild TBI (mTBI) mice fed a semi-synthetic or grain-based diet. NDEV miRNAs encoded dietary differences based on injury condition, suggesting that NDEVs are sensitive to dietary changes and may be able to track diet's effect on TBI recovery. Additionally, we found that diet influenced injury biomarkers, underscoring diet as a confounding variable for NDEV miRNA biomarkers. Together, these findings highlight NDEVs as a promising tool for monitoring the effects of subtle dietary differences on brain health and the importance of diet reporting to improve study reproducibility.
    Keywords:  Biomarkers; Diet; mTBI
    DOI:  https://doi.org/10.1016/j.neulet.2025.138318