bims-mikwok Biomed News
on Mitochondrial quality control
Issue of 2025–11–09
48 papers selected by
Gavin McStay, Liverpool John Moores University



  1. Syst Biol Reprod Med. 2025 Dec;71(1): 549-573
      In recent years, the incidence of male infertility has increased to approximately 10%, with a continued upward trend. Therefore, understanding the mechanisms underlying male infertility and developing effective treatment strategies have become essential areas of focus. Mitochondria are regulated by a complex quality control system including mitochondrial dynamics, mitophagy and biogenesis, which not only maintains mitochondrial structural and functional integrity, but also supports the stability of testicular tissue and the intracellular environment necessary for male fertility. Several studies have demonstrated that dysfunction in mitochondrial dynamics and mitophagy is closely associated with a decline in male fertility. Disruptions caused by excessive external stimuli or gene mutations can impair these processes, resulting in oxidative damage, apoptosis, inflammation, and ferroptosis. These pathological changes ultimately damage testicular cells and tissues. Consequently, this review will focus on the two key mechanisms: mitochondrial dynamics and mitophagy. Furthermore, mitochondrial biogenesis-responsible for producing new mitochondria and regulating the number of mitochondria-also plays an important role in maintaining male fertility. Related studies have shown that mitochondrial biogenesis dysfunction can trigger a cascade of pathological events that lead to testicular tissue damage. In summary, this review systematically examines the roles of mitochondrial dynamics and mitophagy in regulating male fertility. It provides an in-depth analysis of the pathological mechanisms by which dysfunction in these processes leads to male infertility. Additionally, this review summarizes current therapeutic agents targeting mitochondrial dynamics and mitophagy, aiming to identify potential strategies for the clinical treatment of male infertility.
    Keywords:  infertility treatments; male fertility; mitochondrial dynamics; mitophagy
    DOI:  https://doi.org/10.1080/19396368.2025.2574003
  2. J Physiol Biochem. 2025 Nov 03.
      Myocardial infarction (MI) is characterized by sudden interruption of coronary blood flow, leading to ischemic damage and cardiomyocyte death. Evidence for new molecular targets remains limited. Here, we investigated the role of Schlafen 4 (Slfn4), identified via bioinformatic screening, in MI pathogenesis. We analyzed GSE46395 microarray data and observed elevated Slfn4 expression in ischemic cardiac tissue. An MI mouse model further confirmed Slfn4 upregulation, which was abrogated by AAV9-mediated shRNA knockdown. Silencing Slfn4 reduced inflammatory cell infiltration and cardiomyocyte apoptosis, leading to lower serum levels of ANP, BNP, cTnT, cTnI, IL-1β, and TNF-α. Notably, Slfn4 knockdown augmented BNIP3-dependent mitophagy, evidenced by upregulated LC3 I/II, decreased P62, and reduced mitochondrial proteins (COX IV, TOMM20), while also suppressing DRP1-mediated mitochondrial fission. In cultured H9C2 cells subjected to hypoxia, Slfn4 knockdown likewise diminished apoptosis and enhanced BNIP3-associated mitophagy, whereas BNIP3 silencing reversed these protective effects, underscoring the importance of BNIP3-mediated mitophagy in Slfn4-driven cardioprotection. These findings indicate that Slfn4 promotes MI-induced damage by inhibiting BNIP3-mediated mitophagy and exacerbating mitochondrial fission. By contrast, Slfn4 knockdown fosters cardiomyocyte survival, highlighting its therapeutic potential for MI. Overall, our data suggest that modulating Slfn4 expression may preserve mitochondrial quality control, attenuate inflammation and apoptosis, and improve cardiac function following ischemic injury. .
    Keywords:  BNIP3; Hypoxia; Mitophagy; Myocardial infarction; Slfn4
    DOI:  https://doi.org/10.1007/s13105-025-01131-5
  3. Kaohsiung J Med Sci. 2025 Nov 05. e70131
      Osteoarthritis (OA) is a prevalent degenerative joint disease. This study combines bioinformatics analysis with in vivo and in vitro experiments to elucidate the molecular mechanisms through which melatonin (MT) regulates mitophagy to alleviate OA. Rat and chondrocyte OA models were established via anterior cruciate ligament transection or interleukin (IL)-1β induction, followed by treatment with MT, Cyclosporine A (a mitophagy inhibitor), and 740Y-P (a phosphatidylinositol-3 kinase [PI3K] activator). Pathological changes in cartilage, histological scores, and cell apoptosis were evaluated alongside chondrocyte viability, apoptosis, mitochondrial morphology, mitochondrial membrane potential, and mitophagy using H&E and Safranin O-fast green staining, Osteoarthritis Research Society International scoring (OARSI), TUNEL staining, CCK-8, flow cytometry, transmission electron microscopy, JC-1 staining, and immunofluorescence. Levels of inflammatory factors and mitophagy-related protein levels were determined by ELISA and western blot. Bioinformatics analysis was applied to investigate the regulatory mechanisms of MT on mitophagy in OA. In vivo, MT mitigated OA by enhancing mitophagy and reducing apoptosis of cartilage cells. In vitro, MT attenuated IL-1β-induced chondrocyte apoptosis through mitophagy activation, and this effect was partially reversed by mitophagy inhibition. Mechanistically, the PI3K/protein kinase B (AKT)/forkhead box O3 (FoxO3) axis appeared to play a central role. MT suppressed PI3K/AKT signaling, thereby upregulating FoxO3 expression and promoting mitophagy, ultimately reducing chondrocyte apoptosis. Collectively, these findings suggest that MT enhances mitophagy via inhibition of the PI3K/AKT pathway, and subsequent upregulation of FoxO3, leading to reduced apoptosis of cartilage cells and attenuation of OA progression in rats.
    Keywords:  chondrocytes; melatonin; mitophagy; osteoarthritis; phosphatidylinositol‐3 kinase/protein kinase B/forkhead box O3
    DOI:  https://doi.org/10.1002/kjm2.70131
  4. Int Endod J. 2025 Nov 07.
       AIMS: To investigate the effects of Porphyromonas endodontalis on the viability of stem cells from the apical papilla (SCAP) and to explore the role of mitochondrial function in supporting SCAP survival.
    METHODOLOGY: SCAP were isolated from immature third molars, and P. endodontalis was added to the cell culture medium. Cell proliferation and apoptosis were assessed using the CCK8 assay and flow cytometry, with vascular endothelial cells (VECs) serving as a comparative group. Transmission electron microscopy was utilised to observe the mitochondrial morphology and bacterial ultrastructure. Changes in mitochondrial membrane potential (MMP) were assessed using laser scanning confocal microscopy. Additionally, the effects of mitophagy or mitochondrial fission inhibitors on SCAP cell viability and MMP were evaluated. Transcriptomic high-throughput sequencing was conducted to analyse differentially expressed mRNAs in SCAP and VECs after P. endodontalis infection using gene set enrichment analysis. Western blotting was performed to detect the expression of proteins associated with mitophagy and mitochondrial fission. The co-localisation of microtubule-associated protein light chain 3 with translocase of the outer mitochondrial membrane 20 (TOM20), as well as dynamin-related protein 1 with TOM20, was examined using laser scanning confocal microscopy. Data were analysed using SPSS 22.0, and a p-value < 0.05 was considered statistically significant.
    RESULTS: Following 6 h of P. endodontalis infection, SCAP exhibited higher cell viability and lower cell apoptosis rates than VECs. In addition, SCAP maintained more stable mitochondrial morphology and higher MMP compared to VECs. Transcriptomic analysis revealed that differentially expressed mRNAs between infected SCAP and VECs were enriched in terms related to mitochondrial membrane, depolarisation and mitochondrial fission. P. endodontalis infection also upregulated the expression of proteins associated with mitophagy and mitochondrial fission in SCAP. Inhibition of either mitophagy or mitochondrial fission in SCAP decreased the viability of SCAP.
    CONCLUSIONS: SCAP could maintain cell viability after short-term P. endodontalis infection. The underlying mechanism may involve mitochondrial fission and mitophagy, which appear to function as key protective processes enabling SCAP to counteract infection-induced cellular damage. This study helps elucidate the molecular basis of SCAP survival under bacterial infection.
    Keywords:   Porphyromonas endodontalis ; cell viability; mitochondrial fission; mitophagy; stem cells from the apical papilla
    DOI:  https://doi.org/10.1111/iej.70058
  5. J Hypertens. 2025 Dec 01. 43(12): 1972-1983
       BACKGROUND AND PURPOSE: Myocardial fibrosis is a key pathological feature of hypertension, closely associated with mitochondrial dysfunction and calcium overload. The calcium-sensing receptor (CaSR) has emerged as a potential mediator in this process, but its mechanistic role remains unclear. This study aimed to investigate whether Calhex231, a selective CaSR antagonist, could attenuate myocardial fibrosis in Dahl salt-sensitive (SS) rats by restoring mitochondrial dynamics and intracellular calcium homeostasis.
    METHODS: Hypertension was induced in Dahl SS rats using an 8% NaCl diet. From week 5, rats were treated with Calhex231 (10 μmol/kg/day) for 6 weeks. In vitro, cardiac fibroblasts (CFs) were stimulated with TGF-β1 (10 ng/ml) and treated with either Calhex231 or Mdivi-1 (a Drp1 inhibitor). Assessments included echocardiography, histological staining (Masson, HE), immunohistochemistry, Western blotting, and fluorescence-based analyses of mitochondrial membrane potential (JC-1), oxidative stress (Dihydroethidium, SOD1/2), intracellular Ca2+ (Fluo-4 AM), and fibrosis markers (α-SMA, Collagen I/III, MMP-2/9).
    RESULTS: Calhex231 significantly reduced blood pressure and myocardial fibrosis in hypertensive rats, accompanied by improved cardiac structure and diastolic function. Mechanistically, Calhex231 suppressed mitochondrial fission proteins (Drp1, Fis1) and upregulated fusion proteins (MFN2, OPA1), restoring mitochondrial homeostasis. In TGF-β1-stimulated CFs, Calhex231 alleviated calcium overload, preserved mitochondrial membrane potential, reduced ROS production, and downregulated fibrotic markers. Similar protective effects were observed with Mdivi-1, highlighting the involvement of Drp1-mediated fission in CaSR-induced fibrosis.
    CONCLUSION: Inhibition of CaSR with Calhex231 exerts cardioprotective effects by suppressing Drp1-dependent mitochondrial fission, thereby mitigating oxidative stress and calcium overload. These findings support CaSR as a promising therapeutic target for myocardial fibrosis in salt-sensitive hypertension.
    Keywords:  calcium-sensing receptor; mitochondrial dynamics; myocardial fibrosis; oxidative stress; salt-sensitive hypertension
    DOI:  https://doi.org/10.1097/HJH.0000000000004137
  6. Front Physiol. 2025 ;16 1695681
       Background: Neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, etc., are a group of complex and heterogeneous disorders characterized by progressive synaptic loss and pathological protein alterations. Mitochondria are the main source of energy produced by neurons and support the high energy consumption of the nervous system. Mitochondrial quality control, involving processes like mitophagy and mitochondrial biogenesis, is crucial for mitochondrial homeostasis, and mitochondrial dysfunction is closely related to neurodegenerative diseases pathogenesis, making targeting mitochondrial quality control a potential therapeutic strategy. Natural products offer benefits such as cost-effectiveness, fewer side effects, and other positive qualities, making them suitable choices as supplements or alternatives to traditional drugs for treating neurodegenerative diseases.
    Methods: A thorough search was conducted on many databases including Web of Science, PubMed, EMBASE, and MEDLINE to investigate the role of mitochondria in neurodegenerative diseases and the therapeutic effects of natural products.
    Results: By searching the relevant studies on neurodegenerative diseases and mitochondria in recent years, we observed a rise in the number of studies examining the functional characteristics and biological events of mitochondrial quality control systems in neurodegenerative diseases pathogenesis and the potential for natural products regulating mitochondrial quality control to improve neurodegenerative diseases.
    Conclusion: This review summarizes the functional characteristics and biological events of mitochondrial quality control systems in neurodegenerative diseases pathogenesis, and comprehensively analyzes the pharmacological mechanisms by which natural products regulate mitochondrial quality control to improve neurodegenerative diseases, aiming to provide a scientific basis for further research and new clinical drug development.
    Keywords:  mitochondrial quality control; molecular mechanisms; natural products; neurodegenerative diseases; toxicology and adverse effects
    DOI:  https://doi.org/10.3389/fphys.2025.1695681
  7. Biochim Biophys Acta Mol Basis Dis. 2025 Nov 03. pii: S0925-4439(25)00444-2. [Epub ahead of print] 168094
      Pulmonary hypertension (PH) is a lethal vascular disorder characterized by hypoxia-driven vascular remodeling, yet the molecular mechanisms underlying endothelial dysfunction remain poorly understood. Here, we identify peroxiredoxin-1 (PRDX1) as a critical mediator of PH pathogenesis through redox-independent regulation of mitochondrial dynamics. Hypoxia upregulated PRDX1 expression in human pulmonary artery endothelial cells (HPAECs) via HIF1α-dependent transcriptional activation, as demonstrated by chromatin immunoprecipitation and promoter-luciferase assays. PRDX1 silencing attenuated hypoxia-induced HPAEC proliferation and apoptosis resistance, while its overexpression mimicked hypoxic effects independently of its antioxidant activity. Mechanistically, PRDX1 directly interacted with Dynamin-related protein 1 (DRP1) and facilitated sentrin-specific protease 3 (SENP3) binding, suppressing DRP1 SUMO2/3 conjugation to promote mitochondrial fission. In vivo, endothelial-specific PRDX1 knockdown in a hypoxia/SU5416(SuHx)-induced PH rat model significantly reduced right ventricular systolic pressure, vascular wall thickening, and endothelial hyperproliferation, while improving exercise tolerance. These findings reveal a novel HIF1α-PRDX1-DRP1 axis driving mitochondrial fragmentation and vascular remodeling in PH, positioning PRDX1 as a promising therapeutic target for halting disease progression.
    Keywords:  DRP1; HIF1α; PRDX1; Pulmonary hypertension; SUMOylation
    DOI:  https://doi.org/10.1016/j.bbadis.2025.168094
  8. Mol Carcinog. 2025 Nov 02.
      This study explored the effect of ubiquitin-specific protease 4 (USP4) on mitochondrial dynamics in esophageal squamous cell carcinoma (ESCC). USP4 and phosphoglycerate mutase 5 (PGAM5) expression in ESCC tissues was measured. ESCC cells were subjected to gain- and loss-of-function experiments, followed by examinations of proliferation, invasion, migration, apoptosis, light chain 3 (LC3), P62, Bcl2-associated X (Bax), B-cell lymphoma 2 (Bcl2), Cytochrome c (CytC), caspase3, mitochondrial membrane potential (MMP), mitochondrial reactive oxygen species (mtROS), and ROS. An in vivo tumor formation model of ESCC was established for in vivo verification. The relationship between USP4 and PGAM5 was analyzed. USP4 and PGAM5 expression was high in ESCC tissues. Mechanistically, USP4 eliminated K48-linked ubiquitin chains to maintain PGAM5 stability. PGAM5 knockdown impaired malignant behaviors of ESCC cells, reduced LC-3I-to-LC-3II conversion, increased mtROS, ROS, and P62 levels, activated the caspase-dependent mitochondrial pathway, and decreased MMP. Overexpressing USP4 increased MMP and Bcl2 expression while lowering Bax, CytoC, cleaved caspase3, and mtROS levels in ESCC cells, which was nullified by PGAM5 knockdown. Ectopic USP4 accelerated tumor growth in mice, which was reversed by PGAM5 knockdown. Collectively, USP4 promotes cell growth and excessive mitophagy in ESCC by stabilizing PGAM5.
    Keywords:  PGAM5; USP4; apoptosis; esophageal squamous cell carcinoma; mitochondria; mitophagy
    DOI:  https://doi.org/10.1002/mc.70055
  9. Acta Biochim Biophys Sin (Shanghai). 2025 Nov 03.
      Mitochondrial dysfunction is closely related to tumor development. Adenine nucleotide translocator 1 (ANT1), which promotes ADP/ATP translocation across the inner mitochondrial membrane, is an important protein involved in mitochondrial function and plays a role in a variety of diseases, including cancers. However, its role in colorectal cancer (CRC) progression remains poorly understood. This study aims to explore the potential role of ANT1 in CRC and its relationship with mitophagy. Through immunohistochemical analysis, we find that ANT1 expression is significantly higher in the tumor tissues of CRC patients than in adjacent normal tissues and that its overexpression is associated with poor prognosis. Further experiments demonstrate that ANT1 knockdown significantly inhibits CRC cell proliferation, migration, and invasion and leads to mitochondrial dysfunction, increased ROS production, and apoptosis by suppressing mitophagy. Mechanistically, ANT1 knockdown downregulates the PINK1/Parkin pathway, thereby inhibiting mitophagy activity. Notably, PINK1 overexpression partially rescues the cellular dysfunction induced by ANT1 knockdown, suggesting a potential role for PINK1 in reversing the suppression of mitophagy. In vivo xenograft models also show that ANT1 knockdown markedly inhibits tumor growth. In conclusion, ANT1 may play a critical role in CRC progression by regulating mitophagy, providing a basis for its potential as a therapeutic target.
    Keywords:  PINK1; Parkin; adenine nucleotide translocator 1; colorectal cancer; mitophagy
    DOI:  https://doi.org/10.3724/abbs.2025154
  10. Mol Biol Rep. 2025 Nov 06. 53(1): 56
       BACKGROUND: Overexposure to hepatotoxins is a frequent cause of acute liver injury (ALI). This study aimed to investigate the protective effect of resveratrol (RSV) against carbon tetrachloride (CCl₄)-induced ALI and its underlying mechanisms.
    METHODS: In vitro, four hepatic cell lines (HepG2, Huh7, Hepa1-6, and AML12) were pretreated with RSV (10, 20, or 40 µg/mL) before CCl₄ exposure. Cell viability, reactive oxygen species (ROS) levels, and mitophagy-related protein expression were assessed. In vivo, mice were orally administered RSV (10-40 mg/kg) for 7 days prior to CCl₄-induced ALI. Liver histopathological, liver function, oxidative stress markers, and apoptosis were measured.
    RESULTS: RSV significantly attenuated CCl₄-induced cytotoxicity and ROS overproduction in vitro. It activated the expression of PTEN-induced putative kinase 1 (PINK1)/parkin RBR E3 ubiquitin protein ligase (Parkin)-dependent mitophagy, as indicated by upregulated Parkin, PINK1 and LC3-II (microtubule associated protein 1 light chain 3 beta), and reduced p62 expression. In vivo, RSV ameliorated CCl₄-induced ALI, reducing histopathological damage and serum alanine aminotransferase (ALT) / aspartate aminotransferase (AST) levels. RSV enhanced hepatic antioxidant capacity and decreased lipid peroxidation. Furthermore, RSV reduced hepatocyte apoptosis and consistently activated the PINK1/Parkin mitophagy pathway in liver tissues. Both in vitro and in vivo, RSV with the high dose exhibits the most potent effects.
    CONCLUSIONS: These findings demonstrate that RSV, particularly at the high dose exerts hepatoprotective effects against CCl₄-induced ALI, partly by activating PINK1/Parkin-dependent mitophagy and alleviating oxidative stress. This highlights mitophagy's critical role in RSV-mediated protection and offers novel insights into its therapeutic mechanisms.
    Keywords:  Acute liver injury; Mitophagy; Oxidative stress; ROS; Resveratrol
    DOI:  https://doi.org/10.1007/s11033-025-11223-6
  11. Geroscience. 2025 Nov 04.
      Non-lethal exposure to mitochondrial stress has been shown to have beneficial effects due to activation of signaling pathways, including the mitochondrial unfolded protein response (UPRmt). Activation of UPRmt restores the function of the mitochondria and improves general health and longevity in multiple model systems, termed mitohormesis. In C. elegans, mitohormesis can be accomplished by electron transport chain inhibition, a decline in mitochondrial translation, decreased mitochondrial import, and numerous other methods that activate UPRmt. However, not all methods that activate UPRmt promote longevity. These and other studies have started to question whether UPRmt is directly correlated with longevity. Here, we attempt to address this controversy by unraveling the complex molecular regulation of longevity of the nematode under different mitochondrial stressors that induce mitochondrial stress by performing RNA sequencing to profile transcriptome changes. Using this comprehensive and unbiased approach, we aim to determine whether specific transcriptomic changes can reveal a correlation between UPRmt and longevity. Altogether, this study will provide mechanistic insights on mitohormesis and how it correlates with the lifespan of C. elegans.
    Keywords:  Aging; Caenorhabditis elegans; Mitohormesis; UPRmt
    DOI:  https://doi.org/10.1007/s11357-025-01912-2
  12. Biomed J. 2025 Oct 31. pii: S2319-4170(25)00101-5. [Epub ahead of print] 100927
       BACKGROUND: Mitochondrial autophagy is linked to neuropathic pain. This study explores how Fu's subcutaneous needling (FSN) affects sciatica via mitochondrial autophagy modulation.
    METHODS: 40 male SD rats were divided into four groups. Except for the control group (COT), the other three groups were utilized to establish a chronic sciatic nerve injury model. Mechanical pain thresholds were measured. FSN and acupuncture (ACP) groups received treatments every other day for four sessions. Mitochondrial quantity and morphology were examined under a transmission electron microscope, and the expression levels of PINK1, Parkin, and P62 proteins in the rat pyriform were analyzed through immunohistochemistry and immunofluorescence. Furthermore, the levels of TNF-α and IL-6 inflammatory factors in the serum of all groups were measured using ELISA, and the expression levels of midbrain opioid receptors κ and μ mRNA in each group of rats were determined via qPCR.
    RESULTS: FSN and ACP groups showed higher pain thresholds than the model group from the second intervention. FSN outperformed ACP after the fourth intervention. Transmission electron microscope showed improved mitochondrial morphology in FSN and ACP groups, with FSN showing better morphology. FSN upregulated PINK1/Parkin and downregulated P62. FSN also reduced TNF-α and IL-6 levels.
    CONCLUSIONS: FSN alleviates neuropathic pain by enhancing mitochondrial autophagy, restoring mitochondrial dynamics, and reducing inflammation. It shows promise as a therapeutic strategy for neuropathic pain.
    Keywords:  Fu's subcutaneous needling; acupuncture; chronic constriction injury; mitochondrial autophagy; neuropathic pain; sciatica
    DOI:  https://doi.org/10.1016/j.bj.2025.100927
  13. World J Stem Cells. 2025 Oct 26. 17(10): 109369
       BACKGROUND: Osteoarthritis (OA) remains a challenging degenerative joint disease with limited therapeutic interventions.
    AIM: To investigate the potential of synovial mesenchymal stem cell (SMSC)-derived exosomes (SMSCs-Exos) delivering GrpE-like 1 (GRPEL1) in promoting cartilage repair through phosphatase and tensin homolog-induced putative kinase 1 (PINK1)-mediated mitophagy activation.
    METHODS: A comprehensive research approach was employed, including bioinformatics analysis of gene expression datasets (GSE169077 and GSE114007), in vitro experiments with CHON-001 chondrocytes, and in vivo rat knee OA models. Experimental techniques encompassed gene expression profiling, immunofluorescence staining, western blot analysis, co-immunoprecipitation, cell proliferation and migration assays, and histological examinations. Exosomes were genetically modified to overexpress or knockdown GRPEL1, and their effects on cellular function and mitochondrial dynamics were systematically evaluated.
    RESULTS: Bioinformatics analysis revealed GRPEL1 as a critical mitophagy-related gene with significantly altered expression in OA. In vitro studies demonstrated that GRPEL1-loaded SMSCs-Exos effectively counteracted interleukin-1 beta-induced cellular damage by enhancing chondrocyte proliferation and migration, preserving extracellular matrix integrity. Mechanistic investigations confirmed direct interaction between GRPEL1 and PINK1, leading to enhanced mitophagy activation. In vivo rat models substantiated these findings, showing significantly reduced cartilage damage, restored proteoglycan content, and improved joint structure in groups receiving GRPEL1-overexpressing exosomes. Key molecular changes included decreased reactive oxygen species, improved mitochondrial membrane potential, and increased mitophagy markers.
    CONCLUSION: This study provides compelling evidence that SMSCs-Exos delivering GRPEL1 can effectively activate PINK1-mediated mitophagy, offering a promising therapeutic strategy for cartilage repair in OA. The research unveils a novel molecular mechanism for targeting mitochondrial dysfunction and presents a potential disease-modifying approach beyond current symptomatic treatments.
    Keywords:  Cartilage repair; Exosomes; GrpE-like 1; Osteoarthritis; Phosphatase and tensin homolog-induced putative kinase 1; Synovial mesenchymal stem cell
    DOI:  https://doi.org/10.4252/wjsc.v17.i10.109369
  14. Phytomedicine. 2025 Oct 30. pii: S0944-7113(25)01123-7. [Epub ahead of print]148 157486
       BACKGROUND: This study explored the protective effects of kaempferol (KAE) against neurotoxicity caused by microglial activation and its underlying mechanisms.
    METHODS: Microglia BV2 cells were directly stimulated by LPS to assay the effect of KAE. More, C57BL/6 N mice received intraperitoneal LPS injections to prepare a mouse model of neuroinflammation. Autophagy inhibitors 3-MA and Mdivi-1 were utilized to evaluate the effect of KAE on mitophagy.
    RESULTS: The results demonstrated that KAE effectively suppressed LPS-induced the production of inflammatory factors, cell proliferation, phagocytic activity and NF-κB signaling activation in microglia BV2 cells, showing the inhibitory effects on microglial activation and inflammation response. Mechanically, the ability of KAE to inhibit NLRP3 inflammasome activation was associated with its enhancement of mitophagy to reduce mitochondrial DNA (mtDNA) resynthesis and leakage. Inhibition of mitophagy significantly promotes mtDNA resynthesis and release, almost completely counteracting this anti-inflammatory effect of KAE. Next, we demonstrated that KAE treatment protected neurons against LPS-induced microglial neurotoxicity and attenuated depressive-like behavior in neuroinflammation mice. This effect was associated with the interference of KAE on NLRP3 inflammasomes activation mediated by reduced mitophagy in microglia, suggesting that promoting mitophagy was one of the mechanisms by which KAE exerted neuroprotective effects on neuroinflammatory mice.
    CONCLUSION: This study showed that KAE exerted neuroprotective effects by inhibiting LPS-induced neuroinflammation mediated by microglial activation. The underlying mechanism was that KAE promoted mitophagy in microglia exposed to LPS, thereby inhibiting mtDNA leakage-induced NLRP3 inflammasome activation. It pointed to KAE as a promising therapeutic candidate for neuroinflammation-related diseases.
    Keywords:  Kaempferol; Mitophagy; NLRP3 inflammasome; Neuroinflammation; mtDNA
    DOI:  https://doi.org/10.1016/j.phymed.2025.157486
  15. Int Immunopharmacol. 2025 Nov 05. pii: S1567-5769(25)01781-3. [Epub ahead of print]168(Pt 1): 115793
      Traumatic brain injury (TBI) significantly influences neurological outcomes, and its pathogenesis is primarily attributed to neuronal apoptosis and oxidative stress. However, the precise role of Bcl2/adenovirus E1B protein-interacting protein 3 (BNIP3)-mediated mitophagy in this process remains poorly understood. Therefore, this study was designed to elucidate the function of BNIP3-dependent mitophagy in neuronal fate following TBI. Furthermore, an in vitro TBI model was established by promoting oxygen-glucose deprivation/reperfusion in HT22 neuronal cells. The BNIP3 expression in TBI model cells was systematically manipulated via siRNA-mediated knockdown and plasmid-based overexpression. Moreover, mitophagy activity was assessed by monitoring key mitochondria-associated proteins, including mitochondrial adaptor protein P62, microtubule-associated protein 1 light chain 3B (LC3B), the translocase of outer mitochondrial membrane 20 (TOMM20), and cytochrome c oxidase IV (COX IV). Immunofluorescence microscopy and transmission electron microscopy were carried out to analyze mitochondrial-lysosomal colocalization and autophagosome formation, respectively. Similarly, neuronal apoptosis was quantified via TUNEL assay, while oxidative stress was evaluated by measuring superoxide dismutase (SOD) activity and malondialdehyde (MDA) levels to elucidate redox imbalance. The results indicated that TBI substantially upregulated BNIP3 expression and enhanced mitophagy in HT22 cells. Furthermore, it increased neuronal apoptosis and MDA levels, but reduced SOD activity. BNIP3 knockdown significantly attenuated TBI-induced mitophagy while exacerbating apoptotic cell death and oxidative stress. Whereas BNIP3 overexpression elevated mitophagy and conferred robust neuroprotection against TBI-induced neuronal apoptosis and oxidative damage. In conclusion, these in vitro findings indicated that BNIP3-mediated mitophagy is a crucial endogenous neuroprotective mechanism in neurons, suggesting its potential in promoting neuronal survival following TBI, which warrants further in vivo validation.
    Keywords:  Apoptosis; Bcl2/adenovirus E1B protein-interacting protein 3; Mitophagy; Oxidative stress; Traumatic brain injury
    DOI:  https://doi.org/10.1016/j.intimp.2025.115793
  16. Biol Direct. 2025 Nov 06. 20(1): 108
      
    Keywords:  Biomolecular condensate; Galectin-3; Liquid-liquid phase separation (LLPS); Mitochondrial outer membrane (OMM) rupture ; PINK1/Parkin-dependent mitophagy
    DOI:  https://doi.org/10.1186/s13062-025-00692-1
  17. Sci Rep. 2025 Nov 04. 15(1): 38530
      Mitochondrial dysfunction drives intervertebral disc degeneration, and mitochondrial dynamics are regulated by mitofusins (MFNs). In this study, we evaluated the roles of MFN1 and MFN2 in mitochondrial quality control and their responses to inflammation and antioxidant treatment in grade I and III disc nucleus pulposus cells (NPCs). Human NPCs were isolated from intervertebral disc tissues of patients. Tumor necrosis factor-α (TNF-α)-induced inflammation was treated with vitamin E (Vit E) or saponin. Mitochondrial quality control was evaluated via quantitative polymerase chain reaction, western blotting, and immunocytochemistry. Sulfated glycosaminoglycan levels were quantified to assess extracellular matrix (ECM) integrity. Mitochondrial morphology and function were assessed via transmission electron microscopy and a xenograft model using MFN1-knockout NPCs. TNF-α significantly upregulated MFN1 and MFN2, with MFN1 showing heightened sensitivity in grade III disc NPCs, leading to mitochondrial fragmentation and ECM degradation. Antioxidants mitigated these effects, with Vit E proving more effective than saponin in reducing MFN1 expression, preserving mitochondrial structure, and stabilizing ECM composition. Vit E maintained mitochondrial integrity, whereas TNF-α induced mitochondrial swelling. In vivo, MFN1-knockout NPCs exhibited reduced ECM proteoglycan levels, reinforcing its role in disc homeostasis. These findings suggest that although MFN1 and MFN2 respond to TNF-α, MFN1 reacts more robustly, making it a more promising target under inflammatory stress.
    Keywords:  Degenerative disc diseases; Inflammation; Mitochondrial function; Mitochondrial quality control; Mitofusin; Nucleus pulposus cells
    DOI:  https://doi.org/10.1038/s41598-025-19540-3
  18. Exp Neurol. 2025 Nov 04. pii: S0014-4886(25)00399-1. [Epub ahead of print] 115534
      Alzheimer's disease (AD) is characterized by amyloid-β (Aβ) accumulation, synaptic dysfunction, and chronic neuroinflammation. Here, we demonstrate that hyperbaric oxygen therapy (HBOT) has multiple therapeutic effects in 5xFAD transgenic mice. HBOT significantly improved cognitive function. Treated mice increasingly moved to the center of the open field in Y-maze tests and preferred a new arm. Longitudinal 18F-AV-45 PET-MR scans showed progressive reduction in amyloid tracer uptake, which was corroborated by histologically verified reduced plaque burden and upregulation of LRP1, a key Aβ clearance transporter. HBOT preserved neuronal density and enhanced synaptic proteins. Mechanistically, HBOT promoted mitochondrial quality control by upregulating PINK1 and parkin expression, enhancing autophagosome formation, and modulating mitophagy-associated pathways. The transition of microglia to a surveillance phenotype was reflected in decreased soma area and increased branching. The coordinated improvement in amyloid clearance, mitochondrial quality control and synaptic maintenance, and modulation of neuroinflammation suggest that the ability of HBOT to simultaneously act on multiple pathological cascades-in combination with its noninvasive nature and favorable safety profile-makes it a uniquely promising therapeutic strategy. Furthermore, these results suggest that HBOT may be particularly effective at an early stage of the disease. These studies will be critical in establishing the clinical applicability of HBOT in the treatment of Alzheimer's disease.
    Keywords:  Alzheimer's disease; Amyloid-β clearance; Hyperbaric oxygen therapy; Mitophagy; Neuroinflammation; Synaptic plasticity
    DOI:  https://doi.org/10.1016/j.expneurol.2025.115534
  19. Biochem Biophys Res Commun. 2025 Nov 04. pii: S0006-291X(25)01635-3. [Epub ahead of print]790 152919
       BACKGROUND: Gestational diabetes mellitus (GDM) is characterized by the onset of abnormal glucose tolerance during pregnancy, with placental mitochondrial dysfunction implicated in its pathogenesis. This study investigated the role of dynamin-related protein 1 (Drp1) and its phosphorylation at Ser637 in regulating placental mitochondrial function in GDM.
    METHODS: Placental tissues were collected from normal glucose tolerance (NGT) pregnancies and GDM patients. Protein levels of Drp1 and pDrp1(Ser637), along with mitochondrial function markers, were analyzed. Human placental trophoblasts were treated with a PKA activator to examine its role in modulating Drp1 phosphorylation. Additionally, Drp1 overexpression and inhibition (via Mdivi-1) were employed to assess mitochondrial dynamics. Associations between Drp1/pDrp1(Ser637) and oral glucose tolerance test (OGTT) results were evaluated using multivariate linear regression.
    RESULTS: Compared to controls, GDM placentas exhibited significantly elevated Drp1 expression and reduced pDrp1(Ser637) levels, accompanied by impaired mitochondrial function. In trophoblasts under high-glucose conditions, PKA activation effectively restored pDrp1(Ser637) phosphorylation and alleviated Drp1-mediated mitochondrial dysfunction. Overexpression of Drp1 in normal trophoblasts induced mitochondrial abnormalities, while Mdivi-1 treatment markedly attenuated high glucose-induced Drp1 activation and improved mitochondrial homeostasis. Multivariate analysis further identified upregulated Drp1 and downregulated pDrp1(Ser637) as independent risk factors for GDM progression.
    CONCLUSION: Our findings demonstrate that PKA-dependent phosphorylation of Drp1 at Ser637 regulates placental mitochondrial homeostasis in GDM. Targeting this pathway may offer a novel therapeutic strategy for GDM management.
    Keywords:  Drp1; Gestational diabetes mellitus; Mitochondrial dysfunction; Placenta; pDrp1(Ser637)
    DOI:  https://doi.org/10.1016/j.bbrc.2025.152919
  20. Cardiovasc Drugs Ther. 2025 Nov 03.
       PURPOSE: While reperfusion is essential for restoring blood flow, it can paradoxically exacerbate myocardial injury by disrupting energy metabolism, leading to cell necrosis, apoptosis, and structural damage. Despite the significance of ischemia-reperfusion (I/R) injury, effective treatments remain scarce, highlighting the need for a deeper understanding of its underlying mechanisms to develop targeted therapies.
    METHODS: A rat model of myocardial I/R injury and a H9c2 cell-based hypoxia/reoxygenation (H/R) model were utilized for primary validation. HOXB5 was overexpressed several days prior to I/R or H/R induction. Gene and protein expression were assessed by RT-qPCR, Western blotting, immunohistochemistry, and immunofluorescence. Cell viability and apoptosis were evaluated using CCK-8 and flow cytometry. Mitochondrial function and ferroptosis were analyzed by commercial kits. The regulatory relationship among Lin28a, HOXB5, and Sirt5 was examined using RIP, ChIP-PCR, and dual-luciferase assays.
    RESULTS: HOXB5 expression was significantly reduced in infarcted myocardial tissue following I/R injury, of which overexpression attenuated H/R-trigged apoptosis to motivate H9c2 cell survival. Meanwhile, HOXB5 overexpression decreased mitochondrial ROS production, improved mitochondrial respiration, mitochondrial complexes (I, II, III, and V) activity, and reduced mitophagy under H/R conditions in H9c2 cells. Mechanistically, Lin28A was identified as a regulator of HOXB5, which in turn transcriptionally activated Sirt5. Furthermore, the protective effects of HOXB5 overexpression on myocardial histological damage and cardiac function through modulating ferroptosis in rats' I/R injury was abrogated by Sirt5 knockdown.
    CONCLUSION: Our study reveals the crucial role of HOXB5 in maintaining mitochondrial homeostasis and regulating mitophagy, which in turn protects cardiomyocytes from I/R injury through Sirt5. These findings underscore the HOXB5-Sirt5 pathway as a promising therapeutic target for mitigating I/R injury.
    Keywords:  Ferroptosis; HOXB5; Lin28A; Mitochondrial protection; Mitophagy; Myocardial ischemia–reperfusion injury; Sirt5
    DOI:  https://doi.org/10.1007/s10557-025-07785-z
  21. Immunol Lett. 2025 Nov 03. pii: S0165-2478(25)00140-3. [Epub ahead of print] 107107
       OBJECTIVE: To investigate the pro-inflammatory role and underlying mechanism of Perilipin 2 (PLIN2) in Allergic Rhinitis (AR), focusing on its regulation of PINK1/Parkin-mediated mitophagy and the subsequent impact on lipid metabolism and oxidative stress.
    METHODS: Single-cell RNA sequencing (scRNA-seq) analysis was performed using GSE261706 from the GEO database, involving nasal mucosa from AR patients and healthy controls. A murine AR model was induced by ovalbumin (OVA), and human nasal epithelial cells (HNEpCs) were stimulated with Der p1. Interventions included AAV-mediated PLIN2 knockdown in vivo and siRNA-mediated knockdown in vitro. Techniques included Western blotting, qRT-PCR, flow cytometry, ELISA, and immunofluorescence/histological staining to assess PLIN2 expression, mitophagy, lipid accumulation, oxidative stress, and inflammatory responses.
    RESULTS: scRNA-seq analysis identified PLIN2 as a significantly upregulated gene in AR epithelial cells, which correlated with dysfunctional autophagy pathways. In both OVA-induced mice and Der p1-treated HNEpCs, PLIN2 expression was significantly elevated, accompanied by inhibited mitophagy (decreased LC3-II/I ratio, reduced PINK1/Parkin levels, and p62 accumulation), increased lipid deposition, and elevated ROS levels. PLIN2 knockdown markedly ameliorated AR pathology in mice, reducing inflammatory infiltration and serum levels of IgE, IL-4, and IL-5. Mechanistically, PLIN2 knockdown restored PINK1/Parkin-mediated mitophagy, decreased lipid accumulation, and attenuated ROS-induced cellular damage in HNEpCs.
    CONCLUSIONS: PLIN2 exacerbates AR pathogenesis by inhibiting PINK1/Parkin-mediated mitophagy, promoting lipid accumulation and oxidative stress, and ultimately causing cellular damage in nasal epithelial cells. PLIN2 acts as a pivotal mediator linking metabolic dysregulation to inflammation, highlighting it as a promising therapeutic target for AR treatment.
    Keywords:  Allergic Rhinitis; Lipid Accumulation; Mitophagy; PINK1/Parkin Pathway; Perilipin 2
    DOI:  https://doi.org/10.1016/j.imlet.2025.107107
  22. Geroscience. 2025 Nov 07.
      The disposable soma theory posits that there is a trade-off between reproduction and somatic maintenance. In support of this theory, we previously identified that pharmacological inhibition of the germline has widespread protective cell non-autonomous effects on cellular protein homeostasis in the model organism Caenorhabditis elegans. However, the cell non-autonomous effects of the germline on mitochondrial protein homeostasis are not well defined. Here, we use pharmacological or genetic inhibition of the germline to determine its effects on intestinal mitochondrial protein homeostasis as measured by the mitochondrial unfolded protein response (UPRmt). We find that pharmacological inhibition of germline proliferation by 5-fluoro-2-deoxyuridine (FUdR), a DNA synthesis inhibitor, potently inhibits activation of the intestinal UPRmt as well as reverses lifespan effects induced by mitochondrial dysfunction. We find similar results with the genetic mutant (glp-1), which lacks germline proliferation. To further identify the reproductive processes required to regulate the intestinal UPRmt, we examined the genetic mutant fem-1, which contains an intact gonad with oocytes but lacks sperm. Like glp-1 mutants, fem-1 mutants do not activate the intestinal UPRmt due to mitochondrial dysfunction caused by loss of OXPHOS subunits. Restoring reproduction in fem-1 mutants by mating them with wild type males is sufficient to reactivate the intestinal UPRmt. Furthermore, loss of the FOXO transcription factor daf-16 is sufficient to reactivate the intestinal UPRmt in fem-1 mutants and partially in glp-1 mutants. These findings suggest that FOXO/daf-16 acts to limit UPRmt activation in the intestine. These findings also suggest that late-stage reproductive signals that include the maturation of oocytes and fertilization may play a critical role in cell non-autonomous intestinal UPRmt activation.
    Keywords:   Caenorhabditis elegans ; Cell non-autonomous; Fertilization; Germline; Intestines; Mitochondrial unfolded protein response; Reproduction
    DOI:  https://doi.org/10.1007/s11357-025-01890-5
  23. Phytomedicine. 2025 Oct 24. pii: S0944-7113(25)01093-1. [Epub ahead of print]148 157456
       BACKGROUND: Postmenopausal osteoporosis (PMOP) is the predominant form of primary osteoporosis. Its etiology remains incompletely elucidated, and the senescence of bone marrow mesenchymal stem cells (BMSCs) is potentially a contributing factor. Kaempferol (Ka), a common flavonoid with antioxidant and anti-inflammatory effects, is a prospective candidate for therapeutic intervention in osteoporosis. However, its impact on BMSC senescence and the underlying mechanisms remains to be elucidated.
    PURPOSE: This study sought to elucidate the impact of Ka on BMSCs and to ascertain its potential targets and regulatory mechanisms.
    METHODS: BMSCs were extracted from ovariectomized (OVX) rats to observe the impact of Ka on cellular senescence, mitochondrial function, and mitophagy. Proteomics was utilized to investigate the potential mechanisms. Molecular docking, cellular thermal shift assay, small molecule pull-down assay, and surface plasmon resonance were employed to evaluate the relationship between Ka and its direct target Sp1. Dual-luciferase reporter gene assay and chromatin immunoprecipitation (ChIP) were used to investigate the transcriptional regulatory relationship between Sp1 and FUNDC1. Moreover, we performed detailed imaging and histological observations using micro-CT, hematoxylin and eosin (H&E) staining, Masson staining, immunofluorescence, and immunohistochemistry to assess the role of Ka in rat model of PMOP.
    RESULTS: Our results indicated that Ka counteracts senescent BMSCs and osteogenic differentiation deficit caused by postmenopausal, which may be achieved by restoring the basal mitophagy level of OVX-BMSCs, mitochondrial function and reducing reactive oxygen species. Furthermore, Ka treatment enhanced bone density and strength, upregulated Sp1 and FUNDC1 expression in the distal femoral region, and the number of γH2AX positive BMSCs decreased. In terms of mechanism, Ka directly binds to Sp1, promoting its transcriptional activation of FUNDC1-mediated mitophagy. Inhibiting mitophagy or Sp1, or FUNDC1 eliminated the therapeutic effect of Ka.
    CONCLUSIONS: Our findings suggest that Ka ameliorates the senescence of OVX-BMSCs by activating the Sp1/FUNDC1 signaling pathway, promoting mitophagy, and providing a new therapeutic strategy and molecular basis for PMOP.
    Keywords:  Bone marrow mesenchymal stem cells; FUNDC1; Kaempferol; Mitophagy; Postmenopausal osteoporosis; Senescence
    DOI:  https://doi.org/10.1016/j.phymed.2025.157456
  24. Cell Cycle. 2025 Nov 05. 1-17
      Cisplatin (DDP) resistance substantially compromises treatment efficacy in lung adenocarcinoma (LUAD). This study investigates the role of mitochondrial long non-coding RNA (lncRNA) H19 in mediating DDP resistance. High-throughput sequencing and RT-qPCR analyses revealed pronounced H19 upregulation in DDP-resistant A549 (A549/DDP) cells relative to parental A549 cells. Subcellular localization studies indicated that H19 is primarily nuclear in A549 cells but translocates to mitochondria in A549/DDP cells. Functional assays demonstrated that H19 silencing in resistant cells attenuated chemoresistance, suppressed proliferation, migration, invasion, and colony formation in vitro, and delayed tumor growth in vivo. H19 knockdown impaired mitophagy and promoted apoptosis, mirroring autophagy inhibition and restoring DDP sensitivity. In contrast, H19 overexpression in A549 cells did not significantly alter mitophagy or cellular behavior. Furthermore, H19 silencing induced its relocalization from mitochondria back to the nucleus in resistant cells, while overexpression did not affect its nuclear localization. These findings establish that H19 translocation to mitochondria promotes DDP resistance, and its downregulation reverses this process by inhibiting mitophagy and resensitizing cells to DDP. As a nucleus-encoded mitochondria-associated lncRNA (ntmtlncRNA), H19 mediates intercompartmental communication, highlighting its potential as a therapeutic target for overcoming DDP resistance in LUAD.
    Keywords:  LUAD; cisplatin resistance; lncRNA H19; mitophagy
    DOI:  https://doi.org/10.1080/15384101.2025.2581634
  25. Nat Commun. 2025 Nov 07. 16(1): 9845
      Mitophagy is crucial for the selective autophagic degradation of damaged mitochondria, helping to maintain both mitochondrial and cellular homeostasis. Here, we report a fluoroalkylated polypyridinium that specifically targets mitochondria and exhibits high activity in mitophagy induction. The polymer effectively restores mitochondrial function and alleviates the inflammatory response in foam cells by activating mitophagy, and displays inherent red fluorescence under physiological conditions, allowing for direct tracing of its biodistribution in cells and in vivo. Besides, the polymer nanoparticle shows high serum stability due to the antifouling properties of fluoroalkyl tags. After intravenous administration, the nanoparticle reduces oxidative stress, promotes mitophagy, and decreases cellular senescence in atherosclerotic plaques, contributing to high therapeutic efficacy. This study presents an innovative and effective strategy for the treatment of atherosclerosis and other mitochondrial dysfunction-related inflammatory conditions.
    DOI:  https://doi.org/10.1038/s41467-025-64813-0
  26. J Intensive Care. 2025 Nov 04. 13(1): 61
      Heatstroke is characterized by systemic inflammation, immune dysregulation, and multiorgan failure, in which mitochondrial damage in leukocytes plays a pivotal role. This review examines the mechanisms by which heat stress induces leukocyte mitochondrial dysfunction and its downstream effects on immunity, coagulation, and organ integrity. Exposure to heat stress activates leukocytes through damage-associated molecular patterns (DAMPs), triggering the release of proinflammatory cytokines, reactive oxygen species (ROS), and neutrophil extracellular traps (NETs). These responses disrupt endothelial integrity, promote microvascular thrombosis, and contribute to the development of disseminated intravascular coagulation (DIC). Prolonged heat exposure further shifts the immune landscape toward immunosuppression, marked by monocyte deactivation and lymphocyte apoptosis. Mitochondrial dysfunction is central to this biphasic immune response. Heat stress reduces mitochondrial membrane potential, increases ROS production, and promotes the release of mitochondrial DNA and cytochrome c, amplifying inflammation and initiating cell death pathways, including apoptosis, pyroptosis, and ferroptosis. Biomarkers such as reduced mitochondrial membrane potential (ΔΨm), elevated mitochondrial ROS, cytochrome c, circulating mitochondrial DNA (mtDNA), and altered expression of mitophagy regulators (e.g., PINK1 and Parkin) provide insights into mitochondrial integrity and function in leukocytes. In addition to immune disruption, mitochondrial injury exacerbates coagulation abnormalities by promoting platelet activation and endothelial dysfunction, fostering a prothrombotic environment. In the microcirculation, leukocyte adhesion, NET formation, and endothelial damage create a self-amplifying cycle of ischemia and inflammation, ultimately leading to organ dysfunction, including hepatic failure, acute kidney injury, acute lung injury, and gastrointestinal barrier breakdown. Therapeutic strategies aimed at preserving mitochondrial function include antioxidants (e.g., N-acetylcysteine and MitoQ), mitochondrial biogenesis inducers (e.g., PGC-1α activators), and mitophagy enhancers. Understanding the central role of leukocyte mitochondrial damage in heat stress provides a foundation for the development of targeted diagnostics and interventions to prevent organ failure and improve clinical outcomes.
    Keywords:  Cell death; Heat stress; Leukocyte; Mitochondria; Organ dysfunction
    DOI:  https://doi.org/10.1186/s40560-025-00832-9
  27. Phytomedicine. 2025 Oct 26. pii: S0944-7113(25)01098-0. [Epub ahead of print]148 157461
       BACKGROUND: Hepatic fibrosis manifests with impaired extracellular matrix (ECM) deposition and inflammation within the liver. This study probes deep into the hepatoprotective effects and underlying mechanisms of syringin (SY) against hepatic fibrosis and inflammation.
    METHODS: A model of hepatic fibrosis in mice was established through intraperitoneal injection of thioacetamide (TAA) and silencing of SIRT3. RNA sequencing was employed to elaborate upon the SY-associated signaling network. Hepatic stellate cells (HSCs) were treated with transforming growth factor-beta (TGF-β) and lipopolysaccharide/adenosine triphosphate (LPS/ATP) was utilized to induce bone marrow-derived macrophages (BMDMs) for conditioned medium collection. To comprehensively assess the role of SIRT3 in SY-mediated effects, LX-2 cells and BMDMs underwent SIRT3 knockdown (siRNA-SIRT3).
    RESULTS: RNA sequencing analysis suggested that SY exerts its hepatoprotective effects primarily through modulation of the SIRT3/NOD-like receptor pathway. After knocking down SIRT3 via shRNA, the protein and mRNA levels of P2×7r in mouse livers increased significantly, and inflammation was exacerbated. Furthermore, SY up-regulated SIRT3 protein expression while down-regulating PINK1 and Parkin, suggesting a role in mitochondrial quality control. In particular, SIRT3 deficiency in LX-2 cells and BMDMs attenuated SY's regulatory effects on ECM remodeling and inflammation. SY also inhibited BMDM activation, thereby lessening the extracellular release of IL-1β and IL-18.
    CONCLUSIONS: Collectively, the above-summarized results suggest that SY mitigates hepatic inflammation and fibrogenesis by modulating the SIRT3-P2×7r/NLRP3 signaling axis, positioning it as a prospective therapeutic alternative for hepatic fibrosis. SY might be a prospective therapeutic candidate, and its mechanism would be a new direction or strategy for hepatic fibrosis.
    Keywords:  Hepatic fibrosis; Mitophagy; P2×7r; SIRT3; Syringin
    DOI:  https://doi.org/10.1016/j.phymed.2025.157461
  28. Neurochem Res. 2025 Nov 03. 50(6): 345
      Up-to-date data on roles of ATP‑sensitive potassium (KATP) channels indicate their emerging roles in neurodegeneration. The aim of present study was to evaluate the significance of KATP channels on cell viability, calcium dynamics, and mitochondrial morphology with the accent on their intracellular localization. We distinguished between whole-cell KATP effects and specific effects of mitochondrial KATP under both physiological conditions and pathological conditions simulating in vitro Parkinson´s-type neurodegeneration. SH‑SY5Y cells with its high fidelity to dopaminergic neurons were treated for 24 h with the non‑selective KATP opener pinacidil and blocker glibenclamide, or with the mitochondrial KATP opener diazoxide and blocker 5‑hydroxydecanoate (5HD). The effects of modulators were analysed alone or alongside with rotenone, which is widely used as an inducer of Parkinson´s-type neurodegeneration. Intracellular calcium distribution and mitochondrial rebuild pattern was evaluated using the cell segmentation performed by fluorescent confocal microscopy. Although none of the KATP modulators reversed the negative effects of rotenone, significant and selective effects of mitochondrial KATP modulation on calcium homeostasis and mitochondrial morphology were observed. For antagonists, both compounds showed consistent effects, with non-selective glibenclamide exerting stronger effects, particularly in elevating calcium. More distinctive results were obtained for agonists: both reduced calcium concentration; however, pinacidil tended to induce mitochondrial fragmentation, an effect absent in diazoxide-treated cells. Furthermore, strong correlations were identified between calcium levels and several mitochondrial and cell viability parameters.
    Keywords:  5-Hydroxydecanoate; Cell viability; Diazoxide; Glibenclamide; KATP; Mitochondrial dynamics; Neurodegeneration; Pinacidil
    DOI:  https://doi.org/10.1007/s11064-025-04598-2
  29. Cell Death Differ. 2025 Nov 01.
      The transfer of information and metabolites between the mitochondria and the endoplasmic reticulum (ER) is mediated by mitochondria-ER contact sites (MERCS), allowing adaptations in response to changes in cellular homeostasis. MERCS are dynamic structures essential for maintaining cell homeostasis through the modulation of calcium transfer, redox signalling, lipid transfer, autophagy and mitochondrial dynamics. Under stress conditions such as ER protein misfolding, the Unfolded Protein Response (UPRER) mediates PERK and IRE1 activation, both of which localise at MERCS. Adaptive UPRER signalling enhances mitochondrial function and calcium import, whereas maladaptive responses lead to excessive calcium influx and apoptosis. In this study, induction of mild acute ER stress with tunicamycin (TM) in myoblasts promoted myogenesis that required PERK for increased MERCS assembly, mitochondrial turnover and function. Similarly, treatment of C. elegans embryos with an acute low concentration of TM, promoted an extension in lifespan and health-span. The adaptive ER stress response following a low dose of TM in both myoblasts and C. elegans, increased MERCS assembly and activated autophagy machinery, ultimately promoting an increase in mitochondrial remodelling. However, these beneficial adaptations were dependent on the developmental stage, as treatment of myotubes or adult C. elegans resulted in a maladaptive response. In both models the adaptations to UPRER activation were dependent on PERK signalling and its interaction with the UPRmt. The results demonstrate PERK is required for the increased mitochondrial ER communication in response to adaptive UPR signalling, promoting mitochondrial remodelling and improved physiological function.
    DOI:  https://doi.org/10.1038/s41418-025-01603-7
  30. Sci Rep. 2025 Nov 06. 15(1): 38885
      Neuroinflammation plays a central role in a wide spectrum of neurological diseases, driven generally by reactive microglia and astrocytes. Inflammatory stimulation of microglia and astrocytes leads to a metabolic shift from oxidative phosphorylation (OXPHOS) to glycolysis, which is required to support pro-inflammatory effector functions. This metabolic reprogramming is associated with impaired mitochondrial dynamics, including reduced biogenesis, increased fragmentation, and loss of membrane potential. Targeting microglia and astrocyte metabolism may offer a novel therapeutic approach for modulating neuroinflammation and restoring homeostatic immune functions. Here, we examined the potential of 2-Deoxy-D-Glucose (2DG), a glycolysis inhibitor, to attenuate neuroinflammation by restoring mitochondrial dynamics. In BV2 and primary glial cultures, low-dose 2DG reversed LPS-induced metabolic reprogramming, restoring OXPHOS, reducing mitochondrial fragmentation, and enhancing biogenesis. In vivo, it preserved spare respiratory capacity and increased complex-V activity in brain mitochondria from LPS-treated mice without affecting oxidative stress. At a mechanistic level, 2DG restored activation of AMP-activated protein kinase, a master regulator of mitochondrial dynamics. In conjunction with these metabolic effects, 2DG suppressed LPS-induced pro-inflammatory gene expression while enhancing markers associated with the resolution of inflammation and tissue repair. Critically, systemic low-dose 2DG reduced neuroinflammation and restored immune homeostasis in two LPS-induced mouse models, highlighting its therapeutic potential in neurological disorders.
    Keywords:  2-Deoxy-D-Glucose; Immunometabolism; Mitochondrial dynamics; Mitochondrial function; Neuroinflammation
    DOI:  https://doi.org/10.1038/s41598-025-22677-w
  31. Free Radic Biol Med. 2025 Oct 31. pii: S0891-5849(25)01334-6. [Epub ahead of print]242 345-363
      Cepharanthine (Cep), a natural alkaloid from Stephania (Menispermaceae), exhibits broad-spectrum anti-cancer activity. In the present study, Cep was found to induce ferroptosis and mitophagy, for which the relationship and upstream targets remain unelucidated. Herein, the role of Cep in the induction of mitophagy was deeply investigated. Cep showed robust anti-lung cancer effects, as confirmed by decreased cell viability, elevated apoptosis, suppressed colony formation and inhibited growth of tumor grafts in Lewis cell-bearing mice. RNA-sequencing analysis revealed that Cep treatment significantly enriched differentially expressed genes (DEGs) in mitophagy and ferroptosis pathways, which were demonstrated in in vitro and in vivo experiments as well. In-depth investigations showed that inhibition of autophagy abolished Cep-mediated ferroptosis, but not vice versa. Moreover, genetic knockdown of BNIP3 dampened the mitophagy and ferroptosis of lung cancer cells induced by Cep. Additional data confirmed that Cep could bind to and thereby inhibit FBXL4, which attenuated the ubiquitination of BNIP3. FBXL4-mediated BNIP3 activation promoted the recruitment of LC3 to mitochondria and autophagic flux in the presence of Cep. Collectively, Our study elucidates a complete mechanistic pathway wherein Cep activates BNIP3-mediated mitophagy by inhibiting FBXL4, ultimately driving ferroptosis and offering a new therapeutic avenue for lung cancer.
    Keywords:  Cepharanthine (Cep); FBXL4-BNIP3 axis; Ferroptosis; Lung cancer; Mitophagy
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.10.302
  32. Biochem Pharmacol. 2025 Nov 02. pii: S0006-2952(25)00777-4. [Epub ahead of print]243(Pt 1): 117512
      Autophagy plays a critical role in clearance of Mycobacterium tuberculosis. It has emerged as a promising target for host-directed therapies against drug-resistant tuberculosis (TB). This insight opens up promising therapeutic avenues, suggesting that pharmacological activation of autophagy could effectively combat this highly persistent and harmful bacterium. The current study investigates the anti-mycobacterial properties of the anthracene-dione compound Mitoxantrone (MTX) through the activation of autophagy in differentiated THP-1 cells. The non-cytotoxic dose of MTX reduced the intracellular viability of mycobacteria compared to the control cells, and inhibition of autophagy reversed the effect of MTX on intracellular bacterial burden. Through multiparametric approaches, our investigation established the effect of MTX on mitochondria, the principal source of endogenous reactive oxygen species (ROS), acting as essential signal transducers that promote autophagy. Further, we have demonstrated that MTX decreased ATP production, which caused disruption of mitochondrial membrane proteins and increased mitochondrial ROS generation, resulting in mitochondrial fission and accelerating the initiation of mitophagy, leading to the elimination of intracellular mycobacteria. Our findings collectively demonstrated that MTX-induced mitochondrial dysfunction triggered interplay between two selective autophagic responses, diminishing mycobacterial infection and promoting its clearance. This study highlights MTX as a potential host-directed therapeutic candidate against TB through modulation of mitochondrial signaling pathways and autophagic responses.
    Keywords:  Autophagy; Mitochondria; Mitophagy; Mitoxantrone; Mycobacteria
    DOI:  https://doi.org/10.1016/j.bcp.2025.117512
  33. Sci Total Environ. 2025 Nov 06. pii: S0048-9697(25)02507-0. [Epub ahead of print]1005 180867
      Bisphenol A (BPA), recognized as an endocrine disruptor, is widely utilized in the manufacturing of various household products. Research has demonstrated that maternal exposure to BPA during pregnancy is associated with intestinal disorders and can adversely affect offspring. Furthermore, previous studies have identified that melatonin (MT), secreted by the pineal gland, plays a crucial role in maintaining intestinal health and exhibits detoxification properties. Therefore, this study investigates the therapeutic effects of MT on BPA-induced intestinal damage in pregnant mice. The findings indicate that BPA exposure compromises gut mucosal barrier function, evidenced by a reduction in the integrity and height of small intestinal villi, a decrease in goblet cell numbers, and diminished levels of MUC2 and tight junction proteins, alongside increased apoptosis, oxidative stress, and mitochondrial dysfunction. Additionally, BPA exposure significantly downregulated the levels of tryptophan-derived bacteria, tryptophan, and the aryl hydrocarbon receptor (AhR). Conversely, melatonin supplementation exerted beneficial effects on gut homeostasis, enhanced the gut mucosal barrier, and restored gut microbiota balance. Furthermore, in vitro experiments demonstrated that tryptophan can enhance mitophagy through the activation of the AhR, thereby improving intestinal barrier function in Caco-2 cells. In conclusion, our findings provide a novel theoretical foundation for the future application of melatonin in promoting intestinal health. They also offer innovative insights into the relationship between melatonin and the intestinal tryptophan/AhR/mitochondria axis, emphasizing its potential for enteroprotection against BPA-induced enterotoxicity.
    Keywords:  Bisphenol a; Enterotoxicity; Intestinal microbiota; Melatonin; Mitophagy
    DOI:  https://doi.org/10.1016/j.scitotenv.2025.180867
  34. Adv Healthc Mater. 2025 Nov 06. e03530
      Metal ion dyshomeostasis represents a therapeutic vulnerability in cancer, yet simultaneous targeting of multiple metal-dependent death pathways remains challenging. Herein, a pH-responsive copper-based metal-organic framework nanoplatform (Cu-MOF@DPCPX) is engineered to co-trigger cuproptosis, ferroptosis, and mitophagy through tumor-specific copper overload. The system leverages acidic tumor microenvironments for targeted degradation, releasing Cu2⁺. The liberated Cu2⁺ depletes overexpressed glutathione (GSH) to disrupt redox homeostasis and generates toxic Cu⁺ that initiates dual catalytic cycles. 1) Cu⁺ accumulation promotes lipoylated protein aggregation and Fe-S cluster loss, driving cuproptosis; 2) Cu⁺-mediated Fenton-like reactions convert endogenous H2O2 into hydroxyl radicals (·OH) and downregulate GPX4 to induce ferroptosis. Crucially, mitochondrial damage from these pathways activates mitophagy, which releases sequestered copper to establish a self-amplifying death cascade. In vivo, Cu-MOF@DPCPX demonstrates potent tumor suppression across multiple tumor models (4T1-breast, LLC-lung, PAN02-pancreatic, GL261-glioblastoma), while reprogramming immunosuppressive microenvironments via increased CD8⁺ T-cell infiltration and M1 macrophage polarization. This triple-pathway activation strategy overcomes monotherapy limitations and establishes a paradigm for metal-ion-based multimodal oncotherapy.
    Keywords:  copper‐based metal organic framework; metal ion dyshomeostasis; nanocatalyst; pH‐responsive; tumor therapy
    DOI:  https://doi.org/10.1002/adhm.202503530
  35. Brain Res. 2025 Oct 31. pii: S0006-8993(25)00571-2. [Epub ahead of print] 150008
       OBJECTIVE: Parkinson's disease (PD) is a highly prevalent degenerative disease of the nervous system, with symptoms of motor retardation and balance disorders. This study aimed to investigate the Erbai decoction on PD rats and explore the underlying mechanism.
    METHODS: SD rats were divided into five groups, including control, PD, PD + L-Erbai decoction, PD + H-Erbai decoction, and PD + Madopar. Drug treatment was employed with PD rats for 30 days. Firstly, behavioral experiments including the open field test (OFT), pole climbing, tail suspension test (TST), and Morris Water Maze (MWM) were performed. Next, the substantia nigra pars compacta (SNpc) of rats was obtained for H&E and Immunofluorescence (IF) staining, Tunnel, Transmission electron microscopy (TEM), Enzyme-linked immunosorbent assay (ELISA), Flow cytometry, Real-time fluorescence quantitative PCR (RT-qPCR), and Western blotting (WB) assays. Finally, the components of Erbai serum were determined by HPLC-QTOF/MS/MS assay.
    RESULTS: For the behavioral experiments, it was found that Erbai decoction could enhance the balance and recognition abilities of PD rats. The results of HE and Tunnel revealed that Erbai decoction reversed the rotenone-induced pathological injury of SNpc. In addition, compared with the PD group, the PD + H-Erbai decoction group showed a higher level of autophagy and an inhibition of mitochondrial damage. Moreover, Erbai decoction treatment reversed PD modeling-induced p38 MAPK up-regulation and Parkin down-regulation, and the decrease of Parkin recruitment in mitochondria. Importantly, Erbai decoction and Madopar have similar pharmacological effects. 320 unique components of Erbai serum were identified.
    CONCLUSION: Erbai decoction could alleviate the SNpc injury by activating mitophagy in PD rats, and it may relate to the regulation of the p38 MAPK/Parkin signaling pathway.
    Keywords:  Erbai decoction; Mitophagy; Parkinson’s disease; The SNpc area; The p38 MAPK/Parkin signaling pathway
    DOI:  https://doi.org/10.1016/j.brainres.2025.150008
  36. Neurobiol Dis. 2025 Nov 04. pii: S0969-9961(25)00395-X. [Epub ahead of print] 107178
      Elevated intraocular pressure (IOP) imposes a substantial glycolytic demand on retinal tissue, driving excessive lactate production to sustain neuronal energy homeostasis. In the present study, we demonstrate that this metabolic stress markedly increases histone H4 lysine 8 lactylation (H4K8la) throughout the glaucomatous retina. Genome-wide profiling using Cleavage Under Targets and Tagmentation (CUT&Tag) revealed pronounced and selective H4K8la enrichment within the promoter region of mitofusin-2 (MFN2), a key mitochondrial fusion GTPase. Functionally, suppression of MFN2 mediated by H4K8la reciprocally enhances β-catenin expression, particularly under oxidative stress, indicating the existence of a lactate-responsive epigenetic circuit that couples mitochondrial dynamics to activation of the Wnt/β-catenin signaling pathway. In vivo experiments demonstrated that activated Müller glia orchestrate synchronous mitochondrial remodeling in retinal ganglion cells (RGCs) and photoreceptors. We propose that MFN2 functions as a molecular conduit, enabling intercellular mitochondrial transfer between Müller glia and neurons to sustain elevated glycolytic flux. This metabolic reprogramming increases lactate release, stimulates Wnt/β-catenin pathway activation, and ultimately leads to RGC apoptosis. Single-cell RNA sequencing (scRNA-seq) further revealed intensified Müller-amacrine/bipolar cell communication under ocular hypertension, accompanied by a marked reduction in neurexin- and VEGF-mediated interactions. Collectively, these findings identify a lactylation-governed epigenetic checkpoint that links mitochondrial dysfunction to glial-neuronal metabolic coupling in glaucoma, nominating MFN2 as a potential neuroprotective therapeutic target.
    Keywords:  Glaucoma; Histone lactylation; MFN2; Mitochondrial dysfunction; Wnt/β-catenin pathway
    DOI:  https://doi.org/10.1016/j.nbd.2025.107178
  37. Free Radic Biol Med. 2025 Nov 02. pii: S0891-5849(25)01330-9. [Epub ahead of print]242 418-430
      Mutations in glycoprotein, zona pellucida glycoprotein 3 (ZP3), cause human ZP loss and empty follicle syndrome (EFS), yet the underlying mechanisms remain unknown. In this study, we generated Zp3-knockout rats (rZp3-/-) using CRISPR-Cas9, which recapitulated the EFS phenotype with follicular arrest and oocyte degeneration. Transmission electron microscopy revealed complete absence of the zona pellucida and transzonal projections, alongside shortened microvilli that correlated with radixin downregulation, indicating impaired oocyte-granulosa communication. Single-oocyte transcriptomics revealed dysregulated mitochondria-associated signaling pathways, such as glutathione metabolism and oxidative phosphorylation, consistent with energy deprivation and oxidative stress resulting from impaired cell dialogs between oocyte and granulosa cells. In oocytes of rZp3-/-, mitochondrial dynamics were severely disrupted, featuring reduced mitochondrial area, cristae hyperfusion. In addition, electron transport chain activity was impaired in oocytes of rZp3-/-, driving mitochondrial reactive oxygen species (ROS) overproduction. Consequently, rZp3-/- oocytes displayed ferroptosis hallmarks, including elevated lipid peroxidation, iron dysregulation, and glutathione peroxidase 4 (GPX4) inactivation. Pharmacological inhibition of ferroptosis in vitro using deferiprone partially rescued oocyte defects by restoring GPX4, attenuating mitochondrial ROS and lipid peroxidation, therefore inhibit apoptosis in rZp3-/-.This study establishes ZP3 as a guardian of mitochondrial homeostasis and implicates ferroptosis as a key pathway in EFS pathogenesis, offering novel therapeutic targets.
    Keywords:  Ferroptosis; Follicular atresia; Mitochondrial dysfunction; Oocyte-granulosa cell communication; Zp3
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.10.304
  38. J Cell Sci. 2025 Nov 03. pii: jcs.263920. [Epub ahead of print]
      The endoplasmic reticulum (ER) and mitochondria are known to affect myriad cellular mechanisms. More recently, dynamic association between them has been identified in different eukaryotes; these interactions vary in their composition and involvement in regulation of intracellular machineries. FAM134B or RETREG1, originally identified as an oncogene, regulates ER membrane shape and curvature. It is a key ER-phagy or reticulophagy receptor, which promotes autophagy of not only the ER but also simultaneous dual autophagy of ER and mitochondria. While it is known that FAM134B can potentiate contact with mitochondria, its direct involvement in affecting mitochondrial dynamics remains unexplored. Here we show that FAM134B can interact with the canonical fission-promoting protein, DRP1. Functional depletion of FAM134B leads to local Actin rearrangement and reduced DRP1 recruitment onto mitochondria, resulting in hyperfusion. A decrease in FAM134B levels is observed with aging in rat brains, cell and mouse models of Parkinson's disease and patient-derived samples. Our study establishes FAM134B as the ER partner that helps in maintaining mitochondrial morphology and dynamics.
    Keywords:  DRP1; FAM134B; Fission; Mitochondrial hyperfusion
    DOI:  https://doi.org/10.1242/jcs.263920
  39. Arch Gerontol Geriatr. 2025 Nov 03. pii: S0167-4943(25)00335-8. [Epub ahead of print]141 106078
      Chronic physical exercise is a promising non-pharmacological strategy to mitigate the progression of Alzheimer's disease (AD), yet the underlying molecular mechanisms remain incompletely understood. This study investigated the effects of chronic treadmill exercise on behavioral deficits and key molecular pathways in a d-galactose and AlCl3-induced rat model of sporadic AD. Animals were assigned to control, AD, exercise and AD + exercise (AE) groups for a ten-week intervention. Behavioral assessments included the elevated plus maze and Morris Water Maze, followed by molecular and biochemical analyses (RT-qPCR, ELISA) of the hippocampus, skeletal muscle, and plasma. Our results demonstrate that the AD model induced profound cognitive impairments, diminished locomotor activity, heightened anxiety-like behavior, and elevated plasma tau levels. These pathological changes were accompanied by a significant downregulation of the AMPK/SIRT1/PGC-1α mitochondrial biogenesis pathway and, notably, a marked suppression of the membrane repair protein Mitsugumin 53 (MG53) in both the hippocampus and skeletal muscle. The physical exercise regimen successfully ameliorated these behavioral deficits and normalized plasma tau. Mechanistically, physical exercise potently upregulated the AMPK/PGC-1α/FNDC5/BDNF axis in both central and peripheral tissues. Crucially, this study reveals for the first time that physical exercise also triggers a robust upregulation of MG53 at both the gene and protein levels in the brain, muscle, and circulation. These findings identify the physical exercise-induced mobilization of MG53 as a novel and powerful neuroprotective mechanism, linking systemic cellular repair capacity to the enhancement of cognitive resilience against AD.
    Keywords:  Alzheimer's Disease; Exerkines; Hippocampal Signaling; Mitsugumin 53; Physical Exercise
    DOI:  https://doi.org/10.1016/j.archger.2025.106078
  40. Front Physiol. 2025 ;16 1684729
      Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are a group of conditions characterized by acute episodes of pulmonary inflammation and increased pulmonary vascular permeability. These conditions often result in severe morbidity and high mortality rates. Increased alveolar-capillary barrier permeability is a pivotal factor in the pathogenesis of ALI/ARDS, and diffuse alveolar epithelial cell (AEC) death is a salient feature of ALI/ARDS. Alveolar epithelium is composed of alveolar type I epithelial cells (AECI) and alveolar type II epithelial cells (AECII), with AECII playing a more critical role. These cells contain a high density of mitochondria in their cytoplasm, and their function depends on mitochondrial quality control (MQC). Existing reviews either focus solely on the mechanisms of AECs and their relationship to lung injury/fibrosis or broadly explore the role of mitochondrial dynamics in lung diseases. However, neither review comprehensively addresses AECII's MQC and related molecules and signaling pathways. The objective of this study is to investigate the MQC characteristics of AECII in ALI/ARDS, elucidate their role as a regulatory hub for oxidative stress, inflammation, and fibrosis, summarize progress in related clinical trials, and highlight the need for further research to develop effective therapies.
    Keywords:  acute lung injury; acute respiratory distress syndrome; alveolar type II epithelial cells; fibrosis; inflammation; mitochondrial quality control; oxidative stress
    DOI:  https://doi.org/10.3389/fphys.2025.1684729
  41. Anticancer Drugs. 2025 Oct 16.
      Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer that is characterized by rapid growth. PLCG2 is an enzyme that plays a crucial role in intracellular signal transduction pathways. This study aims to discover the role of PLCG2 in SCLC and the underlying mechanism. Relative expression of PLCG2 was detected by reverse transcription-quantitative PCR and Western blot. Cell viability, proliferation, and apoptosis were assessed by cell counting kit-8, colony formation, and flow cytometry assays. Mitophagy-related protein levels were analyzed by Western blot. RNA immunoprecipitation and dual-luciferase reporter assays were used to analyze the interaction between PLCG2 and VCP. A xenograft mouse model was established to analyze the role of PLCG2 in vivo. Results showed that PLCG2 was upregulated in SCLC tissues and cells, with high diagnostic potential. Besides, PLCG2 deficiency inhibited cell survival and mitophagy and promoted apoptosis in SCLC cells. In addition, PLCG2 interacted with VCP, and VCP overexpression reversed the inhibitory effects of PLCG2 silencing. In vivo, PLCG2 silencing suppressed SCLC tumor growth. In conclusion, PLCG2 is a promising biomarker for SCLC diagnosis and might be a potential therapeutic target, with its interaction with VCP playing a role in SCLC cell survival and mitophagy.
    Keywords:  PLCG2; VCP; apoptosis; cell viability; mitophagy; small cell lung cancer
    DOI:  https://doi.org/10.1097/CAD.0000000000001770
  42. Research (Wash D C). 2025 ;8 0957
      Mitophagy alleviates intervertebral disc degeneration (IVDD) by suppressing cGAS-STING and NLRP3 inflammasome-mediated pyroptosis pathways; however, its metabolic regulatory mechanism remains unexplored. Herein, we discovered that mitophagy activator TJ0113 drives metabolic reprogramming characterized by substantially reduced lactate production in senescent nucleus pulposus (NP) cells. This decline directly diminishes histone H3 lysine 18 lactylation (H3K18la), consequently suppressing transcription of the pro-inflammatory gene thrombospondin-1 (THBS1) and blocking downstream inflammatory cascades in IVDD progress. Through combined genetic silencing of THBS1 and pharmacological inhibition of lactate generation, we establish the lactate-H3K18la-THBS1 axis as the essential mechanism mediating mitophagy's anti-inflammatory effects. Our work provides the first evidence that mitophagy orchestrates a metabolic-epigenetic regulatory axis (lactate-H3K18la-THBS1), unveiling novel therapeutic targets for IVDD and paving the way for epigenetic therapies against disc degeneration.
    DOI:  https://doi.org/10.34133/research.0957
  43. Mol Neurobiol. 2025 Nov 07. 63(1): 9
      Puerarin has demonstrated protective effects against Alzheimer's disease (AD) and diabetes. This study aimed to explore the therapeutic potential and mechanistic basis of puerarin against high-fat high-sugar (HFHS) diet-induced AD-like pathology and insulin resistance, with a specific focus on p35/cyclin-dependent kinase 5 (CDK5) signaling. SH-SY5Y cells were cultured under high glucose (HG) conditions to induce AD-like pathology, and mice were fed with an HFHS diet to establish an AD model. Both models were treated with various concentrations of puerarin. The expression and activity of p35/CDK5 were examined by Western blot and kinase assays. Rescue experiments were conducted by transfecting SH-SY5Y cells with p35 overexpression plasmids. The protein levels of amyloid-beta (Aβ)42, amyloid precursor protein (APP), and phosphorylated-Tau were assessed by immunofluorescence. The protein levels in the insulin signal-related signaling pathway were examined. Mitochondrial dysfunction mediated by CDK5/dynamin-related protein 1 (DRP1) was determined. Additionally, cognitive function in mice was evaluated using behavioral tests, including the open field test, novel object recognition test, and Morris water maze. Insulin resistance in mice was assessed using biochemical assays. Puerarin inhibited HG-induced p35/CDK5 activation in SH-SY5Y cells. It also decreased the HG-induced upregulation of Aβ42, APP, and p-Tau in SH-SY5Y cells. Moreover, puerarin ameliorated HG-induced insulin resistance and mitochondrial dysfunction in SH-SY5Y cells, as evidenced by improved insulin signaling and restored mitochondrial ultrastructure. However, p35 overexpression abrogated these protective effects. In vivo, puerarin alleviated HFHS diet-induced cognitive impairment, Aβ deposition, and Tau phosphorylation in mice. Furthermore, puerarin ameliorated HFHS diet-induced insulin resistance and mitochondrial dysfunction in mice. Puerarin ameliorated HFHS diet-induced cognitive impairment, insulin resistance, and mitochondrial dysfunction by inhibiting p35/CDK5 activity. Our findings highlight the therapeutic potential of puerarin in the management of diet-induced AD.
    Keywords:  Alzheimer's disease; High-fat high-sugar diet; Insulin resistance; Mitochondrial dysfunction; P35/CDK5; Puerarin
    DOI:  https://doi.org/10.1007/s12035-025-05300-x
  44. Adv Sci (Weinh). 2025 Nov 07. e18323
      Activation of cytosolic nucleic acid-sensing pathways represents a promising strategy to convert immunologically "cold" tumors into inflamed ones. Iron-sulfur (Fe-S) enzymes are critical regulators of innate immunity and nucleic acid sensing, yet their roles in cancer remain poorly defined. Here, ferredoxin-1 (FDX1), a mitochondrial Fe-S protein frequently downregulated in clear cell renal cell carcinoma (ccRCC), is identified as a dual regulator of ferroptosis and antitumor immunity. FDX1 overexpression triggers mitochondrial permeability transition pore opening, leading to cytosolic release of mitochondrial DNA (mtDNA) and double-stranded RNA (mt-dsRNA). This reveals an independent function of FDX1 as a tumor-intrinsic immunity activator linked to mitochondrial stress signaling. These damage-associated molecular patterns (DAMPs) engage cytosolic nucleic acid sensors-specifically cGAS and RIG-I/MDA5-triggering TBK1 phosphorylation and a robust type I interferon response that occurs prior to overt ferroptosis. This innate immune cascade reshapes the tumor microenvironment by enhancing MHC I/II antigen presentation, recruiting CD8+ T cells, and suppressing tumor growth and metastasis in orthotopic syngeneic models. These findings uncover a previously unrecognized antitumor axis through which FDX1 synergizes with mitochondrial nucleic acid release with ferroptosis to promote immunogenic inflammation and T cell infiltration in ccRCC, offering novel therapeutic opportunities targeting mitochondrial-immune crosstalk.
    Keywords:  FDX1; ccRCC; dsRNA; ferroptosis; innate immunity; mitophagy; mtDNA
    DOI:  https://doi.org/10.1002/advs.202518323
  45. Regen Biomater. 2025 ;12 rbaf101
      Calcium silicate (CS)-based bioactive materials were widely utilized to promote the therapeutic potential of bone marrow mesenchymal stem cells (BMSCs) in bone tissue engineering. The activation of numerous classic bone formation modulators, including the BMP, Wnt, and MAPK/ERK signaling pathways, contributes to the CS-induced osteogenesis of BMSCs. Mitochondrial metabolic patterns have emerged as key contributors to the osteogenic differentiation of mesenchymal stem cells. However, whether CS affects the mitochondrial metabolic profiles of BMSCs is mostly unclear. Herein, we showed that CS induced the osteogenic differentiation of human BMSCs (hBMSCs) mainly via silicon (Si) ion release. Moreover, CS-stimulated hBMSCs underwent metabolic reprogramming accompanied by increased mitochondrial oxidative phosphorylation (OXPHOS) activity. The inhibition of OXPHOS hindered the CS-induced osteogenic differentiation of hBMSCs and bone regeneration, indicating that CS-induced OXPHOS mediated the observed increase in osteogenesis. Mechanistically, CS induced mitophagy and autophagic flux by increasing the formation of autolysosomes and lysosomal degradation to eliminate dysfunctional mitochondria and mitochondrial reactive oxygen species production, leading to enhanced OXPHOS and osteogenesis in hBMSCs. Furthermore, CS promoted mitochondrial fusion in hBMSCs, which may contribute to OXPHOS activation. Our investigation reveals a previously unclear function of CS in regulating the osteogenesis of BMSCs by inducing mitophagy-mediated metabolic shifts toward OXPHOS.
    Keywords:  bone marrow mesenchymal stem cells; bone regeneration; calcium silicate; mitochondria; oxidative phosphorylation
    DOI:  https://doi.org/10.1093/rb/rbaf101
  46. Free Radic Biol Med. 2025 Oct 31. pii: S0891-5849(25)01320-6. [Epub ahead of print]
      X-linked adrenoleukodystrophy (X-ALD) is caused by pathogenic ABCD1 variants, leading to a dysfunctional peroxisomal ABCD1 transporter, crucial for β-oxidation of very long chain fatty acids (VLCFA). The clinical manifestation ranges from asymptomatic carriers to severe childhood cerebral ALD (CALD). The underlying pathophysiology remains unclear, and while elevated oxidative stress and signs of mitochondrial dysfunction have been observed in X-ALD cells and tissues, their precise roles are still uncertain. This study aims to elucidate the interplay among excess VLCFA, mitochondrial function and oxidative stress in fibroblasts derived from CALD and non-CALD patients. Therefore, we measured reactive oxygen species (ROS) using the 2',7'-dichlorofluorescein diacetate assay, mitochondrial function with the Seahorse XFe24 flux analyzer and assessed the regulation of stress homeostasis on the genetic level by qPCR of NRF2-dependent genes NQO1, AR1B10 and AKR1C1. Additional stress was induced by exposure to tert-butyl hydroperoxide (TBHP) and hexacosanoic acid (C26:0). Scanning confocal microscopy and STED super-resolution microscopy was implemented for evaluation of mitochondrial structure and peroxisomal-mitochondrial crosstalk. Our findings indicate that non-CALD cell lines exhibit an overall compromised oxidative status under basal conditions, characterized by significantly reduced oxygen consumption rates (OCR) relative to both CALD and healthy controls, along with diminished expression of NRF2-regulated genes. Notably, ROS levels in non-CALD cells are comparable to those observed in CALD cells. However, when exposed to additional stress, these non-CALD cells show greater potential of defense mechanisms and compensation compared to CALD cells. These findings significantly improve our understanding of metabolic changes in X-ALD, focusing on the ability of different X-ALD phenotypes to cope with oxidative stress. They pave the way for further investigations to understand the different phenotypes and their disease progression, to find reliant biomarkers, and to develop therapeutic approaches and preventive measures for individual patients.
    Keywords:  ABCD1; NRF2; X-linked Adrenoleukodystrophy; mitochondrial dynamics; reactive oxygen species
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2025.10.289
  47. Biofactors. 2025 Nov-Dec;51(6):51(6): e70054
      Bromelain, a combination of pineapple-derived enzymes, has demonstrated neuroprotective effects owing to its antioxidant and anti-inflammatory attributes. The current study intended to explore bromelain's neuroprotective impact against 3-nitropropionic acid (3-NP)-induced Huntington's disease (HD)-like symptoms in rats. Four groups of rats were randomly allocated: group 1 received saline; bromelain (40 mg/kg/i.p.) was given to groups 2 and 4 daily, whereas groups 3 and 4 received daily doses of 3-NP (10 mg/kg/i.p.) for 14 days. On the molecular level, bromelain administration resulted in upregulation of striatal SIRT-1, PGC-1α, TFAM and Nrf2 expression, enhancement of Akt phosphorylation, elevation of FOXO3a deacetylation and lessening of striatal oxidative stress damage by lowering MDA concentrations as well as raising the levels of NQO1, TAC, SOD and GSH. These effects were further substantiated by improvements in muscle strength, locomotor coordination, cognitive performance, and neuronal integrity. In conclusion, bromelain could be a beneficial neuroprotective candidate against HD-like aberrations experimentally induced by 3-NP.
    Keywords:   PGC‐1α/Nrf2 ; SIRT1/FOXO3a ; 3‐nitropropionic acid; bromelain; mitochondrial biogenesis; neuronal survival
    DOI:  https://doi.org/10.1002/biof.70054
  48. J Control Release. 2025 Oct 30. pii: S0168-3659(25)00971-X. [Epub ahead of print]388(Pt 2): 114357
      Myocardial ischemia-reperfusion injury (IRI) poses a significant and escalating threat to global public health. We present a breakthrough strategy using self-propelled lipase@CeO₂ nanomotors that actively target and penetrate damaged myocardium, launching a precise molecular defense via the PPARα/Tfam pathway. Engineered with P-selectin ligands, these nanomotors overcome vascular barriers to accumulate at injury sites. There, they perform dual actions: CeO₂ effectively ROS, with a particular emphasis on suppressing lipid peroxidation. Concurrently, lipase catalyzes the hydrolysis of reduced lipids into free fatty acids(FAs). Critically, these FAs act as endogenous ligands that activate PPARα, triggering its nuclear translocation. This initiates transcription of Tfam, a master regulator of mitochondrial biogenesis. This synergistic molecular regulation effectively suppresses cardiomyocyte ferroptosis, the key driver of IRI. In vivo, LCNMs-P treatment resulted in dramatically improved cardiac function (Ejection fraction: 64.48 ± 3.72 % in LCNMs-P + HF + IRI mice vs 42.71 ± 2.33 % in IRI mice). This work pioneers a self-fueled, pathway-specific nanotherapeutic, transforming enzyme-powered motors into intelligent regulators of cardiac repair.
    Keywords:  Enzyme@nanozyme Nanomotors; Ischemia-reperfusion injury (IRI); Lipid decomposition; Mitochondrial biogenesis; Peroxisome proliferator-activated receptor alpha (PPARα)
    DOI:  https://doi.org/10.1016/j.jconrel.2025.114357