bims-micgli Biomed News
on Microglia
Issue of 2025–10–26
thirteen papers selected by
Matheus Garcia Fragas, Universidade de São Paulo



  1. PLoS Biol. 2025 Oct;23(10): e3003426
      Recent advances in single-cell transcriptomics have led to the identification of disease-associated microglia (DAM) as a distinct, conserved microglia state associated with mouse models of Alzheimer's disease (AD) and amyotrophic lateral sclerosis, and with aging. DAM are characterized by downregulation of homeostatic genes and upregulation of lipid metabolism and phagocytosis genes, including key risk factors for AD in humans. Although characterized in models of AD, whether DAM acts as universal sensor across all neurodegenerative diseases remains unknown. This Essay discusses the dynamics, origins, and therapeutic potential of DAM in neurodegeneration, alongside evidence supporting a protective role for them in regulating disease processes.
    DOI:  https://doi.org/10.1371/journal.pbio.3003426
  2. Sci Adv. 2025 Oct 24. 11(43): eads9181
      Microglial functions rely on their morphodynamic versatility and inflammatory response, yet the molecular determinants, particularly ion channels and receptors, remain poorly understood. Here, we identify chloride intracellular channel 1 (CLIC1), a protein known to exist in both soluble and membrane-associated forms, as highly enriched in human and murine microglia, with minimal expression in other brain cells. Acute blockade or genetic deletion of CLIC1 markedly attenuates microglial surveillance by reducing ramification and motility, without affecting chemotaxis. This phenotype is recapitulated in xenografted human microglia and human brain tissue. Mechanistically, CLIC1 effects involve interactions with actin-binding ezrin, radixin, and moesin (ERM) proteins, suggesting a role in linking the plasma membrane to the cytoskeleton. Contrary to its name, CLIC1 functions are chloride-independent and thus unlikely to reflect ion channel activity. This is supported by patch-clamp electrophysiology revealing lack of chloride conductance in surveillant microglia. Following ATP-evoked activation, CLIC1 blockade strongly suppresses NLRP3-dependent interleukin-1β release, suggesting therapeutic potential against neuroinflammation.
    DOI:  https://doi.org/10.1126/sciadv.ads9181
  3. Alzheimers Dement. 2025 Oct;21(10): e70838
       INTRODUCTION: Alzheimer's disease (AD), the leading cause of dementia, is more common in females. Although sex differences in tau pathology have been reported in AD, findings remain inconsistent. Down syndrome (DS), caused by trisomy 21, is the most common genetic cause of AD (DS-AD) and features tau pathology, but sex effects in DS-AD remain unclear.
    METHODS: We examined post mortem brain samples from individuals with DS-AD, DS without AD, and a rare partial trisomy 21 (PT) case with only two amyloid precursor protein (APP) gene copies. PHF1 tau, total tau, and sarkosyl-soluble and insoluble fractions were quantified by group and sex.
    RESULTS: PHF1 tau was significantly elevated in DS-AD, especially in females. Lower total tau in DS-AD males explained the absence of sex differences after normalization. Sarkosyl-insoluble tau was also higher in DS-AD females. DS without AD, and the PT case showed minimal pathology.
    DISCUSSION: These findings suggest sex-specific tau dynamics in DS-AD and support a role for APP dosage.
    HIGHLIGHTS: Tau pathology is significantly elevated in individuals with DS-AD, especially in females. Female DS-AD brains show markedly higher PHF1 (S396/404) and sarkosyl-insoluble tau levels compared to males. The observed sex difference in phosphorylated tau is driven by lower total tau in DS-AD males. Minimal tau pathology is present in DS without AD and in a rare partial trisomy 21 case. These findings implicate APP gene dosage in tau pathology in DS-AD.
    Keywords:  APP; Alzheimer's disease; Down syndrome; PHF1 tau; sarkosyl‐insoluble; tau
    DOI:  https://doi.org/10.1002/alz.70838
  4. Nat Neurosci. 2025 Oct 23.
      While circadian rhythm disruption may promote neurodegenerative disease, the impact of aging and neurodegenerative pathology on circadian gene expression patterns in different brain cell types remains unknown. Here we used a translating ribosome affinity purification to identify the circadian translatomes of astrocytes, microglia and bulk tissue in healthy mouse cortex and in the settings of amyloid-β plaque pathology or aging. We show that glial circadian translatomes are highly cell-type-specific and exhibit profound, context-dependent reprogramming in response to amyloid pathology or aging. Transcripts involved in glial reactivity, immunometabolism and proteostasis, as well as nearly half of all Alzheimer's disease risk genes, displayed circadian oscillations, many of which were altered by pathology. Microglial oxidative stress and amyloid phagocytosis showed temporal variation in gene expression and function. Thus, circadian rhythms in gene expression are cell-dependent and context dependent, and provide important insights into glial function in health, Alzheimer's disease and aging.
    DOI:  https://doi.org/10.1038/s41593-025-02067-1
  5. Neuron. 2025 Oct 17. pii: S0896-6273(25)00745-7. [Epub ahead of print]
    Alzheimer’s Disease Neuroimaging Initiative (ADNI)
      We previously reported that T96K is a gain-of-function mutation in TREM2 based on its ability to increase ligand-dependent activation. Here, we show that TREM2T96K increases risk for Alzheimer's disease (AD) in a whole-genome sequencing dataset comprised of family-based and case-control samples. Trem2T96K also reduced clustering of microglia around β-amyloid (Aβ) plaques exclusively in female 5xFAD mice. Furthermore, T96K decreased levels of soluble Trem2 in female 5xFAD mice and human microglial cell cultures. We also observed impaired uptake of Aβ in Trem2T96K knockin microglial cells. Moreover, Trem2T96K reduced total area of phagocytic microglia, specifically in female 5xFAD mice. Single-cell RNA sequencing (scRNA-seq) profiling of microglia revealed that Trem2T96K impairs the transition of homeostatic microglia into disease-associated microglia (DAM) in female 5xFAD mice. Downregulated inflammatory pathways associated with Trem2T96K included interleukin (IL)-6/JAK/STAT3, complement, and interferon (IFN)-γ response. Collectively, our results indicate that, like the loss-of-function mutation R47H, Trem2T96K adversely affects microglial function in a sex-dependent manner.
    Keywords:  Alzheimer’s; T96K; TREM2; amyloid; gain of function; innate immunity; microglia; neuroinflammation; single-cell RNA-seq; uptake
    DOI:  https://doi.org/10.1016/j.neuron.2025.09.032
  6. Proc Natl Acad Sci U S A. 2025 Oct 28. 122(43): e2509903122
      In Alzheimer's disease (AD), hyperactivated microglia produce inflammatory mediators that contribute to neuroinflammation and neuronal damage. Amyloid precursor protein (APP), a transmembrane protein expressed in many cell types, including neurons and microglia, plays a critical role in AD pathogenesis via its secretase-mediated processing to release the C-terminal 99-residue transmembrane fragment (C99) that is further cleaved to yield amyloid-β peptides. Voltage-gated proton channels (Hv1) have been implicated in microglial activation and release of inflammatory mediators, but the potential role of these channels in human microglia and AD pathogenesis remains unclear. Here, we demonstrate that human induced pluripotent stem cell-derived microglia (iMG) express native Hv1 channels with biophysical and pharmacological attributes determined by their coassembly with APP and that APP knockdown decreases Hv1 currents, suppressing cytokine and reactive oxygen species release. In HEK293T cells, APP is shown to increase current by favoring channel opening at more negative membrane potentials. C99 is sufficient to assemble with Hv1 and alters channel function even more significantly than APP. Coimmunoprecipitation, total internal reflection fluorescence microscopy, and altered pharmacology further demonstrate that C99 forms stable complexes with Hv1 in the plasma membrane. In addition, we find that two early-onset AD mutations in APP (E682K and D694N) that reside within C99 significantly increase voltage-dependent channel activity beyond that induced by wild type C99, rationalizing their enhanced mediation of neuroinflammation.
    Keywords:  APP; Alzheimer’s disease; C99; Hv1; voltage-gated proton channel
    DOI:  https://doi.org/10.1073/pnas.2509903122
  7. Mol Neurodegener Adv. 2025 ;1(1): 4
    Dominantly Inherited Alzheimer Network
       Background: Over 300 mutations in PSEN1 have been identified as causes of early-onset Alzheimer's disease (EOAD). While these include missense mutations and a few insertions, deletions, or duplications, none result in open reading frame shifts, and all alter γ-secretase function to increase the long/short Aβ ratio.
    Methods: We identified a novel heterozygous PSEN1 nonsense variant, c.325A > T, in a patient and his father, both presenting with EOAD, resulting in the substitution of lysine 109 with a premature stop codon at position (p.K109*). This produces a truncated 109 amino acid (aa) N-terminal PSEN1 fragment. Functional characterization was performed using overexpression models and a heterozygous mouse model (Psen1K109*/+).
    Results: In overexpression models, downstream ATGs serve as alternative starting codons, generating a > 37 kDa and a > 27 kDa PSEN1 C-terminal fragment (PSEN1-CTFA and PSEN1-CTFB, respectively) that retain the two catalytic aspartates of γ-secretase. Heterozygous Psen1K109*/+ mice exhibited subtle phenotypic defects, including reduced Pen2 expression and mild APP-CTF accumulation. Notably, aged mice demonstrated significantly increased Psen2 protein expression, potentially contributing to an elevated Aβ42/Aβ38 ratio.
    Conclusions: These findings indicate that PSEN1 c.325A > T (p.K109*) is not a complete loss-of-function mutation. However, to what extent and by what mechanism it contributes to EOAD pathogenesis remains unclear.
    Supplementary Information: The online version contains supplementary material available at 10.1186/s44477-025-00004-x.
    Keywords:  Alzheimer’s disease; PSEN1; loss-of-function; γ-secretase
    DOI:  https://doi.org/10.1186/s44477-025-00004-x
  8. Sci Adv. 2025 Oct 24. 11(43): eado4330
      Golgi fragmentation is an early and common feature of neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS) and Alzheimer's disease (AD). However, whether a shared mechanism drives Golgi fragmentation across different neurodegenerative conditions remains unclear. Here, we identify the E3 ubiquitin-protein ligase Itchy homolog (ITCH) as a key regulator of proteotoxicity through its role in inducing Golgi fragmentation. Disease-associated accumulation of ITCH promotes fragmentation of both the cis- and trans-Golgi networks, disrupting protein sorting and impairing lysosomal functions. The ITCH-dependent lysosomal dysfunction compromises the clearance of misfolded proteins associated with several neurodegenerative diseases. Inhibition of ITCH protects against proteotoxicity in both mammalian neurons and Drosophila models of neurodegeneration. The accumulation of ITCH in patients with ALS and AD is attributed to up-regulation of the ubiquitin-specific protease USP11, which deubiquitinates and stabilizes ITCH. These results uncover a pathogenic pathway regulating Golgi integrity and contributing to the development of neurodegenerative diseases.
    DOI:  https://doi.org/10.1126/sciadv.ado4330
  9. J Neuroinflammation. 2025 Oct 22. 22(1): 239
      Multiple sclerosis (MS) is an autoimmune and neuroinflammation disease characterized by axonal damage, inflammatory demyelination, and neurodegeneration. However, the precise mechanisms underlying MS pathogenesis remain largely unclear. Here, we identify ZNRF1, an E3 ubiquitin ligase, as a critical regulator of experimental autoimmune encephalomyelitis (EAE), a murine model that recapitulates the autoimmune demyelination features of MS. Mice lacking ZNRF1 exhibit increased susceptibility to EAE progression. Notably, ZNRF1 depletion in peripheral myeloid cells, but not in microglia, leads to enhanced immune cell infiltration into the central nervous system, resulting in demyelination and exacerbated disease severity. The heightened EAE severity in Znrf1-deficient mice is associated with increased polarization of Th1 and Th17 cells, elevated antigen-specific T helper cell proliferation, and amplified immune responses. Furthermore, following EAE induction, macrophages from Znrf1-deficient mice display elevated surface expression of MHC class II (MHC-II) molecules. Collectively, our findings suggest that ZNRF1 in peripheral myeloid cells plays a suppressive role in neuroinflammation by regulating MHC-II surface expression, thereby controlling antigen-specific T-cell proliferation and activation.
    Keywords:  Experimental autoimmune encephalomyelitis; Macrophage; Multiple Sclerosis; Neuroinflammation; ZNRF1
    DOI:  https://doi.org/10.1186/s12974-025-03550-z
  10. Acta Neuropathol. 2025 Oct 23. 150(1): 44
      Parkinson's disease (PD) pathogenic mutations in leucine-rich repeat kinase 2 (LRRK2) are associated with endolysosomal dysfunction across cell types, and carriers of LRRK2 mutations variably present with phosphorylated tau and α-synuclein deposits in post-mortem analysis. LRRK2 mutations increase the phosphorylation of Rab substrates including Rab12 and Rab10. Rab12 and Rab10 are expressed in neuronal and non-neuronal cells with localization to membranes in the endolysosomal compartment, and lysosomal stress activates LRRK2 phosphorylation of Rabs. In this study, using antibodies directed to the LRRK2-mediated phosphorylation sites on Rab12 at amino acid Ser106 (pS106-Rab12) and Rab10 at amino acid Thr73 (pT73-Rab10), we test whether aberrant LRRK2 phosphorylation is associated with tau and/or α-synuclein pathology across clinically distinct neurodegenerative diseases. Analysis of brain tissue lysates and immunohistochemistry of pathology-susceptible brain regions demonstrate that pS106-Rab12 levels are increased in Alzheimer's disease (AD) and Lewy body disease (LBD), including PD with and without G2019S LRRK2 mutation. At early pathological stages, phosphorylated Rab12 localizes to granulovacuolar degeneration bodies (GVBs), which are thought to be active lysosomal-like structures, in neurons. pS106-Rab12-positive GVBs accumulate with pathological tau across brain tissues in AD and LBD, and in G2019S LRRK2 mutation carriers. In a mouse model of tauopathy, pS106-Rab12 localizes to GVBs during early tau deposition in an age-dependent manner. While GVBs are largely absent in neurons with mature protein pathology, subsets of both tau and α-synuclein inclusions appear to incorporate pS106-Rab12 at later pathological stages. Further, pS106-Rab12 labels GVBs in neurons and shows co-pathology with tau inclusions in primary tauopathies including Pick's disease, progressive supranuclear palsy, and corticobasal degeneration. Finally, pT73-Rab10 is elevated and localizes to GVBs, but not tau and α-synuclein inclusions, in AD and LBD, including G2019S LRRK2 mutation carriers. These results implicate LRRK2 kinase activity and Rab phosphorylation in endolysosomal dysfunction in tau- and α-synuclein-associated neurodegenerative diseases.
    Keywords:  Alzheimer’s disease; LRRK2; Parkinson’s disease; Rab12; Synuclein; Tau
    DOI:  https://doi.org/10.1007/s00401-025-02951-x
  11. Lancet Neurol. 2025 Nov;pii: S1474-4422(25)00313-8. [Epub ahead of print]24(11): 931-944
       BACKGROUND: Understanding the neuropathological effects of amyloid β (Aβ)-targeting therapies and amyloid-related imaging abnormalities (ARIA) in Alzheimer's disease is critical for optimising treatment efficacy and patient outcomes. Comparing Aβ PET imaging with neuropathological assessments provides context for evaluating the extent of Aβ clearance and interpreting in-vivo biomarkers. We aimed to assess clinicopathological changes and ARIA-related effects in aducanumab-treated versus untreated Alzheimer's disease.
    METHODS: This retrospective case-control study included five aducanumab-treated participants from clinical trials conducted at the Mayo Clinic (2016-21) who underwent autopsy (2020-23). Treated participants were matched by autosomal dominant Alzheimer's disease mutation or APOE genotype, age at cognitive symptom onset, and sex to 12 untreated participants from the Mayo Clinic Alzheimer's Disease Research Center and Mayo Clinic Study of Aging cohorts in the Mayo Clinic brain bank (Jacksonville, FL, USA). Cognitive, imaging, and neuropathological outcomes were compared using descriptive analyses and Mann-Whitney U tests.
    FINDINGS: Aducanumab-treated participants comprised four males and one female, all carrying at least one APOE ∊4 allele, with two harbouring a PSEN1 mutation. Cumulative dosages of aducanumab ranged from 5 mg/kg to 241 mg/kg; all participants cognitively declined during treatment, and two exhibited ARIA. Reductions in [18F]florbetapir PET Centiloid values ranged from -6% to -81% compared with baseline. Treatment-to-death intervals ranged from 5 months to 41 months. Neuropathological analyses revealed clearance of Aβaa1-8 and Aβ42 localised to cortical layer I in treated participants, with no significant clearance in deeper cortical layers. Regions corresponding to ARIA on MRI showed microinfarcts with haemosiderin, complement activation, and CD68-positive vessel walls originating from Aβ-laden leptomeningeal and penetrating vessels.
    INTERPRETATION: Disproportionate Aβ clearance and ARIA-associated neuropathology localised to superficial cortical layers suggest a distinctive pattern of target engagement by aducanumab. These findings inform understanding and monitoring of similar Aβ-targeting therapies.
    FUNDING: Alzheimer Nederland, National Institute on Aging, and Alzheimer's Association.
    DOI:  https://doi.org/10.1016/S1474-4422(25)00313-8
  12. Cell Mol Neurobiol. 2025 Oct 21. 45(1): 89
      Degeneration of myelinating oligodendrocytes and the resulting breakdown of the myelin sheath are key drivers of neurodegeneration and disability across numerous central nervous system pathologies. Thus, a compelling strategy to preserve neuronal function is to promote endogenous myelin repair by oligodendrocyte precursor cells (OPCs). Extracellular vesicles (EVs) secreted by pro-regenerative microglia have been shown to enhance OPC maturation and remyelination across different experimental models. Yet, the mechanisms by which microglia-derived EVs exert their beneficial effects on OPCs are not fully understood. In this study, we performed transcriptomic profiling of primary murine OPCs treated during differentiation with EVs obtained from donor microglia following stimulation with pro-inflammatory cytokines (i-EVs), interleukin-4 (IL4-EVs), or mesenchymal stem cells in the presence of the inflammatory cocktail (MSC-EVs; GEO accession number: GSE304130). Compared to controls, IL4-EVs and MSC-EVs induced robust changes in gene expression, whereas i-EVs elicited far fewer alterations. Using bioinformatic analyses, we identified the molecular pathways significantly modulated by microglial EVs, revealing a large overlap between IL4-EV and MSC-EV targets. Notably, many of these shared pathways centered on mitochondrial function and bioenergetic metabolism. Upstream regulatory network inference further pinpointed candidate transcription factors and kinases that may drive EV-induced transcriptional reprogramming in OPCs. Hence, our findings indicate that rewiring mitochondria-associated pathways is a core mechanism underlying the pro-differentiation effects of microglial EVs on OPCs. Elucidating these intracellular circuits will open new avenues for developing EV-based or mitochondria-targeted remyelinating therapies.
    Keywords:  Extracellular vesicles; Microglia; Mitochondria; Neuroinflammation; OPCs; Oligodendrocytes; Remyelination
    DOI:  https://doi.org/10.1007/s10571-025-01612-7
  13. Glia. 2025 Oct 22.
      Neuroinflammation mediated by microglia and astrocytes is a major component of traumatic brain injury (TBI) pathophysiology. The sterile alpha and TIR motif containing 1 (SARM1) protein has been identified to play a key role in neurodegeneration and inflammatory cascades. Therefore, we hypothesized that the inhibition of SARM1 would prevent glial reactivity following TBI and could be targeted for therapeutic intervention. TBI was modeled in wild type (WT) and SARM1 knock-out (SARM1-KO) mice of both biological sexes by midline fluid percussion injury. At 7 or 28 days post-injury, brains were collected to examine glial reactivity via immunohistochemistry and compared to naïve controls. The density of microglia and glial fibrillary acidic protein (GFAP) immunoreactivity of astrocytes was significantly increased across time post-injury. Furthermore, microglial morphological changes and increased colocalization with a surrogate marker of phagocytosis (CD68) were evident at 7 days post-injury. In the absence of SARM1, microglial density and colocalization with CD68 was greater compared with WT animals, regardless of TBI. However, there were no differences in GFAP immunoreactivity with the genetic deletion of SARM1. When investigating biological sexes, the TBI-induced increase in microglial density and cell volume was greater in male mice at 7 days post-injury; however, microglia were more deramified in females. There were no significant differences in GFAP immunoreactivity between male and female mice. These results indicate that the genetic deletion of SARM1 is not sufficient to alter GFAP-labeling of astrocytes; however, SARM1 appears to impact microglial density and CD68 colocalization in the naïve and injured brain.
    Keywords:  SARM1; TBI; astrocyte; biological sex; microglia; neurodegeneration
    DOI:  https://doi.org/10.1002/glia.70095