bims-mihora Biomed News
on Mitohormesis, repair and aging
Issue of 2025–12–14
twenty papers selected by
Lisa Patel, Istesso



  1. Mitochondrion. 2025 Dec 04. pii: S1567-7249(25)00104-7. [Epub ahead of print]87 102107
      Mitochondrial protein import is necessary to ensure the proper functioning of the organelle of the cell as a whole. More than 1000 proteins are synthesized on cytosolic ribosomes and then imported into mitochondria through translocases such as TOMM and TIMM complexes. Upon entry, they can reach their final mitochondrial compartment, namely the outer mitochondrial membrane (OMM), the intermembrane space (IMS), the inner mitochondrial membrane (IMM), and the matrix. In this review, we will first explore the main mitochondrial protein import mechanisms. Then, we will focus on how import deficiencies may trigger stress paradigms. Stress response pathways are activated to restore correct cellular homeostasis. We will explore four interconnected pathways at the cellular or mitochondrial scale, which can compensate for import alterations. These are the DELE1-HRI axis combined with the ISR, the UPRam, the UPRmt, and mitophagy. Their activation depends on the extent of import alteration, with ISR and UPRmt pathways activated in conditions of low stress. If stress levels are too high, the elimination of dysfunctional mitochondria by mitophagy is triggered. Last, we will explore how mitochondrial import deficiencies are a feature common to multifaceted pathologies, such as neurodegenerative diseases and cancer. We will also present pharmacological compounds mimicking stress response mechanisms and that could be used as a therapeutic option in the near future to restore efficient mitochondrial protein import rates. Overall, this review highlights the critical role of mitochondrial protein import in cellular and mitochondrial stress response, and in disease pathogenesis. It also emphasizes the potential of mitochondrial protein import as a therapeutic target, despite the surprising absence of direct pharmacological treatments to date.
    Keywords:  DELE1/HRI; ISR; Mitochondrial protein import; Pharmacological modulation; UPRam; UPRmt
    DOI:  https://doi.org/10.1016/j.mito.2025.102107
  2. J Thorac Dis. 2025 Nov 30. 17(11): 9586-9597
       Background: Cigarette smoke extract (CSE) induces reactive oxygen species (ROS) generation in human bronchial epithelial cells, leading to mitochondrial dysfunction and subsequently triggering the mitochondrial unfolded protein response (UPRmt) mediated by the bZIP transcription factor activating transcription factor 4 (ATF-4). This study aimed to investigate whether UPRmt and ATF-4 are involved in the pathway of mitochondrial function impairment in BEAS-2B cells caused by CSE.
    Methods: BEAS-2B bronchial epithelial cells were treated with different concentrations of CSE, and mitochondrial function was detected by JC-1 staining and MitoSoxRed staining. The expression and localisation of translocase of inner mitochondrial membrane 23 (Tim23) and ATF-4 were detected by Western blot method and immunofluorescence staining with laser confocal microscopy.
    Results: The study showed decreased membrane potential and mitochondrial ROS accumulation in BEAS-2B cells after CSE treatment, which indicated that CSE caused mitochondrial dysfunction and oxidative stress. The expression of Tim23 was up-regulated after CSE exposure; this alteration hints at a potential activation of mitochondrial stress pathways. The expression of ATF-4 showed a positive correlation with the time of CSE treatment, and the expression in the nucleus was increased, which indicated that CSE altered the expression and localisation of Tim23 and ATF-4.
    Conclusions: CSE leads to a decrease in mitochondrial membrane potential and an increase in ROS generation, and triggers alterations in the expression of Tim23 and ATF-4. These findings are consistent with the model that CSE may impair mitochondrial homeostasis, potentially through mechanisms involving mitochondrial stress responses (such as UPRmt) and ATF-4 signaling.
    Keywords:  BEAS-2B; Cigarette smoke extract (CSE); activating transcription factor 4 (ATF-4); mitochondrial unfolded protein response (UPRmt)
    DOI:  https://doi.org/10.21037/jtd-2025-460
  3. Mol Neurobiol. 2025 Dec 10. 63(1): 279
      Tauopathies are neurodegenerative diseases characterized by the abnormal accumulation of tau protein in neurons, leading to cognitive impairment. A common feature of these disorders is mitochondrial dysfunction, which leads to bioenergetic deficits and contributes to neuronal cell death. As neurons have high energy demands, impaired mitochondrial function directly affects their viability and function. Thus, mitochondria represent an attractive target for neuroprotective strategies in tauopathies. Mitochondrial transplantation (MT) is an emerging therapeutic approach to restoring cellular bioenergetics. Although MT has shown promise in various models of brain diseases, its efficacy has not been evaluated in the context of tau-induced mitochondrial dysfunction. This study examines the impact of MT on healthy cells and in a cellular model of tauopathy. Mitochondria were freshly isolated from astrocytic cells and transplanted into healthy SH-SY5Y neuroblastoma cells and SH-SY5Y cells overexpressing the P301L tau mutation, for 24 and 48 h. Our results demonstrate that MT enhances cell viability, ATP production, mitochondrial membrane potential, and respiration in both healthy and tau-mutant SH-SY5Y cells. In addition, MT reduced mitochondrial superoxide anion levels and promoted neurite outgrowth in both cell lines. Key bioenergetic outcomes were recapitulated in neurons derived from induced pluripotent stem cells (iPSCs) carrying the P301L tau mutation. These findings suggest that MT might be a promising therapeutic strategy to counteract mitochondrial deficits in tauopathies. Importantly, this approach positions mitochondria not as a target but as the therapeutic agent itself. Further studies are warranted to advance MT toward in vivo applications in tau-related neurodegenerative disorders.
    Keywords:  Bioenergetic; Mitochondria; Neurites; P301Ltau mutation; Tauopathies; Transplantation
    DOI:  https://doi.org/10.1007/s12035-025-05604-y
  4. FEBS J. 2025 Dec 07.
      The biogenesis of mitochondria relies on the import of newly synthesized precursor proteins from the cytosol. Tom70 is a mitochondrial surface receptor which recognizes precursors and serves as an interface between mitochondrial protein import and the cytosolic proteostasis network. Mitochondrial import defects trigger a complex stress response, in which compromised protein synthesis rates are a characteristic element. The molecular interplay that connects mitochondrial (dys)function to cytosolic translation rates in yeast cells is however poorly understood. Here, we show that the deletion of the two Tom70 paralogs of yeast (TOM70 and TOM71) leads to defects in mitochondrial biogenesis and slow cell growth. Surprisingly, upon heat stress, the deletion of ZUO1, a chaperone of the ribosome-associated complex (RAC), largely prevented the slow growth and the reduced translation rates in the tom70Δ/tom71Δ double deletion mutant. In contrast, the mitochondrial defects were not cured but even enhanced by ZUO1 deletion. Our study shows that Zuo1 is a critical component in the signaling pathway that mutes protein synthesis upon mitochondrial dysfunction. We propose a novel paradigm according to which RAC serves as a stress-controlled regulatory element of the cytosolic translation machinery.
    Keywords:  Tom70; mitochondria; protein import; proteostasis; ribosome‐associated complex
    DOI:  https://doi.org/10.1111/febs.70356
  5. Adv Biol (Weinh). 2025 Dec 12. e00472
      HK1 and HK2 are increasingly recognized not only as glycolytic enzymes but also as key modulators of mitochondrial function and cell fate through dynamic interactions with VDAC. This review explores how HK-VDAC complexes support metabolic flexibility, regulate apoptosis, and coordinate glycolytic and mitochondrial activity across diverse physiological and pathological conditions. We incorporate recent reinterpretations of the Warburg effect, emphasizing how spatial and functional reorganization of HK supports proliferative metabolism beyond classical models of mitochondrial dysfunction. Importantly, the HK-VDAC interaction is dynamically regulated by post-translational modifications and signaling pathways that control its stability and mitochondrial anchoring. Disruption of these regulatory mechanisms can impair the balance between glycolytic and mitochondrial metabolism, contributing to disease progression. Emerging evidence links altered HK-VDAC interactions to the metabolic and apoptotic imbalances observed in cancer, neurodegeneration, and aging. By integrating insights from structural biology, bioenergetics, and disease models, we highlight mitochondrial HK anchoring as a central hub for metabolic adaptation and stress response.
    Keywords:  HK‐VDAC; Warburg effect; aging; apoptosis; cancer; metabolism; mitochondria
    DOI:  https://doi.org/10.1002/adbi.202500472
  6. bioRxiv. 2025 Nov 12. pii: 2025.10.31.685870. [Epub ahead of print]
      Mutations in PKD1 and PKD2 cause autosomal-dominant polycystic kidney disease (ADPKD), characterized by fluid-filled cysts, aberrant cell proliferation, and widespread genetic and epigenetic remodeling. While mitochondrial dysfunction and metabolic shifts are central to disease progression, the mechanisms linking PKD mutations to these changes remain unclear. Here, we demonstrate that ER-mitochondria connectivity was disrupted in Pkd1- and Pkd2- deleted mice, preceding cyst formation. This disconnection induces mitochondrial stress, triggering epigenetic remodeling and transcriptional activation of pathways driving proliferation and metabolic reprogramming. Remarkably, restoring PKD function in the ER or pharmacologically enhancing ER-mitochondria connection ameliorates mitochondrial dysfunction, epigenetic shifts, and cystogenesis. These findings reveal a critical role for ER-localized PKD in maintaining mitochondrial integrity and transcriptional homeostasis. Mitochondrial dysfunction resulting from ER-mitochondria uncoupling emerges as a key driver of cystogenesis in ADPKD, and correcting this defect may offer a promising therapeutic strategy.
    Significance: Autosomal dominant polycystic kidney disease (ADPKD) is the most prevalent monogenic cause of kidney failure, marked by fluid-filled cysts, aberrant cell proliferation, metabolic reprogramming, and extensive genetic and epigenetic alterations. The mechanisms by which loss-of-function mutations in PKD1 and PKD2 drive disease progression remain poorly understood. Here, we demonstrate that ER-mitochondria contacts are disrupted in Pkd -mutant mice prior to cyst formation. This disconnection induces mitochondrial dysfunction and epigenetic remodeling, which in turn promote metabolic reprogramming and cystogenesis. Restoration of PKD function in the ER or pharmacological enhancement of ER-mitochondria coupling mitigates these pathological changes. Our findings uncover a critical role for ER-mitochondria crosstalk in suppressing cystogenesis and identify a promising therapeutic target for ADPKD.
    DOI:  https://doi.org/10.1101/2025.10.31.685870
  7. Redox Biol. 2025 Dec 09. pii: S2213-2317(25)00479-3. [Epub ahead of print]89 103966
      Mutations in mitochondrial aminoacyl-tRNA synthetases (mtARSs) causes mitochondrial defects and serious, progressive and usually lethal diseases with exceptional heterogeneous and tissue-specific clinical manifestations. However, the pathogenic mechanisms for specific mtARS related diseases are largely unknown and currently there is no highly effective treatment or cure for these diseases. In the present study, we generate Drosophila models with human mitochondrial prolyl-tRNA synthetase (PARS2) deficiency by knocking out or knocking down dPARS2, the Drosophila ortholog of human PARS2, and further characterize the disease-associated defects and explore the molecular basis of these phenotypes. Inactivation of dPARS2 in Drosophila causes developmental delay and seizure, two main clinical features in human PARS2 deficiency-associated patients. Biochemical analysis demonstrates that loss of dPARS2 activity results in reduced mitochondrial tRNAPro aminoacylation, decreased levels of OXPHOS complex proteins, defective assembly and altered enzyme activities of OXPHOS complexes. Interestingly, we discover that dPARS2 deficiency activates the integrated stress response (ISR), which reduces global protein translation and increases activity of ATF4 in our neuronal dPARS2 knockdown model. Importantly, blockade of ISR activation by genetic suppression of GCN2 kinase prevents developmental delay and seizure phenotypes in dPARS2-deficient flies. Furthermore, the genetic suppression of ATF4, the ISR key effector, also reverses these developmental and behavioral abnormalities associated with dPARS2 deficiency. Furthermore, a disease-associated PARS2 V95I variant causes mitochondrial dysfunction and ISR activation in human cells, verifying the findings in the Drosophila models. Together, these results not only provide evidence for PARS2 deficiency associated mitochondrial dysfunction, but also reveal a novel pathogenic mechanism involved in ISR activation in the PARS2 deficiency related disease, indicating a novel disease treatment approach by targeting ISR.
    Keywords:  Developmental delay; Integrated stress response; Mitochondrial aminoacyl-tRNA synthetases; PARS2; Seizures
    DOI:  https://doi.org/10.1016/j.redox.2025.103966
  8. bioRxiv. 2025 Nov 27. pii: 2025.11.24.689866. [Epub ahead of print]
      Glaucoma is a leading cause of irreversible blindness, characterized by progressive retinal ganglion cell (RGC) loss and optic nerve degeneration. Mitochondrial dysfunction plays a central role in this neurodegeneration, yet effective targeted therapies remain limited. Here, we identify the mitochondrial scaffold A-kinase anchoring protein 1 (AKAP1) as a critical regulator of RGC resilience and axon regeneration. AKAP1 expression is diminished in human glaucomatous retinas and experimental glaucoma models, correlating with elevated intraocular pressure, disrupted mitochondrial dynamics, oxidative stress, and synaptic instability. Restoration of AKAP1 via adeno-associated virus serotype 2-mediated gene therapy preserves RGC survival, promotes mitochondrial fusion and cristae integrity, enhances ATP production, and mitigates oxidative and apoptotic stress in mouse models of glaucoma and optic nerve injury. Transcriptomic profiling of AKAP1 knockout retinas reveals widespread dysregulation of mitochondrial and synaptic gene networks. Mechanistically, AKAP1 stabilizes synapses by promoting mitochondrial biogenesis, modulating calcium/calmodulin-dependent kinase II and synapsin phosphorylation, maintaining synaptophysin expression, and suppressing complement component C1q expression, thereby preventing early synaptic loss in glaucomatous neurodegeneration. Moreover, restoring AKAP1 expression facilitates axonal regeneration, preserves the central visual pathway, and maintains visual function. Collectively, these findings establish AKAP1 as a master regulator of mitochondrial and synaptic homeostasis and axonal regeneration and a promising therapeutic target for vision preservation in glaucomatous neurodegeneration.
    One Sentence Summary: AKAP1 protects retinal ganglion cells and preserves vision by restoring mitochondrial and synaptic health in experimental glaucoma models.
    DOI:  https://doi.org/10.1101/2025.11.24.689866
  9. bioRxiv. 2025 Dec 01. pii: 2025.11.27.690982. [Epub ahead of print]
      Aging is the primary risk factor for clonal hematopoiesis and the development of hematologic malignancies ( 1-5 ), yet the selective pressures that shape stem cell behavior and clonal expansion during aging remain poorly defined. Here, we identify proteostasis stress as a central driver of hematopoietic stem cell (HSC) aging and clonal evolution. We show that Heat shock factor 1 (Hsf1) is activated in aging HSCs to preserve proteostasis and sustain self-renewal. However, this physiological, age-associated adaptive mechanism is co-opted by pre-leukemic Dnmt3a -mutant HSCs to resist proteostasis and inflammatory stress required to fuel clonal expansion during aging. In the context of co-occurring Dnmt3a and Nras mutations, which are frequently observed in human acute myeloid leukemia (AML) ( 6-13 ), mutant HSCs and progenitors exhibit heightened dependence on Hsf1 for expansion, malignant transformation and disease progression. Loss of Hsf1 , or disruption of proteostasis, impairs expansion of mutant progenitors, delays leukemia onset, and prolongs survival. Together, these findings reveal proteostasis as a key constraint in the aging hematopoietic system that imposes a selective bottleneck. Hsf1 activation enables both physiological adaptation in aging stem cells and pathological clonal outgrowth in pre-leukemic and leukemic states, establishing proteostasis control as a pivotal mechanism linking stem cell aging to clonal hematopoiesis and malignancy.
    DOI:  https://doi.org/10.1101/2025.11.27.690982
  10. World J Stem Cells. 2025 Nov 26. 17(11): 113032
      Mesenchymal stem cells (MSCs) are multipotent stromal cells that serve as progenitors for connective tissue and have emerged as a crucial resource in the field of tissue engineering owing to their capacity to differentiate into multiple cell lineages. MSCs-based bone regeneration strategies hold immense therapeutic potential, yet their efficacy is critically limited by inefficient osteogenic differentiation. Mounting evidence positions mitochondria as central regulators of this process, extending beyond their traditional role as cellular powerhouses. Mitochondrial regulation not only influences the induction rate of MSCs differentiation, but also determines the differentiation pathway and the ultimate fate of the resulting cells. To date, research in bone regeneration engineering has predominantly focused on the application of stem cell-based biomaterials, with limited attention given to mitochondrial development. We aim to provide a novel research perspective for targeted mitochondrial interventions in bone regeneration engineering by elucidating the mechanisms through which mitochondria regulate osteogenic differentiation of MSCs.
    Keywords:  Bone regeneration; Mesenchymal stem cells; Mitochondrial; Mitophagy; Oxidative stress
    DOI:  https://doi.org/10.4252/wjsc.v17.i11.113032
  11. Biol Chem. 2025 Dec 10.
      The diverse, and sometimes opposing, roles of mitochondria require sophisticated organizational and regulatory strategies. This review examines emerging evidence that mitochondria can solve this challenge through functional specialization - adopting distinct bioenergetic and metabolic programs based on location, contacts, and cellular conditions. We discuss both established principles and recent technological breakthroughs that reveal this hidden complexity. Ongoing advances promise to move the field from describing mitochondrial diversity to uncovering its regulatory mechanisms and therapeutic potential.
    Keywords:  heterogeneity; metabolic specialization; mitochondria
    DOI:  https://doi.org/10.1515/hsz-2025-0210
  12. World J Stem Cells. 2025 Nov 26. 17(11): 114306
      GrpE-like 1 (GRPEL1)-carrying exosomes derived from synovial mesenchymal stem cells (SMSC) prevent mitochondrial dysfunction associated with osteoarthritis (OA) by activating PINK1-mediated mitophagy, restoring chondrocyte function, and preserving the extracellular matrix both in vitro and in vivo. Bioinformatics analysis of human OA datasets identified GRPEL1 as a mitophagy-related gene that is downregulated in OA. Exosomes enriched with GRPEL1 derived from SMSCs enhanced mitochondrial membrane potential and ATP production, reduced lipid peroxidation and reactive oxygen species, increased mitophagy markers (PINK1, Parkin, LC3-II/I), decreased p62 levels, and alleviated cartilage degeneration in a rat destabilization model. A causal role for mitophagy is supported by co-immunoprecipitation experiments confirming a GRPEL1-PINK1 interaction, and by PINK1 knockdown, which diminishes the protective effects of GRPEL1. These findings suggest that exosomes enriched with GRPEL1 derived from SMSCs represents a promising disease-modifying approach for OA by targeting mitochondrial quality control.
    Keywords:  Cartilage repair; Exosomes; GrpE-like 1; Mitochondrial quality control; Mitophagy; Osteoarthritis; PINK1; Synovial mesenchymal stem cell
    DOI:  https://doi.org/10.4252/wjsc.v17.i11.114306
  13. Cell Biosci. 2025 Dec 11.
      Mesenchymal stem cells (MSCs) have many uses in tissue engineering and clinical applications. However, maintaining their stemness during in vitro expansion is challenging. We previously found that Krüppel-like factor 2 (KLF2) plays a crucial role in maintaining the stemness of MSCs. In this study, KLF2 was revealed to be closely linked to mitochondrial oxidative phosphorylation (OXPHOS), and impaired KLF2 expression in MSCs led to mitochondrial dysfunction that ultimately resulted in the loss of stemness. Moreover, decreased KLF2 expression was associated with reduced expression of the mitochondrial electron transport chain components, particularly the accessory subunit of complex I, NDUFC1. Further study demonstrated that KLF2 transcriptionally regulates NDUFC1 expression by binding to its promoter region. In addition, NDUFC1 knockdown largely phenocopied KLF2 knockdown in mitochondrial dysfunction and loss of stemness, and these phenotypes were partially rescued by NDUFC1 overexpression. Taken together, we reveal that KLF2 critically maintains MSCs stemness by transcriptionally promoting the expression of mitochondrial electron transport chain components such as NDUFC1, and KLF2/NDUFC1 axis-regulated mitochondrial oxidative phosphorylation may serve as a novel therapeutic target for improving MSCs stemness.
    Keywords:  KLF2; MSCs; NDUFC1; OXPHOS; Stemness
    DOI:  https://doi.org/10.1186/s13578-025-01501-y
  14. Cells. 2025 Nov 26. pii: 1861. [Epub ahead of print]14(23):
      Mitochondria are essential for β-cell function, coupling glucose metabolism to ATP production and insulin secretion. In diabetes, β-cell mitochondrial dysfunction arises from oxidative stress, impaired quality control and disrupted dynamics, leading to reduced oxidative phosphorylation, defective insulin release and progressive cell loss. Key transcriptional regulators link genetic susceptibility to mitochondrial dysfunction in both type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). These disruptions impair mitophagy, mitochondrial translation and redox homeostasis. Therapeutic strategies that restore mitochondrial function, including mitophagy enhancers, mitochondrial antioxidants, and transcriptional regulators, have shown potential in preserving β-cell integrity. As mitochondrial failure precedes β-cell loss, targeting mitochondrial pathways may represent a critical approach to modifying diabetes progression.
    Keywords:  diabetes; mitochondria; mitochondrial dysfunction; mitophagy; β-cell
    DOI:  https://doi.org/10.3390/cells14231861
  15. Adv Sci (Weinh). 2025 Dec 07. e17586
      Mitochondria-driven macrophage dysregulation contributes significantly to inflammatory disease progression; however, the mechanism underlying bisphosphonate-related osteonecrosis of the jaw (BRONJ) remains unclear. This study demonstrates that zoledronic acid (ZA) disrupts mitochondrial bioenergetic function in macrophages, leading to elevated mitochondrial membrane potential, excessive mitochondrial reactive oxygen species (mtROS), and increased HIF-1α expression, which together promote a pro-inflammatory transition in macrophages. ZA further inhibits autophagy by activating the TLR4-MyD88/PI3K-AKT-mTOR pathway, preventing the clearance of dysfunctional mitochondria and sustaining superoxide production. Genetic loss of Atg5 in innate immune cells disrupts autophagosome maturation and markedly worsens ZA-induced BRONJ development. To restore mitochondrial degradation and biofunction, ZA-loaded nanoparticles incorporating the mTOR inhibitor rapamycin (ZDPR) are developed. ZDPR effectively prevents BRONJ and exerts therapeutic benefits in osteoporosis and osteolysis. These findings highlight bone-targeted mitochondria rescue as a promising strategy to enhance antiresorptive therapy.
    Keywords:  PI3K‐AKT‐mTOR pathway; anti‐resorptive therapy; autophagy; bisphosphonate‐related osteonecrosis of the jaw; macrophage polarization; mitochondrial dysfunction; zoledronic acid
    DOI:  https://doi.org/10.1002/advs.202517586
  16. Cell Signal. 2025 Dec 06. pii: S0898-6568(25)00709-0. [Epub ahead of print]139 112294
      Acute kidney injury (AKI) caused by renal ischemia-reperfusion injury (RIRI) is primarily a mitochondrial disorder characterized by disrupted dynamics, impaired biogenesis, and defective quality control. Excessive DRP1-mediated fission, suppression of the AMPK-SIRT-PGC-1α axis, and failure of the PINK1-Parkin mitophagy system converge to drive tubular dysfunction and ferroptosis. Here, we integrate recent insights into a "mitochondrial reprogramming" framework encompassing three axes-dynamic remodeling, metabolic renewal, and proteostatic reinforcement. Therapeutic strategies targeting these axes, such as DRP1 inhibition, AMPK-SIRT-PGC-1α activation, and reinforcement of mitophagy and MAM integrity by agents like melatonin, puerarin, or Schisandrin B, have shown promise in restoring mitochondrial resilience. Furthermore, mitochondrial biomarkers and imaging tools (mtDNA, mitochondrial peptides, [18F]BCPP-EF PET) may enable phenotype-guided interventions. This review outlines the "RIRI-Mitochondria-AKI-CKD continuum," emphasizing that mitochondrial maladaptation bridges acute injury and chronic fibrosis, highlighting mitochondria as precision therapeutic targets in AKI.
    Keywords:  Biomarkers; Ferroptosis; Mitochondria; Mitophagy; Renal ischemia–reperfusion
    DOI:  https://doi.org/10.1016/j.cellsig.2025.112294
  17. Nat Commun. 2025 Dec 12. 16(1): 11103
      Microglial capacity to adapt to tissue needs is a hallmark feature of these cells. New studies show that mitochondria critically regulate the phenotypic adaptability of macrophages. To determine whether these organelles play similar roles in shaping microglial phenotypes, we generated transgenic mouse crosses to accurately visualize and manipulate microglial mitochondria. We find that brain-region differences in microglial attributes and responses to aging are accompanied by regional differences in mitochondrial mass and aging-associated mitochondrial remodeling. Microglial mitochondria are also altered within hours of LPS injections and microglial expression of inflammation-, trophic-, and phagocytosis-relevant genes is strongly correlated with expression of mitochondria-relevant genes. Finally, direct genetic manipulation of microglial mitochondria alters microglial morphology and leads to brain-region specific effects on microglial gene expression. Overall, this study advances our understanding of microglial mitochondria and supports the idea that mitochondria influence basal microglial phenotypes and phenotypic remodeling that takes place over hours to months.
    DOI:  https://doi.org/10.1038/s41467-025-66709-5
  18. Front Immunol. 2025 ;16 1669054
      Innate immune memory, traditionally underappreciated in contrast to adaptive immunity, is now recognized as a critical component of host defense, particularly in the context of sepsis and sterile inflammatory injury. Recent advances have identified a central role for metabolic and epigenetic reprogramming in driving trained immunity (TRIM), where monocytes, macrophages, and other innate cells develop enhanced or tolerized responses to secondary stimuli. This review synthesizes current knowledge of how damage-associated molecular patterns (DAMPs), including oxidized LDL, HMGB1, heme, urate crystals, and mitochondrial DNA, serve as potent inducers of immunometabolic rewiring, often through the mTOR/HIF-1α axis or alternative pathways such as SYK signaling. We highlight distinct epigenetic mechanisms, such as enhancer priming via H3K4me1/H3K27ac, and metabolic shifts like the Warburg effect, succinate accumulation, and fatty acid synthesis, that define the trained or tolerized states. Particular attention is given to the relevance of these mechanisms in the pathophysiology of sepsis, burns, trauma, and other critical illnesses where persistent DAMP exposure may sustain maladaptive inflammation or immunosuppression. We review data linking central (stem cell-level) and peripheral reprogramming to long-term immune dysfunction in various inflammatory disease models, and explore how DAMPs intersect with PAMPs to shape the immune trajectory. Finally, we identify pressing gaps in the field, including the need for standardized TRIM models, validated biomarkers of innate memory, and mechanistic clarity on mitochondrial DAMPs in immune tolerance. These insights provide a foundation for future therapeutic strategies aimed at modulating trained immunity to improve outcomes in critically ill patients.
    Keywords:  DAMPs; epigenetics; innate immunity; innate training; trauma
    DOI:  https://doi.org/10.3389/fimmu.2025.1669054
  19. Commun Biol. 2025 Dec 11. 8(1): 1759
      Hypermetabolism, a futile cycle of energy production and consumption, has been proposed as an adaptative response to deficiencies in mitochondrial oxidative phosphorylation. However, the cellular costs of hypermetabolism remain largely unknown. Here we studied the consequences of hypermetabolism in human motor neurons harboring a heteroplasmic mutation in MT-ATP6, which impairs ATP synthase assembly. Respirometry, metabolomics, and proteomics analyses of the motor neurons showed that elevated ATP production rates were accompanied with increased demand for acetyl-Coenzyme A (acetyl-CoA) and depleted pantothenate (vitamin B5), and the proteome was remodeled to support the metabolic adaptation. Mitochondrial membrane potential and coupling efficiency remained stable, and the therapeutic agent avanafil did not affect metabolite levels. However, a redistribution of acetyl-CoA usage resulted in metabolic trade-offs, including reduced histone acetylation and altered maintenance of the neurotransmitter acetylcholine, revealing potential vulnerabilities in motor neurons. These findings advance the understanding of cellular metabolic consequences imposed by hypermetabolic conditions.
    DOI:  https://doi.org/10.1038/s42003-025-09149-7
  20. Nat Commun. 2025 Dec 12. 16(1): 11104
      Microglia continually surveil the brain allowing for rapid detection of tissue damage or infection. Microglial metabolism is linked to tissue homeostasis, yet how mitochondria are subcellularly partitioned in microglia and dynamically reorganize during surveillance, injury responses, and phagocytic engulfment in the intact brain are not known. Here, we performed intravital imaging and ultrastructural analyses of microglia mitochondria in mice and human tissue, revealing that microglial processes diverge in their mitochondrial content, with some containing multiple mitochondria while others are completely void. Microtubules and hexokinase 2 mirror this uneven mitochondrial distribution indicating that these cytoskeletal and metabolic components are linked to mitochondrial organization in microglia. Microglial processes that engage in minute-to-minute surveillance typically do not have mitochondria. Moreover, unlike process surveillance, mitochondrial motility does not change with animal anesthesia. Likewise, the processes that acutely chemoattract to a lesion site or initially engage with a neuron undergoing programmed cell death do not contain mitochondria. Rather, microglia mitochondria have a delayed arrival into the responding cell processes. Thus, there is subcellular heterogeneity of mitochondrial partitioning. Moreover, microglial processes that surveil and acutely respond to damage do not contain mitochondria.
    DOI:  https://doi.org/10.1038/s41467-025-66708-6